1
|
Li Y, Tian S, Qiao Y, Cong C, Yang J, Cao S, Zhu X, Zhao L, Li P, Su J. Associations of Handgrip Strength and Testosterone With Cerebral White Matter Hyperintensity and Microstructural Injury. J Cachexia Sarcopenia Muscle 2025; 16:e13833. [PMID: 40464223 PMCID: PMC12134939 DOI: 10.1002/jcsm.13833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 03/26/2025] [Accepted: 04/21/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND White matter hyperintensity (WMH) is one of the key imaging markers of cerebral small vessel disease (CSVD) and white matter microstructural injury may occur earlier than WMH. However, the associations of handgrip strength (HGS) and serum total testosterone (STT) with WMH and microstructural injury have not been thoroughly investigated. Therefore, we aimed to explore the associations of HGS and STT with WMH and microstructural injury, measured by fractional anisotropy (FA), mean diffusivity (MD), intracellular volume fraction (ICVF), and isotropic volume fraction (ISOVF). METHODS A total of 34 832 participants from the UK Biobank (aged 64.15 ± 7.75 years, 50.9% male) were included in the study. Cross-sectional analyses were conducted using three linear regression models to assess the relationships between HGS, STT and neuroimaging outcomes. Sensitivity analyses were performed to validate the robustness of the linear regression results. Longitudinal, mediation and Mendelian randomization (MR) analyses were conducted to explore the longitudinal impact of HGS on WMH, the mediating effects of STT between HGS and neuroimaging outcomes, and probable causal relationships between HGS, STT and WMH. RESULTS A greater HGS was linearly associated with a smaller WMH volume (β = -0.006642, p < 0.001) and fuller microstructure (p < 0.001). These relationships persisted when stratified by sex or age decade and were supported by the results of the longitudinal analysis (β = -26.4, p = 0.0145). In females, STT was found to be linearly negatively related to WMH volume (β = -0.109, p < 0.001), MD and ISOVF, and linearly positively related to FA and ICVF (p < 0.001). In males, STT was linearly negatively linked to WMH volume (β = -0.016, p < 0.001) and MD (β = -1.6× 10 - 7 $$ \times {10}^{-7} $$ , p = 0.007), and positively associated to ICVF (β = 0.00012, p = 0.043). STT mediated the association between HGS and WMH volume (mediation proportion: 1.06% in males and 1.57% in females), and the possible causality was suggested for males through MR analysis: the positive causality between HGS and STT (p = 0.0394) and the negative causality between STT and WMH (p = 0.0787). CONCLUSIONS Our study revealed that greater HGS was linearly associated with reduced WMH volume and less white matter microstructural injury, mediated by STT. Improving muscle function may contribute to deferring white matter damage and preventing stroke and dementia, offering a feasible secondary prevention measure for CSVD.
Collapse
Affiliation(s)
- Yuna Li
- Department of Neurology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shan Tian
- Department of Neurology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yuan Qiao
- Department of Neurology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Chaohua Cong
- Department of Neurology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Junting Yang
- Department of Neurology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Shanshan Cao
- Department of Neurology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xirui Zhu
- School of Electrical and Information EngineeringZhengzhou University of Light IndustryZhengzhouChina
| | - Lei Zhao
- Department of Neurology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Panlong Li
- School of Electrical and Information EngineeringZhengzhou University of Light IndustryZhengzhouChina
- Department of Medical ImagingHenan Provincial People's Hospital & Zhengzhou University People's HospitalZhengzhouChina
| | - Jingjing Su
- Department of Neurology, Shanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
2
|
Wu X, Wan X, Tang Y, Li Y, Ye Z, Peng H, Zhang J, Pan L, Liu P. Causal Associations of Brain Imaging-Derived Phenotypes and Intracranial Aneurysm: A Bidirectional Mendelian Randomization Study. World Neurosurg 2025; 197:123881. [PMID: 40058632 DOI: 10.1016/j.wneu.2025.123881] [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: 01/12/2025] [Accepted: 03/03/2025] [Indexed: 04/06/2025]
Abstract
BACKGROUND Intracranial aneurysms (IAs) represent a significant and potentially fatal category of cerebrovascular disorders that pose serious health risks. Observational studies have indicated a potential link between brain imaging-derived phenotypes (IDPs) and various IAs. Nonetheless, the nature of these relationships remains ambiguous. METHODS Two-sample bidirectional Mendelian randomization (MR) analyses were performed to examine the causal links between IDPs and IAs. We utilized summary statistics from a population comprising 587 brain IDPs (UK Biobank). Additionally, we incorporated data of IAs from the HUNT study. RESULTS We identified a total of 13 IDPs that demonstrated a significant causal influence on the risk of developing IA, subarachnoid hemorrhage, and unruptured IA by forward MR. Specifically, an increase in mean diffusivity in the right external capsule is associated with the risk of IA (odds ratio [OR] = 1.830, 95% confidence interval [CI]: 1.134-2.553, P = 1.04 × 10-7). The results from the reverse MR analysis revealed that there is an association between genetically predicted IA and isotropic or free water volume fraction in body of corpus callosum (OR = 0.323, 95% CI: 0.232-0.461, P = 7.22 × 10-5), and an association between subarachnoid hemorrhage and mean diffusivity in the body of corpus callosum (OR = 0.360, 95% CI: 0.236-0.540, P = 2.50 × 10-4). CONCLUSIONS The findings indicate that these IDPs play a crucial role in the etiology of these IAs and highlight the importance of understanding the mechanisms through which they exert their effects.
Collapse
Affiliation(s)
- Xiao Wu
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xichen Wan
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yue Tang
- Department of Neurosurgery, The Fourth Hospital of Changsha, Changsha, China
| | - Yuxia Li
- Department of Neurology, Tangshan Central Hospital, Tangshan, China
| | - Zhennan Ye
- Department of Neurosurgery, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Haoyang Peng
- Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jincan Zhang
- Department of Neurosurgery, The Fourth Hospital of Changsha, Changsha, China
| | - Li Pan
- Department of Neurosurgery, Yangtze River Shipping General Hospital, Wuhan, China
| | - Peng Liu
- Department of Neurosurgery, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
3
|
Won J, Tarumi T, Tomoto T, Shan K, Rodrigue KM, Kennedy KM, Park DC, Zhang R. Associations of cardiorespiratory fitness with brain white matter microstructural integrity and white matter hyperintensity volume across the adult lifespan. GeroScience 2025:10.1007/s11357-025-01664-z. [PMID: 40299260 DOI: 10.1007/s11357-025-01664-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 04/13/2025] [Indexed: 04/30/2025] Open
Abstract
Higher cardiorespiratory fitness (CRF) is associated with less deterioration in brain microstructural white matter (WM) integrity in older adults assessed with MRI diffusion tensor imaging (DTI) and brain WM hyperintensities (WMH) volume measured with fluid-attenuated-inversion-recovery (FLAIR) imaging. This study investigated associations of CRF measured with peak oxygen consumption (V̇O2peak) with brain WM microstructural integrity and WMH in healthy individuals across the adult lifespan. We hypothesized that higher CRF is associated with less deterioration in WM microstructural integrity and WMH across the adult lifespan, which in turn is associated with better cognitive performance. A total of 177 healthy adults underwent treadmill exercise testing to measure V̇O2peak, MRI scan to measure free water (FW)-corrected DTI metrics and whole-brain WMH volume, and a comprehensive cognitive test battery. Linear regression models were used to examine the associations of DTI metrics and WMH volume with age, V̇O2peak, and age × V̇O2peak interaction. Higher CRF was associated with lower FW. Age × V̇O2peak interactions were observed in FW of the corpus callosum (CC) and WMH volume, such that the positive regression slopes of FW and WMH volume with age were lower in those with higher V̇O2peak than those with lower V̇O2peak. Lower WMH volume and FW in the CC were associated with better fluid cognition composite score. In conclusion, higher CRF is associated with less deteriorations in brain WM integrity and better cognitive function across the adult lifespan. These findings highlight the importance of maintaining and improving CRF to slow brain aging.
Collapse
Affiliation(s)
- Junyeon Won
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital, 7232 Greenville Ave, Dallas, TX, 75231, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Takashi Tarumi
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital, 7232 Greenville Ave, Dallas, TX, 75231, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, Japan
| | - Tsubasa Tomoto
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital, 7232 Greenville Ave, Dallas, TX, 75231, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Ibaraki, Japan
| | - Kevin Shan
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital, 7232 Greenville Ave, Dallas, TX, 75231, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Karen M Rodrigue
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX, USA
| | - Kristen M Kennedy
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX, USA
| | - Denise C Park
- Center for Vital Longevity, School of Behavioral and Brain Sciences, The University of Texas at Dallas, Dallas, TX, USA
| | - Rong Zhang
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital, 7232 Greenville Ave, Dallas, TX, 75231, USA.
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
4
|
Moran KL, Smith CJ, McManus E, Allan SM, Montaldi D, Muhlert N. Cerebrovascular health impacts processing speed through anterior white matter alterations: a UK biobank study. Sci Rep 2025; 15:9860. [PMID: 40119105 PMCID: PMC11928594 DOI: 10.1038/s41598-025-93399-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 03/06/2025] [Indexed: 03/24/2025] Open
Abstract
Cerebrovascular disease is associated with an increased likelihood of developing dementia. Cerebrovascular risk factors are modifiable and may reduce the risk of later-life cognitive dysfunction, however, the relationship between cerebrovascular risk factors, brain integrity and cognition remains poorly characterised. Using a UK Biobank sample of mid-to-old aged adults, without neurological disease, our structural equation mediation models showed that poor cerebrovascular health, indicated by the presence of cerebrovascular risk factors, was associated with slowed processing speed. This effect was best explained by anterior white matter microstructure (e.g. genu, anterior corona radiata), rather than posterior (e.g. splenium, posterior corona radiata)-the mediatory effect of anterior white matter strengthened further with age. Effects were also significantly reduced when considering other forms of cognition, demonstrating both regional- and cognitive-specificity. Our findings also illustrate that cerebrovascular risk factors cross-sectionally predict cognitive processing speed performance, which can be further strengthened by accounting for risk factor duration, particularly hypertensive duration. In summary, our study highlights the vulnerability of anterior regions and sensitivity of processing speed performance to cerebrovascular burden, and show this effect is amplified with age. We also highlight an improved method of cerebrovascular burden quantification, which accounts for factor duration, as well as risk factor presence and degree. Future work will aim to establish the role of medication and effective risk factor control in alleviating or preventing white matter pathology and cognitive dysfunction.
Collapse
Affiliation(s)
- Katie L Moran
- Division of Psychology, Communication and Human Neurosciences, School of Health Sciences, University of Manchester, Manchester, UK.
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, ManchesterManchester, UK.
| | - Craig J Smith
- Division of Cardiovascular Sciences, School of Medical Sciences, University of Manchester, Manchester, UK
- Manchester Centre for Clinical Neurosciences, Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Salford, UK
| | - Elizabeth McManus
- Division of Psychology and Mental Health, School of Health Sciences, University of Manchester, Manchester, UK
| | - Stuart M Allan
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, ManchesterManchester, UK
- Division of Neuroscience, School of Biological Sciences, University of Manchester, Manchester, UK
| | - Daniela Montaldi
- Division of Psychology, Communication and Human Neurosciences, School of Health Sciences, University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, ManchesterManchester, UK
| | - Nils Muhlert
- Division of Psychology, Communication and Human Neurosciences, School of Health Sciences, University of Manchester, Manchester, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, ManchesterManchester, UK
| |
Collapse
|
5
|
Bauer CE, Zachariou V, Pappas C, Maillard P, DeCarli C, Caprihan A, Gold BT. Healthy dietary intake diminishes the effect of cerebral small vessel disease on cognitive performance in older adults. Front Neurol 2025; 16:1508148. [PMID: 40115382 PMCID: PMC11925079 DOI: 10.3389/fneur.2025.1508148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 02/10/2025] [Indexed: 03/23/2025] Open
Abstract
Introduction We evaluated whether regular dietary intake of nutrients commonly found in fish, unsaturated oils, and nuts would moderate the associations between neuroimaging biomarkers of cerebral small vessel disease (cSVD) and cognitive function in older adults. Methods Dietary information, Montreal Cognitive Assessment (MoCA) scores, and magnetic resonance imaging (MRI) scans were collected from 71 older adults without dementia (60-86 years). MRI biomarkers of cSVD were calculated for each participant. Multivariate linear regression models were computed using dietary intake as the moderating variable. Covariates included age, sex, and estimated intracranial volume. Results Dietary intake moderated the association between several cSVD biomarkers and MoCA scores such that the expected negative association between cSVD biomarkers and cognition was seen at low levels of healthy dietary intake, but not at medium or high levels. A dietary intake by age moderation was not observed. Discussion Our findings indicate that healthy dietary intake may confer cognitive reserve against cSVD in older adults.
Collapse
Affiliation(s)
- Christopher E Bauer
- Departments of Neuroscience, University of Kentucky, Lexington, KY, United States
| | | | - Colleen Pappas
- Departments of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Pauline Maillard
- Departments of Neurology, University of California, Davis, Davis, CA, United States
- Center for Neurosciences, University of California, Davis, Davis, CA, United States
| | - Charles DeCarli
- Departments of Neurology, University of California, Davis, Davis, CA, United States
- Center for Neurosciences, University of California, Davis, Davis, CA, United States
| | | | - Brian T Gold
- Departments of Neuroscience, University of Kentucky, Lexington, KY, United States
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
- Magnetic Resonance Imaging and Spectroscopy Center, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
6
|
Xiao Y, Gui Y, Dai J, Zhao H, Rao Z, Luo D, Deng X, Xiao X. Mediation of White Matter Alterations in the Association Between Ventricular Dilation and Cognitive Decline in Hydrocephalus Patients: An MRI Study. J Magn Reson Imaging 2025; 61:737-748. [PMID: 38767272 DOI: 10.1002/jmri.29452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/02/2024] [Accepted: 05/02/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND Cognitive impairment is commonly observed in hydrocephalus patients. Ventricular enlargement compresses brain parenchyma, especially the white matter (WM). PURPOSE To investigate whether the relationship between ventricular dilation and cognitive decline in hydrocephalus patients is mediated by WM alterations. STUDY TYPE Retrospective. POPULATION 51 communicating hydrocephalus patients (median age, 54 years), 50 obstructive hydrocephalus patients (median age, 49 years), and 53 control subjects (median age, 50 years). FIELD STRENGTH/SEQUENCE Diffusion tensors imaging, 3D T1 BRAVO, 3D FIESTA, CUBE T2, and FLAIR sequences at 3T. ASSESSMENT DTI parameters (skeletonized fractional anisotropy (FA), skeletonized mean diffusivity (MD), and peak width of skeletonized mean diffusivity p(PSMD)) were extracted using FSL software. Global, periventricular, and deep white matter hyperintensity (WMH) volumes, degree of ventricular enlargement (Evans index), and other conventional imaging markers (number of lacunes and perivascular spaces, intracranial and brain volume) were extracted using united imaging intelligence. Cognitive tests included Montreal cognitive assessment (MoCA), clock drawing test (CDT), and vocabulary fluency test (VFT). STATISTICAL TESTS Multivariable linear regression analysis, mediation analyses, and dominance analysis. P-value <0.05 was considered significant. RESULTS The degree of ventricular dilation, DTI parameters, and cognitive function scores were interrelated. The skeletonized FA values (β = -0.0917, 95% confidence interval (CI): -0.205, -0.024) and normalized global WMH volume (β = -0.0635, 95% CI: -0.13, -0.0005) together mediated 37.2% of the association between Evans index and MoCA. A comparable causal pathway was found for periventricular WMHs but not for deep WMHs. Dominance analysis indicated skeletonized FA values had a greater impact on cognition than WMH volume. The skeletonized FA values also mediated the association between Evans index and CDT (β = -0.0897, 95% CI: -0.165, -0.026) and VFT (β = -0.1589, 95% CI: -0.27, -0.083). CONCLUSION WM alterations were causal mediators between ventricular dilation and cognitive decline in hydrocephalus patients. EVIDENCE LEVEL 3. TECHNICAL EFFICACY Stage 3.
Collapse
Affiliation(s)
- Yawen Xiao
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yifei Gui
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jiankun Dai
- GE Healthcare, MR Research China, Beijing, China
| | - Heng Zhao
- Department of Radiology, First Affiliated Hospital of University of South China, Hengyang, China
| | - Zhongliang Rao
- Department of Radiology, The Third Hospital of Nanchang, Nanchang, China
| | - Dan Luo
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xinru Deng
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xinlan Xiao
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
7
|
Xu Y, Huang D, Zhang H, Fang Q, Xia Y, Shi F, Gong X. White matter hyperintensities regress at a high rate at three months after minor ischemic stroke or transient ischemic attack. J Neuroradiol 2025; 52:101239. [PMID: 39798248 DOI: 10.1016/j.neurad.2024.101239] [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: 07/28/2024] [Revised: 12/06/2024] [Accepted: 12/16/2024] [Indexed: 01/15/2025]
Abstract
BACKGROUND The potential for early white matter hyperintensities(WMH) regression and associated contributory factors remains uncertain. The purpose of this study is to investigate whether WMH regress at early time of three months after minor ischemic stroke (MIS) or transient ischemic attack (TIA), while also identifying factors that may influence this outcome. METHODS A retrospective analysis of a prospective subcohort from the CHANCE trial comprising individuals with MIS and TIA was conducted. All patients underwent brain MRI at the onset and at three months. Deep learning algorithms were employed for the automatic segmentation of WMH volumes in four distinct regions. Scores for lacunes, cerebral microbleeds (CMB), perivascular spaces (PVS), WMH, and overall cerebral small vessel disease (CSVD) burden were quantified. Patients were divided into the stable, regression and progression groups according to change in WMH volume. The demographic, clinical, and imaging data of the participants in the three groups were collected and statistically analyzed. RESULTS A total of 98 patients with minor ischemic stroke or TIA were included. There were 22 patients in the stable group, 41 patients in the regression group and 35 patients in the progression group. Age and hypertension status were significantly different among the three groups. The lacunes, CMB,WMH, and total CSVD burden scores differed notably among groups, with all the CSVD markers being severely elevated in the progression group, moderately elevated in the regression group, and subtly elevated in the stable group. CONCLUSION The findings suggest that WMH could exhibit regression within three months following minor ischemic stroke or TIA. Patients under the age of 65, without a hypertension history, and with a low CSVD burden are more likely to experience WMH regression.
Collapse
Affiliation(s)
- Yuyun Xu
- Soochow Medical college of Soochow University, Suzhou, PR China; Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, PR China
| | - Danjiang Huang
- Taizhou First People's Hospital, Huangyan Hospital of Wenzhou Medical College, PR China
| | - He Zhang
- Taizhou First People's Hospital, Huangyan Hospital of Wenzhou Medical College, PR China
| | | | - Yuwei Xia
- Shanghai United Imaging Intelligence, Shanghai, PR China
| | - Feng Shi
- Shanghai United Imaging Intelligence, Shanghai, PR China
| | - Xiangyang Gong
- Soochow Medical college of Soochow University, Suzhou, PR China; Center for Rehabilitation Medicine, Department of Radiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, PR China.
| |
Collapse
|
8
|
Thammasart S, Harvey DJ, Maillard P, DeCarli C, Donnay CA, Wheeler GJ, Fan AP. Associations between cerebral blood flow and progression of white matter hyperintensities. FRONTIERS IN NEUROIMAGING 2025; 3:1463311. [PMID: 39906355 PMCID: PMC11790564 DOI: 10.3389/fnimg.2024.1463311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 12/16/2024] [Indexed: 02/06/2025]
Abstract
Introduction In an aging population, white matter hyperintensities (WMHs), observed on FLAIR MRI sequences, are indicators of cognitive decline, motor impairment, and increased vascular risk. However, the pathophysiological mechanisms underlying WMHs, including dynamic changes in cerebral blood flow (CBF) within and adjacent to lesions, remain poorly understood. Methods Our study examined a diverse cohort of 300 elderly participants through arterial spin labeling (ASL) on 3 Tesla MRI, analyzing both cross-sectional and longitudinal data. We characterized the relationship between CBF and WMH development in different lesion locations (based on distance from ventricles) and brain tissue types (WMH lesion, penumbra, and normal white matter). Results Our findings reveal that WMHs exhibit significantly lower relative CBF (rCBF) compared to penumbra, normal-appearing white matter, and gray matter, with juxtaventricular WMHs (JVWMH) displaying the most substantial reductions. Longitudinally, WMHs that increased in size over a two-year period had lower baseline rCBF than those that remained stagnant, particularly in juxtaventricular and periventricular regions. Discussion This study not only highlights the predictive value of rCBF in WMH progression but also provides location-specific hemodynamic information about WMHs that can guide clinical management of WMH-related brain changes and their clinical manifestations.
