1
|
Ackah JA, Li X, Zeng H, Chen X. Imaging-validated correlates and implications of the pathophysiologic mechanisms of ageing-related cerebral large artery and small vessel diseases: a systematic review and meta-analysis. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2025; 21:12. [PMID: 40264233 PMCID: PMC12016073 DOI: 10.1186/s12993-025-00274-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/21/2025] [Indexed: 04/24/2025]
Abstract
BACKGROUND Cerebral large artery and small vessel diseases are considered substrates of neurological disorders. We explored how the mechanisms of neurovascular uncoupling, dysfunctional blood-brain-barrier (BBB), compromised glymphatic pathway, and impaired cerebrovascular reactivity (CVR) and autoregulation, identified through diverse neuroimaging techniques, impact cerebral large artery and small vessel diseases. METHODS Studies (1990-2024) that reported on neuroradiological findings on ageing-related cerebral large artery and small vessel diseases were reviewed. Fifty-two studies involving 23,693 participants explored the disease mechanisms, 9 studies (sample size = 3,729) of which compared metrics of cerebrovascular functions (CF) between participants with cerebral large artery and small vessel diseases (target group) and controls with no vascular disease. Measures of CF included CVR, cerebral blood flow (CBF), blood pressure and arterial stiffness. RESULTS The findings from 9 studies (sample size = 3,729, mean age = 60.2 ± 11.5 years), revealed negative effect sizes of CVR [SMD = - 1.86 (95% CI - 2.80, - 0.92)] and CBF [SMD = - 2.26 (95% CI - 4.16, - 0.35)], respectively indicating a reduction in cerebrovascular functions in the target group compared to their controls. Conversely, there were significant increases in the measures of blood pressure [SMD = 0.32 (95% CI 0.18, 0.46)] and arterial stiffness [SMD = 0.87 (95% CI 0.77, 0.98)], which signified poor cerebrovascular functions in the target group. In the combined model the overall average effect size was negative [SMD = - 0.81 (95% CI - 1.53 to - 0.08), p < 0.001]. Comparatively, this suggests that the negative impacts of CVR and CBF reductions significantly outweighed the effects of blood pressure and arterial stiffness, thereby predominantly shaping the overall model. Against their controls, trends of reduction in CF were observed exclusively among participants with cerebral large artery disease (SMD = - 2.09 [95% CI: - 3.57, - 0.62]), as well as those with small vessel diseases (SMD = - 0.85 [95% CI - 1.34, - 0.36]). We further delineated the underlying mechanisms and discussed their interconnectedness with cognitive impairments. CONCLUSION In a vicious cycle, dysfunctional mechanisms in the glymphatic system, neurovascular unit, BBB, autoregulation, and reactivity play distinct roles that contribute to reduced CF and cognitive risk among individuals with cerebral large artery and/or small vessel diseases. Reduction in CVR and CBF points to reductions in CF, which is associated with increased risk of cognitive impairment among ageing populations ≥ 60 years.
Collapse
Affiliation(s)
- Joseph A Ackah
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Xuelong Li
- Department of Neurology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Huixing Zeng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Xiangyan Chen
- Division of Science, Engineering, and Health Studies, College of Professional and Continuing Education, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China.
| |
Collapse
|
2
|
Wen C, Gan JH, Liu S, Lu H, Wang LC, Wu H, Shi ZH, Ji Y. Enlarged perivascular spaces correlate with blood-brain barrier leakage and cognitive impairment in Alzheimer's disease. J Alzheimers Dis 2025; 104:382-392. [PMID: 39924914 DOI: 10.1177/13872877251317220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
Abstract
BackgroundThe clinical significance of enlarged perivascular spaces (EPVS) in Alzheimer' s disease (AD) was ambiguous.ObjectiveTo investigate whether EPVS contribute to blood-brain barrier (BBB) leakage and cognition in AD.MethodsThe study included a total of 64 participants (26 healthy controls and 38 patients with AD). The evaluation of EPVS and BBB permeability was performed in specific anatomical locations: the centrum semiovale (CSO), basal ganglia, and hippocampus. The EPVS ratings were performed according to Potter's instructions. BBB permeability was evaluated using dynamic contrast-enhanced-MRI. The relationship between EPVS and global cognition (Mini-Mental State Examination and Montreal Cognitive Assessment), cognitive subdomains, and BBB permeability were examined in both groups. Finally, the relationship between CSO BBB permeability and cognition in AD patients was investigated.ResultsHigh-grade CSO EPVS was found associated with AD (OR: 3.40, 95% CI: 1.11-11.90, p = 0.04). In the AD group, a significant correlation was observed between high-grade CSO EPVS and lower MMSE score (r = -0.36, p = 0.03) and verbal fluency (r = -0.44, p = 0.01). High-grade CSO EPVS positively correlated with BBB leakage (r = 0.58, p < 0.001). The BBB permeability of CSO negatively correlated with verbal fluency (r = -0.52, p < 0.001) and attention (r = -0.40, p = 0.01).ConclusionsHigh-grade CSO EPVS is related to BBB leakage, which contributes to cognitive impairment in AD patients, especially verbal frequency. CSO EPVS can function as a convenient AD marker for intervention and therapy.
Collapse
Affiliation(s)
- Chen Wen
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jing-Huan Gan
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Shuai Liu
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin Dementia Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin, China
| | - Hao Lu
- Department of Radiology, Tianjin Huanhu Hospital, Tianjin, China
| | - Li-Chen Wang
- Department of Radiology, Tianjin Huanhu Hospital, Tianjin, China
| | - Hao Wu
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin Dementia Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin, China
| | - Zhi-Hong Shi
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin Dementia Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin, China
| | - Yong Ji
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, China National Clinical Research Center for Neurological Diseases, Beijing, China
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin Dementia Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin, China
| |
Collapse
|
3
|
Stringer MS, Blair GW, Kopczak A, Kerkhofs D, Thrippleton MJ, Chappell FM, Maniega SM, Brown R, Shuler K, Hamilton I, Garcia DJ, Doubal FN, Clancy U, Sakka E, Poliakova T, Janssen E, Duering M, Ingrisch M, Staals J, Backes WH, van Oostenbrugge R, Biessels GJ, Dichgans M, Wardlaw JM. Cerebrovascular Function in Sporadic and Genetic Cerebral Small Vessel Disease. Ann Neurol 2025; 97:483-498. [PMID: 39552538 PMCID: PMC11831873 DOI: 10.1002/ana.27136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/19/2024]
Abstract
OBJECTIVE Cerebral small vessel diseases (SVDs) are associated with cerebrovascular dysfunction, such as increased blood-brain barrier leakage (permeability surface area product), vascular pulsatility, and decreased cerebrovascular reactivity (CVR). No studies assessed all 3 functions concurrently. We assessed 3 key vascular functions in sporadic and genetic SVD to determine associations with SVD severity, subtype, and interrelations. METHODS In this prospective, cross-sectional, multicenter INVESTIGATE-SVDs study, we acquired brain magnetic resonance imaging in patients with sporadic SVD/cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), including structural, quantitative microstructural, permeability surface area product, blood plasma volume fraction, vascular pulsatility, and CVR (in response to CO2) scans. We determined vascular function and white matter hyperintensity (WMH) associations, using covariate-adjusted linear regression; normal-appearing white matter and WMH differences, interrelationships between vascular functions, using linear mixed models; and major sources of variance using principal component analyses. RESULTS We recruited 77 patients (45 sporadic/32 CADASIL) at 3 sites. In adjusted analyses, patients with worse WMH had lower CVR (B = -1.78, 95% CI -3.30, -0.27) and blood plasma volume fraction (B = -0.594, 95% CI -0.987, -0.202). CVR was worse in WMH than normal-appearing white matter (eg, CVR: B = -0.048, 95% CI -0.079, -0.017). Adjusting for WMH severity, SVD subtype had minimal influence on vascular function (eg, CVR in CADASIL vs sporadic: B = 0.0169, 95% CI -0.0247, 0.0584). Different vascular function mechanisms were not generally interrelated (eg, permeability surface area product~CVR: B = -0.85, 95% CI -4.72, 3.02). Principal component analyses identified WMH volume/quantitative microstructural metrics explained most variance in CADASIL and arterial pulsatility in sporadic SVD, but similar main variance sources. INTERPRETATION Vascular function was worse with higher WMH, and in WMH than normal-appearing white matter. Sporadic SVD-CADASIL differences largely reflect disease severity. Limited vascular function interrelations may suggest disease stage-specific differences. ANN NEUROL 2025;97:483-498.