Collapse
Affiliation(s)
- Siriluk Thammasart
- Biological Engineering Program, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok, Thailand
| | - Danielle J. Harvey
- Public Health Sciences, University of California Davis School of Medicine, Davis, CA, United States
| | - Pauline Maillard
- Neurology, University of California, Davis, Davis, CA, United States
| | - Charles DeCarli
- Neurology, University of California, Davis, Davis, CA, United States
| | - Corinne A. Donnay
- Neurology, University of California, Davis, Davis, CA, United States
| | - Gregory J. Wheeler
- Biomedical Engineering, University of California, Davis, Davis, CA, United States
| | - Audrey P. Fan
- Neurology, University of California, Davis, Davis, CA, United States
- Biomedical Engineering, University of California, Davis, Davis, CA, United States
| |
Collapse
|
9
|
Won J, Tomoto T, Shan K, Tarumi T, Zhang R. Associations of Central Arterial Stiffness With Brain White Matter Integrity and Gray Matter Volume in MRI Across the Adult Lifespan. J Magn Reson Imaging 2025. [PMID: 39792583 DOI: 10.1002/jmri.29713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/20/2024] [Accepted: 12/28/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Central arterial stiffening is associated with brain white matter (WM) damage and gray matter (GM) volume loss in older adults, but little is known about this association from an adult lifespan perspective. PURPOSE To investigate the associations of central arterial stiffness with WM microstructural organization, WM lesion load, cortical thickness, and GM volume in healthy adults across the lifespan. STUDY TYPE This is a cross-sectional study. SUBJECTS A total of 173 healthy adults (22-81 years) were included in this study. FIELD STRENGTH/SEQUENCE 3-T, T1-weighted magnetization prepared rapid gradient echo (MPRAGE), single-shot echo-planar imaging diffusion-weighted, and T2-weighted fluid-attenuated inversion recovery (FLAIR) sequences. ASSESSMENT The participants underwent measurements of central arterial stiffness using carotid-femoral pulse wave velocity (cfPWV), diffusion tensor imaging (DTI) to measure whole-brain WM microstructural organization with free water (FW) and FW-corrected fractional anisotropy (FACOR), FLAIR to measure whole-brain WM hyperintensities (WMH), and MPRAGE to measure whole-brain cortical thickness and GM volume. The associations of age and cfPWV with MRI measures were assessed. STATISTICAL TESTS Linear regression models to examine the associations of brain WM and GM measures with age, cfPWV, and age × cfPWV interaction after adjusting for sex, education, and intracranial volume (ICV) (voxel-wise and cluster threshold P < 0.05). To understand the direction of the interaction result, the sample was stratified into lower and higher cfPWV groups using a median split of cfPWV. RESULTS Age × cfPWV interactions were observed in WM FW, WMH volume, cortical thickness, and GM volume (P < 0.01) such that the positive regression slopes between age, FW, and WMH volume were higher, while the negative regression slopes between age, cortical thickness, and GM volume were lower in those who had higher cfPWV relative to those who had lower cfPWV. DATA CONCLUSION Central arterial stiffening may accelerate the age-related deteriorations in GM and WM structure across the adult lifespan. PLAIN LANGUAGE SUMMARY Central arterial stiffening is associated with brain white matter (WM) damage and gray matter (GM) volume loss in older adults. We extended this investigation into an adult lifespan perspective by examining the associations of central arterial stiffening with brain structure in adults across age. A total of 172 healthy adults (22-81 years) underwent central arterial stiffening measure using applanation tonometry and brain measurement using MRI. We observed that higher central arterial stiffening may accelerate the age-related deterioration in brain WM and GM structure. These results suggest the importance of maintaining vascular health to slow age-related brain structural changes from an adult lifespan perspective. LEVEL OF EVIDENCE 4 TECHNICAL EFFICACY: Stage 5.
Collapse
Affiliation(s)
- Junyeon Won
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital, Dallas, Texas, USA
| | - Tsubasa Tomoto
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital, Dallas, Texas, USA
- Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Japan
| | - Kevin Shan
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital, Dallas, Texas, USA
| | - Takashi Tarumi
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital, Dallas, Texas, USA
- Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Japan
| | - Rong Zhang
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital, Dallas, Texas, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
10
|
Boons JHC, Vinke EJ, Dingemanse G, Kremer B, Goedegebure A, Vernooij MW. Hearing loss and its relation to longitudinal changes in white matter microstructure in older adults: The Rotterdam Study. Neurobiol Aging 2025; 145:24-31. [PMID: 39447491 DOI: 10.1016/j.neurobiolaging.2024.10.003] [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: 04/30/2024] [Revised: 10/08/2024] [Accepted: 10/13/2024] [Indexed: 10/26/2024]
Abstract
Hearing loss is considered a potentially modifiable risk factor for dementia. The sensory deprivation theory postulates that hearing loss adversely affects cognition in older adults through structural brain changes, but longitudinal studies are scarce. To find evidence for a possible detrimental effect of hearing loss on white matter microstructure, we carried out a longitudinal study in the population-based Rotterdam Study. A total of 1877 participants with a median age at baseline of 56.4 years (IQR: [52.2-60.0]) underwent audiometry and had longitudinal diffusion imaging data available with a mean follow-up of 4.0 years. A lower level of hearing acuity was associated with worse white matter microstructure in the left uncinate fasciculus and superior longitudinal fasciculus at baseline. Poorer hearing acuity was also associated with faster microstructural deterioration over time in the left superior longitudinal fasciculus. The strongest effects were observed for low-frequency hearing thresholds, while the high-frequency thresholds showed the weakest associations. These results suggest that hearing loss may contribute to the age-related decline in brain structure, consistent with the sensory deprivation theory.
Collapse
Affiliation(s)
- Jordi H C Boons
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC, Rotterdam, The Netherlands
| | - Elisabeth J Vinke
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands; Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Gertjan Dingemanse
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC, Rotterdam, The Netherlands
| | - Bernd Kremer
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC, Rotterdam, The Netherlands
| | - André Goedegebure
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus MC, Rotterdam, The Netherlands
| | - Meike W Vernooij
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands; Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
11
|
Chan K, Fischer C, Maralani PJ, Black SE, Moody AR, Khademi A. Stratifying vascular disease patients into homogeneous subgroups using machine learning and FLAIR MRI biomarkers. NPJ IMAGING 2024; 2:56. [PMID: 39749287 PMCID: PMC11688236 DOI: 10.1038/s44303-024-00063-x] [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: 08/03/2024] [Accepted: 12/03/2024] [Indexed: 01/04/2025]
Abstract
This study proposes a framework to stratify vascular disease patients based on brain health and cerebrovascular disease (CVD) risk using regional FLAIR biomarkers. Intensity and texture biomarkers were extracted from FLAIR volumes of 379 atherosclerosis patients. K-Means clustering identified five homogeneous subgroups. The 15 most important biomarkers for subgroup differentiation, identified via Random Forest classification, were used to generate biomarker profiles. ANOVA tests showed age and white matter lesion volume were significantly (p < 0.05) different across subgroups, while Fisher's tests revealed significant (p < 0.05) differences in the prevalence of several vascular risk factors across subgroup. Based on biomarker and clinical profiles, Subgroup 4 was characterized with neurodegeneration unrelated to CVD, Subgroup 3 identified patients with high CVD risk requiring aggressive intervention, and Subgroups 1, 2, and 5 identified patients with varying levels of moderate risk, suitable for long-term lifestyle interventions. This study supports personalized treatment and risk stratification based on FLAIR biomarkers.
Collapse
Affiliation(s)
- Karissa Chan
- Department of Electrical, Computer and Biomedical Engineering, Toronto Metropolitan University, Toronto, ON Canada
- Institute for Biomedical Engineering, Science Tech (iBEST), a Partnership between St. Michael’s Hospital and Toronto Metropolitan University, Toronto, ON Canada
| | - Corinne Fischer
- Keenan Research Center for Biomedical Science, St. Michael’s Hospital, Unity Health Network, Toronto, ON Canada
| | | | - Sandra E. Black
- Institute of Medical Science, University of Toronto, Toronto, ON Canada
- Horvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON Canada
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, ON Canada
- L.C. Campbell Cognitive Neurology Research Unit, Sunnybrook Health Sciences Centre, Toronto, ON Canada
| | - Alan R. Moody
- Department of Medical Imaging, University of Toronto, Toronto, ON Canada
| | - April Khademi
- Department of Electrical, Computer and Biomedical Engineering, Toronto Metropolitan University, Toronto, ON Canada
- Institute for Biomedical Engineering, Science Tech (iBEST), a Partnership between St. Michael’s Hospital and Toronto Metropolitan University, Toronto, ON Canada
- Keenan Research Center for Biomedical Science, St. Michael’s Hospital, Unity Health Network, Toronto, ON Canada
- Department of Medical Imaging, University of Toronto, Toronto, ON Canada
- Institute of Medical Science, University of Toronto, Toronto, ON Canada
- Vector Institute for Artificial Intelligence, Toronto, ON Canada
| |
Collapse
|
12
|
McGill MB, Clark AL, Schnyer DM. Traumatic brain injury, posttraumatic stress disorder, and vascular risk are independently associated with white matter aging in Vietnam-Era veterans. J Int Neuropsychol Soc 2024; 30:923-934. [PMID: 39558525 DOI: 10.1017/s1355617724000626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
OBJECTIVE Traumatic brain injury (TBI), mental health conditions (e.g., posttraumatic stress disorder [PTSD]), and vascular comorbidities (e.g., hypertension, diabetes) are highly prevalent in the Veteran population and may exacerbate age-related changes to cerebral white matter (WM). Our study examined (1) relationships between health conditions-TBI history, PTSD, and vascular risk-and cerebral WM micro- and macrostructure, and (2) associations between WM measures and cognition. METHOD We analyzed diffusion tensor images from 183 older male Veterans (mean age = 69.18; SD = 3.61) with (n = 95) and without (n = 88) a history of TBI using tractography. Generalized linear models examined associations between health conditions and diffusion metrics. Total WM hyperintensity (WMH) volume was calculated from fluid-attenuated inversion recovery images. Robust regression examined associations between health conditions and WMH volume. Finally, elastic net regularized regression examined associations between WM measures and cognitive performance. RESULTS Veterans with and without TBI did not differ in severity of PTSD or vascular risk (p's >0.05). TBI history, PTSD, and vascular risk were independently associated with poorer WM microstructural organization (p's <0.5, corrected), however the effects of vascular risk were more numerous and widespread. Vascular risk was positively associated with WMH volume (p = 0.004, β=0.200, R2 = 0.034). Higher WMH volume predicted poorer processing speed (R2 = 0.052). CONCLUSIONS Relative to TBI history and PTSD, vascular risk may be more robustly associated with WM micro- and macrostructure. Furthermore, greater WMH burden is associated with poorer processing speed. Our study supports the importance of vascular health interventions in mitigating negative brain aging outcomes in Veterans.
Collapse
Affiliation(s)
- Makenna B McGill
- Department of Psychology, The University of Texas at Austin, Austin, TX, USA
| | - Alexandra L Clark
- Department of Psychology, The University of Texas at Austin, Austin, TX, USA
| | - David M Schnyer
- Department of Psychology, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
13
|
Rosbergen MT, Wolters FJ, Vinke EJ, Mattace-Raso FUS, Roshchupkin GV, Ikram MA, Vernooij MW. Cluster-Based White Matter Signatures and the Risk of Dementia, Stroke, and Mortality in Community-Dwelling Adults. Neurology 2024; 103:e209864. [PMID: 39255426 PMCID: PMC11399066 DOI: 10.1212/wnl.0000000000209864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Markers of white matter (WM) injury on brain MRI are important indicators of brain health. Different patterns of WM atrophy, WM hyperintensities (WMHs), and microstructural integrity could reflect distinct pathologies and disease risks, but large-scale imaging studies investigating WM signatures are lacking. This study aims to identify distinct WM signatures using brain MRI in community-dwelling adults, determine underlying risk factor profiles, and assess risks of dementia, stroke, and mortality associated with each signature. METHODS Between 2005 and 2016, we measured WMH volume, WM volume, fractional anisotropy (FA), and mean diffusivity (MD) using automated pipelines on structural and diffusion MRI in community-dwelling adults aged older than 45 years of the Rotterdam study. Continuous surveillance was conducted for dementia, stroke, and mortality. We applied hierarchical clustering to identify separate WM injury clusters and Cox proportional hazard models to determine their risk of dementia, stroke, and mortality. RESULTS We included 5,279 participants (mean age 65.0 years, 56.0% women) and identified 4 distinct data-driven WM signatures: (1) above-average microstructural integrity and little WM atrophy and WMH; (2) above-average microstructural integrity and little WMH, but substantial WM atrophy; (3) poor microstructural integrity and substantial WMH, but little WM atrophy; and (4) poor microstructural integrity with substantial WMH and WM atrophy. Prevalence of cardiovascular risk factors, lacunes, and cerebral microbleeds was higher in clusters 3 and 4 than in clusters 1 and 2. During a median 10.7 years of follow-up, 291 participants developed dementia, 220 had a stroke, and 910 died. Compared with cluster 1, dementia risk was increased for all clusters, notably cluster 3 (hazard ratio [HR] 3.06, 95% CI 2.12-4.42), followed by cluster 4 (HR 2.31, 95% CI 1.58-3.37) and cluster 2 (HR 1.67, 95% CI 1.17-2.38). Compared with cluster 1, risk of stroke was higher only for clusters 3 (HR 1.55, 95% CI 1.02-2.37) and 4 (HR 1.94, 95% CI 1.30-2.89), whereas mortality risk was increased in all clusters (cluster 2: HR 1.27, 95% CI 1.06-1.53, cluster 3: HR 1.65, 95% CI 1.35-2.03, cluster 4: HR 1.76, 95% CI 1.44-2.15), compared with cluster 1. Models including clusters instead of an individual imaging marker showed a superior goodness of fit for dementia and mortality, but not for stroke. DISCUSSION Clustering can derive WM signatures that are differentially associated with dementia, stroke, and mortality risk. Future research should incorporate spatial information of imaging markers.
Collapse
Affiliation(s)
- Mathijs T Rosbergen
- From the Department of Epidemiology (M.T.R., F.J.W., E.J.V., F.U.S.M.-R., G.V.R., M.A.I., M.W.V.), Department of Radiology and Nuclear Medicine (M.T.R., F.J.W., E.J.V., G.V.R., M.W.V.), Department of Internal Medicine (F.U.S.M.-R.), and Department of Medical Informatics (G.V.R.), Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Frank J Wolters
- From the Department of Epidemiology (M.T.R., F.J.W., E.J.V., F.U.S.M.-R., G.V.R., M.A.I., M.W.V.), Department of Radiology and Nuclear Medicine (M.T.R., F.J.W., E.J.V., G.V.R., M.W.V.), Department of Internal Medicine (F.U.S.M.-R.), and Department of Medical Informatics (G.V.R.), Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Elisabeth J Vinke
- From the Department of Epidemiology (M.T.R., F.J.W., E.J.V., F.U.S.M.-R., G.V.R., M.A.I., M.W.V.), Department of Radiology and Nuclear Medicine (M.T.R., F.J.W., E.J.V., G.V.R., M.W.V.), Department of Internal Medicine (F.U.S.M.-R.), and Department of Medical Informatics (G.V.R.), Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Francesco U S Mattace-Raso
- From the Department of Epidemiology (M.T.R., F.J.W., E.J.V., F.U.S.M.-R., G.V.R., M.A.I., M.W.V.), Department of Radiology and Nuclear Medicine (M.T.R., F.J.W., E.J.V., G.V.R., M.W.V.), Department of Internal Medicine (F.U.S.M.-R.), and Department of Medical Informatics (G.V.R.), Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Gennady V Roshchupkin
- From the Department of Epidemiology (M.T.R., F.J.W., E.J.V., F.U.S.M.-R., G.V.R., M.A.I., M.W.V.), Department of Radiology and Nuclear Medicine (M.T.R., F.J.W., E.J.V., G.V.R., M.W.V.), Department of Internal Medicine (F.U.S.M.-R.), and Department of Medical Informatics (G.V.R.), Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Mohammad Arfan Ikram
- From the Department of Epidemiology (M.T.R., F.J.W., E.J.V., F.U.S.M.-R., G.V.R., M.A.I., M.W.V.), Department of Radiology and Nuclear Medicine (M.T.R., F.J.W., E.J.V., G.V.R., M.W.V.), Department of Internal Medicine (F.U.S.M.-R.), and Department of Medical Informatics (G.V.R.), Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Meike W Vernooij
- From the Department of Epidemiology (M.T.R., F.J.W., E.J.V., F.U.S.M.-R., G.V.R., M.A.I., M.W.V.), Department of Radiology and Nuclear Medicine (M.T.R., F.J.W., E.J.V., G.V.R., M.W.V.), Department of Internal Medicine (F.U.S.M.-R.), and Department of Medical Informatics (G.V.R.), Erasmus MC University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
14
|
Schmitzer L, Kaczmarz S, Göttler J, Hoffmann G, Kallmayer M, Eckstein HH, Hedderich DM, Kufer J, Zimmer C, Preibisch C, Hyder F, Sollmann N. Macro- and microvascular contributions to cerebral structural alterations in patients with asymptomatic carotid artery stenosis. J Cereb Blood Flow Metab 2024; 44:1629-1642. [PMID: 38506325 PMCID: PMC11418673 DOI: 10.1177/0271678x241238935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 01/22/2024] [Accepted: 01/29/2024] [Indexed: 03/21/2024]
Abstract
Atherosclerosis can underly internal carotid artery stenosis (ICAS), a major risk factor for ischemic stroke, as well as small vessel disease (SVD). This study aimed to investigate hemodynamics and structural alterations associated with SVD in ICAS patients. 28 patients with unilateral asymptomatic ICAS and 30 age-matched controls underwent structural (T1-/T2-weighted and diffusion tensor imaging [DTI]) and hemodynamic (pseudo-continuous arterial spin labeling and dynamic susceptibility contrast) magnetic resonance imaging. SVD-related alterations were assessed using free water (FW), FW-corrected DTI, and peak-width of skeletonized mean diffusivity (PSMD). Furthermore, cortical thickness, cerebral blood flow (CBF), and capillary transit time heterogeneity (CTH) were analyzed. Ipsilateral to the stenosis, cortical thickness was significantly decreased in the posterior dorsal cingulate cortex (p = 0.024) and temporal pole (p = 0.028). ICAS patients exhibited elevated PSMD (p = 0.005), FW (p < 0.001), and contralateral alterations in FW-corrected DTI metrics. We found significantly lateralized CBF (p = 0.011) and a tendency for lateralized CTH (p = 0.067) in the white matter (WM) related to ICAS. Elevated PSMD and FW may indicate a link between SVD and WM changes. Contralateral alterations were seen in FW-corrected DTI, whereas hemodynamic and cortical changes were mainly ipsilateral, suggesting SVD might influence global brain changes concurrent with ICAS-related hemodynamic alterations.
Collapse
Affiliation(s)
- Lena Schmitzer
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Department of Radiology & Biomedical Imaging, Magnetic Resonance Research Center (MRRC), Yale University, New Haven, CT, USA
- TUM-Neuroimaging Center, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Stephan Kaczmarz
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Department of Radiology & Biomedical Imaging, Magnetic Resonance Research Center (MRRC), Yale University, New Haven, CT, USA
- TUM-Neuroimaging Center, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Philips GmbH Market DACH, Hamburg, Germany
| | - Jens Göttler
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Department of Radiology & Biomedical Imaging, Magnetic Resonance Research Center (MRRC), Yale University, New Haven, CT, USA
- TUM-Neuroimaging Center, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Gabriel Hoffmann
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- TUM-Neuroimaging Center, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Michael Kallmayer
- Department for Vascular and Endovascular Surgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Hans-Henning Eckstein
- Department for Vascular and Endovascular Surgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Dennis Martin Hedderich
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Jan Kufer
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Department of Radiology & Biomedical Imaging, Magnetic Resonance Research Center (MRRC), Yale University, New Haven, CT, USA
- TUM-Neuroimaging Center, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Claus Zimmer
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Christine Preibisch
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Department of Neurology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Fahmeed Hyder
- Department of Radiology & Biomedical Imaging, Magnetic Resonance Research Center (MRRC), Yale University, New Haven, CT, USA
| | - Nico Sollmann
- Department of Diagnostic and Interventional Neuroradiology, School of Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- TUM-Neuroimaging Center, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Department of Diagnostic and Interventional Radiology, University Hospital Ulm, Ulm, Germany
| |
Collapse
|
15
|
Rimmele DL, Petersen EL, Affolderbach S, Petersen M, Cheng B, Mayer C, Nägele FL, Harth V, Terschüren C, Kühn S, Zeller T, Gerloff C, Thomalla G. Differences in impact of current and former shift work on cardiovascular risk factors, carotid atherosclerosis, and white matter integrity. SLEEP ADVANCES : A JOURNAL OF THE SLEEP RESEARCH SOCIETY 2024; 5:zpae056. [PMID: 39156216 PMCID: PMC11329802 DOI: 10.1093/sleepadvances/zpae056] [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: 10/08/2023] [Revised: 07/11/2024] [Indexed: 08/20/2024]
Abstract
Study Objectives The association of shift work (SW) and disrupted circadian rhythm with markers of large artery atherosclerosis and cerebral small vessel disease is uncertain. We aimed to study the separate association of current and former SW with these markers. Methods We included participants from the population-based Hamburg City Health Study. SW was defined by monthly working hours between 06:00 pm and 07:00 am containing night shifts for at least 12 months. Cross-sectional data were obtained from structured questionnaires, laboratory analyses, physical examinations, brain magnetic resonance imaging, and carotid ultrasound. We performed multivariable regression analysis with carotid intima-media thickness (CIMT), and peak-width skeletonized mean diffusivity (PSMD) as dependent variables. Results Three hundred and forty-four current, 238 former, and 7162 never-shift workers were included. The median age was 60 years for both current and former shift workers, and total duration of SW was comparable for the two groups. Current shift workers were less frequently female (27.3% vs. 44.5%; p < .001), had more frequent hyperlipidemia (31.5% vs. 22.3%; p = .024), and diabetes (16.2% vs. 3.2%; p < .001). After adjustment for age and sex, reduced quality of sleep (β = 1.61, p = .001) and low education (β = 2.63, p < .001) were associated with current but not former SW. Adjusted for age and sex, the current SW was associated with higher CIMT (β = 0.02, p = .001) and PSMD (β = 9.06e-06, p = .006), whereas former SW was not. Adjusted for risk factors, current SW remained associated with PSMD (β = 9.91e-06, p = .006) but not with CIMT. Conclusions Current SW was associated with CIMT and with PSMD, with the latter association remaining after adjustment for risk factors. Former SW showed no associations with CIMT or PSMD. This may indicate that current SW is linked with increased neurovascular risk through disrupted circadian rhythms. Trial Registration Information The trial was submitted at http://www.clinicaltrials.gov, under NCT03934957 on January 4, 2019. The first participant was enrolled in February 2016.
Collapse
Affiliation(s)
- David Leander Rimmele
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Neurology, Luzerner Kantonsspital, Lucerne, Switzerland
| | - Elina L Petersen
- Epidemiological Study Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sarah Affolderbach
- Institute for Occupational and Maritime Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marvin Petersen
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Bastian Cheng
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carola Mayer
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Felix Leonard Nägele
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Volker Harth
- Institute for Occupational and Maritime Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Claudia Terschüren
- Institute for Occupational and Maritime Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simone Kühn
- Department of Psychiatry, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tanja Zeller
- University Center of Cardiovascular Science, University Heart and Vascular Center Hamburg, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), University Heart and Vascular Center Hamburg, Hamburg, Germany
| | - Christian Gerloff
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Götz Thomalla
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
16
|
Neto S, Reis A, Pinheiro M, Ferreira M, Neves V, Castanho TC, Santos N, Rodrigues AJ, Sousa N, Santos MAS, Moura GR. Unveiling the molecular landscape of cognitive aging: insights from polygenic risk scores, DNA methylation, and gene expression. Hum Genomics 2024; 18:75. [PMID: 38956648 PMCID: PMC11221141 DOI: 10.1186/s40246-024-00640-6] [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: 04/23/2024] [Accepted: 06/17/2024] [Indexed: 07/04/2024] Open
Abstract
BACKGROUND Aging represents a significant risk factor for the occurrence of cerebral small vessel disease, associated with white matter (WM) lesions, and to age-related cognitive alterations, though the precise mechanisms remain largely unknown. This study aimed to investigate the impact of polygenic risk scores (PRS) for WM integrity, together with age-related DNA methylation, and gene expression alterations, on cognitive aging in a cross-sectional healthy aging cohort. The PRSs were calculated using genome-wide association study (GWAS) summary statistics for magnetic resonance imaging (MRI) markers of WM integrity, including WM hyperintensities, fractional anisotropy (FA), and mean diffusivity (MD). These scores were utilized to predict age-related cognitive changes and evaluate their correlation with structural brain changes, which distinguish individuals with higher and lower cognitive scores. To reduce the dimensionality of the data and identify age-related DNA methylation and transcriptomic alterations, Sparse Partial Least Squares-Discriminant Analysis (sPLS-DA) was used. Subsequently, a canonical correlation algorithm was used to integrate the three types of omics data (PRS, DNA methylation, and gene expression data) and identify an individual "omics" signature that distinguishes subjects with varying cognitive profiles. RESULTS We found a positive association between MD-PRS and long-term memory, as well as a correlation between MD-PRS and structural brain changes, effectively discriminating between individuals with lower and higher memory scores. Furthermore, we observed an enrichment of polygenic signals in genes related to both vascular and non-vascular factors. Age-related alterations in DNA methylation and gene expression indicated dysregulation of critical molecular features and signaling pathways involved in aging and lifespan regulation. The integration of multi-omics data underscored the involvement of synaptic dysfunction, axonal degeneration, microtubule organization, and glycosylation in the process of cognitive aging. CONCLUSIONS These findings provide valuable insights into the biological mechanisms underlying the association between WM coherence and cognitive aging. Additionally, they highlight how age-associated DNA methylation and gene expression changes contribute to cognitive aging.
Collapse
Affiliation(s)
- Sonya Neto
- Institute for Biomedicine (iBiMED) and Department of Medical Sciences, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Andreia Reis
- Institute for Biomedicine (iBiMED) and Department of Medical Sciences, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Miguel Pinheiro
- Institute for Biomedicine (iBiMED) and Department of Medical Sciences, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Margarida Ferreira
- Institute for Biomedicine (iBiMED) and Department of Medical Sciences, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Vasco Neves
- Institute for Biomedicine (iBiMED) and Department of Medical Sciences, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Teresa Costa Castanho
- ICVS - School of Medicine, Campus Gualtar, University of Minho, 4710-057, Braga, Portugal
- Clinical Academic Center - Braga (2CA-B), Braga, Portugal
| | - Nadine Santos
- ICVS - School of Medicine, Campus Gualtar, University of Minho, 4710-057, Braga, Portugal
- Clinical Academic Center - Braga (2CA-B), Braga, Portugal
| | - Ana João Rodrigues
- ICVS - School of Medicine, Campus Gualtar, University of Minho, 4710-057, Braga, Portugal
- Clinical Academic Center - Braga (2CA-B), Braga, Portugal
| | - Nuno Sousa
- ICVS - School of Medicine, Campus Gualtar, University of Minho, 4710-057, Braga, Portugal
- Clinical Academic Center - Braga (2CA-B), Braga, Portugal
- P5 Medical Center, Braga, Portugal
| | - Manuel A S Santos
- Institute for Biomedicine (iBiMED) and Department of Medical Sciences, University of Aveiro, 3810-193, Aveiro, Portugal
- Multidisciplinary Institute of Aging, MIA-Portugal, Faculty of Medicine, University of Coimbra, Rua Largo 2, 3º, 3000-370, Coimbra, Portugal
| | - Gabriela R Moura
- Institute for Biomedicine (iBiMED) and Department of Medical Sciences, University of Aveiro, 3810-193, Aveiro, Portugal.
| |
Collapse
|
17
|
Kasai S, Watanabe K, Ide S, Ishimoto Y, Sasaki M, Umemura Y, Tatsuo S, Kakeda S, Mikami T, Tamada Y, Miki Y, Wakabayashi K, Tomiyama M, Kakeda S. FLAIR Hyperintensities in the Anterior Part of the Callosal Splenium in the Elderly Population: A Large Cohort Study. Acad Radiol 2024; 31:2922-2929. [PMID: 38413313 DOI: 10.1016/j.acra.2024.02.001] [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: 10/03/2023] [Revised: 01/24/2024] [Accepted: 02/01/2024] [Indexed: 02/29/2024]
Abstract
RATIONALE AND OBJECTIVES Although hyperintensity in the anterior portion of the callosal splenium on FLAIR (aCS-hyperintensity) is a common finding in elderly adults, no previous studies have examined the clinical significance. In this large elderly population study, we aimed to investigate the associations of aCS-hyperintensity with vascular risk factors, cognitive decline, and other MRI measurements. MATERIALS AND METHODS This cross-sectional study included 2110 participants (median age, 69 years; 61.1% females) who underwent 3 T MRI. The participants were grouped as 215 with mild cognitive impairment (MCI) and 1895 cognitively normal older adults (NOAs). Two neuroradiologists evaluated aCS-hyperintensity by using a four-point scale (none, mild, moderate, and severe). Periventricular hyperintensities (PVHs) were also rated on a four-point scale according to the Fazekas scale. The total intracranial volume (ICV), total brain volume, choroid plexus volume (CPV), and lateral ventricle volume (LVV) were calculated. RESULTS Logistic regression analysis showed diabetes was the main predictor of aCS-hyperintensity after adjusting for potential confounders (age, sex, hypertension, and hyperlipidemia) (p < 0.01), whereas PVH was associated with hypertension (p < 0.01). aCS-hyperintensity rated as "severe" was associated with a presence of MCI (p < 0.01). For the imaging factors, LVV was an independent predictor of aCS-hyperintensity when brain volume and PVH grade were added to the analysis (p < 0.01). CONCLUSION Cerebral small vessel disease due to diabetes is a major contributor to the development of aCS-hyperintensity. Cerebrospinal fluid clearance failure may also relate to aCS-hyperintensity, which may offer new insights into the pathologic processes underlying MCI.
Collapse
Affiliation(s)
- Sera Kasai
- Department of Radiology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Keita Watanabe
- Department of radiology, Kyoto Prefectural University of Medicine, 465 Kajiimachi, Jokyo-ku, Kyoto-shi, Kyoto, Japan.
| | - Satoru Ide
- Department of Radiology, University of Occupational and Environmental Health, School of Medicine, Kitakyushu, Japan
| | - Yuka Ishimoto
- Department of Radiology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Miho Sasaki
- Department of Radiology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Yoshihito Umemura
- Department of Radiology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Soichiro Tatsuo
- Department of Radiology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Sachi Kakeda
- Department of Radiology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Tatsuya Mikami
- Innovation Center for Health Promotion, Hirosaki University, Hirosaki, Japan
| | - Yoshinori Tamada
- Innovation Center for Health Promotion, Hirosaki University, Hirosaki, Japan
| | - Yasuo Miki
- Department of Neuropathology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Koichi Wakabayashi
- Department of Neuropathology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Masahiko Tomiyama
- Department of Neurology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Shingo Kakeda
- Department of Radiology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| |
Collapse
|
18
|
Yaqub A, Vojinovic D, Vernooij MW, Slagboom PE, Ghanbari M, Beekman M, van der Grond J, Hankemeier T, van Duijn CM, Ikram MA, Ahmad S. Plasma trimethylamine N-oxide (TMAO): associations with cognition, neuroimaging, and dementia. Alzheimers Res Ther 2024; 16:113. [PMID: 38769578 PMCID: PMC11103865 DOI: 10.1186/s13195-024-01480-1] [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: 10/23/2023] [Accepted: 05/13/2024] [Indexed: 05/22/2024]
Abstract
BACKGROUND The gut-derived metabolite Trimethylamine N-oxide (TMAO) and its precursors - betaine, carnitine, choline, and deoxycarnitine - have been associated with an increased risk of cardiovascular disease, but their relation to cognition, neuroimaging markers, and dementia remains uncertain. METHODS In the population-based Rotterdam Study, we used multivariable regression models to study the associations between plasma TMAO, its precursors, and cognition in 3,143 participants. Subsequently, we examined their link to structural brain MRI markers in 2,047 participants, with a partial validation in the Leiden Longevity Study (n = 318). Among 2,517 participants, we assessed the risk of incident dementia using multivariable Cox proportional hazard models. Following this, we stratified the longitudinal associations by medication use and sex, after which we conducted a sensitivity analysis for individuals with impaired renal function. RESULTS Overall, plasma TMAO was not associated with cognition, neuroimaging markers or incident dementia. Instead, higher plasma choline was significantly associated with poor cognition (adjusted mean difference: -0.170 [95% confidence interval (CI) -0.297;-0.043]), brain atrophy and more markers of cerebral small vessel disease, such as white matter hyperintensity volume (0.237 [95% CI: 0.076;0.397]). By contrast, higher carnitine concurred with lower white matter hyperintensity volume (-0.177 [95% CI: -0.343;-0.010]). Only among individuals with impaired renal function, TMAO appeared to increase risk of dementia (hazard ratio (HR): 1.73 [95% CI: 1.16;2.60]). No notable differences were observed in stratified analyses. CONCLUSIONS Plasma choline, as opposed to TMAO, was found to be associated with cognitive decline, brain atrophy, and markers of cerebral small vessel disease. These findings illustrate the complexity of relationships between TMAO and its precursors, and emphasize the need for concurrent study to elucidate gut-brain mechanisms.
Collapse
Affiliation(s)
- Amber Yaqub
- Department of Epidemiology, Erasmus MC, University Medical Center, PO Box 2040, Rotterdam, CA, 3000, the Netherlands
| | - Dina Vojinovic
- Department of Epidemiology, Erasmus MC, University Medical Center, PO Box 2040, Rotterdam, CA, 3000, the Netherlands
- Section of Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Meike W Vernooij
- Department of Epidemiology, Erasmus MC, University Medical Center, PO Box 2040, Rotterdam, CA, 3000, the Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - P Eline Slagboom
- Section of Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus MC, University Medical Center, PO Box 2040, Rotterdam, CA, 3000, the Netherlands
| | - Marian Beekman
- Section of Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeroen van der Grond
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Thomas Hankemeier
- Division of Analytical Biosciences, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | | | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC, University Medical Center, PO Box 2040, Rotterdam, CA, 3000, the Netherlands.
| | - Shahzad Ahmad
- Department of Epidemiology, Erasmus MC, University Medical Center, PO Box 2040, Rotterdam, CA, 3000, the Netherlands
| |
Collapse
|
19
|
Won J, Maillard P, Shan K, Ashley J, Cardim D, Zhu DC, Zhang R. Association of Blood Pressure With Brain White Matter Microstructural Integrity Assessed With MRI Diffusion Tensor Imaging in Healthy Young Adults. Hypertension 2024; 81:1145-1155. [PMID: 38487873 PMCID: PMC11023804 DOI: 10.1161/hypertensionaha.123.22337] [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: 11/02/2023] [Accepted: 02/28/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND High blood pressure (BP) in middle-aged and older adults is associated with a brain white matter (WM) microstructural abnormality. However, little evidence is available in healthy young adults. We investigated the associations between high BP and WM microstructural integrity in young adults. METHODS This study included 1015 healthy young adults (542 women, 22-37 years) from the Human Connectome Project. Brachial systolic and diastolic BP were measured using a semiautomatic or manual sphygmomanometer. Diffusion-weighted magnetic resonance imaging was acquired to obtain diffusion tensor imaging metrics of free water (FW) content, FW-corrected WM fractional anisotropy, axial diffusivity, radial diffusivity, and mean diffusivity. Using whole-brain voxel-wise linear regression models and ANCOVA, we examined associations of BP and hypertension stage with diffusion tensor imaging metrics after adjusting for age, sex, education, body mass index, smoking status, alcohol consumption history, and differences in the b value used for diffusion magnetic resonance imaging. RESULTS Systolic and diastolic BP of the sample (mean±SD) were 122.8±13.0 and 76.0±9.9 mm Hg, respectively. Associations of BP with diffusion tensor imaging metrics revealed regional heterogeneity for FW-corrected fractional anisotropy. High BP and high hypertension stage were associated with higher FW and lower FW-corrected axial diffusivity, FW-corrected radial diffusivity, and FW-corrected mean diffusivity. Moreover, associations of high diastolic BP and hypertension stage with high FW were found only in men not in women. CONCLUSIONS High BP in young adults is associated with altered brain WM microstructural integrity, suggesting that high BP may have damaging effects on brain WM microstructural integrity in early adulthood, particularly in men.
Collapse
Affiliation(s)
- Junyeon Won
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital, Dallas, TX
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Pauline Maillard
- Department of Neurology, University of California, Davis, CA, USA
| | - Kevin Shan
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital, Dallas, TX
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX
| | - John Ashley
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital, Dallas, TX
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Danilo Cardim
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital, Dallas, TX
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX
| | - David C. Zhu
- Department of Radiology and Cognitive Imaging Research Center, Michigan State University, East Lansing, Michigan, USA
| | - Rong Zhang
- Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital, Dallas, TX
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
20
|
Wiltgen T, Voon C, Van Leemput K, Wiestler B, Mühlau M. Intensity scaling of conventional brain magnetic resonance images avoiding cerebral reference regions: A systematic review. PLoS One 2024; 19:e0298642. [PMID: 38483873 PMCID: PMC10939249 DOI: 10.1371/journal.pone.0298642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 01/26/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Conventional brain magnetic resonance imaging (MRI) produces image intensities that have an arbitrary scale, hampering quantification. Intensity scaling aims to overcome this shortfall. As neurodegenerative and inflammatory disorders may affect all brain compartments, reference regions within the brain may be misleading. Here we summarize approaches for intensity scaling of conventional T1-weighted (w) and T2w brain MRI avoiding reference regions within the brain. METHODS Literature was searched in the databases of Scopus, PubMed, and Web of Science. We included only studies that avoided reference regions within the brain for intensity scaling and provided validating evidence, which we divided into four categories: 1) comparative variance reduction, 2) comparative correlation with clinical parameters, 3) relation to quantitative imaging, or 4) relation to histology. RESULTS Of the 3825 studies screened, 24 fulfilled the inclusion criteria. Three studies used scaled T1w images, 2 scaled T2w images, and 21 T1w/T2w-ratio calculation (with double counts). A robust reduction in variance was reported. Twenty studies investigated the relation of scaled intensities to different types of quantitative imaging. Statistically significant correlations with clinical or demographic data were reported in 8 studies. Four studies reporting the relation to histology gave no clear picture of the main signal driver of conventional T1w and T2w MRI sequences. CONCLUSIONS T1w/T2w-ratio calculation was applied most often. Variance reduction and correlations with other measures suggest a biologically meaningful signal harmonization. However, there are open methodological questions and uncertainty on its biological underpinning. Validation evidence on other scaling methods is even sparser.
Collapse
Affiliation(s)
- Tun Wiltgen
- Department of Neurology, School of Medicine, Technical University of Munich, Munich, Germany
- TUM-Neuroimaging Center, School of Medicine, Technical University of Munich, Munich, Germany
| | - Cuici Voon
- Department of Neurology, School of Medicine, Technical University of Munich, Munich, Germany
- TUM-Neuroimaging Center, School of Medicine, Technical University of Munich, Munich, Germany
| | - Koen Van Leemput
- Department of Neuroscience and Biomedical Engineering, Aalto University Helsinki, Espoo, Finland
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Benedikt Wiestler
- Department of Neuroradiology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Mark Mühlau
- Department of Neurology, School of Medicine, Technical University of Munich, Munich, Germany
- TUM-Neuroimaging Center, School of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
21
|
Solé-Guardia G, Luijten M, Geenen B, Claassen JAHR, Litjens G, de Leeuw FE, Wiesmann M, Kiliaan AJ. Three-dimensional identification of microvascular pathology and neurovascular inflammation in severe white matter hyperintensity: a case report. Sci Rep 2024; 14:5004. [PMID: 38424226 PMCID: PMC10904845 DOI: 10.1038/s41598-024-55733-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 02/27/2024] [Indexed: 03/02/2024] Open
Abstract
White matter hyperintensities (WMH) are the most prevalent markers of cerebral small vessel disease (SVD), which is the major vascular risk factor for dementia. Microvascular pathology and neuroinflammation are suggested to drive the transition from normal-appearing white matter (NAWM) to WMH, particularly in individuals with hypertension. However, current imaging techniques cannot capture ongoing NAWM changes. The transition from NAWM into WMH is a continuous process, yet white matter lesions are often examined dichotomously, which may explain their underlying heterogeneity. Therefore, we examined microvascular and neurovascular inflammation pathology in NAWM and severe WMH three-dimensionally, along with gradual magnetic resonance imaging (MRI) fluid-attenuated inversion recovery (FLAIR) signal (sub-)segmentation. In WMH, the vascular network exhibited reduced length and complexity compared to NAWM. Neuroinflammation was more severe in WMH. Vascular inflammation was more pronounced in NAWM, suggesting its potential significance in converting NAWM into WMH. Moreover, the (sub-)segmentation of FLAIR signal displayed varying degrees of vascular pathology, particularly within WMH regions. These findings highlight the intricate interplay between microvascular pathology and neuroinflammation in the transition from NAWM to WMH. Further examination of neurovascular inflammation across MRI-visible alterations could aid deepening our understanding on WMH conversion, and therewith how to improve the prognosis of SVD.