Collapse
Affiliation(s)
- Michael S. Stringer
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | - Gordon W. Blair
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | - Anna Kopczak
- Institute for Stroke and Dementia Research (ISD)University HospitalMunichGermany
| | - Danielle Kerkhofs
- Department of Neurology, CARIM School for cardiovascular diseasesMaastricht University Medical CenterMaastrichtthe Netherlands
| | - Michael J. Thrippleton
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | - Francesca M. Chappell
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | - Susana Muñoz Maniega
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | - Rosalind Brown
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | - Kirsten Shuler
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | - Iona Hamilton
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | - Daniela Jaime Garcia
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | - Fergus N. Doubal
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | - Una Clancy
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | - Eleni Sakka
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | - Tetiana Poliakova
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | - Esther Janssen
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | - Marco Duering
- Institute for Stroke and Dementia Research (ISD)University HospitalMunichGermany
- Medical Image Analysis Center (MIAC AG) and Department of Biomedical EngineeringUniversity of BaselBaselSwitzerland
| | | | - Julie Staals
- Department of Neurology, CARIM School for cardiovascular diseasesMaastricht University Medical CenterMaastrichtthe Netherlands
| | - Walter H. Backes
- Department of Radiology & Nuclear MedicineMaastricht University Medical Center, Schools for Mental Health & Neuroscience and Cardiovascular DiseaseMaastrichtthe Netherlands
| | - Robert van Oostenbrugge
- Department of Neurology, CARIM School for cardiovascular diseasesMaastricht University Medical CenterMaastrichtthe Netherlands
| | - Geert Jan Biessels
- Department of Neurology and Neurosurgery, UMC Utrecht Brain CenterUniversity Medical Center UtrechtUtrechtNetherlands
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD)University HospitalMunichGermany
- German Center for Neurodegenerative Diseases (DZNE, Munich)MunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | - Joanna M. Wardlaw
- Brain Research Imaging Center, Center for Clinical Brain SciencesUK Dementia Institute Center at the University of EdinburghEdinburghUK
| | | |
Collapse
|
4
|
Jiang YQ, Chen QZ, Yang Y, Zang CX, Ma JW, Wang JR, Dong YR, Zhou N, Yang X, Li FF, Bao XQ, Zhang D. White matter lesions contribute to motor and non-motor disorders in Parkinson's disease: a critical review. GeroScience 2025; 47:591-609. [PMID: 39576561 PMCID: PMC11872850 DOI: 10.1007/s11357-024-01428-1] [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: 07/09/2024] [Accepted: 11/08/2024] [Indexed: 03/04/2025] Open
Abstract
Parkinson's disease (PD) is a prevalent neurodegenerative disease, characterized by movement disorders and non-motor symptoms like cognitive impairment and depression. Degeneration of dopaminergic neurons in the substantia nigra and Lewy bodies have long been considered as main neuropathological changes. However, recent magnetic resonance imaging (MRI) studies have shown that white matter lesions (WMLs) were present in PD patients. WMLs are characterized by loss or impairment of myelin sheath in central nerve fibers, which are closely correlated with motor and cognitive dysfunction in PD. WMLs alterations precede nigrostriatal neuronal losses and can independently affect the clinical severity or characteristics of motor coordination in PD patients. Currently, the exact mechanism of WMLs involvement in the occurrence and development of PD remains unclear. It is speculated that WMLs may participate in the pathogenesis of PD by disrupting important connections in brain or promoting axonal degeneration. In this review, we will discuss the pathological changes and mechanisms of WMLs, elaborate the impact of WMLs on the progression of PD, clarify the importance of WMLs in PD pathogenesis, and thus provide novel targets for PD treatments.
Collapse
Affiliation(s)
- Yue-Qi Jiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Qiu-Zhu Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Yang Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Cai-Xia Zang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Jing-Wei Ma
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Jin-Rong Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Yi-Rong Dong
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Ning Zhou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Xing Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Fang-Fang Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China
| | - Xiu-Qi Bao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China.
| | - Dan Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, People's Republic of China.
| |
Collapse
|
5
|
Chen JXY, Vipin A, Sandhu GK, Leow YJ, Zailan FZ, Tanoto P, Lee ES, Lee KL, Cheung C, Kandiah N. Blood-brain barrier integrity disruption is associated with both chronic vascular risk factors and white matter hyperintensities. J Prev Alzheimers Dis 2025; 12:100029. [PMID: 39863325 DOI: 10.1016/j.tjpad.2024.100029] [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: 10/01/2024] [Revised: 11/21/2024] [Accepted: 12/02/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND Cardiovascular risk factors (CRFs) like hypertension, high cholesterol, and diabetes mellitus are increasingly linked to cognitive decline and dementia, especially in cerebral small vessel disease (cSVD). White matter hyperintensities (WMH) are closely associated with cognitive impairment, but the mechanisms behind their development remain unclear. Blood-brain barrier (BBB) dysfunction may be a key factor, particularly in cSVD. OBJECTIVE This study explores the relationship between CRFs, BBB integrity, and WMH burden. DESIGN, SETTING, AND PARTICIPANTS The study included 155 participants from the Biomarkers and Cognition Study, Singapore (BIOCIS). CRFs were assessed through blood tests for glucose and lipid profiles, and blood pressure measurements. WMH volumes were quantified using MRI. MEASUREMENTS BBB integrity was evaluated using a Transendothelial Electrical Resistance (TEER) assay with human brain microvascular endothelial cells (hBMEC) exposed to participant plasma. RESULTS Plasma from individuals with a higher WMH burden was associated with increased BBB disruption in hBMEC. Higher systolic and diastolic blood pressure, as well as body mass index, were correlated with greater BBB disruption. Regression analyses revealed that elevated blood glucose and lipid levels were linked to increased BBB disruption. Both periventricular and subcortical WMH burdens were associated with increased BBB disruption. CONCLUSION This study highlights a relationship between CRFs, BBB disruption, and WMH burden, suggesting that CRFs may impair BBB integrity and contribute to WMH and cognitive decline in cSVD.
Collapse
Affiliation(s)
- James Xiao Yuan Chen
- Dementia Research Centre (Singapore), Lee Kong Chian School of Medicine - Nanyang Technological University, Singapore
| | - Ashwati Vipin
- Dementia Research Centre (Singapore), Lee Kong Chian School of Medicine - Nanyang Technological University, Singapore
| | - Gurveen Kaur Sandhu
- Dementia Research Centre (Singapore), Lee Kong Chian School of Medicine - Nanyang Technological University, Singapore
| | - Yi Jin Leow
- Dementia Research Centre (Singapore), Lee Kong Chian School of Medicine - Nanyang Technological University, Singapore
| | - Fatin Zahra Zailan
- Dementia Research Centre (Singapore), Lee Kong Chian School of Medicine - Nanyang Technological University, Singapore
| | - Pricilia Tanoto
- Dementia Research Centre (Singapore), Lee Kong Chian School of Medicine - Nanyang Technological University, Singapore
| | - Ee Soo Lee
- Lee Kong Chian School of Medicine - Nanyang Technological University, Singapore; School of Pharmacy, University of Nottingham Malaysia, Selangor, Malaysia
| | - Khang Leng Lee
- Lee Kong Chian School of Medicine - Nanyang Technological University, Singapore
| | - Christine Cheung
- Lee Kong Chian School of Medicine - Nanyang Technological University, Singapore; Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Nagaendran Kandiah
- Dementia Research Centre (Singapore), Lee Kong Chian School of Medicine - Nanyang Technological University, Singapore.
| |
Collapse
|
6
|
Cai L, Tozer DJ, Markus HS. Cerebral Microbleeds and Their Association With Inflammation and Blood-Brain Barrier Leakage in Small Vessel Disease. Stroke 2025; 56:427-436. [PMID: 39744850 PMCID: PMC11771357 DOI: 10.1161/strokeaha.124.048974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/18/2024] [Accepted: 11/15/2024] [Indexed: 01/29/2025]
Abstract
BACKGROUND How cerebral microbleeds (CMBs) are formed, and how they cause tissue damage is not fully understood, but it has been suggested they are associated with inflammation, and they could also be related to increased blood-brain barrier (BBB) leakage. We investigated the relationship of CMBs with inflammation and BBB leakage in cerebral small vessel disease, and in particular, whether these 2 processes were increased in the vicinity of CMBs. METHODS In 54 patients with sporadic cerebral small vessel disease presenting with lacunar stroke, we simultaneously assessed microglial activation using the positron emission tomography ligand [11C]PK11195 and BBB leakage using dynamic contrast enhanced magnetic resonance imaging, on a positron emission tomography-magnetic resonance imaging system. To assess local inflammation and BBB leakage, 3 one-voxel concentric shells were generated around each CMB on susceptibility-weighted imaging and resampled to positron emission tomography and T1 mapping images, respectively. In these 3 shells, we calculated the mean of PK11195 nondisplaceable binding potential (BPND) as a marker of microglial activation, as well as the mean influx rate as a marker of BBB leakage. In addition, 93 blood biomarkers related to cardiovascular disease, inflammation, and endothelial activation were measured to quantify systemic inflammation. RESULTS No significant associations were found between the number of CMBs and the measures for microglial activation (β=2.6×10-5, P=0.050) and BBB leakage (β=-0.0001, P=0.400) in the white matter. There was no difference in measures of microglial activation (P=0.403) or BBB leakage (P=0.423) across the 3 shells surrounding the CMBs. Furthermore, after correcting for multiple comparisons, no associations were observed between systemic inflammation biomarkers and the number of CMBs. CONCLUSIONS We found no evidence that CMBs are associated with either microglial activation assessed by [11]CPK11195 positron emission tomography or BBB leakage assessed by dynamic contrast enhanced magnetic resonance imaging, either globally or locally, in sporadic cerebral small vessel disease. There was also no association with markers of systemic inflammation.
Collapse
Affiliation(s)
- Lupei Cai
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, United Kingdom
| | - Daniel J. Tozer
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, United Kingdom
| | - Hugh S. Markus
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, United Kingdom
| |
Collapse
|
7
|
Zhilan T, Zengyu Z, Pengpeng J, Hualan Y, Chao L, Yan X, Zimin G, Shuangxing H, Weiwei L. Salidroside promotes pro-angiogenesis and repair of blood brain barrier via Notch/ITGB1 signal path in CSVD Model. J Adv Res 2025; 68:429-444. [PMID: 38417575 PMCID: PMC11785571 DOI: 10.1016/j.jare.2024.02.019] [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: 10/26/2023] [Revised: 12/05/2023] [Accepted: 02/25/2024] [Indexed: 03/01/2024] Open
Abstract
INTRODUCTION Salidroside (SAL), extracted from Rhodiola rosea, has been widely used in coronary heart disease and myocardial ischemia for decades. Previous studies have demonstrated that SAL could reduce arteriosclerosis, and thus combat ischemic brain damage. However, the in-depth function of the salidroside in Cerebral Small Vascular Disease (CSVD) has not been discovered, and related molecular mechanism is still unclear. OBJECTIVES The present study aims to explore the effects of salidroside in angiogenesis as well as repair of blood brain barrier (BBB) and its possible mechanisms. METHODS We established a rat model of SHR via 2-vessel gradual occlusion (SHR-2VGO) to mimic the CSVD. Subsequently, the MRI, pathomorphism, as well as Morriss water maze test were conducted to determine CSVD-related indicators. 8 weeks post-surgery, animals were randomly administered SAL, DAPT, ATN161 or saline.The aim was to explore the protective effects of SAL in CSVD as well as its possible mechanism. RESULTS Here we found that SAL could attenuate cerebral hypoperfusion-induced BBB disruption, promote the pro-angiogenesis through enhancing the cell budding. Further investigations demonstrated that SAL could significantly increase the expression of Notch1, Hes1, Hes5, and ITGB1. In addition, we confirmed that SAL could activate Notch signal path, and then up-regulate ITGB1 to promote pro-angiogenesis and thus protect BBB from disruption. CONCLUSION The aforementioned findings demonstrated that SAL could protect BBB integrity through Notch-ITGB1 signaling path in CSVD, which indicated that SAL could be a potential medicine candidate for CSVD treatment.