Collapse
Affiliation(s)
- Gemma Solé-Guardia
- Department of Medical Imaging, Anatomy, Donders Institute for Brain, Cognition & Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Radboud university medical center, 6525 EZ, Nijmegen, PO Box 9101, The Netherlands
| | - Matthijs Luijten
- Department of Medical Imaging, Anatomy, Donders Institute for Brain, Cognition & Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Radboud university medical center, 6525 EZ, Nijmegen, PO Box 9101, The Netherlands
| | - Bram Geenen
- Department of Medical Imaging, Anatomy, Donders Institute for Brain, Cognition & Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Radboud university medical center, 6525 EZ, Nijmegen, PO Box 9101, The Netherlands
| | - Jurgen A H R Claassen
- Department of Geriatrics, Donders Institute for Brain, Cognition & Behavior, Radboud Alzheimer Center, Radboud university medical center, Nijmegen, The Netherlands
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Geert Litjens
- Department of Pathology, Radboud university medical center, Nijmegen, The Netherlands
- Computational Pathology Group, Research Institute for Medical Innovation, Radboud university medical center, Nijmegen, The Netherlands
| | - Frank-Erik de Leeuw
- Department of Neurology, Donders Institute for Brain, Cognition & Behavior, Radboud university medical center, Nijmegen, The Netherlands
| | - Maximilian Wiesmann
- Department of Medical Imaging, Anatomy, Donders Institute for Brain, Cognition & Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Radboud university medical center, 6525 EZ, Nijmegen, PO Box 9101, The Netherlands
| | - Amanda J Kiliaan
- Department of Medical Imaging, Anatomy, Donders Institute for Brain, Cognition & Behavior, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Radboud university medical center, 6525 EZ, Nijmegen, PO Box 9101, The Netherlands.
| |
Collapse
|
22
|
Jochems ACC, Muñoz Maniega S, Clancy U, Arteaga C, Jaime Garcia D, Chappell FM, Hewins W, Locherty R, Backhouse EV, Barclay G, Jardine C, McIntyre D, Gerrish I, Kampaite A, Sakka E, Valdés Hernández M, Wiseman S, Bastin ME, Stringer MS, Thrippleton MJ, Doubal FN, Wardlaw JM. Magnetic Resonance Imaging Tissue Signatures Associated With White Matter Changes Due to Sporadic Cerebral Small Vessel Disease Indicate That White Matter Hyperintensities Can Regress. J Am Heart Assoc 2024; 13:e032259. [PMID: 38293936 PMCID: PMC11056146 DOI: 10.1161/jaha.123.032259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 12/21/2023] [Indexed: 02/01/2024]
Abstract
BACKGROUND White matter hyperintensities (WMHs) might regress and progress contemporaneously, but we know little about underlying mechanisms. We examined WMH change and underlying quantitative magnetic resonance imaging tissue measures over 1 year in patients with minor ischemic stroke with sporadic cerebral small vessel disease. METHODS AND RESULTS We defined areas of stable normal-appearing white matter, stable WMHs, progressing and regressing WMHs based on baseline and 1-year brain magnetic resonance imaging. In these areas we assessed tissue characteristics with quantitative T1, fractional anisotropy (FA), mean diffusivity (MD), and neurite orientation dispersion and density imaging (baseline only). We compared tissue signatures cross-sectionally between areas, and longitudinally within each area. WMH change masks were available for N=197. Participants' mean age was 65.61 years (SD, 11.10), 59% had a lacunar infarct, and 68% were men. FA and MD were available for N=195, quantitative T1 for N=182, and neurite orientation dispersion and density imaging for N=174. Cross-sectionally, all 4 tissue classes differed for FA, MD, T1, and Neurite Density Index. Longitudinally, in regressing WMHs, FA increased with little change in MD and T1 (difference estimate, 0.011 [95% CI, 0.006-0.017]; -0.002 [95% CI, -0.008 to 0.003] and -0.003 [95% CI, -0.009 to 0.004]); in progressing and stable WMHs, FA decreased (-0.022 [95% CI, -0.027 to -0.017] and -0.009 [95% CI, -0.011 to -0.006]), whereas MD and T1 increased (progressing WMHs, 0.057 [95% CI, 0.050-0.063], 0.058 [95% CI, 0.050 -0.066]; stable WMHs, 0.054 [95% CI, 0.045-0.063], 0.049 [95% CI, 0.039-0.058]); and in stable normal-appearing white matter, MD increased (0.004 [95% CI, 0.003-0.005]), whereas FA and T1 slightly decreased and increased (-0.002 [95% CI, -0.004 to -0.000] and 0.005 [95% CI, 0.001-0.009]). CONCLUSIONS Quantitative magnetic resonance imaging shows that WMHs that regress have less abnormal microstructure at baseline than stable WMHs and follow trajectories indicating tissue improvement compared with stable and progressing WMHs.
Collapse
Affiliation(s)
- Angela C. C. Jochems
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
- UK Dementia Research Institute at the University of EdinburghEdinburghUnited Kingdom
| | - Susana Muñoz Maniega
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
- UK Dementia Research Institute at the University of EdinburghEdinburghUnited Kingdom
| | - Una Clancy
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
- UK Dementia Research Institute at the University of EdinburghEdinburghUnited Kingdom
| | - Carmen Arteaga
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
- UK Dementia Research Institute at the University of EdinburghEdinburghUnited Kingdom
| | - Daniela Jaime Garcia
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
- UK Dementia Research Institute at the University of EdinburghEdinburghUnited Kingdom
| | - Francesca M. Chappell
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
- UK Dementia Research Institute at the University of EdinburghEdinburghUnited Kingdom
| | - Will Hewins
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
- UK Dementia Research Institute at the University of EdinburghEdinburghUnited Kingdom
| | - Rachel Locherty
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
- UK Dementia Research Institute at the University of EdinburghEdinburghUnited Kingdom
| | - Ellen V. Backhouse
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
- UK Dementia Research Institute at the University of EdinburghEdinburghUnited Kingdom
| | - Gayle Barclay
- Edinburgh Imaging Facility, Royal Infirmary of EdinburghEdinburghUnited Kingdom
| | - Charlotte Jardine
- Edinburgh Imaging Facility, Royal Infirmary of EdinburghEdinburghUnited Kingdom
| | - Donna McIntyre
- Edinburgh Imaging Facility, Royal Infirmary of EdinburghEdinburghUnited Kingdom
| | - Iona Gerrish
- Edinburgh Imaging Facility, Royal Infirmary of EdinburghEdinburghUnited Kingdom
| | - Agniete Kampaite
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
| | - Eleni Sakka
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
| | - Maria Valdés Hernández
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
- UK Dementia Research Institute at the University of EdinburghEdinburghUnited Kingdom
| | - Stewart Wiseman
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
- UK Dementia Research Institute at the University of EdinburghEdinburghUnited Kingdom
| | - Mark E. Bastin
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
| | - Michael S. Stringer
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
- UK Dementia Research Institute at the University of EdinburghEdinburghUnited Kingdom
| | - Michael J. Thrippleton
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
- UK Dementia Research Institute at the University of EdinburghEdinburghUnited Kingdom
- Edinburgh Imaging Facility, Royal Infirmary of EdinburghEdinburghUnited Kingdom
| | - Fergus N. Doubal
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
- UK Dementia Research Institute at the University of EdinburghEdinburghUnited Kingdom
| | - Joanna M. Wardlaw
- Centre for Clinical Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
- UK Dementia Research Institute at the University of EdinburghEdinburghUnited Kingdom
- Edinburgh Imaging Facility, Royal Infirmary of EdinburghEdinburghUnited Kingdom
| |
Collapse
|
23
|
Rivier CA, Renedo DB, de Havenon A, Sunmonu NA, Gill TM, Payabvash S, Sheth KN, Falcone GJ. Association of Poor Oral Health With Neuroimaging Markers of White Matter Injury in Middle-Aged Participants in the UK Biobank. Neurology 2024; 102:e208010. [PMID: 38165331 PMCID: PMC10870735 DOI: 10.1212/wnl.0000000000208010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 10/03/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Poor oral health is a modifiable risk factor that is associated with clinically observed cardiovascular disease. However, the relationship between oral and brain health is not well understood. We tested the hypothesis that poor oral health is associated with worse neuroimaging brain health profiles in middle-aged persons without stroke or dementia. METHODS We performed a 2-stage cross-sectional neuroimaging study using UK Biobank data. First, we tested for association between self-reported poor oral health and MRI neuroimaging markers of brain health. Second, we used Mendelian randomization (MR) analyses to test for association between genetically determined poor oral health and the same neuroimaging markers. Poor oral health was defined as the presence of dentures or loose teeth. As instruments for the MR analysis, we used 116 independent DNA sequence variants linked to increased composite risk of dentures or teeth that are decayed, missing, or filled. Neuroimaging markers of brain health included white matter hyperintensity (WMH) volume and aggregate measures of fractional anisotropy (FA) and mean diffusivity (MD), 2 metrics indicative of white matter tract disintegrity obtained through diffusion tensor imaging across 48 brain regions. RESULTS We included 40,175 persons (mean age 55 years, female sex 53%) enrolled from 2006 to 2010, who underwent a dedicated research brain MRI between 2014 and 2016. Among participants, 5,470 (14%) had poor oral health. Poor oral health was associated with a 9% increase in WMH volume (β = 0.09, SD = 0.014, p < 0.001), 10% change in aggregate FA score (β = 0.10, SD = 0.013, p < 0.001), and 5% change in aggregate MD score (β = 0.05, SD = 0.013, p < 0.001). Genetically determined poor oral health was associated with a 30% increase in WMH volume (β = 0.30, SD = 0.06, p < 0.001), 43% change in aggregate FA score (β = 0.43, SD = 0.06, p < 0.001), and 10% change in aggregate MD score (β = 0.10, SD = 0.03, p < 0.01). DISCUSSION Among middle age Britons without stroke or dementia, poor oral health was associated with worse neuroimaging brain health profiles. Genetic analyses confirmed these associations, supporting a potentially causal association. Because the neuroimaging markers evaluated in this study precede and are established risk factors of stroke and dementia, our results suggest that oral health, an easily modifiable process, may be a promising target for very early interventions focused on improving brain health.
Collapse
Affiliation(s)
- Cyprien A Rivier
- From the Departments of Neurology (C.A.R., D.B.R., A.d.H., N.A.S., K.N.S., G.J.F.), Internal Medicine (T.M.G.), and Radiology (S.P.), Yale University School of Medicine; and Yale Center for Brain and Mind Health (C.A.R., A.d.H., S.P., K.N.S., G.J.F.), New Haven, CT
| | - Daniela B Renedo
- From the Departments of Neurology (C.A.R., D.B.R., A.d.H., N.A.S., K.N.S., G.J.F.), Internal Medicine (T.M.G.), and Radiology (S.P.), Yale University School of Medicine; and Yale Center for Brain and Mind Health (C.A.R., A.d.H., S.P., K.N.S., G.J.F.), New Haven, CT
| | - Adam de Havenon
- From the Departments of Neurology (C.A.R., D.B.R., A.d.H., N.A.S., K.N.S., G.J.F.), Internal Medicine (T.M.G.), and Radiology (S.P.), Yale University School of Medicine; and Yale Center for Brain and Mind Health (C.A.R., A.d.H., S.P., K.N.S., G.J.F.), New Haven, CT
| | - N Abimbola Sunmonu
- From the Departments of Neurology (C.A.R., D.B.R., A.d.H., N.A.S., K.N.S., G.J.F.), Internal Medicine (T.M.G.), and Radiology (S.P.), Yale University School of Medicine; and Yale Center for Brain and Mind Health (C.A.R., A.d.H., S.P., K.N.S., G.J.F.), New Haven, CT
| | - Thomas M Gill
- From the Departments of Neurology (C.A.R., D.B.R., A.d.H., N.A.S., K.N.S., G.J.F.), Internal Medicine (T.M.G.), and Radiology (S.P.), Yale University School of Medicine; and Yale Center for Brain and Mind Health (C.A.R., A.d.H., S.P., K.N.S., G.J.F.), New Haven, CT
| | - Seyedmehdi Payabvash
- From the Departments of Neurology (C.A.R., D.B.R., A.d.H., N.A.S., K.N.S., G.J.F.), Internal Medicine (T.M.G.), and Radiology (S.P.), Yale University School of Medicine; and Yale Center for Brain and Mind Health (C.A.R., A.d.H., S.P., K.N.S., G.J.F.), New Haven, CT
| | - Kevin N Sheth
- From the Departments of Neurology (C.A.R., D.B.R., A.d.H., N.A.S., K.N.S., G.J.F.), Internal Medicine (T.M.G.), and Radiology (S.P.), Yale University School of Medicine; and Yale Center for Brain and Mind Health (C.A.R., A.d.H., S.P., K.N.S., G.J.F.), New Haven, CT
| | - Guido J Falcone
- From the Departments of Neurology (C.A.R., D.B.R., A.d.H., N.A.S., K.N.S., G.J.F.), Internal Medicine (T.M.G.), and Radiology (S.P.), Yale University School of Medicine; and Yale Center for Brain and Mind Health (C.A.R., A.d.H., S.P., K.N.S., G.J.F.), New Haven, CT
| |
Collapse
|
24
|
Caçoilo A, Dortdivanlioglu B, Rusinek H, Weickenmeier J. A multiphysics model to predict periventricular white matter hyperintensity growth during healthy brain aging. BRAIN MULTIPHYSICS 2023; 5:100072. [PMID: 37546181 PMCID: PMC10399513 DOI: 10.1016/j.brain.2023.100072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/08/2023] Open
Abstract
Periventricular white matter hyperintensities (WMH) are a common finding in medical images of the aging brain and are associated with white matter damage resulting from cerebral small vessel disease, white matter inflammation, and a degeneration of the lateral ventricular wall. Despite extensive work, the etiology of periventricular WMHs remains unclear. We pose that there is a strong coupling between age-related ventricular expansion and the degeneration of the ventricular wall which leads to a dysregulated fluid exchange across this brain-fluid barrier. Here, we present a multiphysics model that couples cerebral atrophy-driven ventricular wall loading with periventricular WMH formation and progression. We use patient data to create eight 2D finite element models and demonstrate the predictive capabilities of our damage model. Our simulations show that we accurately capture the spatiotemporal features of periventricular WMH growth. For one, we observe that damage appears first in both the anterior and posterior horns and then spreads into deeper white matter tissue. For the other, we note that it takes up to 12 years before periventricular WMHs first appear and derive an average annualized periventricular WMH damage growth rate of 15.2 ± 12.7 mm2/year across our models. A sensitivity analysis demonstrated that our model parameters provide sufficient sensitivity to rationalize subject-specific differences with respect to onset time and damage growth. Moreover, we show that the septum pellucidum, a membrane that separates the left and right lateral ventricles, delays the onset of periventricular WMHs at first, but leads to a higher WMH load in the long-term.
Collapse
Affiliation(s)
- Andreia Caçoilo
- Department of Mechanical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, United States of America
| | - Berkin Dortdivanlioglu
- Civil, Architectural, and Environmental Engineering, The University of Texas at Austin, Austin, TX 78712, United States of America
| | - Henry Rusinek
- Department of Radiology, New York University Grossman School of Medicine, New York, NY 10016, United States of America
| | - Johannes Weickenmeier
- Department of Mechanical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, United States of America
| |
Collapse
|
25
|
Parent O, Bussy A, Devenyi GA, Dai A, Costantino M, Tullo S, Salaciak A, Bedford S, Farzin S, Béland ML, Valiquette V, Villeneuve S, Poirier J, Tardif CL, Dadar M, Chakravarty MM. Assessment of white matter hyperintensity severity using multimodal magnetic resonance imaging. Brain Commun 2023; 5:fcad279. [PMID: 37953840 PMCID: PMC10636521 DOI: 10.1093/braincomms/fcad279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/05/2023] [Accepted: 10/17/2023] [Indexed: 11/14/2023] Open
Abstract
White matter hyperintensities are radiological abnormalities reflecting cerebrovascular dysfunction detectable using MRI. White matter hyperintensities are often present in individuals at the later stages of the lifespan and in prodromal stages in the Alzheimer's disease spectrum. Tissue alterations underlying white matter hyperintensities may include demyelination, inflammation and oedema, but these are highly variable by neuroanatomical location and between individuals. There is a crucial need to characterize these white matter hyperintensity tissue alterations in vivo to improve prognosis and, potentially, treatment outcomes. How different MRI measure(s) of tissue microstructure capture clinically-relevant white matter hyperintensity tissue damage is currently unknown. Here, we compared six MRI signal measures sampled within white matter hyperintensities and their associations with multiple clinically-relevant outcomes, consisting of global and cortical brain morphometry, cognitive function, diagnostic and demographic differences and cardiovascular risk factors. We used cross-sectional data from 118 participants: healthy controls (n = 30), individuals at high risk for Alzheimer's disease due to familial history (n = 47), mild cognitive impairment (n = 32) and clinical Alzheimer's disease dementia (n = 9). We sampled the median signal within white matter hyperintensities on weighted MRI images [T1-weighted (T1w), T2-weighted (T2w), T1w/T2w ratio, fluid-attenuated inversion recovery (FLAIR)] as well as the relaxation times from quantitative T1 (qT1) and T2* (qT2*) images. qT2* and fluid-attenuated inversion recovery signals within white matter hyperintensities displayed different age- and disease-related trends compared to normal-appearing white matter signals, suggesting sensitivity to white matter hyperintensity-specific tissue deterioration. Further, white matter hyperintensity qT2*, particularly in periventricular and occipital white matter regions, was consistently associated with all types of clinically-relevant outcomes in both univariate and multivariate analyses and across two parcellation schemes. qT1 and fluid-attenuated inversion recovery measures showed consistent clinical relationships in multivariate but not univariate analyses, while T1w, T2w and T1w/T2w ratio measures were not consistently associated with clinical variables. We observed that the qT2* signal was sensitive to clinically-relevant microstructural tissue alterations specific to white matter hyperintensities. Our results suggest that combining volumetric and signal measures of white matter hyperintensity should be considered to fully characterize the severity of white matter hyperintensities in vivo. These findings may have implications in determining the reversibility of white matter hyperintensities and the potential efficacy of cardio- and cerebrovascular treatments.
Collapse
Affiliation(s)
- Olivier Parent
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - Aurélie Bussy
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - Gabriel Allan Devenyi
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - Alyssa Dai
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec H3A 1A1, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Quebec H3A 2B4, Canada
| | - Manuela Costantino
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
| | - Stephanie Tullo
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - Alyssa Salaciak
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
| | - Saashi Bedford
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - Sarah Farzin
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
| | - Marie-Lise Béland
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
| | - Vanessa Valiquette
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - Sylvia Villeneuve
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
- Center for the Studies in the Prevention of Alzheimer's Disease, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Quebec H3A 2B4, Canada
| | - Judes Poirier
- Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
- Center for the Studies in the Prevention of Alzheimer's Disease, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
- Molecular Neurobiology Unit, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
- Department of Medicine, McGill University, Montreal, Quebec H4A 3J1, Canada
| | - Christine Lucas Tardif
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Quebec H3A 2B4, Canada
- Department of Biomedical Engineering, McGill University, Montreal, Quebec H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - Mahsa Dadar
- Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - M Mallar Chakravarty
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec H3A 1A1, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
- Department of Biomedical Engineering, McGill University, Montreal, Quebec H3A 2B4, Canada
| |
Collapse
|
26
|
Acosta JN, Haider SP, Rivier C, Leasure AC, Sheth KN, Falcone GJ, Payabvash S. Blood pressure-related white matter microstructural disintegrity and associated cognitive function impairment in asymptomatic adults. Stroke Vasc Neurol 2023; 8:358-367. [PMID: 36878613 PMCID: PMC10647862 DOI: 10.1136/svn-2022-001929] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 02/13/2023] [Indexed: 03/08/2023] Open
Abstract
BACKGROUND AND OBJECTIVES We aimed to investigate the white matter (WM) microstructural/cytostructural disintegrity patterns related to higher systolic blood pressure (SBP), and whether they mediate SBP effects on cognitive performance in middle-aged adults. METHODS Using the UK Biobank study of community-dwelling volunteers aged 40-69 years, we included participants without a history of stroke, dementia, demyelinating disease or traumatic brain injury. We investigated the association of SBP with MRI diffusion metrics: fractional anisotropy (FA), mean diffusivity (MD), intracellular volume fraction (a measure of neurite density), isotropic (free) water volume fraction (ISOVF) and orientation dispersion across WM tracts. Then, we determined whether WM diffusion metrics mediated the effects of SBP on cognitive function. RESULTS We analysed 31 363 participants-mean age of 63.8 years (SD: 7.7), and 16 523 (53%) females. Higher SBP was associated with lower FA and neurite density, but higher MD and ISOVF. Among different WM tracts, diffusion metrics of the internal capsule anterior limb, external capsule, superior and posterior corona radiata were most affected by higher SBP. Among seven cognitive metrics, SBP levels were only associated with 'fluid intelligence' (adjusted p<0.001). In mediation analysis, the averaged FA of external capsule, internal capsule anterior limb and superior cerebellar peduncle mediated 13%, 9% and 13% of SBP effects on fluid intelligence, while the averaged MD of external capsule, internal capsule anterior and posterior limbs, and superior corona radiata mediated 5%, 7%, 7% and 6% of SBP effects on fluid intelligence, respectively. DISCUSSION Among asymptomatic adults, higher SBP is associated with pervasive WM microstructure disintegrity, partially due to reduced neuronal count, which appears to mediate SBP adverse effects on fluid intelligence. Diffusion metrics of select WM tracts, which are most reflective of SBP-related parenchymal damage and cognitive impairment, may serve as imaging biomarkers to assess treatment response in antihypertensive trials.
Collapse
Affiliation(s)
- Julián N Acosta
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Stefan P Haider
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Otorhinolaryngology, Ludwig Maximilians University Munich, Munchen, Germany
| | - Cyprien Rivier
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Audrey C Leasure
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Kevin N Sheth
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Guido J Falcone
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Seyedmehdi Payabvash
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
27
|
Shrestha S, Zhu X, Sullivan KJ, Blackshear C, Deal JA, Sharrett AR, Kamath V, Schneider ALC, Jack CR, Huang J, Palta P, Reid RI, Knopman DS, Gottesman RF, Chen H, Windham BG, Griswold ME, Mosley TH. Association of Olfaction and Microstructural Integrity of Brain Tissue in Community-Dwelling Adults: Atherosclerosis Risk in Communities Neurocognitive Study. Neurology 2023; 101:e1328-e1340. [PMID: 37541841 PMCID: PMC10558165 DOI: 10.1212/wnl.0000000000207636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 05/30/2023] [Indexed: 08/06/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Research on olfaction and brain neuropathology may help understand brain regions associated with normal olfaction and dementia pathophysiology. To identify early regional brain structures affected in poor olfaction, we examined cross-sectional associations of microstructural integrity of the brain with olfaction in the Atherosclerosis Risk in Communities Neurocognitive Study. METHODS Participants were selected from a prospective cohort study of community-dwelling adults; selection criteria included the following: evidence of cognitive impairment, participation in a previous MRI study, and a random sample of cognitively normal participants. Microstructural integrity was measured by 2 diffusion tensor imaging (DTI) measures, fractional anisotropy (FA) and mean diffusivity (MD), and olfaction by a 12-item odor identification test at the same visit. Higher FA and MD values indicate better and worse microstructural integrity, respectively, and higher odor identification scores indicate better olfaction. We used brain region-specific linear regression models to examine associations between DTI measures and olfaction, adjusting for potential confounders. RESULTS Among 1,418 participants (mean age 76 ± 5 years, 41% male, 21% Black race, 59% with normal cognition), the mean olfaction score was 9 ± 2.3. Relevant to olfaction, higher MD in the medial temporal lobe (MTL) regions, namely the hippocampus (β -0.79 [95% CI -0.94 to -0.65] units lower olfaction score per 1 SD higher MD), amygdala, entorhinal area, and some white matter (WM) tracts connecting to these regions, was associated with olfaction. We also observed associations with MD and WM FA in multiple atlas regions that were not previously implicated in olfaction. The associations between MD and olfaction were particularly stronger in the MTL regions among individuals with mild cognitive impairment (MCI) compared with those with normal cognition (e.g., βhippocampus -0.75 [95% CI -1.02 to -0.49] and -0.44 [95% CI -0.63 to -0.26] for MCI and normal cognition, respectively, p interaction = 0.004). DISCUSSION Neuronal microstructural integrity in multiple brain regions, particularly the MTL (the regions known to be affected in early Alzheimer disease), is associated with odor identification ability. Differential associations in the MTL regions among cognitively normal individuals compared with those with MCI may reflect the earlier vs later effects of the dementia pathogenesis. It is likely that some of the associated regions may not have any functional relevance to olfaction.
Collapse
Affiliation(s)
- Srishti Shrestha
- From the The Memory Impairment and Neurodegenerative Dementia (MIND) Center (S.S., X.Z., K.J.S., C.B., J.H., B.G.W., M.E.G., T.H.M.), University of Mississippi Medical Center, Jackson; Department of Epidemiology (J.A.D., A.R.S.), Johns Hopkins University Bloomberg School of Public Health; Department of Psychiatry and Behavioral Sciences (V.K.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology (A.L.C.S.), and Department of Biostatistics, Epidemiology, and Informatics (A.L.C.S.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Department of Radiology (C.R.J., R.I.R.), Mayo Clinic, Rochester, MN; Department of Neurology (J.H.), University of Mississippi Medical Center, Jackson; Department of Neurology (P.P.), University of North Carolina at Chapel Hill; Department of Neurology (D.S.K.), Mayo Clinic, Rochester, MN; Stroke Branch (R.F.G.), National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, MD; and Department of Epidemiology and Biostatistics (H.C.), Michigan State University, East Lansing.
| | - Xiaoqian Zhu
- From the The Memory Impairment and Neurodegenerative Dementia (MIND) Center (S.S., X.Z., K.J.S., C.B., J.H., B.G.W., M.E.G., T.H.M.), University of Mississippi Medical Center, Jackson; Department of Epidemiology (J.A.D., A.R.S.), Johns Hopkins University Bloomberg School of Public Health; Department of Psychiatry and Behavioral Sciences (V.K.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology (A.L.C.S.), and Department of Biostatistics, Epidemiology, and Informatics (A.L.C.S.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Department of Radiology (C.R.J., R.I.R.), Mayo Clinic, Rochester, MN; Department of Neurology (J.H.), University of Mississippi Medical Center, Jackson; Department of Neurology (P.P.), University of North Carolina at Chapel Hill; Department of Neurology (D.S.K.), Mayo Clinic, Rochester, MN; Stroke Branch (R.F.G.), National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, MD; and Department of Epidemiology and Biostatistics (H.C.), Michigan State University, East Lansing
| | - Kevin J Sullivan
- From the The Memory Impairment and Neurodegenerative Dementia (MIND) Center (S.S., X.Z., K.J.S., C.B., J.H., B.G.W., M.E.G., T.H.M.), University of Mississippi Medical Center, Jackson; Department of Epidemiology (J.A.D., A.R.S.), Johns Hopkins University Bloomberg School of Public Health; Department of Psychiatry and Behavioral Sciences (V.K.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology (A.L.C.S.), and Department of Biostatistics, Epidemiology, and Informatics (A.L.C.S.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Department of Radiology (C.R.J., R.I.R.), Mayo Clinic, Rochester, MN; Department of Neurology (J.H.), University of Mississippi Medical Center, Jackson; Department of Neurology (P.P.), University of North Carolina at Chapel Hill; Department of Neurology (D.S.K.), Mayo Clinic, Rochester, MN; Stroke Branch (R.F.G.), National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, MD; and Department of Epidemiology and Biostatistics (H.C.), Michigan State University, East Lansing
| | - Chad Blackshear
- From the The Memory Impairment and Neurodegenerative Dementia (MIND) Center (S.S., X.Z., K.J.S., C.B., J.H., B.G.W., M.E.G., T.H.M.), University of Mississippi Medical Center, Jackson; Department of Epidemiology (J.A.D., A.R.S.), Johns Hopkins University Bloomberg School of Public Health; Department of Psychiatry and Behavioral Sciences (V.K.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology (A.L.C.S.), and Department of Biostatistics, Epidemiology, and Informatics (A.L.C.S.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Department of Radiology (C.R.J., R.I.R.), Mayo Clinic, Rochester, MN; Department of Neurology (J.H.), University of Mississippi Medical Center, Jackson; Department of Neurology (P.P.), University of North Carolina at Chapel Hill; Department of Neurology (D.S.K.), Mayo Clinic, Rochester, MN; Stroke Branch (R.F.G.), National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, MD; and Department of Epidemiology and Biostatistics (H.C.), Michigan State University, East Lansing
| | - Jennifer A Deal
- From the The Memory Impairment and Neurodegenerative Dementia (MIND) Center (S.S., X.Z., K.J.S., C.B., J.H., B.G.W., M.E.G., T.H.M.), University of Mississippi Medical Center, Jackson; Department of Epidemiology (J.A.D., A.R.S.), Johns Hopkins University Bloomberg School of Public Health; Department of Psychiatry and Behavioral Sciences (V.K.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology (A.L.C.S.), and Department of Biostatistics, Epidemiology, and Informatics (A.L.C.S.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Department of Radiology (C.R.J., R.I.R.), Mayo Clinic, Rochester, MN; Department of Neurology (J.H.), University of Mississippi Medical Center, Jackson; Department of Neurology (P.P.), University of North Carolina at Chapel Hill; Department of Neurology (D.S.K.), Mayo Clinic, Rochester, MN; Stroke Branch (R.F.G.), National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, MD; and Department of Epidemiology and Biostatistics (H.C.), Michigan State University, East Lansing
| | - A Richey Sharrett
- From the The Memory Impairment and Neurodegenerative Dementia (MIND) Center (S.S., X.Z., K.J.S., C.B., J.H., B.G.W., M.E.G., T.H.M.), University of Mississippi Medical Center, Jackson; Department of Epidemiology (J.A.D., A.R.S.), Johns Hopkins University Bloomberg School of Public Health; Department of Psychiatry and Behavioral Sciences (V.K.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology (A.L.C.S.), and Department of Biostatistics, Epidemiology, and Informatics (A.L.C.S.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Department of Radiology (C.R.J., R.I.R.), Mayo Clinic, Rochester, MN; Department of Neurology (J.H.), University of Mississippi Medical Center, Jackson; Department of Neurology (P.P.), University of North Carolina at Chapel Hill; Department of Neurology (D.S.K.), Mayo Clinic, Rochester, MN; Stroke Branch (R.F.G.), National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, MD; and Department of Epidemiology and Biostatistics (H.C.), Michigan State University, East Lansing
| | - Vidyulata Kamath
- From the The Memory Impairment and Neurodegenerative Dementia (MIND) Center (S.S., X.Z., K.J.S., C.B., J.H., B.G.W., M.E.G., T.H.M.), University of Mississippi Medical Center, Jackson; Department of Epidemiology (J.A.D., A.R.S.), Johns Hopkins University Bloomberg School of Public Health; Department of Psychiatry and Behavioral Sciences (V.K.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology (A.L.C.S.), and Department of Biostatistics, Epidemiology, and Informatics (A.L.C.S.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Department of Radiology (C.R.J., R.I.R.), Mayo Clinic, Rochester, MN; Department of Neurology (J.H.), University of Mississippi Medical Center, Jackson; Department of Neurology (P.P.), University of North Carolina at Chapel Hill; Department of Neurology (D.S.K.), Mayo Clinic, Rochester, MN; Stroke Branch (R.F.G.), National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, MD; and Department of Epidemiology and Biostatistics (H.C.), Michigan State University, East Lansing
| | - Andrea L C Schneider
- From the The Memory Impairment and Neurodegenerative Dementia (MIND) Center (S.S., X.Z., K.J.S., C.B., J.H., B.G.W., M.E.G., T.H.M.), University of Mississippi Medical Center, Jackson; Department of Epidemiology (J.A.D., A.R.S.), Johns Hopkins University Bloomberg School of Public Health; Department of Psychiatry and Behavioral Sciences (V.K.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology (A.L.C.S.), and Department of Biostatistics, Epidemiology, and Informatics (A.L.C.S.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Department of Radiology (C.R.J., R.I.R.), Mayo Clinic, Rochester, MN; Department of Neurology (J.H.), University of Mississippi Medical Center, Jackson; Department of Neurology (P.P.), University of North Carolina at Chapel Hill; Department of Neurology (D.S.K.), Mayo Clinic, Rochester, MN; Stroke Branch (R.F.G.), National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, MD; and Department of Epidemiology and Biostatistics (H.C.), Michigan State University, East Lansing
| | - Clifford R Jack
- From the The Memory Impairment and Neurodegenerative Dementia (MIND) Center (S.S., X.Z., K.J.S., C.B., J.H., B.G.W., M.E.G., T.H.M.), University of Mississippi Medical Center, Jackson; Department of Epidemiology (J.A.D., A.R.S.), Johns Hopkins University Bloomberg School of Public Health; Department of Psychiatry and Behavioral Sciences (V.K.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology (A.L.C.S.), and Department of Biostatistics, Epidemiology, and Informatics (A.L.C.S.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Department of Radiology (C.R.J., R.I.R.), Mayo Clinic, Rochester, MN; Department of Neurology (J.H.), University of Mississippi Medical Center, Jackson; Department of Neurology (P.P.), University of North Carolina at Chapel Hill; Department of Neurology (D.S.K.), Mayo Clinic, Rochester, MN; Stroke Branch (R.F.G.), National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, MD; and Department of Epidemiology and Biostatistics (H.C.), Michigan State University, East Lansing
| | - Juebin Huang
- From the The Memory Impairment and Neurodegenerative Dementia (MIND) Center (S.S., X.Z., K.J.S., C.B., J.H., B.G.W., M.E.G., T.H.M.), University of Mississippi Medical Center, Jackson; Department of Epidemiology (J.A.D., A.R.S.), Johns Hopkins University Bloomberg School of Public Health; Department of Psychiatry and Behavioral Sciences (V.K.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology (A.L.C.S.), and Department of Biostatistics, Epidemiology, and Informatics (A.L.C.S.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Department of Radiology (C.R.J., R.I.R.), Mayo Clinic, Rochester, MN; Department of Neurology (J.H.), University of Mississippi Medical Center, Jackson; Department of Neurology (P.P.), University of North Carolina at Chapel Hill; Department of Neurology (D.S.K.), Mayo Clinic, Rochester, MN; Stroke Branch (R.F.G.), National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, MD; and Department of Epidemiology and Biostatistics (H.C.), Michigan State University, East Lansing
| | - Priya Palta
- From the The Memory Impairment and Neurodegenerative Dementia (MIND) Center (S.S., X.Z., K.J.S., C.B., J.H., B.G.W., M.E.G., T.H.M.), University of Mississippi Medical Center, Jackson; Department of Epidemiology (J.A.D., A.R.S.), Johns Hopkins University Bloomberg School of Public Health; Department of Psychiatry and Behavioral Sciences (V.K.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology (A.L.C.S.), and Department of Biostatistics, Epidemiology, and Informatics (A.L.C.S.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Department of Radiology (C.R.J., R.I.R.), Mayo Clinic, Rochester, MN; Department of Neurology (J.H.), University of Mississippi Medical Center, Jackson; Department of Neurology (P.P.), University of North Carolina at Chapel Hill; Department of Neurology (D.S.K.), Mayo Clinic, Rochester, MN; Stroke Branch (R.F.G.), National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, MD; and Department of Epidemiology and Biostatistics (H.C.), Michigan State University, East Lansing
| | - Robert I Reid
- From the The Memory Impairment and Neurodegenerative Dementia (MIND) Center (S.S., X.Z., K.J.S., C.B., J.H., B.G.W., M.E.G., T.H.M.), University of Mississippi Medical Center, Jackson; Department of Epidemiology (J.A.D., A.R.S.), Johns Hopkins University Bloomberg School of Public Health; Department of Psychiatry and Behavioral Sciences (V.K.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology (A.L.C.S.), and Department of Biostatistics, Epidemiology, and Informatics (A.L.C.S.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Department of Radiology (C.R.J., R.I.R.), Mayo Clinic, Rochester, MN; Department of Neurology (J.H.), University of Mississippi Medical Center, Jackson; Department of Neurology (P.P.), University of North Carolina at Chapel Hill; Department of Neurology (D.S.K.), Mayo Clinic, Rochester, MN; Stroke Branch (R.F.G.), National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, MD; and Department of Epidemiology and Biostatistics (H.C.), Michigan State University, East Lansing
| | - David S Knopman
- From the The Memory Impairment and Neurodegenerative Dementia (MIND) Center (S.S., X.Z., K.J.S., C.B., J.H., B.G.W., M.E.G., T.H.M.), University of Mississippi Medical Center, Jackson; Department of Epidemiology (J.A.D., A.R.S.), Johns Hopkins University Bloomberg School of Public Health; Department of Psychiatry and Behavioral Sciences (V.K.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology (A.L.C.S.), and Department of Biostatistics, Epidemiology, and Informatics (A.L.C.S.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Department of Radiology (C.R.J., R.I.R.), Mayo Clinic, Rochester, MN; Department of Neurology (J.H.), University of Mississippi Medical Center, Jackson; Department of Neurology (P.P.), University of North Carolina at Chapel Hill; Department of Neurology (D.S.K.), Mayo Clinic, Rochester, MN; Stroke Branch (R.F.G.), National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, MD; and Department of Epidemiology and Biostatistics (H.C.), Michigan State University, East Lansing
| | - Rebecca F Gottesman
- From the The Memory Impairment and Neurodegenerative Dementia (MIND) Center (S.S., X.Z., K.J.S., C.B., J.H., B.G.W., M.E.G., T.H.M.), University of Mississippi Medical Center, Jackson; Department of Epidemiology (J.A.D., A.R.S.), Johns Hopkins University Bloomberg School of Public Health; Department of Psychiatry and Behavioral Sciences (V.K.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology (A.L.C.S.), and Department of Biostatistics, Epidemiology, and Informatics (A.L.C.S.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Department of Radiology (C.R.J., R.I.R.), Mayo Clinic, Rochester, MN; Department of Neurology (J.H.), University of Mississippi Medical Center, Jackson; Department of Neurology (P.P.), University of North Carolina at Chapel Hill; Department of Neurology (D.S.K.), Mayo Clinic, Rochester, MN; Stroke Branch (R.F.G.), National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, MD; and Department of Epidemiology and Biostatistics (H.C.), Michigan State University, East Lansing
| | - Honglei Chen
- From the The Memory Impairment and Neurodegenerative Dementia (MIND) Center (S.S., X.Z., K.J.S., C.B., J.H., B.G.W., M.E.G., T.H.M.), University of Mississippi Medical Center, Jackson; Department of Epidemiology (J.A.D., A.R.S.), Johns Hopkins University Bloomberg School of Public Health; Department of Psychiatry and Behavioral Sciences (V.K.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology (A.L.C.S.), and Department of Biostatistics, Epidemiology, and Informatics (A.L.C.S.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Department of Radiology (C.R.J., R.I.R.), Mayo Clinic, Rochester, MN; Department of Neurology (J.H.), University of Mississippi Medical Center, Jackson; Department of Neurology (P.P.), University of North Carolina at Chapel Hill; Department of Neurology (D.S.K.), Mayo Clinic, Rochester, MN; Stroke Branch (R.F.G.), National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, MD; and Department of Epidemiology and Biostatistics (H.C.), Michigan State University, East Lansing
| | - B Gwen Windham
- From the The Memory Impairment and Neurodegenerative Dementia (MIND) Center (S.S., X.Z., K.J.S., C.B., J.H., B.G.W., M.E.G., T.H.M.), University of Mississippi Medical Center, Jackson; Department of Epidemiology (J.A.D., A.R.S.), Johns Hopkins University Bloomberg School of Public Health; Department of Psychiatry and Behavioral Sciences (V.K.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology (A.L.C.S.), and Department of Biostatistics, Epidemiology, and Informatics (A.L.C.S.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Department of Radiology (C.R.J., R.I.R.), Mayo Clinic, Rochester, MN; Department of Neurology (J.H.), University of Mississippi Medical Center, Jackson; Department of Neurology (P.P.), University of North Carolina at Chapel Hill; Department of Neurology (D.S.K.), Mayo Clinic, Rochester, MN; Stroke Branch (R.F.G.), National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, MD; and Department of Epidemiology and Biostatistics (H.C.), Michigan State University, East Lansing
| | - Michael E Griswold
- From the The Memory Impairment and Neurodegenerative Dementia (MIND) Center (S.S., X.Z., K.J.S., C.B., J.H., B.G.W., M.E.G., T.H.M.), University of Mississippi Medical Center, Jackson; Department of Epidemiology (J.A.D., A.R.S.), Johns Hopkins University Bloomberg School of Public Health; Department of Psychiatry and Behavioral Sciences (V.K.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology (A.L.C.S.), and Department of Biostatistics, Epidemiology, and Informatics (A.L.C.S.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Department of Radiology (C.R.J., R.I.R.), Mayo Clinic, Rochester, MN; Department of Neurology (J.H.), University of Mississippi Medical Center, Jackson; Department of Neurology (P.P.), University of North Carolina at Chapel Hill; Department of Neurology (D.S.K.), Mayo Clinic, Rochester, MN; Stroke Branch (R.F.G.), National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, MD; and Department of Epidemiology and Biostatistics (H.C.), Michigan State University, East Lansing
| | - Thomas H Mosley
- From the The Memory Impairment and Neurodegenerative Dementia (MIND) Center (S.S., X.Z., K.J.S., C.B., J.H., B.G.W., M.E.G., T.H.M.), University of Mississippi Medical Center, Jackson; Department of Epidemiology (J.A.D., A.R.S.), Johns Hopkins University Bloomberg School of Public Health; Department of Psychiatry and Behavioral Sciences (V.K.), Johns Hopkins University School of Medicine, Baltimore, MD; Department of Neurology (A.L.C.S.), and Department of Biostatistics, Epidemiology, and Informatics (A.L.C.S.), University of Pennsylvania Perelman School of Medicine, Philadelphia; Department of Radiology (C.R.J., R.I.R.), Mayo Clinic, Rochester, MN; Department of Neurology (J.H.), University of Mississippi Medical Center, Jackson; Department of Neurology (P.P.), University of North Carolina at Chapel Hill; Department of Neurology (D.S.K.), Mayo Clinic, Rochester, MN; Stroke Branch (R.F.G.), National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, MD; and Department of Epidemiology and Biostatistics (H.C.), Michigan State University, East Lansing
| |
Collapse
|
28
|
Kern KC, Zagzoug MS, Gottesman RF, Wright CB, Leigh R. Diffusion tensor free water MRI predicts progression of FLAIR white matter hyperintensities after ischemic stroke. Front Neurol 2023; 14:1172031. [PMID: 37808483 PMCID: PMC10559725 DOI: 10.3389/fneur.2023.1172031] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 08/23/2023] [Indexed: 10/10/2023] Open
Abstract
Background The progression of FLAIR white matter hyperintensities (WMHs) on MRI heralds vascular-mediated cognitive decline. Even before FLAIR WMH progression, adjacent normal appearing white matter (NAWM) already demonstrates microstructural deterioration on diffusion tensor imaging (DTI). We hypothesized that elevated DTI free water (FW) would precede FLAIR WMH progression, implicating interstitial fluid accumulation as a key pathological step in the progression of cerebral small vessel disease. Methods Participants at least 3 months after an ischemic stroke or TIA with WMH on MRI underwent serial brain MRIs every 3 months over the subsequent year. For each participant, the WMHs were automatically segmented, serial MRIs were aligned, and a region of WMH penumbra tissue at risk was defined by dilating lesions at any time point and subtracting baseline lesions. Penumbra voxels were classified as either stable or progressing to WMH if they were segmented as new lesions and demonstrated increasing FLAIR intensity over time. Aligned DTI images included FW and FW-corrected fractional anisotropy (FATissue) and mean diffusivity (MDTissue). Logistic regression and area under the receiver-operator characteristic curve (AUC) were used to test whether baseline DTI predicted voxel-wise classification of stable penumbra or progression to WMH while covarying for clinical risk factors. Results In the included participants (n = 26, mean age 71 ± 9 years, 31% female), we detected a median annual voxel-wise WMH growth of 2.9 ± 2.6 ml. Each baseline DTI metric was associated with lesion progression in the penumbra, but FW had the greatest AUC of 0.732 (0.730 - 0.733) for predicting voxel-wise WMH progression pooled across participants. Discussion Baseline increased interstitial fluid, estimated as FW on DTI, predicted the progression of NAWM to WMH over the following year. These results implicate the presence of FW in the pathogenesis of cerebral small vessel disease progression.