Collapse
Affiliation(s)
- Tu Zhilan
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Zhang Zengyu
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China; Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jin Pengpeng
- Department of Chronic Disease Management, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Yang Hualan
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Li Chao
- Vasculocardiology Department, Change County Hospital of Traditional Chinese Medicine, Shandong Province 261300, China
| | - Xi Yan
- Department of Radiology, Shanghai TCM-Integrated Hospital, 200082 Shanghai, China
| | - Guo Zimin
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Hou Shuangxing
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China.
| | - Li Weiwei
- Institute of Pediatrics, Children's Hospital of Fudan University, Fudan University, Shanghai 201102, China.
| |
Collapse
|
8
|
Jansen JFA. Editorial for "White Matter Hyperintensity is Associated with Malignant Cerebral Edema in Ischemic Stroke Treated with Thrombectomy". J Magn Reson Imaging 2025; 61:450-451. [PMID: 38751098 DOI: 10.1002/jmri.29421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 12/15/2024] Open
Affiliation(s)
- Jacobus F A Jansen
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- Mental Health and Neuroscience Research Institute, Maastricht University, Maastricht, The Netherlands
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
9
|
Tu Z, Xi Y, Zhang Y, Jin P, Yang H, Li C, Zhang Z, Wang H, Hou S. Assessment of blood-brain barrier injury in hypertensive CSVD by 11.7TMR T1mapping and microvascular pathologic changes. Metab Brain Dis 2024; 40:66. [PMID: 39680228 DOI: 10.1007/s11011-024-01483-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 11/13/2024] [Indexed: 12/17/2024]
Abstract
We used spontaneously hypertensive rats (SHR) as a hypertensive cerebral small vessel disease (CSVD) model to quantify blood-brain barrier (BBB) disruption by 11.7TMR T1mapping and to investigate white matter lesions and microangiopathy in CSVD. Male SHR were used as a hypertensive CSVD animal model and normotensive Wistar-Kyoto rats (WKY) were used as a control model. After 18 weeks, the rats did the Morris water maze test were evaluated, blood-brain barrier (BBB) integrity were evaluated by using Bruker 11.7T MR T1 mapping. ITK-SNAP software was used to measure hippocampal volume. Then, pathological analysis was carried out on rats, myelin integrity, vascular permeability and microvessel density were assessed by immunohistochemistry. Our data showed that hypertensive CSVD model exhibited decreased memory function, BBB leakage could be detected differently in different brain regions, and T1 values of the hippocampus showed the greatest drop than other areas. Furthermore, the pathological changes in small vessels were more extensive, average optical density of myelin basic protein (MBP) in the white matter of SHR group was significantly reduced, moreover, VEGFR2 immunoreactivity scores (IRS) and CD34-assessed MVD in SHR group were significantly higher than WKY group. We find different parts of the brain tissues have different degrees of BBB leakage, hippocampal atrophy and hippocampal volume were decreased in hypertensive CSVD by using T1 Mapping. Loss of myelin integrity, vascular permeability increased and microangiopathy may contribute to hypertensive-related BBB functional deficits in CSVD model.
Collapse
Affiliation(s)
- Zhilan Tu
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai, 201399, China
| | - Yan Xi
- Department of Radiology, Shanghai TCM-Integrated Hospital, 230 Baoding Road, Shanghai, 200000, China
| | - Yuwen Zhang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Pengpen Jin
- Clinical Center for Intelligent Rehabilitation Research, Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, 201613, China
| | - Hualan Yang
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai, 201399, China
| | - Chao Li
- School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei, 437100, China
| | - Zengyu Zhang
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong, Shanghai, 201399, China
| | - He Wang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Human Phenome Institute, Fudan University, Shanghai, China.
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, 220 Handan Road, 200082, Shanghai, China.
| | - Shuangxing Hou
- Department of Neurology, Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, 201613, Shanghai, China.
| |
Collapse
|
10
|
Alotaibi FF, Mohamed G, Bakry SS, Alqahtani M, BinAmir H, AlKawi A, Alreshaid AA, AlZawahmaha M, Alhazzani A, Shuaib A, Al-Ajlan FS. Association of white matter hyperintensities with lipoprotein (a) levels: insights from a cohort study. Front Neurol 2024; 15:1476005. [PMID: 39711788 PMCID: PMC11663010 DOI: 10.3389/fneur.2024.1476005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 11/25/2024] [Indexed: 12/24/2024] Open
Abstract
Background Little is known about the relationship between lipoprotein (a) [Lp(a)] and cerebral white matter hyperintensities (WMH). The aim of the study was to examine if elevated Lp(a) levels are associated with higher burden of WMH. Methods We retrospectively investigated associations between Lp(a) and the burden of WMH among patients with confirmed diagnosis of acute ischemic stroke or transient ischemic attacks. WMH burden was assessed using 3-Tesla brain MRI and graded according to the Fazekas score. Multivariable models were generated to determine the contribution of Lp(a) to the presence and extent of WMH. Results One hundred and fifty-three patients were included (mean age, 45.9 years; 35.9% women). When the study population was stratified by Lp(a) level into three categories, low (<75 nmol/L), moderate (75 to <125 nmol/L), and high (≥125 nmol/L), the distribution of the three groups was 60.8, 15.0 and 24.2%, respectively. High Lp(a) Level was associated with higher burden of both periventricular WMH and deep WMH compared to the lower level (odds ratio [OR], 4.4; 95% confidence interval [CI], 1.60-12.07; p = 0.004; and OR, 5.6; CI, 1.69-14.7; p = 0.001, respectively). Conclusion We show in this cohort of patients that a higher burden of WMH was observed in patients with higher level of Lp(a). Further studies are needed to confirm this observation and assess whether lowering Lp(a) level may be a potential therapeutic target for mitigating the development of WMH.
Collapse
Affiliation(s)
- Fawaz F. Alotaibi
- Neuroscience Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Gamal Mohamed
- Department of Biostatistics, Epidemiology, and Scientific Computing, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Sawsan S. Bakry
- Neuroscience Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Mohammed Alqahtani
- Neuroscience Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Hussain BinAmir
- Neuroscience Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Ammar AlKawi
- Neuroscience Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
- Neuroscience Center, King Faisal Specialist Hospital, Alfaisal University, Riyadh, Saudi Arabia
| | - Abdulrahman A. Alreshaid
- Neuroscience Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
- Neuroscience Center, King Faisal Specialist Hospital, Alfaisal University, Riyadh, Saudi Arabia
| | - Mohamed AlZawahmaha
- Neuroscience Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Adel Alhazzani
- Neuroscience Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Ashfaq Shuaib
- Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Fahad S. Al-Ajlan
- Neuroscience Center, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
- Neuroscience Center, King Faisal Specialist Hospital, Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
11
|
Cui M, Jin Z, Wang Y, Jiang J, Peng S, Wei Q, Zhang S, Tuo Q, Xie J, Leng H, Wang H, Zhao Y, Lei P, Xu J, Wang K, Zhang J, Jiang Y, Ding D, Xie F, Yu J, Dong Q. Imaging, biomarkers, and vascular cognitive impairment in China: Rationale and design for the VICA study. Alzheimers Dement 2024; 20:8898-8909. [PMID: 39535340 DOI: 10.1002/alz.14352] [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: 06/28/2024] [Revised: 09/26/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION Vascular cognitive impairment (VCI) is highly heterogeneous, with unclear pathogenesis. Individuals with vascular risk factors (VRF), cerebral small vessel disease (CSVD), and stroke are all at risk of developing VCI. To address the growing challenges posed by VCI, the "Vascular, Imaging and Cognition Association of China" (VICA) was established. METHODS VICA aims to recruit 10,000 participants, including 2000 with VRF, 3000 with CSVD, and 5000 stroke patients, to form a nationwide multicenter cohort. The study integrates clinical, neuroimaging, and multi-omics data to better understand VCI heterogeneity, improve disease prediction, and ensure timely diagnosis. RESULTS VICA has screened 2045 eligible VRF participants from six communities in Wuhan, Shanghai, and Taizhou, along with 602 CSVD and 1269 stroke patients from 135 hospitals nationwide. Baseline enrollment and follow-up work are still ongoing. DISCUSSION Establishing a high-quality longitudinal cohort is crucial for understanding VCI pathogenesis and developing novel markers for early screening and diagnosis. HIGHLIGHTS Establish a large-scale prospective longitudinal cohort comprising 10,000 participants, focusing on the high-risk population of vascular cognitive impairment (VCI) in China. Establish a nationwide three-tier medical network, make full use of resources, and achieve extensive enrollment of patients with cerebral small vessel disease and stroke patients. Utilize multimodal imaging and biomarkers to lay the foundation for constructing more-precise risk models. Introduce eye movement and gait analysis as new methods for assessing cognitive function. Use positron emission tomography to further investigate the interaction between vascular factors and neurodegeneration.