Collapse
Affiliation(s)
- Kyle C. Kern
- Stroke Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Marwah S. Zagzoug
- Stroke Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Rebecca F. Gottesman
- Stroke Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Clinton B. Wright
- Stroke Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Richard Leigh
- Stroke Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
- Department of Neurology, Johns Hopkins Medicine, Baltimore, MD, United States
| |
Collapse
|
29
|
Yu K, Chen XF, Guo J, Wang S, Huang XT, Guo Y, Dong SS, Yang TL. Assessment of bidirectional relationships between brain imaging-derived phenotypes and stroke: a Mendelian randomization study. BMC Med 2023; 21:271. [PMID: 37491271 PMCID: PMC10369749 DOI: 10.1186/s12916-023-02982-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 07/17/2023] [Indexed: 07/27/2023] Open
Abstract
BACKGROUND Stroke is a major cause of mortality and long-term disability worldwide. Whether the associations between brain imaging-derived phenotypes (IDPs) and stroke are causal is uncertain. METHODS We performed two-sample bidirectional Mendelian randomization (MR) analyses to explore the causal associations between IDPs and stroke. Summary data of 587 brain IDPs (up to 33,224 individuals) from the UK Biobank and five stroke types (sample size range from 301,663 to 446,696, case number range from 5,386 to 40,585) from the MEGASTROKE consortium were used. RESULTS Forward MR indicated 14 IDPs belong to projection fibers or association fibers were associated with stroke. For example, higher genetically determined mean diffusivity (MD) in the right external capsule was causally associated with an increased risk of small vessel stroke (IVW OR = 2.76, 95% CI 2.07 to 3.68, P = 5.87 × 10-12). Reverse MR indicated that genetically determined higher risk of any ischemic stroke was associated with increased isotropic or free water volume fraction (ISOVF) in body of corpus callosum (IVW β = 0.23, 95% CI 0.14 to 0.33, P = 3.22 × 10-7). This IDP is a commissural fiber and it is not included in the IDPs identified by forward MR. CONCLUSIONS We identified 14 IDPs with statistically significant evidence of causal effects on stroke or stroke subtypes. We also identified potential causal effects of stroke on one IDP of commissural fiber. These findings might guide further work toward identifying preventative strategies at the brain imaging levels.
Collapse
Affiliation(s)
- Ke Yu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Xiao-Feng Chen
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Jing Guo
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Sen Wang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Xiao-Ting Huang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Yan Guo
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Shan-Shan Dong
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China.
| | - Tie-Lin Yang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, Biomedical Informatics & Genomics Center, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China.
| |
Collapse
|
30
|
Visser VL, Caçoilo A, Rusinek H, Weickenmeier J. Mechanical loading of the ventricular wall as a spatial indicator for periventricular white matter degeneration. J Mech Behav Biomed Mater 2023; 143:105921. [PMID: 37269602 PMCID: PMC10266836 DOI: 10.1016/j.jmbbm.2023.105921] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/15/2023] [Accepted: 05/18/2023] [Indexed: 06/05/2023]
Abstract
Progressive white matter degeneration in periventricular and deep white matter regions appears as white matter hyperintensities (WMH) on MRI scans. To date, periventricular WMHs are often associated with vascular dysfunction. Here, we demonstrate that ventricular inflation resulting from cerebral atrophy and hemodynamic pulsation with every heartbeat leads to a mechanical loading state of periventricular tissues that significantly affects the ventricular wall. Specifically, we present a physics-based modeling approach that provides a rationale for ependymal cell involvement in periventricular WMH formation. Building on eight previously created 2D finite element brain models, we introduce novel mechanomarkers for ependymal cell loading and geometric measures that characterize lateral ventricular shape. We show that our novel mechanomarkers, such as maximum ependymal cell deformations and maximum curvature of the ventricular wall, spatially overlap with periventricular WMH locations and are sensitive predictors for WMH formation. We also explore the role of the septum pellucidum in mitigating mechanical loading of the ventricular wall by constraining the radial expansion of the lateral ventricles during loading. Our models consistently show that ependymal cells are stretched thin only in the horns of the ventricles irrespective of ventricular shape. We therefore pose that periventricular WMH etiology is strongly linked to the deterioration of the over-stretched ventricular wall resulting in CSF leakage into periventricular white matter. Subsequent secondary damage mechanisms, including vascular degeneration, exacerbate lesion formation and lead to progressive growth into deep white matter regions.
Collapse
Affiliation(s)
- Valery L Visser
- Department of Mechanical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, United States of America; Department of Biomedical Engineering, Eindhoven University of Technology, 5600 MB Eindhoven, The Netherlands
| | - Andreia Caçoilo
- Department of Mechanical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, United States of America
| | - Henry Rusinek
- Department of Radiology, New York University Grossman School of Medicine, New York, NY 10016, United States of America
| | - Johannes Weickenmeier
- Department of Mechanical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, United States of America.
| |
Collapse
|
31
|
Taghvaei M, Cook P, Sadaghiani S, Shakibajahromi B, Tackett W, Dolui S, De D, Brown C, Khandelwal P, Yushkevich P, Das S, Wolk DA, Detre JA. Young versus older subject diffusion magnetic resonance imaging data for virtual white matter lesion tractography. Hum Brain Mapp 2023; 44:3943-3953. [PMID: 37148501 PMCID: PMC10258527 DOI: 10.1002/hbm.26326] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 04/12/2023] [Accepted: 04/17/2023] [Indexed: 05/08/2023] Open
Abstract
White matter hyperintensity (WMH) lesions on T2 fluid-attenuated inversion recovery (FLAIR) magnetic resonance imaging (MRI) and changes in adjacent normal-appearing white matter can disrupt computerized tract reconstruction and result in inaccurate measures of structural brain connectivity. The virtual lesion approach provides an alternative strategy for estimating structural connectivity changes due to WMH. To assess the impact of using young versus older subject diffusion MRI data for virtual lesion tractography, we leveraged recently available diffusion MRI data from the Human Connectome Project (HCP) Lifespan database. Neuroimaging data from 50 healthy young (39.2 ± 1.6 years) and 46 healthy older (74.2 ± 2.5 years) subjects were obtained from the publicly available HCP-Aging database. Three WMH masks with low, moderate, and high lesion burdens were extracted from the WMH lesion frequency map of locally acquired FLAIR MRI data. Deterministic tractography was conducted to extract streamlines in 21 WM bundles with and without the WMH masks as regions of avoidance in both young and older cohorts. For intact tractography without virtual lesion masks, 7 out of 21 WM pathways showed a significantly lower number of streamlines in older subjects compared to young subjects. A decrease in streamline count with higher native lesion burden was found in corpus callosum, corticostriatal tract, and fornix pathways. Comparable percentages of affected streamlines were obtained in young and older groups with virtual lesion tractography using the three WMH lesion masks of increasing severity. We conclude that using normative diffusion MRI data from young subjects for virtual lesion tractography of WMH is, in most cases, preferable to using age-matched normative data.
Collapse
Affiliation(s)
- Mohammad Taghvaei
- Department of NeurologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Philip Cook
- Department of RadiologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Shokufeh Sadaghiani
- Department of NeurologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | | | - William Tackett
- Department of NeurologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Sudipto Dolui
- Department of RadiologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Debarun De
- Department of Computer EngineeringUniversity of IllinoisUrbanaIllinoisUSA
| | - Christopher Brown
- Department of NeurologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Pulkit Khandelwal
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Paul Yushkevich
- Department of RadiologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Sandhitsu Das
- Department of NeurologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - David A. Wolk
- Department of NeurologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - John A. Detre
- Department of NeurologyUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
32
|
Chan K, Fischer C, Maralani PJ, Black SE, Moody AR, Khademi A. Alzheimer's and vascular disease classification using regional texture biomarkers in FLAIR MRI. Neuroimage Clin 2023; 38:103385. [PMID: 36989851 PMCID: PMC10074987 DOI: 10.1016/j.nicl.2023.103385] [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: 12/03/2022] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023]
Abstract
Interactions between subcortical vascular disease and dementia due to Alzheimer's disease (AD) are unclear, and clinical overlap between the diseases makes diagnosis challenging. Existing studies have shown regional microstructural changes specific to each disease, and that textures in fluid-attenuated inversion recovery (FLAIR) MRI images may characterize abnormalities in tissue microstructure. This work aims to investigate regional FLAIR biomarkers that can differentiate dementia cohorts with and without subcortical vascular disease. FLAIR and diffusion MRI (dMRI) volumes were obtained in 65 mild cognitive impairment (MCI), 21 AD, 44 subcortical vascular MCI (scVMCI), 22 Mixed etiology, and 48 healthy elderly patients. FLAIR texture and intensity biomarkers were extracted from the normal appearing brain matter (NABM), WML penumbra, blood supply territory (BST), and white matter tract regions of each patient. All FLAIR biomarkers were correlated to dMRI metrics in each region and global WML load, and biomarker means between groups were compared using ANOVA. Binary classifications were performed using Random Forest classifiers to investigate the predictive nature of the regional biomarkers, and SHAP feature analysis was performed to further investigate optimal regions of interest for differentiating disease groups. The regional FLAIR biomarkers were strongly correlated to MD, while all biomarker regions but white matter tracts were strongly correlated to WML burden. Classification between Mixed disease and healthy, AD, and scVMCI patients yielded accuracies of 97%, 81%, and 72% respectively using WM tract biomarkers. Classification between scVMCI and healthy, MCI, and AD patients yielded accuracies of 89%, 84%, and 79% respectively using penumbra biomarkers. Only the classification between AD and healthy patients had optimal results using NABM biomarkers. This work presents novel regional FLAIR biomarkers that may quantify white matter degeneration related to subcortical vascular disease, and which indicate that investigating degeneration in specific regions may be more important than assessing global WML burden in vascular disease groups.
Collapse
Affiliation(s)
- Karissa Chan
- Electrical, Computer and Biomedical Engineering Department, Toronto Metropolitan University, 350 Victoria St., Toronto, ON M5B 2K3, Canada; Institute for Biomedical Engineering, Science Tech (iBEST), A Partnership Between St. Michael's Hospital and Toronto Metropolitan University, 209 Victoria St., Toronto, ON M5B 1T8, Canada.
| | - Corinne Fischer
- Institute of Medical Science, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Network, 30 Bond St., Toronto, ON M5B 1W8, Canada; Department of Psychiatry, Faculty of Medicine, University of Toronto, 250 College Street, Toronto, ON M5T 1R8, Canada.
| | - Pejman Jabehdar Maralani
- Department of Medical Imaging, University of Toronto, 263 McCaul St., Toronto, ON M5T 1W7, Canada.
| | - Sandra E Black
- Institute of Medical Science, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada; Horvitz Brain Sciences Research Program, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada; Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada; L.C. Campbell Cognitive Neurology Research Unit, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada.
| | - Alan R Moody
- Department of Medical Imaging, University of Toronto, 263 McCaul St., Toronto, ON M5T 1W7, Canada.
| | - April Khademi
- Electrical, Computer and Biomedical Engineering Department, Toronto Metropolitan University, 350 Victoria St., Toronto, ON M5B 2K3, Canada; Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Network, 30 Bond St., Toronto, ON M5B 1W8, Canada; Institute for Biomedical Engineering, Science Tech (iBEST), A Partnership Between St. Michael's Hospital and Toronto Metropolitan University, 209 Victoria St., Toronto, ON M5B 1T8, Canada; Rotman Research Institute, Baycrest Hospital, 3560 Bathurst Street, Toronto, ON M6A 2E1, Canada.
| |
Collapse
|
33
|
Rivier CA, Renedo D, de Havenon A, Gill TM, Payabvash S, Sheth KN, Falcone GJ. Poor Oral Health Is Associated with Worse Brain Imaging Profiles. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.03.18.23287435. [PMID: 36993472 PMCID: PMC10055602 DOI: 10.1101/2023.03.18.23287435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Importance Poor oral health is a modifiable risk factor that is associated with a variety of health outcomes. However, the relationship between oral and brain health is not well understood. Objective To test the hypothesis that poor oral health is associated with worse neuroimaging brain health profiles in persons without stroke or dementia. Design We conducted a 2-stage cross-sectional neuroimaging study using data from the UK Biobank (UKB). First, we tested for association between self-reported poor oral health and MRI neuroimaging markers of brain health. Second, we used Mendelian Randomization (MR) analyses to test for association between genetically-determined poor oral health and the same neuroimaging markers. Setting Ongoing population study in the United Kingdom. The UKB enrolled participants between 2006 and 2010. Data analysis was performed from September 1, 2022, to January 10, 2023. Participants 40,175 persons aged 40 to 70 enrolled between 2006 to 2010 who underwent a dedicated research brain MRI between 2012 and 2013. Exposures During MRI assessment, poor oral health was defined as the presence of dentures or loose teeth. As instruments for the MR analysis, we used 116 independent DNA sequence variants known to significantly increase the composite risk of decayed, missing, or filled teeth and dentures. Main Outcomes and Measures As neuroimaging markers of brain health, we assessed the volume of white matter hyperintensities (WMH), as well as aggregate measures of fractional anisotropy (FA) and mean diffusivity (MD), two metrics indicative of white matter tract disintegrity obtained through diffusion tensor imaging. These measurements were evaluated across 48 distinct brain regions, with FA and MD values for each region also considered as individual outcomes for the MR method. Results Among study participants, 5,470 (14%) had poor oral health. We found that poor oral health was associated with a 9% increase in WMH volume (beta = 0.09, standard deviation (SD) = 0.014, p P< 0.001), a 10% change in the aggregate FA score (beta = 0.10, SD = 0.013, P < 0.001), and a 5% change in the aggregate MD score (beta = 0.05, SD = 0.013, P < 0.001). Genetically-determined poor oral health was associated with a 30% increase in WMH volume (beta = 0.30, SD = 0.06, P < 0.001), a 43% change in aggregate FA score (beta = 0.42, SD = 0.06, P < 0.001), and an 10% change in aggregate MD score (beta = 0.10, SD = 0.03, P = 0.01). Conclusions and Relevance Among middle age Britons without stroke or dementia enrolled in a large population study, poor oral health was associated with worse neuroimaging brain health profiles. Genetic analyses confirmed these associations, supporting a potential causal association. Because the neuroimaging markers evaluated in the current study are established risk factors for stroke and dementia, our results suggest that oral health may be a promising target for interventions focused on improving brain health.
Collapse
Affiliation(s)
- Cyprien A. Rivier
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Daniela Renedo
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Adam de Havenon
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
- Yale Center for Brain and Mind Health, New Haven, CT, USA
| | - Thomas M. Gill
- Department of Internal Medicine, Yale School of Medicine, 06510, New Haven, CT, United States
| | - Sam Payabvash
- Department of Radiology, Yale University School of Medicine, New Haven, CT, USA
- Yale Center for Brain and Mind Health, New Haven, CT, USA
| | - Kevin N. Sheth
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
- Yale Center for Brain and Mind Health, New Haven, CT, USA
| | - Guido J. Falcone
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
- Yale Center for Brain and Mind Health, New Haven, CT, USA
| |
Collapse
|
34
|
Haddad SMH, Pieruccini-Faria F, Montero-Odasso M, Bartha R. Localized White Matter Tract Integrity Measured by Diffusion Tensor Imaging Is Altered in People with Mild Cognitive Impairment and Associated with Dual-Task and Single-Task Gait Speed. J Alzheimers Dis 2023; 92:1367-1384. [PMID: 36911933 DOI: 10.3233/jad-220476] [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: 03/12/2023]
Abstract
BACKGROUND Altered white matter (WM) tract integrity may contribute to mild cognitive impairment (MCI) and gait abnormalities. OBJECTIVE The purpose of this study was to determine whether diffusion tensor imaging (DTI) metrics were altered in specific portions of WM tracts in people with MCI and to determine whether gait speed variations were associated with the specific DTI metric changes. METHODS DTI was acquired in 44 people with MCI and 40 cognitively normal elderly controls (CNCs). Fractional anisotropy (FA) and radial diffusivity (RD) were measured along 18 major brain WM tracts using probabilistic tractography. The average FA and RD along the tracts were compared between the groups using MANCOVA and post-hoc tests. The tracts with FA or RD differences between the groups were examined using an along-tract exploratory analysis to identify locations that differed between the groups. Associations between FA and RD in whole tracts and in the segments of the tracts that differed between the groups and usual/dual-task gait velocities and gross cognition were examined. RESULTS Lower FA and higher RD was observed in right cingulum-cingulate gyrus endings (rh.ccg) of the MCI group compared to the CNC group. These changes were localized to the posterior portions of the rh.ccg and correlated with gait velocities. CONCLUSION Lower FA and higher RD in the posterior portion of the rh.ccg adjacent to the posterior cingulate suggests decreased microstructural integrity in the MCI group. The correlation of these metrics with gait velocities suggests an important role for this tract in maintaining normal cognitive-motor function.
Collapse
Affiliation(s)
- Seyyed M H Haddad
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, University of Western Ontario, London, Canada
| | - Frederico Pieruccini-Faria
- Department of Medicine, Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada.,Gait and Brain Lab, Parkwood Institute, Lawson Health Research Institute, London, Canada
| | - Manuel Montero-Odasso
- Department of Medicine, Division of Geriatric Medicine, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada.,Gait and Brain Lab, Parkwood Institute, Lawson Health Research Institute, London, Canada.,Department of Epidemiology and Biostatistics, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Canada
| | - Robert Bartha
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, University of Western Ontario, London, Canada.,Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Canada
| |
Collapse
|
35
|
Badr MY, Elkholy AA, Shoeib SM, Bahey MG, Mohamed EA, Reda AM. Assessment of incidence of cerebral vascular diseases and prediction of stroke risk in chronic obstructive pulmonary disease patients using multimodal biomarkers. THE CLINICAL RESPIRATORY JOURNAL 2023; 17:211-228. [PMID: 36696969 PMCID: PMC9978912 DOI: 10.1111/crj.13587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 12/31/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND Early assessment of cerebrovascular disease in chronic obstructive pulmonary disease (COPD) patients is an important issue for a favorable influence on the quality of life. METHODOLOGY This cross-sectional case-control study was conducted on 38 eligible COPD patients (mean age 55.5 ± 11.5, 25 males, and 13 females) and 26 age-/sex-matched healthy controls. All participants were subjected to stroke risk screening instruments that included the Stroke Riskometer™, the Framingham 10-Year Risk Score, the stroke risk screening tool (the Department of Disease Control of Thailand), the My Risk Stroke Calculator, and Q Stroke. Radiologically, diffusion tensor imaging (DTI) and echo-gradient MRI (T2 star) T2 star imaging were done. Color-coded duplex sonography was done. Laboratory investigations included C-reactive protein (CRP), serum amyloid A, plasma fibrinogen level, serum IL6, 8-Isoprostane, vWF and urinary albumin creatinine ratio. RESULTS Stroke risk screening instruments revealed a significant increase in COPD patients. DTI showed a significant bilateral reduction in fractional isotropy and a significant bilateral increase in mean diffusivity of white matter through many areas in COPD patients. Patients also had a significant increase of intima-media thickness, presence of atherosclerotic focal thicknesses or plaques on duplex sonography. There was a significant elevation of CRP, serum amyloid A, plasma fibrinogen level, serum IL6, 8-isoprostane, von Willebrand factor (vWF), and urinary albumin creatinine ratio in COPD patients. CONCLUSION COPD patients had an increased risk for stroke that could be assessed on stroke risk screening instruments, DTI, T2 star, duplex sonography, and laboratory investigation and could be correlated with the severity of the disease.