Collapse
Affiliation(s)
- Mei Cui
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- MOE Frontiers Center for Brain Science, National Center for Neurological Disorders, Shanghai, China
| | - Zishuo Jin
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yingzhe Wang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiwei Jiang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Sisi Peng
- Department of Neuropsychology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qiang Wei
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Shuting Zhang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qingzhang Tuo
- Department of Geriatrics and State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Junchao Xie
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haixia Leng
- Division of Neuropsychiatry and Psychosomatics, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Hongxing Wang
- Division of Neuropsychiatry and Psychosomatics, Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yanxin Zhao
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Peng Lei
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jun Xu
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Kai Wang
- Department of Neurology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Junjian Zhang
- Department of Neurology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yanfeng Jiang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, China
| | - Ding Ding
- Institute of Neurology, National Center for Neurological Disorders, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Fang Xie
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Jintai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- MOE Frontiers Center for Brain Science, National Center for Neurological Disorders, Shanghai, China
| |
Collapse
|
12
|
Hannawi Y. Cerebral Small Vessel Disease: a Review of the Pathophysiological Mechanisms. Transl Stroke Res 2024; 15:1050-1069. [PMID: 37864643 DOI: 10.1007/s12975-023-01195-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/02/2023] [Accepted: 09/18/2023] [Indexed: 10/23/2023]
Abstract
Cerebral small vessel disease (cSVD) refers to the age-dependent pathological processes involving the brain small vessels and leading to vascular cognitive impairment, intracerebral hemorrhage, and acute lacunar ischemic stroke. Despite the significant public health burden of cSVD, disease-specific therapeutics remain unavailable due to the incomplete understanding of the underlying pathophysiological mechanisms. Recent advances in neuroimaging acquisition and processing capabilities as well as findings from cSVD animal models have revealed critical roles of several age-dependent processes in cSVD pathogenesis including arterial stiffness, vascular oxidative stress, low-grade systemic inflammation, gut dysbiosis, and increased salt intake. These factors interact to cause a state of endothelial cell dysfunction impairing cerebral blood flow regulation and breaking the blood brain barrier. Neuroinflammation follows resulting in neuronal injury and cSVD clinical manifestations. Impairment of the cerebral waste clearance through the glymphatic system is another potential process that has been recently highlighted contributing to the cognitive decline. This review details these mechanisms and attempts to explain their complex interactions. In addition, the relevant knowledge gaps in cSVD mechanistic understanding are identified and a systematic approach to future translational and early phase clinical research is proposed in order to reveal new cSVD mechanisms and develop disease-specific therapeutics.
Collapse
Affiliation(s)
- Yousef Hannawi
- Division of Cerebrovascular Diseases and Neurocritical Care, Department of Neurology, The Ohio State University, 333 West 10th Ave, Graves Hall 3172C, Columbus, OH, 43210, USA.
| |
Collapse
|
13
|
Chen JL, Wang R, Ma PQ, Wang YM, Tang QQ. Association between intercellular adhesion molecule-1 to depression and blood-brain barrier penetration in cerebellar vascular disease. World J Psychiatry 2024; 14:1661-1670. [PMID: 39564172 PMCID: PMC11572681 DOI: 10.5498/wjp.v14.i11.1661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/10/2024] [Accepted: 09/18/2024] [Indexed: 11/07/2024] Open
Abstract
BACKGROUND Cerebral small vessel disease (CSVD) is a prevalent cerebrovascular disease in clinical practice that is often associated with macrovascular disease. A clear understanding of the underlying causes of CSVD remains elusive. AIM To explore the association between intercellular adhesion molecule-1 (ICAM-1) and blood-brain barrier (BBB) penetration in CSVD. METHODS This study included patients admitted to Fuyang People's Hospital and Fuyang Community (Anhui, China) between December 2021 and March 2022. The study population comprised 142 patients, including 80 in the CSVD group and 62 in the control group. Depression was present in 53 out of 80 patients with CSVD. Multisequence magnetic resonance imaging (MRI) and dynamic contrast-enhanced MRI were applied in patients to determine the brain volume, cortical thickness, and cortical area of each brain region. Moreover, neuropsychological tests including the Hamilton depression scale, mini-mental state examination, and Montreal cognitive assessment basic scores were performed. RESULTS The multivariable analysis showed that age [P = 0.011; odds ratio (OR) = 0.930, 95% confidence interval (CI): 0.880-0.983] and ICAM-1 levels (P = 0.023; OR = 1.007, 95%CI: 1.001-1.013) were associated with CSVD. Two regions of interest (ROIs; ROI3 and ROI4) in the white matter showed significant (both P < 0.001; 95%CI: 0.419-0.837 and 0.366-0.878) differences between the two groups, whereas only ROI1 in the gray matter showed significant difference (P = 0.046; 95%CI: 0.007-0.680) between the two groups. ICAM-1 was significantly correlated (all P < 0.05) with cortical thickness in multiple brain regions in the CSVD group. CONCLUSION This study revealed that ICAM-1 levels were independently associated with CSVD. ICAM-1 may be associated with cortical thickness in the brain, predominantly in the white matter, and a significant increase in BBB permeability, proposing the involvement of ICAM-1 in BBB destruction.
Collapse
Affiliation(s)
- Ju-Luo Chen
- Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui Province, China
- Department of Neurology, Fuyang People’s Hospital, Fuyang 236000, Anhui Province, China
| | - Rui Wang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui Province, China
| | - Pei-Qi Ma
- Department of Neurology, Fuyang People’s Hospital, Fuyang 236000, Anhui Province, China
| | - You-Meng Wang
- Department of Neurology, Fuyang People’s Hospital, Fuyang 236000, Anhui Province, China
| | - Qi-Qiang Tang
- Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, Anhui Province, China
| |
Collapse
|
14
|
Yan X, Zhang Y, He R, Chen X, Lin M. A bibliometric analysis of cerebral small vessel disease. Front Aging Neurosci 2024; 16:1400844. [PMID: 39435188 PMCID: PMC11492496 DOI: 10.3389/fnagi.2024.1400844] [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: 03/14/2024] [Accepted: 09/16/2024] [Indexed: 10/23/2024] Open
Abstract
Background Cerebral small vessel disease (CSVD) is a significant contributor to both stroke and dementia. While numerous studies on CSVD have been published, herein, we have conducted a bibliometric examination of the literature on CSVD, revealing its hot spots and emerging patterns. Methods We used the Web of Science Core Collection as our primary database and conducted a literature search from January 2008 to January 2023. CiteSpace, VOSviewer, online bibliometric platform, and R-bibliometrix were employed to conduct bibliometric analysis and network visualization, including the number of publications, countries, institutions, journals, citations, authors, references, and keywords. Results A total of 4891 publications on CSVD were published in 790 journals by 19,066 authors at 3,862 institutions from 84 countries. The United States produced the most written works and had a significant impact in this field of study. The University of Edinburgh had the highest publication count overall. The journal with the most publications and co-citations was Stroke. Wardlaw, Joanna was the most prolific author and commonly cited in the field. The current areas of research interest revolved around "MRI segmentation" and "Enlarged perivascular spaces in the basal ganglia." Conclusion We conducted a bibliometric analysis to examine the advancements, focal points, and cutting-edge areas in the field of CSVD to reveal potential future research opportunities. Research on CSVD is currently rapidly advancing, with a consistent rise in publications on the topic since 2008. At the same time, we identified leading countries, institutions, and leading scholars in the field and analyzed journals and representative literature. Keyword co-occurrence analysis and burst graph emergence detection identified MRI segmentation and Basal ganglia enlarged perivascular spaces as the most recent areas of research interest.
Collapse
Affiliation(s)
- Xiaoxiao Yan
- Department of Neurology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yongyin Zhang
- Department of Neurology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ruqian He
- Department of Neurology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiachan Chen
- Department of Neurology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Mian Lin
- Department of Orthopedics, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
15
|
Swiatek VM, Schreiber S, Amini A, Hasan D, Rashidi A, Stein KP, Neyazi B, Sandalcioglu IE. Intracranial Aneurysms and Cerebral Small Vessel Disease: Is There an Association between Large- and Small-Artery Diseases? J Clin Med 2024; 13:5864. [PMID: 39407924 PMCID: PMC11476928 DOI: 10.3390/jcm13195864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Background/Objectives: Intracranial aneurysms (IAs) may be connected to interactions between large and small intracranial vessels. We aimed to investigate the association between IAs and cerebral small-vessel disease (CSVD) and assess CSVD impact on IA patient management. Methods: This retrospective study analyzed clinical data and MRI features of CSVD in 192 subarachnoid hemorrhage (SAH) patients: 136 with incidental IA, 147 with severe CSVD without SAH/IA, and 50 controls without SAH, IA, or severe CSVD. MRI assessments followed the Standards for Reporting Vascular Changes on Neuroimaging (STRIVE), with a total burden of small-vessel disease (TBSVD) score calculated. Statistical analyses included forward selection and binary logistic regression. Results: TBSVD differed significantly across groups (p < 0.001), except between SAH and IA groups (p = 0.8). Controls had the lowest TBSVD (1.00; 1.22 ± 0.996), followed by SAH (2.00; 2.08 ± 1.013) and IA groups (2.00; 2.04 ± 1.141), with the highest in the CSVD group (1.00; 1.22 ± 0.996). White-matter hyperintensity (WMH) patterns varied with IA rupture status (p = 0.044); type A was prevalent in SAH patients and type D in the IA group. Incorporating MRI CSVD features and TBSVD into risk assessments did not enhance IA prediction or outcome models. Conclusions: IA patients exhibit a higher CSVD burden than controls, suggesting a link between small and large intracranial vessels. WMH patterns distinguish between ruptured and unruptured IA patients, offering potential markers for IA rupture risk assessment and signaling a paradigm shift in understanding IAs and CSVD.