Collapse
Affiliation(s)
- Marwa Y Badr
- Neurology unit, Neuropsychiatry Department, Faculty of Medicine, Tanta University, Egypt
| | - Amira A Elkholy
- Pulmonology Department, Faculty of Medicine, Tanta University, Egypt
| | - Sara M Shoeib
- Clinical Pathology Department, Faculty of Medicine, Tanta University, Egypt
| | - Marwa G Bahey
- Medical Microbiology and immunology Department, Faculty of Medicine, Tanta University, Egypt
| | - Esraa A Mohamed
- Medical Microbiology and immunology Department, Faculty of Medicine, Tanta University, Egypt
| | - Alaa M Reda
- Diagnostic Radiology Department, Faculty of Medicine, Tanta University, Egypt
| |
Collapse
|
36
|
Heterogeneity of White Matter Hyperintensity and Cognitive Impairment in Patients with Acute Lacunar Stroke. Brain Sci 2022; 12:brainsci12121674. [PMID: 36552134 PMCID: PMC9776102 DOI: 10.3390/brainsci12121674] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/29/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Background: The severity of white matter hyperintensity (WMH) in patients with acute lacunar stroke (ALS) may be not completely parallel to cognitive impairment. Controversies persist about the effects of WMH on cognitive dysfunction. It is vital to explore whether the association may be affected by certain factors and whether a subsequent subgroup analysis is necessary. The aim of this study was to evaluate the relationship between WMH and cognitive impairment in acute lacunar stroke patients and the possible causal factors. Methods: We continuously enrolled patients with ALS who were hospitalized at the First Affiliated Hospital of Soochow University between October 2017 and June 2022. The cognitive function of all patients was assessed by using the Montreal Cognitive Assessment (MoCA) scale 14 ± 2 days after the onset of AIS, and the results were adjusted to the education level. The MoCA scale was reevaluated at the 6-month (day 182 ± 7) follow-up by outpatient visit or video. Demographic and clinical data were collected. The manifestations of chronic cerebral small-vessel disease (CSVD), including the total Fazekas score and total CSVD burden score, were assessed with an MRI scan. A mismatch refers to an inconsistency between the severity of WMH and cognitive dysfunction. A Type 1 mismatch refers to cognitive impairment with mild WMH (total Fazekas score = 0−1), and a Type 2 mismatch refers to severe WMH (total Fazekas score = 5−6) in patients with normal cognitive function. Results: Among 213 enrolled ALS patients, 66 patients (31.0%) had cognitive dysfunction, and 40 patients (18.8%) had mismatches. Twenty-seven cases (12.7%) were Type 1 mismatched, and seventeen cases (8.0%) were Type 2 mismatched. Age, gender, fibrinogen and cerebral infarction history were independent risk factors for cognitive impairment in ALS patients. Imaging features, including moderate to severe WMH, deep WMH and the total CSVD burden score, were also independently associated with cognitive impairment. The patients in the mismatched group were older, had more severe deep WMH and had a higher occurrence of depression (p < 0.05). The NIHSS score, depression and microbleeds were significantly different between the Type 1 mismatched group and the matched group (p = 0.018, p = 0.012 and p = 0.047). Patients in the Type 2 mismatched group were male (p = 0.04), had a lower level of fibrinogen (p = 0.005), a lower incidence of CMBs (p = 0.003), a lower total CSVD burden score (p = 0.017), more severe paraventricular WMH (p = 0.035) and milder deep WMH (p = 0.026). Conclusions: Our study examined a homogeneous study cohort of recruited patients with symptomatic ALS. We found heterogeneity between WMH and cognitive function in ALS patients. Despite a similar WMH severity, some baseline clinical features and other conventional CSVD imaging characteristics may account for this heterogeneity phenomenon. Our findings provide data for the early diagnosis and prevention of cognitive impairment in ALS patients and suggest that the severity of WMH is not completely parallel to cognitive impairment. The white matter microstructural injury and remote WMH effects may account for the mismatch phenomenon. More attention should be paid to understanding the underlying mechanisms and finding new imaging markers.
Collapse
|
37
|
Haddad SMH, Scott CJM, Ozzoude M, Berezuk C, Holmes M, Adamo S, Ramirez J, Arnott SR, Nanayakkara ND, Binns M, Beaton D, Lou W, Sunderland K, Sujanthan S, Lawrence J, Kwan D, Tan B, Casaubon L, Mandzia J, Sahlas D, Saposnik G, Hassan A, Levine B, McLaughlin P, Orange JB, Roberts A, Troyer A, Black SE, Dowlatshahi D, Strother SC, Swartz RH, Symons S, Montero-Odasso M, ONDRI Investigators, Bartha R. Comparison of Diffusion Tensor Imaging Metrics in Normal-Appearing White Matter to Cerebrovascular Lesions and Correlation with Cerebrovascular Disease Risk Factors and Severity. Int J Biomed Imaging 2022; 2022:5860364. [PMID: 36313789 PMCID: PMC9616672 DOI: 10.1155/2022/5860364] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 04/21/2022] [Accepted: 06/01/2022] [Indexed: 11/13/2023] Open
Abstract
Alterations in tissue microstructure in normal-appearing white matter (NAWM), specifically measured by diffusion tensor imaging (DTI) fractional anisotropy (FA), have been associated with cognitive outcomes following stroke. The purpose of this study was to comprehensively compare conventional DTI measures of tissue microstructure in NAWM to diverse vascular brain lesions in people with cerebrovascular disease (CVD) and to examine associations between FA in NAWM and cerebrovascular risk factors. DTI metrics including fractional anisotropy (FA), mean diffusivity (MD), axial diffusivity (AD), and radial diffusivity (RD) were measured in cerebral tissues and cerebrovascular anomalies from 152 people with CVD participating in the Ontario Neurodegenerative Disease Research Initiative (ONDRI). Ten cerebral tissue types were segmented including NAWM, and vascular lesions including stroke, periventricular and deep white matter hyperintensities, periventricular and deep lacunar infarcts, and perivascular spaces (PVS) using T1-weighted, proton density-weighted, T2-weighted, and fluid attenuated inversion recovery MRI scans. Mean DTI metrics were measured in each tissue region using a previously developed DTI processing pipeline and compared between tissues using multivariate analysis of covariance. Associations between FA in NAWM and several CVD risk factors were also examined. DTI metrics in vascular lesions differed significantly from healthy tissue. Specifically, all tissue types had significantly different MD values, while FA was also found to be different in most tissue types. FA in NAWM was inversely related to hypertension and modified Rankin scale (mRS). This study demonstrated the differences between conventional DTI metrics, FA, MD, AD, and RD, in cerebral vascular lesions and healthy tissue types. Therefore, incorporating DTI to characterize the integrity of the tissue microstructure could help to define the extent and severity of various brain vascular anomalies. The association between FA within NAWM and clinical evaluation of hypertension and disability provides further evidence that white matter microstructural integrity is impacted by cerebrovascular function.
Collapse
Affiliation(s)
- Seyyed M. H. Haddad
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, University of Western Ontario, London, Canada
| | - Christopher J. M. Scott
- L.C. Campbell Cognitive Neurology Research Unit, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Medicine (Neurology), Sunnybrook Health Sciences Centre and University of Toronto, Toronto, Canada
| | - Miracle Ozzoude
- L.C. Campbell Cognitive Neurology Research Unit, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Medicine (Neurology), Sunnybrook Health Sciences Centre and University of Toronto, Toronto, Canada
| | | | - Melissa Holmes
- L.C. Campbell Cognitive Neurology Research Unit, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Medicine (Neurology), Sunnybrook Health Sciences Centre and University of Toronto, Toronto, Canada
| | - Sabrina Adamo
- Clinical Neurosciences, University of Toronto, Toronto, Canada
| | - Joel Ramirez
- L.C. Campbell Cognitive Neurology Research Unit, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Medicine (Neurology), Sunnybrook Health Sciences Centre and University of Toronto, Toronto, Canada
| | - Stephen R. Arnott
- Rotman Research Institute, Baycrest Health Sciences, Toronto, Canada
| | - Nuwan D. Nanayakkara
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, University of Western Ontario, London, Canada
| | - Malcolm Binns
- Rotman Research Institute, Baycrest Health Sciences, Toronto, Canada
| | - Derek Beaton
- Rotman Research Institute, Baycrest Health Sciences, Toronto, Canada
| | - Wendy Lou
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Kelly Sunderland
- Rotman Research Institute, Baycrest Health Sciences, Toronto, Canada
| | | | - Jane Lawrence
- Thunder Bay Regional Health Research Institute, Thunder Bay, Canada
| | | | - Brian Tan
- Rotman Research Institute, Baycrest Health Sciences, Toronto, Canada
| | - Leanne Casaubon
- Department of Medicine, University of Toronto, Toronto, Canada
| | - Jennifer Mandzia
- Department of Medicine, Division of Neurology, University of Western Ontario, London, Canada
| | - Demetrios Sahlas
- Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, Canada
| | | | - Ayman Hassan
- Thunder Bay Regional Research Institute, Thunder Bay, Canada
| | - Brian Levine
- Rotman Research Institute, Baycrest Health Sciences, Toronto, Canada
| | | | - J. B. Orange
- School of Communication Sciences and Disorders, Western University, London, Canada
| | - Angela Roberts
- Roxelyn and Richard Pepper Department of Communication Sciences and Disorder, Northwestern University, Evanston, USA
| | - Angela Troyer
- Rotman Research Institute, Baycrest Health Sciences, Toronto, Canada
| | - Sandra E. Black
- L.C. Campbell Cognitive Neurology Research Unit, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Medicine (Neurology), Sunnybrook Health Sciences Centre and University of Toronto, Toronto, Canada
- Rotman Research Institute, Baycrest Health Sciences, Toronto, Canada
- Sunnybrook Health Sciences Centre, University of Toronto, Stroke Research Program, Toronto, Canada
| | | | - Stephen C. Strother
- Rotman Research Institute, Baycrest Health Sciences, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Richard H. Swartz
- Sunnybrook Health Sciences Centre, University of Toronto, Stroke Research Program, Toronto, Canada
| | - Sean Symons
- Department of Medical Imaging, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Manuel Montero-Odasso
- Department of Medicine, Division of Geriatric Medicine, Parkwood Hospital, St. Joseph's Health Care London, London, Canada
| | - ONDRI Investigators
- Ontario Neurodegenerative Disease Initiative, Ontario Brain Institute, Toronto, Canada
| | - Robert Bartha
- Centre for Functional and Metabolic Mapping, Robarts Research Institute, University of Western Ontario, London, Canada
- Department of Medical Biophysics, University of Western Ontario, London, Canada
| |
Collapse
|
38
|
Caçoilo A, Rusinek H, Weickenmeier J. 3D finite-element brain modeling of lateral ventricular wall loading to rationalize periventricular white matter hyperintensity locations. ENGINEERING WITH COMPUTERS 2022; 38:3939-3955. [PMID: 37485473 PMCID: PMC10361695 DOI: 10.1007/s00366-022-01700-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 06/19/2022] [Indexed: 07/25/2023]
Abstract
Aging-related periventricular white matter hyperintensities (pvWMHs) are a common observation in medical images of the aging brain. The underlying tissue damage is part of the complex pathophysiology associated with age-related microstructural changes and cognitive decline. PvWMH formation is linked to blood-brain barrier dysfunction from cerebral small vessel disease as well as the accumulation of cerebrospinal fluid in periventricular tissue due to progressive denudation of the ventricular wall. In need of a unifying theory for pvWMH etiology, image-based finite-element modeling is used to demonstrate that ventricular expansion from age-related cerebral atrophy and hemodynamic loading leads to maximum mechanical loading of the ventricular wall in the same locations that show pvWMHs. Ventricular inflation, induced via pressurization of the ventricular wall, creates significant ventricular wall stretch and stress on the ependymal cells lining the wall, that are linked to cerebrospinal fluid leaking from the lateral ventricles into periventricular white matter tissue. Eight anatomically accurate 3D brain models of cognitively healthy subjects with a wide range of ventricular shapes are created. For all models, our simulations show that mechanomarkers of mechanical wall loading are consistently highest in pvWMHs locations (p < 0.05). Maximum principal strain, the ependymal cell thinning ratio, and wall curvature are on average 14%, 8%, and 24% higher in pvWMH regions compared to the remaining ventricular wall, respectively. Computational modeling provides a powerful framework to systematically study pvWMH formation and growth with the goal to develop pharmacological interventions in the future.
Collapse
Affiliation(s)
- Andreia Caçoilo
- Department of Mechanical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Henry Rusinek
- Department of Radiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Johannes Weickenmeier
- Department of Mechanical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| |
Collapse
|
39
|
Sun Y, Hu Y, Qiu Y, Zhang Y, Jiang C, Lu P, Xu Q, Shi Y, Wei H, Zhou Y. Characterization of white matter over 1–2 years in small vessel disease using MR-based quantitative susceptibility mapping and free-water mapping. Front Aging Neurosci 2022; 14:998051. [PMID: 36247993 PMCID: PMC9562046 DOI: 10.3389/fnagi.2022.998051] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/07/2022] [Indexed: 11/13/2022] Open
Abstract
PurposeThe aim of this study was to investigate alterations in white matter lesions (WMLs) and normal-appearing white matter (NAWM) with small vessel disease (SVD) over 1–2 years using quantitative susceptibility mapping (QSM) and free-water (FW) mapping.MethodsFifty-one SVD patients underwent MRI brain scans and neuropsychological testing both at baseline and follow-up. The main approach for treating these patients is the management of risk factors. Quantitative susceptibility (QS), fractional anisotropy (FA), mean diffusivity (MD), FW, FW-corrected FA (FAT), and FW-corrected MD (MDT) maps within WMLs and NAWM were generated. Furthermore, the JHU-ICBM-DTI label atlas was used as an anatomic guide, and the measurements of the segmented NAWMs were calculated. The average regional values were extracted, and a paired t-test was used to analyze the longitudinal change. Partial correlations were used to assess the relationship between the MRI indices changes (e.g., ΔQSfollowup − baseline/QSbaseline) and the cognitive function changes (e.g., ΔMoCAfollowup − baseline/MoCAbaseline).ResultsAfter SVD risk factor control, no gradual cognitive decline occurred during 1–2 years. However, we still found that the QS values (index of demyelination) increased in the NAWM at follow-up, especially in the NAWM part of the left superior frontal blade (SF), left occipital blade, right uncinate fasciculus, and right corticospinal tract (CST). FW (index of neuroinflammation/edema) analysis revealed that the follow-up group differed from the baseline group in the NAWM part of the right CST and inferior frontal blade (IF). Decreased FAT (index of axonal loss) was observed in the NAWM part of the right SF and IF at follow-up. In addition, the FAT changes in the NAWM part of the right IF were associated with overall cognitive performance changes. In contrast, no significant differences were found in the WMLs.ConclusionThe NAWM was still in the progressive injury process over time, while WMLs remained relatively stable, which supports the notion that SVD is a chronic progressive disease. The process of axonal loss in the NAWM part of the prefrontal lobe might be a biomarker of cognitive changes in the evolution of SVD.
Collapse
Affiliation(s)
- Yawen Sun
- Department of Radiology, School of Medicine, Ren Ji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Hu
- Department of Radiology, School of Medicine, Ren Ji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yage Qiu
- Department of Radiology, School of Medicine, Ren Ji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yuyao Zhang
- School of Information and Science and Technology, ShanghaiTech University, Shanghai, China
| | - Changhao Jiang
- School of Information and Science and Technology, ShanghaiTech University, Shanghai, China
| | - Peiwen Lu
- Department of Neurology, School of Medicine, Ren Ji Hospital, Shanghai Jiao Tong University, Shanghai, China
- Ren Ji-UNSW CHeBA Neurocognitive Center, School of Medicine, Ren Ji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Qun Xu
- Department of Neurology, School of Medicine, Ren Ji Hospital, Shanghai Jiao Tong University, Shanghai, China
- Ren Ji-UNSW CHeBA Neurocognitive Center, School of Medicine, Ren Ji Hospital, Shanghai Jiao Tong University, Shanghai, China
- Department of Health Manage Center, School of Medicine, Ren Ji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yuting Shi
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Hongjiang Wei
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Yan Zhou
| | - Yan Zhou
- Department of Radiology, School of Medicine, Ren Ji Hospital, Shanghai Jiao Tong University, Shanghai, China
- Hongjiang Wei
| |
Collapse
|
40
|
Yuan X, Li X, Xu Y, Zhong L, Yan Z, Chen Z. Microstructural changes of the vestibulocochlear nerve in patients with Ménière's disease using diffusion tensor imaging. Front Neurol 2022; 13:915826. [PMID: 36226092 PMCID: PMC9548978 DOI: 10.3389/fneur.2022.915826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 08/22/2022] [Indexed: 12/01/2022] Open
Abstract
Objective To evaluate the microstructural changes of the vestibulocochlear nerve in patients with Ménière's disease. Methods A total of 26 subjects, 13 patients with MD and 13 healthy controls, underwent diffusion tensor imaging (DTI) on a 3T scanner. The independent sample t-test was used to compare the differences in fractional anisotropy (FA) and apparent diffusion coefficient (ADC) between the two groups. A Pearson correlation was used between DTI and the dizziness handicap inventory (DHI) scores. Results There was a significant decrease in FA and an increase in ADC of the vestibulocochlear nerve in MD patients compared with healthy controls (P = 0.04, P = 0.001). FA had negative correlations with the DHI score (r = −0.62, P = 0.02) and DHI-functional score (r = −0.64, P = 0.02). Conclusion These results are the first evidence of possible changes in the microstructure of the vestibulocochlear nerves in patients with MD. DTI is a potential technique for evaluating the vestibulocochlear nerve in patients with MD.
Collapse
Affiliation(s)
- Xiaojia Yuan
- Department of Chinese Medicine, Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Nan Yang, China
- Department of Radiology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaozhen Li
- Department of Radiology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Xiaozhen Li
| | - Yu Xu
- Department of Radiology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Liqun Zhong
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zhanfeng Yan
- Department of Otolaryngology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zhengguang Chen
- Department of Radiology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Zhengguang Chen
| |
Collapse
|
41
|
Mayer C, Nägele FL, Petersen M, Frey BM, Hanning U, Pasternak O, Petersen E, Gerloff C, Thomalla G, Cheng B. Free-water diffusion MRI detects structural alterations surrounding white matter hyperintensities in the early stage of cerebral small vessel disease. J Cereb Blood Flow Metab 2022; 42:1707-1718. [PMID: 35410517 PMCID: PMC9441727 DOI: 10.1177/0271678x221093579] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In cerebral small vessel disease (CSVD), both white matter hyperintensities (WMH) of presumed vascular origin and the normal-appearing white matter (NAWM) contain microstructural brain alterations on diffusion-weighted MRI (DWI). Contamination of DWI-derived metrics by extracellular free-water can be corrected with free-water (FW) imaging. We investigated the alterations in FW and FW-corrected fractional anisotropy (FA-t) in WMH and surrounding tissue and their association with cerebrovascular risk factors. We analysed 1,000 MRI datasets from the Hamburg City Health Study. DWI was used to generate FW and FA-t maps. WMH masks were segmented on FLAIR and T1-weighted MRI and dilated repeatedly to create 8 NAWM masks representing increasing distance from WMH. Linear models were applied to compare FW and FA-t across WMH and NAWM masks and in association with cerebrovascular risk. Median age was 64 ± 14 years. FW and FA-t were altered 8 mm and 12 mm beyond WMH, respectively. Smoking was significantly associated with FW in NAWM (p = 0.008) and FA-t in WMH (p = 0.008) and in NAWM (p = 0.003) while diabetes and hypertension were not. Further research is necessary to examine whether FW and FA-t alterations in NAWM are predictors for developing WMH.
Collapse
Affiliation(s)
- Carola Mayer
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Felix L Nägele
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marvin Petersen
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Benedikt M Frey
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Uta Hanning
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ofer Pasternak
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, USA.,Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, USA
| | - Elina Petersen
- Clinical for Cardiology, University Heart and Vascular Center, Germany.,Population Health Research Department, University Heart and Vascular Center, Hamburg, Germany
| | - Christian Gerloff
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Götz Thomalla
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Bastian Cheng
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
42
|
The negative affectivity dimension of Type D personality associated with increased risk for acute ischemic stroke and white matter hyperintensity. J Psychosom Res 2022; 160:110973. [PMID: 35749831 DOI: 10.1016/j.jpsychores.2022.110973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 05/24/2022] [Accepted: 06/13/2022] [Indexed: 11/21/2022]
Abstract
OBJECTIVE This study was conducted to examine the relationship among type D personality, acute ischemic stroke (AIS), and white matter hyperintensity (WMH). METHODS In a cross-sectional study conducted between September 2020 and June 2021, 235 patients aged 50-85 years with first-ever ischemic cerebrovascular disease, including 146 males and 89 females, were enrolled. All participants underwent the Type D Scale-14 test containing negative affectivity (NA) and social inhibition (SI) subscales. Clinical and laboratory data were also collected for analysis. The patients were divided into the AIS group (n = 148) and the transient ischemic attack (TIA) group (n = 87) according to whether there was an acute lesion. RESULTS Patients with type D personality had a higher frequency of AIS and LAA and a higher level of WMH. Multiple logistic regression showed that the NA score was related to a 1.11-fold increase in the odds of AIS (95% CI: 1.03-1.19). Neither NA nor SI showed a clear association with a higher frequency of LAA. Higher scores of NA (OR = 1.07, 95% CI: 1.01-1.15), SI (OR = 1.11, 95% CI: 1.03-1.19), and the interaction between the two dimensions (OR = 1.03, 95% CI: 1.01-1.05) were independently associated with an increased load of WMH. CONCLUSION Type D personality was related to AIS and WMH. In particular, it was NA, not SI, affected the occurrence of AIS. Our findings may provide new insights regarding behavioral vulnerability for the development of cerebrovascular disorders.