Collapse
Affiliation(s)
- Vanessa M. Swiatek
- Department of Neurosurgery, Otto-von-Guericke University, 39120 Magdeburg, Germany; (V.M.S.); (A.A.); (A.R.); (K.-P.S.); (I.E.S.)
| | - Stefanie Schreiber
- Department of Neurology, Otto-von-Guericke University, 39120 Magdeburg, Germany;
- German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
- Center for Behavioral Brain Sciences, Otto-von-Guericke University, 39120 Magdeburg, Germany
| | - Amir Amini
- Department of Neurosurgery, Otto-von-Guericke University, 39120 Magdeburg, Germany; (V.M.S.); (A.A.); (A.R.); (K.-P.S.); (I.E.S.)
| | - David Hasan
- Department of Neurosurgery, Duke University, Durham, NC 27707, USA;
| | - Ali Rashidi
- Department of Neurosurgery, Otto-von-Guericke University, 39120 Magdeburg, Germany; (V.M.S.); (A.A.); (A.R.); (K.-P.S.); (I.E.S.)
| | - Klaus-Peter Stein
- Department of Neurosurgery, Otto-von-Guericke University, 39120 Magdeburg, Germany; (V.M.S.); (A.A.); (A.R.); (K.-P.S.); (I.E.S.)
| | - Belal Neyazi
- Department of Neurosurgery, Otto-von-Guericke University, 39120 Magdeburg, Germany; (V.M.S.); (A.A.); (A.R.); (K.-P.S.); (I.E.S.)
| | - I. Erol Sandalcioglu
- Department of Neurosurgery, Otto-von-Guericke University, 39120 Magdeburg, Germany; (V.M.S.); (A.A.); (A.R.); (K.-P.S.); (I.E.S.)
| |
Collapse
|
16
|
van Dinther M, Voorter PHM, Zhang E, van Kuijk SMJ, Jansen JFA, van Oostenbrugge RJ, Backes WH, Staals J. The neurovascular unit and its correlation with cognitive performance in patients with cerebral small vessel disease: a canonical correlation analysis approach. GeroScience 2024; 46:5061-5073. [PMID: 38888875 PMCID: PMC11335703 DOI: 10.1007/s11357-024-01235-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 05/31/2024] [Indexed: 06/20/2024] Open
Abstract
Growing evidence indicates an important role of neurovascular unit (NVU) dysfunction in the pathophysiology of cerebral small vessel disease (cSVD). Individually measurable functions of the NVU have been correlated with cognitive function, but a combined analysis is lacking. We aimed to perform a unified analysis of NVU function and its relation with cognitive performance. The relationship between NVU function in the white matter and cognitive performance (both latent variables composed of multiple measurable variables) was investigated in 73 patients with cSVD (mean age 70 ± 10 years, 41% women) using canonical correlation analysis. MRI-based NVU function measures included (1) the intravoxel incoherent motion derived perfusion volume fraction (f) and microvascular diffusivity (D*), reflecting cerebral microvascular flow; (2) the IVIM derived intermediate volume fraction (fint), indicative of the perivascular clearance system; and (3) the dynamic contrast-enhanced MRI derived blood-brain barrier (BBB) leakage rate (Ki) and leakage volume fraction (VL), reflecting BBB integrity. Cognitive performance was composed of 13 cognitive test scores. Canonical correlation analysis revealed a strong correlation between the latent variables NVU function and cognitive performance (r 0.73; p = 0.02). For the NVU, the dominating variables were D*, fint, and Ki. Cognitive performance was driven by multiple cognitive tests comprising different cognitive domains. The functionality of the NVU is correlated with cognitive performance in cSVD. Instead of focusing on individual pathophysiological mechanisms, future studies should target NVU dysfunction as a whole to acquire a coherent understanding of the complex disease mechanisms that occur in the NVU in cSVD.Trial registration: NTR3786 (Dutch Trial Register).
Collapse
Affiliation(s)
- Maud van Dinther
- Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands.
- CARIM-School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands.
| | - Paulien H M Voorter
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- MHeNs-School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Eleana Zhang
- Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Sander M J van Kuijk
- Department of Epidemiology and Medical Technology Assessment (KEMTA), Maastricht University, Maastricht, the Netherlands
| | - Jacobus F A Jansen
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- MHeNs-School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Robert J van Oostenbrugge
- Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands
- CARIM-School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
- MHeNs-School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Walter H Backes
- CARIM-School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- MHeNs-School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Julie Staals
- Department of Neurology, Maastricht University Medical Center, Maastricht, The Netherlands
- CARIM-School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
17
|
Dörner M, Tyndall A, Hainc N, von Känel R, Neumann K, Euler S, Schreiber F, Arndt P, Fuchs E, Garz C, Glanz W, Butryn M, Schulze JB, Schiebler SLF, John AC, Hildebrand A, Hofmann AB, Machetanz L, Kirchebner J, Tacik P, Grimm A, Jansen R, Pawlitzki M, Henneicke S, Bernal J, Perosa V, Düzel E, Meuth SG, Vielhaber S, Mattern H, Schreiber S. Neuropsychiatric symptoms and lifelong mental activities in cerebral amyloid angiopathy - a cross-sectional study. Alzheimers Res Ther 2024; 16:196. [PMID: 39232823 PMCID: PMC11375846 DOI: 10.1186/s13195-024-01519-3] [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: 04/23/2024] [Accepted: 06/25/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND While several studies in cerebral amyloid angiopathy (CAA) focus on cognitive function, data on neuropsychiatric symptoms (NPS) and lifelong mental activities in these patients are scarce. Since NPS are associated with functional impairment, faster cognitive decline and faster progression to death, replication studies in more diverse settings and samples are warranted. METHODS We prospectively recruited n = 69 CAA patients and n = 18 cognitively normal controls (NC). The number and severity of NPS were assessed using the Alzheimer's Disease (AD) Assessment Scale's (ADAS) noncognitive subscale. We applied different regression models exploring associations between NPS number or severity and group status (CAA vs. NC), CAA severity assessed with magnetic resonance imaging (MRI) or cognitive function (Mini-Mental State Examination (MMSE), ADAS cognitive subscale), adjusting for age, sex, years of education, arterial hypertension, AD pathology, and apolipoprotein E status. Mediation analyses were performed to test indirect effects of lifelong mental activities on CAA severity and NPS. RESULTS Patients with CAA had 4.86 times (95% CI 2.20-10.73) more NPS and 3.56 units (95% CI 1.94-5.19) higher expected NPS severity than NC. Higher total CAA severity on MRI predicted 1.14 times (95% CI 1.01.-1.27) more NPS and 0.57 units (95% CI 0.19-0.95) higher expected NPS severity. More severe white matter hyperintensities were associated with 1.21 times more NPS (95% CI 1.05-1.39) and 0.63 units (95% CI 0.19-1.08) more severe NPS. NPS number (MMSE mean difference - 1.15, 95% CI -1.67 to -0.63; ADAS cognitive mean difference 1.91, 95% CI 1.26-2.56) and severity (MMSE - 0.55, 95% CI -0.80 to -0.30; ADAS cognitive mean difference 0.89, 95% CI 0.57-1.21) predicted lower cognitive function. Greater lifelong mental activities partially mediated the relationship between CAA severity and NPS (indirect effect 0.05, 95% CI 0.0007-0.13), and greater lifelong mental activities led to less pronounced CAA severity and thus to less NPS (indirect effect - 0.08, 95% CI -0.22 to -0.002). DISCUSSION This study suggests that NPS are common in CAA, and that this relationship may be driven by CAA severity. Furthermore, NPS seem to be tied to lower cognitive function. However, lifelong mental activities might mitigate the impact of NPS in CAA.
Collapse
Affiliation(s)
- Marc Dörner
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, 39120, Magdeburg, Germany.
- Department of Consultation-Liaison-Psychiatry and Psychosomatic Medicine, University Hospital Zurich, University of Zurich, Culmannstrasse 8, Zurich, 8091, Switzerland.
| | - Anthony Tyndall
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, 8091, Switzerland
| | - Nicolin Hainc
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, 8091, Switzerland
| | - Roland von Känel
- Department of Consultation-Liaison-Psychiatry and Psychosomatic Medicine, University Hospital Zurich, University of Zurich, Culmannstrasse 8, Zurich, 8091, Switzerland
| | - Katja Neumann
- Department of Neurology, Otto-von-Guericke University, 39120, Magdeburg, Germany
| | - Sebastian Euler
- Department of Consultation-Liaison-Psychiatry and Psychosomatic Medicine, University Hospital Zurich, University of Zurich, Culmannstrasse 8, Zurich, 8091, Switzerland
| | - Frank Schreiber
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, 39120, Magdeburg, Germany
- Department of Neurology, Otto-von-Guericke University, 39120, Magdeburg, Germany
| | - Philipp Arndt
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, 39120, Magdeburg, Germany
- Department of Neurology, Otto-von-Guericke University, 39120, Magdeburg, Germany
| | - Erelle Fuchs
- Department of Neuroradiology, Otto-von-Guericke University, 39120, Magdeburg, Germany
| | - Cornelia Garz
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, 39120, Magdeburg, Germany
- Department of Neurology, Otto-von-Guericke University, 39120, Magdeburg, Germany
| | - Wenzel Glanz
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, 39120, Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, 39120, Magdeburg, Germany
| | - Michaela Butryn
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, 39120, Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, 39120, Magdeburg, Germany
| | - Jan Ben Schulze
- Department of Consultation-Liaison-Psychiatry and Psychosomatic Medicine, University Hospital Zurich, University of Zurich, Culmannstrasse 8, Zurich, 8091, Switzerland
| | - Sarah Lavinia Florence Schiebler
- Department of Consultation-Liaison-Psychiatry and Psychosomatic Medicine, University Hospital Zurich, University of Zurich, Culmannstrasse 8, Zurich, 8091, Switzerland
| | - Anna-Charlotte John
- Department of Neurology, Otto-von-Guericke University, 39120, Magdeburg, Germany
| | - Annkatrin Hildebrand
- Department of Neurology, Otto-von-Guericke University, 39120, Magdeburg, Germany
| | - Andreas B Hofmann
- Department of Psychiatry, Psychotherapy, and Psychosomatics, University Hospital of Psychiatry Zurich, University of Zurich, Zurich, 8032, Switzerland
| | - Lena Machetanz
- Department of Forensic Psychiatry, University Hospital of Psychiatry Zurich, University of Zurich, Zurich, 8032, Switzerland
| | - Johannes Kirchebner
- Department of Forensic Psychiatry, University Hospital of Psychiatry Zurich, University of Zurich, Zurich, 8032, Switzerland
| | - Pawel Tacik
- Department of Parkinson's Disease, Sleep and Movement Disorders, University Hospital Bonn, 53127, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, 53127, Bonn, Germany
| | - Alexander Grimm
- Center for Neurology, Tuebingen University Hospital and Hertie-Institute for Clinical Brain Research, Eberhard Karls University, 72076, Tuebingen, Tuebingen, Germany
| | - Robin Jansen
- Department of Neurology, Heinrich Heine University, 40225, Düsseldorf, Germany
| | - Marc Pawlitzki
- Department of Neurology, Heinrich Heine University, 40225, Düsseldorf, Germany
| | - Solveig Henneicke
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, 39120, Magdeburg, Germany
- Department of Neurology, Otto-von-Guericke University, 39120, Magdeburg, Germany
| | - Jose Bernal
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, 39120, Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, 39120, Magdeburg, Germany
| | - Valentina Perosa
- J. Philip Kistler Stroke Research Center, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Emrah Düzel
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, 39120, Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, 39120, Magdeburg, Germany
| | - Sven G Meuth
- Department of Neurology, Heinrich Heine University, 40225, Düsseldorf, Germany
| | - Stefan Vielhaber
- Department of Neurology, Otto-von-Guericke University, 39120, Magdeburg, Germany
| | - Hendrik Mattern
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, 39120, Magdeburg, Germany
- Center for Behavioural Brain Sciences (CBBS), 39120, Magdeburg, Germany
- Biomedical Magnetic Resonance, Otto-von-Guericke University, 39120, Magdeburg, Germany
| | - Stefanie Schreiber
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, 39120, Magdeburg, Germany.
- Department of Neurology, Otto-von-Guericke University, 39120, Magdeburg, Germany.
- Center for Behavioural Brain Sciences (CBBS), 39120, Magdeburg, Germany.
| |
Collapse
|
18
|
Muir RT, Smith EE. The Spectrum of Cerebral Small Vessel Disease: Emerging Pathophysiologic Constructs and Management Strategies. Neurol Clin 2024; 42:663-688. [PMID: 38937035 DOI: 10.1016/j.ncl.2024.03.003] [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] [Indexed: 06/29/2024]
Abstract
Cerebral small vessel disease (CSVD) is a spectrum of disorders that affect small arterioles, venules, cortical and leptomeningeal vessels, perivascular spaces, and the integrity of neurovascular unit, blood brain barrier, and surrounding glia and neurons. CSVD is an important cause of lacunar ischemic stroke and sporadic hemorrhagic stroke, as well as dementia-which will constitute some of the most substantive population and public health challenges over the next century. This article provides an overview of updated pathophysiologic frameworks of CSVD; discusses common and underappreciated clinical and neuroimaging manifestations of CSVD; and reviews emerging genetic risk factors linked to sporadic CSVD.
Collapse
Affiliation(s)
- Ryan T Muir
- Calgary Stroke Program, Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta T2N 1N4, Canada; Department of Community Health Sciences, University of Calgary, Calgary, Alberta T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Eric E Smith
- Calgary Stroke Program, Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta T2N 1N4, Canada; Department of Community Health Sciences, University of Calgary, Calgary, Alberta T2N 1N4, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta T2N 1N4, Canada.
| |
Collapse
|
19
|
Dupré N, Drieu A, Joutel A. Pathophysiology of cerebral small vessel disease: a journey through recent discoveries. J Clin Invest 2024; 134:e172841. [PMID: 38747292 PMCID: PMC11093606 DOI: 10.1172/jci172841] [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] [Indexed: 05/19/2024] Open
Abstract
Cerebral small vessel disease (cSVD) encompasses a heterogeneous group of age-related small vessel pathologies that affect multiple regions. Disease manifestations range from lesions incidentally detected on neuroimaging (white matter hyperintensities, small deep infarcts, microbleeds, or enlarged perivascular spaces) to severe disability and cognitive impairment. cSVD accounts for approximately 25% of ischemic strokes and the vast majority of spontaneous intracerebral hemorrhage and is also the most important vascular contributor to dementia. Despite its high prevalence and potentially long therapeutic window, there are still no mechanism-based treatments. Here, we provide an overview of the recent advances in this field. We summarize recent data highlighting the remarkable continuum between monogenic and multifactorial cSVDs involving NOTCH3, HTRA1, and COL4A1/A2 genes. Taking a vessel-centric view, we discuss possible cause-and-effect relationships between risk factors, structural and functional vessel changes, and disease manifestations, underscoring some major knowledge gaps. Although endothelial dysfunction is rightly considered a central feature of cSVD, the contributions of smooth muscle cells, pericytes, and other perivascular cells warrant continued investigation.
Collapse
Affiliation(s)
- Nicolas Dupré
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Paris, France
| | - Antoine Drieu
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Paris, France
| | - Anne Joutel
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Paris, France
- GHU-Paris Psychiatrie et Neurosciences, Hôpital Sainte Anne, Paris, France
| |
Collapse
|
20
|
Reiländer A, Engel M, Nöth U, Deichmann R, Shrestha M, Wagner M, Gracien RM, Seiler A. Cortical microstructural involvement in cerebral small vessel disease. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2024; 6:100218. [PMID: 38510580 PMCID: PMC10951897 DOI: 10.1016/j.cccb.2024.100218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Background In cerebral small vessel disease (CSVD), cortical atrophy occurs at a later stage compared to microstructural abnormalities and therefore cannot be used for monitoring short-term disease progression. We aimed to investigate whether cortical diffusion tensor imaging (DTI) and quantitative (q) magnetic resonance imaging (MRI) are able to detect early microstructural involvement of the cerebral cortex in CSVD. Materials and Methods 33 CSVD patients without significant cortical or whole-brain atrophy and 16 healthy control subjects were included and underwent structural MRI, DTI and high-resolution qMRI with T2, T2* and T2' mapping at 3 T as well as comprehensive cognitive assessment. After tissue segmentation and reconstruction of the cortical boundaries with the Freesurfer software, DTI and qMRI parameters were saved as surface datasets and averaged across all vertices. Results Cortical diffusivity and quantitative T2 values were significantly increased in patients compared to controls (p < 0.05). T2 values correlated significantly positively with white matter hyperintensity (WMH) volume (p < 0.01). Both cortical diffusivity and T2 showed significant negative associations with axonal damage to the white matter fiber tracts (p < 0.05). Conclusions Cortical diffusivity and quantitative T2 mapping are suitable to detect microstructural involvement of the cerebral cortex in CSVD and represent promising imaging biomarkers for monitoring disease progression and effects of therapeutical interventions in clinical studies.
Collapse
Affiliation(s)
- Annemarie Reiländer
- Department of Neurology, Goethe University Hospital, Frankfurt, Germany
- Brain Imaging Center, Goethe University, Frankfurt, Germany
| | - Marlene Engel
- Department of Neurology, Goethe University Hospital, Frankfurt, Germany
| | - Ulrike Nöth
- Brain Imaging Center, Goethe University, Frankfurt, Germany
| | - Ralf Deichmann
- Brain Imaging Center, Goethe University, Frankfurt, Germany
| | - Manoj Shrestha
- Brain Imaging Center, Goethe University, Frankfurt, Germany
| | - Marlies Wagner
- Brain Imaging Center, Goethe University, Frankfurt, Germany
- Institute of Neuroradiology, Goethe University Hospital, Frankfurt, Germany
| | - René-Maxime Gracien
- Department of Neurology, Goethe University Hospital, Frankfurt, Germany
- Brain Imaging Center, Goethe University, Frankfurt, Germany
| | - Alexander Seiler
- Brain Imaging Center, Goethe University, Frankfurt, Germany
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel, Germany
- Neurovascular Center, University Hospital Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
21
|
Chen DH, Huang JR, Su SL, Chen Q, Wu BY. Therapeutic potential of mesenchymal stem cells for cerebral small vessel disease. Regen Ther 2024; 25:377-386. [PMID: 38414558 PMCID: PMC10899004 DOI: 10.1016/j.reth.2023.11.002] [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: 08/23/2023] [Revised: 10/12/2023] [Accepted: 11/05/2023] [Indexed: 02/29/2024] Open
Abstract
Cerebral small vessel disease (CSVD), as the most common, chronic and progressive vascular disease on the brain, is a serious neurological disease, whose pathogenesis remains unclear. The disease is a leading cause of stroke and vascular cognitive impairment and dementia, and contributes to about 20% of strokes, including 25% of ischemic strokes and 45% of dementias. Undoubtedly, the high incidence and poor prognosis of CSVD have brought a heavy economic and medical burden to society. The present treatment of CSVD focuses on the management of vascular risk factors. Although vascular risk factors may be important causes or accelerators of CSVD and should always be treated in accordance with best clinical practice, controlling risk factors alone could not curb the progression of CSVD brain injury. Therefore, developing safer and more effective treatment strategies for CSVD is urgently needed. Recently, mesenchymal stem cells (MSCs) therapy has become an emerging therapeutic modality for the treatment of central nervous system disease, given their paracrine properties and immunoregulatory. Herein, we discussed the therapeutic potential of MSCs for CSVD, aiming to enable clinicians and researchers to understand of recent progress and future directions in the field.