Collapse
|
43
|
Could salt intake directly affect the cerebral microvasculature in hypertension? J Stroke Cerebrovasc Dis 2022; 31:106632. [PMID: 35870266 DOI: 10.1016/j.jstrokecerebrovasdis.2022.106632] [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: 04/08/2022] [Revised: 06/26/2022] [Accepted: 06/30/2022] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVES Excess dietary salt and chronic kidney disease (CKD) are acknowledged stroke risk factors. The development of small vessel disease, similarly affecting the cerebral and renal microvasculatures, may be an important mechanistic link underlying this interaction. Therefore, we aimed to evaluate if the dietary salt intake and markers of CKD (estimated glomerular filtration rate, albuminuria) relate to transcranial Doppler (TCD) markers of cerebral small vessel disease (CSVD) in hypertensive patients. MATERIALS AND METHODS Fifty-six hypertensive patients (57% with diabetes) underwent TCD monitoring in the middle (MCA) and posterior (PCA) cerebral arteries for evaluating neurovascular coupling (NVC), dynamic cerebral autoregulation (dCA), and vasoreactivity to carbon dioxide (VRCO2). We investigated the relation between renal parameters and TCD studies using Pearson's correlation coefficient and linear regression analyses. RESULTS There were no associations between dCA, VRCO2, NVC, and renal function tests. However, there was a negative association between the daily salt intake and the natural frequency during visual stimulation (r2=0.101, ß=-0.340, p=0.035), indicative of increased rigidity of the cerebral resistance vessels that react to cognitive activation. CONCLUSIONS In this cross-sectional study, we found an association between excess dietary salt consumption and CSVD in hypertensive patients. Future research is needed to evaluate whether the natural frequency could be an early, non-invasive, surrogate marker for microvascular dysfunction in hypertension.
Collapse
|
44
|
Ferris JK, Greeley B, Vavasour IM, Kraeutner SN, Rinat S, Ramirez J, Black SE, Boyd LA. In vivo myelin imaging and tissue microstructure in white matter hyperintensities and perilesional white matter. Brain Commun 2022; 4:fcac142. [PMID: 35694147 PMCID: PMC9178967 DOI: 10.1093/braincomms/fcac142] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 03/28/2022] [Accepted: 05/26/2022] [Indexed: 11/12/2022] Open
Abstract
White matter hyperintensities negatively impact white matter structure and relate to cognitive decline in aging. Diffusion tensor imaging detects changes to white matter microstructure, both within the white matter hyperintensity and extending into surrounding (perilesional) normal-appearing white matter. However, diffusion tensor imaging markers are not specific to tissue components, complicating the interpretation of previous microstructural findings. Myelin water imaging is a novel imaging technique that provides specific markers of myelin content (myelin water fraction) and interstitial fluid (geometric mean T2). Here we combined diffusion tensor imaging and myelin water imaging to examine tissue characteristics in white matter hyperintensities and perilesional white matter in 80 individuals (47 older adults and 33 individuals with chronic stroke). To measure perilesional normal-appearing white matter, white matter hyperintensity masks were dilated in 2 mm segments up to 10 mm in distance from the white matter hyperintensity. Fractional anisotropy, mean diffusivity, myelin water fraction, and geometric mean T2 were extracted from white matter hyperintensities and perilesional white matter. We observed a spatial gradient of higher mean diffusivity and geometric mean T2, and lower fractional anisotropy, in the white matter hyperintensity and perilesional white matter. In the chronic stroke group, myelin water fraction was reduced in the white matter hyperintensity but did not show a spatial gradient in perilesional white matter. Across the entire sample, white matter metrics within the white matter hyperintensity related to whole-brain white matter hyperintensity volume; with increasing white matter hyperintensity volume there was increased mean diffusivity and geometric mean T2, and decreased myelin water fraction in the white matter hyperintensity. Normal-appearing white matter adjacent to white matter hyperintensities exhibits characteristics of a transitional stage between healthy white matter and white matter hyperintensities. This effect was observed in markers sensitive to interstitial fluid, but not in myelin water fraction, the specific marker of myelin concentration. Within the white matter hyperintensity, interstitial fluid was higher and myelin concentration was lower in individuals with more severe cerebrovascular disease. Our data suggests white matter hyperintensities have penumbra-like effects in perilesional white matter that specifically reflect increased interstitial fluid, with no changes to myelin concentration. In contrast, within the white matter hyperintensity there are varying levels of demyelination, which vary based on the severity of cerebrovascular disease. Diffusion tensor imaging and myelin imaging may be useful clinical markers to predict white matter hyperintensity formation, and to stage neuronal damage within white matter hyperintensities.
Collapse
Affiliation(s)
- Jennifer K. Ferris
- University of British Columbia Graduate Programs in Rehabilitation Sciences, , Vancouver, Canada
| | - Brian Greeley
- University of British Columbia Department of Physical Therapy, , Vancouver, Canada
| | - Irene M. Vavasour
- The University of British Columbia Department of Radiology, , Vancouver, Canada
- University of British Columbia UBC MRI Research Centre, Faculty of Medicine, , Vancouver, Canada
| | - Sarah N. Kraeutner
- University of British Columbia Department of Psychology, , Okanagan, Kelowna, Canada
| | - Shie Rinat
- University of British Columbia Graduate Programs in Rehabilitation Sciences, , Vancouver, Canada
| | - Joel Ramirez
- LC Campbell Cognitive Neurology Research Unit, Dr Sandra Black Centre for Brain Resilience and Recovery , Toronto, Canada
- Sunnybrook Research Institute, University of Toronto Hurvitz Brain Sciences Research Program, , Toronto, Canada
| | - Sandra E. Black
- LC Campbell Cognitive Neurology Research Unit, Dr Sandra Black Centre for Brain Resilience and Recovery , Toronto, Canada
- Sunnybrook Research Institute, University of Toronto Hurvitz Brain Sciences Research Program, , Toronto, Canada
| | - Lara A. Boyd
- University of British Columbia Graduate Programs in Rehabilitation Sciences, , Vancouver, Canada
- University of British Columbia Department of Physical Therapy, , Vancouver, Canada
| |
Collapse
|
45
|
Abstract
BACKGROUND Blood pressure variability (BPV) has been linked with cognitive impairment and dementia. However, the pathophysiological mechanisms by which BPV affects cognition are unclear. This systematic review aims to assess the links between different BPV measures and white and grey matter structures. METHODS AND RESULTS The following databases were searched from inception through to January 2021; EMBASE, MEDLINE, EMCARE and SCOPUS. Studies that reported on the relationship between within-individual BPV (short, medium or long-term variability) or a circadian blood pressure (BP) measurement and MRI assessed brain structures were included. Overall, 20 studies met the criteria and were included, of which 11 studies looked at short-term BPV, eight articles investigated visit-to-visit BPV and one study looked at a compositional BPV measurement. Due to heterogeneity in study samples, meta-analysis was not possible. Across the included studies, associations between MRI indices and BP dipping patterns were mixed; higher long-term BPV and higher sleep systolic BPV was found to be associated with lower whole brain volume and hippocampal volume. CONCLUSION Increased BPV, in particular systolic long-term and systolic night-time BPV, appears to be associated with lower brain volume and hippocampal volume. This highlights the adverse effect that increased BPV has upon the brain, potentially contributing to cognitive decline, including dementia, in late-life.
Collapse
|
46
|
Bahsoun MA, Khan MU, Mitha S, Ghazvanchahi A, Khosravani H, Jabehdar Maralani P, Tardif JC, Moody AR, Tyrrell PN, Khademi A. FLAIR MRI biomarkers of the normal appearing brain matter are related to cognition. Neuroimage Clin 2022; 34:102955. [PMID: 35180579 PMCID: PMC8857609 DOI: 10.1016/j.nicl.2022.102955] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 01/28/2022] [Accepted: 02/01/2022] [Indexed: 01/04/2023]
Abstract
Normal appearing brain matter (NABM) biomarkers in FLAIR MRI are related to cognition. NABM texture in FLAIR MRI is correlated to mean diffusivity (MD) in dMRI. Analysis conducted on large multicentre FLAIR MRI dataset: 1400 subjects, 87 centers. NABM biomarkers vary differently across age and MoCA categories. Biomarkers showed differences in patients with AD dementia and vascular disease.
A novel biomarker panel was proposed to quantify macro and microstructural biomarkers from the normal-appearing brain matter (NABM) in multicentre fluid-attenuation inversion recovery (FLAIR) MRI. The NABM is composed of the white and gray matter regions of the brain, with the lesions and cerebrospinal fluid removed. The primary hypothesis was that NABM biomarkers from FLAIR MRI are related to cognitive outcome as determined by MoCA score. There were three groups of features designed for this task based on 1) texture: microstructural integrity (MII), macrostructural damage (MAD), microstructural damage (MID), 2) intensity: median, skewness, kurtosis and 3) volume: NABM to ICV volume ratio. Biomarkers were extracted from over 1400 imaging volumes from more than 87 centres and unadjusted ANOVA analysis revealed significant differences in means of the MII, MAD, and NABM volume biomarkers across all cognitive groups. In an adjusted ANCOVA model, a significant relationship between MoCA categories was found that was dependent on subject age for MII, MAD, intensity, kurtosis and NABM volume biomarkers. These results demonstrate that structural brain changes in the NABM are related to cognitive outcome (with different relationships depending on the age of the subjects). Therefore these biomarkers have high potential for clinical translation. As a secondary hypothesis, we investigated whether texture features from FLAIR MRI can quantify microstructural changes related to how “structured” or “damaged” the tissue is. Based on correlation analysis with diffusion weighted MRI (dMRI), it was shown that FLAIR MRI texture biomarkers (MII and MAD) had strong correlations to mean diffusivity (MD) which is related to tissue degeneration in the GM and WM regions. As FLAIR MRI is routinely collected for clinical neurological examinations, novel biomarkers from FLAIR MRI could be used to supplement current clinical biomarkers and for monitoring disease progression. Biomarkers could also be used to stratify patients into homogeneous disease subgroups for clinical trials, or to learn more about mechanistic development of dementia disease.
Collapse
Affiliation(s)
- M-A Bahsoun
- Electrical, Computer and Biomedical Engineering Dept., Ryerson University, Toronto, ON, Canada
| | - M U Khan
- Electrical, Computer and Biomedical Engineering Dept., Ryerson University, Toronto, ON, Canada
| | - S Mitha
- Electrical, Computer and Biomedical Engineering Dept., Ryerson University, Toronto, ON, Canada
| | - A Ghazvanchahi
- Electrical, Computer and Biomedical Engineering Dept., Ryerson University, Toronto, ON, Canada
| | - H Khosravani
- Hurvitz Brain Sciences Program Division of Neurology, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | | | - J-C Tardif
- Montreal Heart Institute, Montreal, QU, Canada; Department of Medicine, Université de Montréal, QU, Canada
| | - A R Moody
- Department of Medical Imaging, University of Toronto, Toronto, ON, Canada
| | - P N Tyrrell
- Department of Medical Imaging, University of Toronto, Toronto, ON, Canada; Department of Statistical Sciences, University of Toronto, Toronto, ON, Canada; Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - A Khademi
- Electrical, Computer and Biomedical Engineering Dept., Ryerson University, Toronto, ON, Canada; Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Network, Toronto, ON, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), a partnership between St. Michael's Hospital and Ryerson University, Toronto, ON, Canada
| |
Collapse
|
47
|
Hsieh S, Yang MH. Potential Diffusion Tensor Imaging Biomarkers for Elucidating Intra-Individual Age-Related Changes in Cognitive Control and Processing Speed. Front Aging Neurosci 2022; 14:850655. [PMID: 35557836 PMCID: PMC9087335 DOI: 10.3389/fnagi.2022.850655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 03/24/2022] [Indexed: 11/24/2022] Open
Abstract
Cognitive aging, especially cognitive control, and processing speed aging have been well-documented in the literature. Most of the evidence was reported based on cross-sectional data, in which inter-individual age effects were shown. However, there have been some studies pointing out the possibility of overlooking intra-individual changes in cognitive aging. To systematically examine whether age-related differences and age-related changes might yield distinctive patterns, this study directly compared cognitive control function and processing speed between different cohorts versus follow-up changes across the adult lifespan. Moreover, considering that cognitive aging has been attributed to brain disconnection in white matter (WM) integrity, this study focused on WM integrity via acquiring diffusion-weighted imaging data with an MRI instrument that are further fitted to a diffusion tensor model (i.e., DTI) to detect water diffusion directionality (i.e., fractional anisotropy, FA; mean diffusivity, MD; radial diffusivity, RD; axial diffusivity, AxD). Following data preprocessing, 114 participants remained for further analyses in which they completed the two follow-up sessions (with a range of 1-2 years) containing a series of neuropsychology instruments and computerized cognitive control tasks. The results show that many significant correlations between age and cognitive control functions originally shown on cross-sectional data no longer exist on the longitudinal data. The current longitudinal data show that MD, RD, and AxD (especially in the association fibers of anterior thalamic radiation) are more strongly correlated to follow-up aging processes, suggesting that axonal/myelin damage is a more robust phenomenon for observing intra-individual aging processes. Moreover, processing speed appears to be the most prominent cognitive function to reflect DTI-related age (cross-sectional) and aging (longitudinal) effects. Finally, converging the results from regression analyses and mediation models, MD, RD, and AxD appear to be the representative DTI measures to reveal age-related changes in processing speed. To conclude, the current results provide new insights to which indicator of WM integrity and which type of cognitive changes are most representative (i.e., potentially to be neuroimaging biomarkers) to reflect intra-individual cognitive aging processes.
Collapse
Affiliation(s)
- Shulan Hsieh
- Cognitive Electrophysiology Laboratory: Control, Aging, Sleep, and Emotion, Department of Psychology, National Cheng Kung University, Tainan, Taiwan
- Institute of Allied Health Sciences, National Cheng Kung University, Tainan, Taiwan
- Department of Public Health, National Cheng Kung University, Tainan, Taiwan
| | - Meng-Heng Yang
- Cognitive Electrophysiology Laboratory: Control, Aging, Sleep, and Emotion, Department of Psychology, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
48
|
Ma Z, Reich DS, Dembling S, Duyn JH, Koretsky AP. Outlier detection in multimodal MRI identifies rare individual phenotypes among more than 15,000 brains. Hum Brain Mapp 2022; 43:1766-1782. [PMID: 34957633 PMCID: PMC8886649 DOI: 10.1002/hbm.25756] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 11/25/2021] [Accepted: 12/04/2021] [Indexed: 01/11/2023] Open
Abstract
Outliers in neuroimaging represent spurious data or the data of unusual phenotypes that deserve special attention such as clinical follow-up. Outliers have usually been detected in a supervised or semi-supervised manner for labeled neuroimaging cohorts. There has been much less work using unsupervised outlier detection on large unlabeled cohorts like the UK Biobank brain imaging dataset. Given its large sample size, rare imaging phenotypes within this unique cohort are of interest, as they are often clinically relevant and could be informative for discovering new processes. Here, we developed a two-level outlier detection and screening methodology to characterize individual outliers from the multimodal MRI dataset of more than 15,000 UK Biobank subjects. In primary screening, using brain ventricles, white matter, cortical thickness, and functional connectivity-based imaging phenotypes, every subject was parameterized with an outlier score per imaging phenotype. Outlier scores of these imaging phenotypes had good-to-excellent test-retest reliability, with the exception of resting-state functional connectivity (RSFC). Due to the low reliability of RSFC outlier scores, RSFC outliers were excluded from further individual-level outlier screening. In secondary screening, the extreme outliers (1,026 subjects) were examined individually, and those arising from data collection/processing errors were eliminated. A representative subgroup of 120 subjects from the remaining non-artifactual outliers were radiologically reviewed, and radiological findings were identified in 97.5% of them. This study establishes an unsupervised framework for investigating rare individual imaging phenotypes within a large neuroimaging cohort.
Collapse
Affiliation(s)
- Zhiwei Ma
- Laboratory of Functional and Molecular ImagingNational Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMarylandUSA
| | - Daniel S. Reich
- Translational Neuroradiology SectionNational Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMarylandUSA
| | - Sarah Dembling
- Laboratory of Functional and Molecular ImagingNational Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMarylandUSA
| | - Jeff H. Duyn
- Laboratory of Functional and Molecular ImagingNational Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMarylandUSA
| | - Alan P. Koretsky
- Laboratory of Functional and Molecular ImagingNational Institute of Neurological Disorders and Stroke, National Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
49
|
Kular L, Ewing E, Needhamsen M, Pahlevan Kakhki M, Covacu R, Gomez-Cabrero D, Brundin L, Jagodic M. DNA methylation changes in glial cells of the normal-appearing white matter in Multiple Sclerosis patients. Epigenetics 2022; 17:1311-1330. [PMID: 35094644 PMCID: PMC9586622 DOI: 10.1080/15592294.2021.2020436] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Multiple Sclerosis (MS), the leading cause of non-traumatic neurological disability in young adults, is a chronic inflammatory and neurodegenerative disease of the central nervous system (CNS). Due to the poor accessibility to the target organ, CNS-confined processes underpinning the later progressive form of MS remain elusive thereby limiting treatment options. We aimed to examine DNA methylation, a stable epigenetic mark of genome activity, in glial cells to capture relevant molecular changes underlying MS neuropathology. We profiled DNA methylation in nuclei of non-neuronal cells, isolated from 38 post-mortem normal-appearing white matter (NAWM) specimens of MS patients (n = 8) in comparison to white matter of control individuals (n = 14), using Infinium MethylationEPIC BeadChip. We identified 1,226 significant (genome-wide adjusted P-value < 0.05) differentially methylated positions (DMPs) between MS patients and controls. Functional annotation of the altered DMP-genes uncovered alterations of processes related to cellular motility, cytoskeleton dynamics, metabolic processes, synaptic support, neuroinflammation and signaling, such as Wnt and TGF-β pathways. A fraction of the affected genes displayed transcriptional differences in the brain of MS patients, as reported by publically available transcriptomic data. Cell type-restricted annotation of DMP-genes attributed alterations of cytoskeleton rearrangement and extracellular matrix remodelling to all glial cell types, while some processes, including ion transport, Wnt/TGF-β signaling and immune processes were more specifically linked to oligodendrocytes, astrocytes and microglial cells, respectively. Our findings strongly suggest that NAWM glial cells are highly altered, even in the absence of lesional insult, collectively exhibiting a multicellular reaction in response to diffuse inflammation.
Collapse
Affiliation(s)
- Lara Kular
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Ewoud Ewing
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Maria Needhamsen
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Majid Pahlevan Kakhki
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Ruxandra Covacu
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - David Gomez-Cabrero
- Department of Medicine, Unit of Computational Medicine, Center for Molecular Medicine, Karolinska Institutet, Solna, Sweden
- Mucosal and Salivary Biology Division, King’s College London Dental Institute, London, UK
- Translational Bioinformatics Unit, Navarrabiomed, Complejo Hospitalario de Navarra (Chn), Universidad Pública de Navarra (Upna), IdiSNA, Pamplona, Spain
- Biological & Environmental Sciences & Engineering Division, King Abdullah University of Science & Technology, Thuwal, Kingdom of Saudi Arabia
| | - Lou Brundin
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Maja Jagodic
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
50
|
Petersen M, Frey BM, Mayer C, Kühn S, Gallinat J, Hanning U, Fiehler J, Borof K, Jagodzinski A, Gerloff C, Thomalla G, Cheng B. Fixel based analysis of white matter alterations in early stage cerebral small vessel disease. Sci Rep 2022; 12:1581. [PMID: 35091684 PMCID: PMC8799636 DOI: 10.1038/s41598-022-05665-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 01/11/2022] [Indexed: 12/13/2022] Open
Abstract
Cerebral small vessel disease (CSVD) is a common cause of morbidity and cognitive decline in the elderly population. However, characterizing the disease pathophysiology and its association with potential clinical sequelae in early stages is less well explored. We applied fixel-based analysis (FBA), a novel framework of investigating microstructural white matter integrity by diffusion-weighted imaging, to data of 921 participants of the Hamburg City Health Study, comprising middle-aged individuals with increased cerebrovascular risk in early stages of CSVD. In individuals in the highest quartile of white matter hyperintensity loads (n = 232, median age 63 years; IQR 15.3 years), FBA detected significantly reduced axonal density and increased atrophy of transcallosal fiber tracts, the bilateral superior longitudinal fasciculus, and corticospinal tracts compared to participants in the lowest quartile of white matter hyperintensities (n = 228, mean age 55 years; IQR 10 years). Analysis of all participants (N = 921) demonstrated a significant association between reduced fiber density and worse executive functions operationalized by the Trail Making Test. Findings were confirmed by complementary analysis of diffusion tensor metrics.
Collapse
Affiliation(s)
- Marvin Petersen
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| | - Benedikt M Frey
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Carola Mayer
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Simone Kühn
- Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jürgen Gallinat
- Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Uta Hanning
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jens Fiehler
- Department of Diagnostic and Interventional Neuroradiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katrin Borof
- Epidemiological Study Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Annika Jagodzinski
- Epidemiological Study Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of General and Interventional Cardiology, University Heart and Vascular Center, Hamburg, Germany
| | - Christian Gerloff
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Götz Thomalla
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Bastian Cheng
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| |
Collapse
|