Collapse
Affiliation(s)
- Dong-Hua Chen
- Neurology Department, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Baiyun District, Guangzhou, 510515, China
| | - Jia-Rong Huang
- Neurology Department, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Baiyun District, Guangzhou, 510515, China
| | - Shuo-Lei Su
- Shaoguan University, No.288 University Road, Xinshaozhen Zhenjiang District, Shaoguan, 512005, China
| | - Qiong Chen
- Medical Research center of Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Baiyun District, Guangzhou, 510515, China
- Precision Medicine Center of Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Baiyun District, Guangzhou, 510515, China
| | - Bing-Yi Wu
- Medical Research center of Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Baiyun District, Guangzhou, 510515, China
- Precision Medicine Center of Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Baiyun District, Guangzhou, 510515, China
| |
Collapse
|
22
|
Voorter PHM, van Dinther M, Jansen WJ, Postma AA, Staals J, Jansen JFA, van Oostenbrugge RJ, van der Thiel MM, Backes WH. Blood-Brain Barrier Disruption and Perivascular Spaces in Small Vessel Disease and Neurodegenerative Diseases: A Review on MRI Methods and Insights. J Magn Reson Imaging 2024; 59:397-411. [PMID: 37658640 DOI: 10.1002/jmri.28989] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/17/2023] [Accepted: 08/17/2023] [Indexed: 09/03/2023] Open
Abstract
Perivascular spaces (PVS) and blood-brain barrier (BBB) disruption are two key features of cerebral small vessel disease (cSVD) and neurodegenerative diseases that have been linked to cognitive impairment and are involved in the cerebral waste clearance system. Magnetic resonance imaging (MRI) offers the possibility to study these pathophysiological processes noninvasively in vivo. This educational review provides an overview of the MRI techniques used to assess PVS functionality and BBB disruption. MRI-visible PVS can be scored on structural images by either (subjectively) counting or (automatically) delineating the PVS. We highlight emerging (diffusion) techniques to measure proxies of perivascular fluid and its movement, which may provide a more comprehensive understanding of the role of PVS in diseases. For the measurement of BBB disruption, we explain the most commonly used MRI technique, dynamic contrast-enhanced (DCE) MRI, as well as a more recently developed technique based on arterial spin labeling (ASL). DCE MRI and ASL are thought to measure complementary characteristics of the BBB. Furthermore, we describe clinical studies that have utilized these MRI techniques in cSVD and neurodegenerative diseases, particularly Alzheimer's disease (AD). These studies demonstrate the role of PVS and BBB dysfunction in these diseases and provide insight into the large overlap, but also into the differences between cSVD and AD. Overall, MRI techniques may provide valuable insights into the pathophysiological mechanisms underlying these diseases and have the potential to be used as markers for disease progression and treatment response. LEVEL OF EVIDENCE: 3 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Paulien H M Voorter
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Maud van Dinther
- School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands
- Department of Neurology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Willemijn J Jansen
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
- Department of Psychiatry and Neuropsychology, Alzheimer Center Limburg, Maastricht University, Maastricht, the Netherlands
| | - Alida A Postma
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Julie Staals
- School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands
- Department of Neurology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Jacobus F A Jansen
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Robert J van Oostenbrugge
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
- School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands
- Department of Neurology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Merel M van der Thiel
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
- Department of Psychiatry and Neuropsychology, Alzheimer Center Limburg, Maastricht University, Maastricht, the Netherlands
| | - Walter H Backes
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
- School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
23
|
Rowsthorn E, Pham W, Nazem-Zadeh MR, Law M, Pase MP, Harding IH. Imaging the neurovascular unit in health and neurodegeneration: a scoping review of interdependencies between MRI measures. Fluids Barriers CNS 2023; 20:97. [PMID: 38129925 PMCID: PMC10734164 DOI: 10.1186/s12987-023-00499-0] [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: 10/03/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023] Open
Abstract
The neurovascular unit (NVU) is a complex structure that facilitates nutrient delivery and metabolic waste clearance, forms the blood-brain barrier (BBB), and supports fluid homeostasis in the brain. The integrity of NVU subcomponents can be measured in vivo using magnetic resonance imaging (MRI), including quantification of enlarged perivascular spaces (ePVS), BBB permeability, cerebral perfusion and extracellular free water. The breakdown of NVU subparts is individually associated with aging, pathology, and cognition. However, how these subcomponents interact as a system, and how interdependencies are impacted by pathology remains unclear. This systematic scoping review identified 26 studies that investigated the inter-relationships between multiple subcomponents of the NVU in nonclinical and neurodegenerative populations using MRI. A further 112 studies investigated associations between the NVU and white matter hyperintensities (WMH). We identify two putative clusters of NVU interdependencies: a 'vascular' cluster comprising BBB permeability, perfusion and basal ganglia ePVS; and a 'fluid' cluster comprising ePVS, free water and WMH. Emerging evidence suggests that subcomponent coupling within these clusters may be differentially related to aging, neurovascular injury or neurodegenerative pathology.
Collapse
Affiliation(s)
- Ella Rowsthorn
- Department of Neuroscience, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC, 3004, Australia
- Turner Institute for Brain and Mental Health & School of Psychological Sciences, Monash University, 18 Innovation Walk, Clayton, VIC, 3168, Australia
| | - William Pham
- Department of Neuroscience, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Mohammad-Reza Nazem-Zadeh
- Department of Neuroscience, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Meng Law
- Department of Neuroscience, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC, 3004, Australia
- Department of Radiology, Alfred Health, 99 Commercial Road, Melbourne, VIC, 3004, Australia
- Department of Electrical and Computer Systems Engineering, Monash University, 14 Alliance Lane, Clayton, VIC, 3168, Australia
| | - Matthew P Pase
- Turner Institute for Brain and Mental Health & School of Psychological Sciences, Monash University, 18 Innovation Walk, Clayton, VIC, 3168, Australia
- Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA, 02115, USA
| | - Ian H Harding
- Department of Neuroscience, Central Clinical School, Monash University, 99 Commercial Road, Melbourne, VIC, 3004, Australia.
- Monash Biomedical Imaging, Monash University, 762-772 Blackburn Road, Clayton, VIC, 3168, Australia.
| |
Collapse
|
24
|
Hao Y, Zhou H, Pan C, Xie G, Hu J, Zhang B, Qian S, Yan S. Prediction factors and clinical significance of different types of hemorrhagic transformation after intravenous thrombolysis. Eur J Med Res 2023; 28:509. [PMID: 37951926 PMCID: PMC10638828 DOI: 10.1186/s40001-023-01503-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 11/03/2023] [Indexed: 11/14/2023] Open
Abstract
BACKGROUND AND PURPOSE Hemorrhagic transformation (HT) after intravenous thrombolysis (IVT) in acute ischemic stroke seriously affects the prognosis of patients. This study aimed to investigate the risk factors of different types of HT and their correlation with prognosis after IVT. METHODS Based on the CASE II registry, we included patients with acute ischemic stroke who received IVT within 4.5 h of onset. HT was further divided into hemorrhagic infarction (HI) and parenchymal hemorrhage (PH). Poor outcome was defined as a modified Rankin Scale (mRS) score of 3-6 at 3 months. Multivariate logistic regression analysis was used to determine the independent influencing factors of HT subtypes and clinical outcome. RESULTS Among 13108 included patients, 541 (4.1%) developed HI and 440 (3.4%) developed PH. In multivariate analysis, age (OR 1.038, 95% CI 1.028 to 1.049, p < 0.001), atrial fibrillation (OR 1.446, 95% CI 1.141 to 1.943, p = 0.002), baseline diastolic pressure (OR 1.012, 95% CI 1.004 to 1.020, p = 0.005), baseline NIHSS score (OR 1.060, 95% CI 1.049 to 1.071, p < 0.001) and onset to treatment time (OR 1.002, 95% CI 1.000 to 1.004, p = 0.020) independently predicted PH after IVT. In the patients with HT, PH (OR 3.611, 95% CI 2.540 to 5.134, p < 0.001) and remote hemorrhage (OR 1.579, 95% CI 1.115 to 2.235, p = 0.010) were independently related to poor outcome. CONCLUSIONS Different types of HT after IVT had different risk factors and clinical significance. The occurrence of PH and remote hemorrhage independently increased the risk of poor outcome.
Collapse
Affiliation(s)
- Yanan Hao
- Department of Neurology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Huan Zhou
- Department of Neurology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Chengzhen Pan
- Department of Neurology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Hangzhou, China
| | - Guomin Xie
- Department of Neurology, Lee Hui-Lee East Hospital, Ningbo, China
| | - Jin Hu
- Department of Neurology, The First Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Bing Zhang
- Department of Neurology, Huzhou Central Hospital, Huzhou, China
| | - Shuxia Qian
- Department of Neurology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China.
| | - Shenqiang Yan
- Department of Neurology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China.
| |
Collapse
|
25
|
Ding L, Hou B, Zang J, Su T, Feng F, Zhu Z, Peng B. Imaging of Angiogenesis in White Matter Hyperintensities. J Am Heart Assoc 2023; 12:e028569. [PMID: 37889177 PMCID: PMC10727415 DOI: 10.1161/jaha.122.028569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 09/19/2023] [Indexed: 10/28/2023]
Abstract
Background White matter hyperintensities (WMHs) are areas of increased signal intensity on T2-weighted magnetic resonance imaging (MRI). WMH penumbra may be a potential target for early intervention in WMHs. We explored the relationship between angiogenesis and WMH penumbra in patients with WMHs. Methods and Results Twenty-one patients with confluent WMHs of Fazekas grade ≥2 were included. All the participants underwent 68Ga-NOTA-PRGD2 positron emission tomography/magnetic resonance imaging. WMH penumbra was analyzed with masks created for the WMH and 7 normal-appearing white matter layers; each layer was dilated away from the WMH by 2 mm. Angiogenesis array and ELISA were used to detect the serum levels of angiogenic factors, inflammatory factors, HIF-1 alpha, and S100B. Fourteen patients with increased 68Ga-NOTA-PRGD2 maximum standardized uptake (>0.17) were classified into group 2. Seven patients with maximum standardized uptake ≤0.17 were classified as group 1. WMH volume and serum levels of integrin αvβ3, vascular endothelial growth factor receptor 22, and interleukin-1β tended to be higher in group 2 than in group 1. In group 2, 68Ga-NOTA-PRGD2 uptake was significantly increased at the border between the WMH and normal-appearing white matter than in WMHs (P=0.004). The structure penumbra, defined by fractional anisotropy, was wider in group 2 (8 mm) than in group 1 (2 mm). The cerebral blood flow penumbra was 12 mm in both groups. Angiogenesis showed a correlation with reduced cerebral blood flow and microstructure integrity. Conclusions Our study provides evidence that angiogenesis occurs in the WMH penumbra. Further studies are warranted to verify the effect of angiogenesis on WMH growth.
Collapse
Affiliation(s)
- Lingling Ding
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Bo Hou
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Jie Zang
- Department of Nuclear MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Tong Su
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Feng Feng
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Zhaohui Zhu
- Department of Nuclear MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Bin Peng
- Department of NeurologyPeking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
- Department of NeurologyState Key Laboratory of Complex Severe and Rare DiseasesBeijingChina
| |
Collapse
|
26
|
Moon Y, Jeon HJ, Han SH, Min-Young N, Kim HJ, Kwon KJ, Moon WJ, Kim SH. Blood-brain barrier breakdown is linked to tau pathology and neuronal injury in a differential manner according to amyloid deposition. J Cereb Blood Flow Metab 2023; 43:1813-1825. [PMID: 37283062 PMCID: PMC10676138 DOI: 10.1177/0271678x231180035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 04/05/2023] [Accepted: 04/18/2023] [Indexed: 06/08/2023]
Abstract
The blood-brain barrier (BBB) breakdown has been suggested as an early marker for Alzheimer's disease (AD); yet the relationship between BBB breakdown and AD-specific biomarkers based on the amyloid/tau/neurodegeneration framework is not clear. This study investigated the relationship between BBB permeability, AD-specific biomarkers, and cognition in patients with cognitive impairment. In this prospective study, we enrolled 62 participants with mild cognitive impairment or dementia between January 2019 and October 2020. All participants were assessed through cognitive tests, amyloid positron emission tomography (PET), dynamic contrast-enhanced magnetic resonance imaging (MRI) for BBB permeability (Ktrans), cerebrospinal fluid studies for Aβ42/40 ratio, phosphorylated-tau Thr181 protein (p-tau), total tau protein (t-tau), and structural MRI for neurodegeneration. In amyloid PET (+) group, higher cortical Ktrans was associated with lower Aβ40 (r = -0.529 p = 0.003), higher Aβ42/40 ratio (r = 0.533, p = 0.003), lower p-tau (r = -0.452, p = 0.014) and lower hippocampal volume (r = -0.438, p = 0.017). In contrast, cortical Ktrans was positively related to t-tau level. (r = 0.489, p = 0.004) in amyloid PET (-) group. Our results suggest that BBB permeability is related to AD-specific biomarkers, but the relationship can vary by the presence of Aβ plaque accumulation.
Collapse
Affiliation(s)
- Yeonsil Moon
- Department of Neurology, Konkuk University Medical Center, Seoul, Republic of Korea
- Research Institute of Medical Science, Konkuk University of Medicine, Seoul, Republic of Korea
| | - Hong Jun Jeon
- Research Institute of Medical Science, Konkuk University of Medicine, Seoul, Republic of Korea
- Department of Psychiatry, Konkuk University Medical Center, Seoul, Republic of Korea
| | - Seol-Heui Han
- Department of Neurology, Konkuk University Medical Center, Seoul, Republic of Korea
- Research Institute of Medical Science, Konkuk University of Medicine, Seoul, Republic of Korea
| | - Noh Min-Young
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Hee-Jin Kim
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Kyoung Ja Kwon
- Center for Geriatric Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Won-Jin Moon
- Research Institute of Medical Science, Konkuk University of Medicine, Seoul, Republic of Korea
- Department of Radiology, Konkuk University Medical Center, Seoul, Republic of Korea
| | - Seung Hyun Kim
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
27
|
Freeze WM, van Veluw SJ, Jansen WJ, Bennett DA, Jacobs HIL. Locus coeruleus pathology is associated with cerebral microangiopathy at autopsy. Alzheimers Dement 2023; 19:5023-5035. [PMID: 37095709 PMCID: PMC10593911 DOI: 10.1002/alz.13096] [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: 10/14/2022] [Revised: 03/10/2023] [Accepted: 03/27/2023] [Indexed: 04/26/2023]
Abstract
INTRODUCTION We investigated the link between locus coeruleus (LC) pathology and cerebral microangiopathy in two large neuropathology datasets. METHODS We included data from the National Alzheimer's Coordinating Center (NACC) database (n = 2197) and Religious Orders Study and Rush Memory and Aging Project (ROSMAP; n = 1637). Generalized estimating equations and logistic regression were used to examine associations between LC hypopigmentation and presence of cerebral amyloid angiopathy (CAA) or arteriolosclerosis, correcting for age at death, sex, cortical Alzheimer's disease (AD) pathology, ante mortem cognitive status, and presence of vascular and genetic risk factors. RESULTS LC hypopigmentation was associated with higher odds of overall CAA in the NACC dataset, leptomeningeal CAA in the ROSMAP dataset, and arteriolosclerosis in both datasets. DISCUSSION LC pathology is associated with cerebral microangiopathy, independent of cortical AD pathology. LC degeneration could potentially contribute to the pathways relating vascular pathology to AD. Future studies of the LC-norepinephrine system on cerebrovascular health are warranted. HIGHLIGHTS We associated locus coeruleus (LC) pathology and cerebral microangiopathy in two large autopsy datasets. LC hypopigmentation was consistently related to arteriolosclerosis in both datasets. LC hypopigmentation was related to cerebral amyloid angiopathy (CAA) presence in the National Alzheimer's Coordinating Center dataset. LC hypopigmentation was related to leptomeningeal CAA in the Religious Orders Study and Rush Memory and Aging Project dataset. LC degeneration may play a role in the pathways relating vascular pathology to Alzheimer's disease.
Collapse
Affiliation(s)
- WM Freeze
- Department of Radiology, Leiden University Medical Center, 2333 ZA, Leiden, the Netherlands
- Department of Psychiatry and Neuropsychology, Alzheimer Center Limburg, School for Mental Health and Neuroscience, Maastricht University, 6229 ET, Maastricht, the Netherlands
| | - SJ van Veluw
- Department of Radiology, Leiden University Medical Center, 2333 ZA, Leiden, the Netherlands
- Department of Neurology, J. Philip Kistler Stroke Research Center, MGH, Boston, MA 02114, USA
| | - WJ Jansen
- Department of Psychiatry and Neuropsychology, Alzheimer Center Limburg, School for Mental Health and Neuroscience, Maastricht University, 6229 ET, Maastricht, the Netherlands
- Banner Alzheimer’s Institute, Phoenix, AZ 85006, USA
| | - DA Bennett
- Department of Neurological Sciences, Rush Alzheimer’s Disease Center, Rush University Medical Center; Chicago, IL 60612, USA
| | - HIL Jacobs
- Department of Psychiatry and Neuropsychology, Alzheimer Center Limburg, School for Mental Health and Neuroscience, Maastricht University, 6229 ET, Maastricht, the Netherlands
- Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
28
|
Bennett J, van Dinther M, Voorter P, Backes W, Barnes J, Barkhof F, Captur G, Hughes AD, Sudre C, Treibel TA. Assessment of Microvascular Disease in Heart and Brain by MRI: Application in Heart Failure with Preserved Ejection Fraction and Cerebral Small Vessel Disease. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1596. [PMID: 37763715 PMCID: PMC10534635 DOI: 10.3390/medicina59091596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/18/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023]
Abstract
The objective of this review is to investigate the commonalities of microvascular (small vessel) disease in heart failure with preserved ejection fraction (HFpEF) and cerebral small vessel disease (CSVD). Furthermore, the review aims to evaluate the current magnetic resonance imaging (MRI) diagnostic techniques for both conditions. By comparing the two conditions, this review seeks to identify potential opportunities to improve the understanding of both HFpEF and CSVD.
Collapse
Affiliation(s)
- Jonathan Bennett
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, UK
- Department of Cardiology, Barts Heart Centre, London EC1A 7BE, UK
| | - Maud van Dinther
- Department of Neurology, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
- School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6211 LX Maastricht, The Netherlands
| | - Paulien Voorter
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
- School for Mental Health & Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Walter Backes
- School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6211 LX Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, 6229 HX Maastricht, The Netherlands
- School for Mental Health & Neuroscience, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200 MD Maastricht, The Netherlands
| | - Josephine Barnes
- Dementia Research Centre, UCL Queens Square Institute of Neurology, University College London, London WC1E 6BT, UK
| | - Frederick Barkhof
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, Vrije University, P.O. Box 7057, 1007 MB Amsterdam, The Netherlands
- Queen Square Institute of Neurology, University College London, London WC1E 6BT, UK
- Centre for Medical Image Computing, University College London, London WC1E 6BT, UK
| | - Gabriella Captur
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, UK
- Medical Research Council Unit for Lifelong Health and Ageing, Department of Population Science and Experimental Medicine, University College London, London WC1E 6BT, UK
- Centre for Inherited Heart Muscle Conditions, Cardiology Department, The Royal Free Hospital, London NW3 2QG, UK
| | - Alun D. Hughes
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, UK
- Medical Research Council Unit for Lifelong Health and Ageing, Department of Population Science and Experimental Medicine, University College London, London WC1E 6BT, UK
| | - Carole Sudre
- Dementia Research Centre, UCL Queens Square Institute of Neurology, University College London, London WC1E 6BT, UK
- Centre for Medical Image Computing, University College London, London WC1E 6BT, UK
- Medical Research Council Unit for Lifelong Health and Ageing, Department of Population Science and Experimental Medicine, University College London, London WC1E 6BT, UK
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London WC2R 2LS, UK
| | - Thomas A. Treibel
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, UK
- Department of Cardiology, Barts Heart Centre, London EC1A 7BE, UK
| |
Collapse
|
29
|
Zhang M, Tang J, Xia D, Xue Y, Ren X, Huang Q, Shi L, Tang W, Fu J. Evaluation of glymphatic-meningeal lymphatic system with intravenous gadolinium-based contrast-enhancement in cerebral small-vessel disease. Eur Radiol 2023; 33:6096-6106. [PMID: 37410111 DOI: 10.1007/s00330-023-09796-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 03/19/2023] [Accepted: 03/23/2023] [Indexed: 07/07/2023]
Abstract
OBJECTIVE To investigate the feasibility of using noninvasive neuroimaging methods in visualizing and evaluating the clearance of the glymphatic-meningeal lymphatic system (GMLS) in patients with arteriosclerotic cerebral small-vessel disease (CSVD) and controls. METHODS This observational study recruited patients with high-burden CSVD and controls (age 50-80 years). At multiple time points before and after intravenous administration of a contrast agent, three-dimensional (3D) brain volume T1-weighted imaging and 3D Cube T2-fluid attenuated inversion recovery imaging were performed to visualize and assess the clearance of the glymphatics and meningeal lymphatic vessels (mLVs). We measured the signal intensity ratio (SIR) of four regions of interest representing the glymphatics and mLVs at each time point. The clearance rate at 24 h (CR24h) and changes in the SIR from baseline to 24 h (∆SIR) were defined as the clearance function. The analysis of variance was used to evaluate the group differences after adjusting for hypertension. RESULTS A total of 20 CSVD patients and 15 controls were included. Cortical periarterial enhancement and the enhancement of enlarged perivascular spaces in the basal ganglia were respectively observed in 11 (55.00%) and 16 (80.00%) CSVD patients, but in none of controls. All CSVD patients and most of controls (80.00%) showed cortical perivenous enhancement. Para-sinus enhancement was observed in all participants. CSVD patients showed lower CR24h and higher ∆SIR of the glymphatics and mLVs (all p < 0.05). CONCLUSION The impaired drainage of the GMLS in patients with high-burden CSVD could be visually evaluated using noninvasive neuroimaging methods with intravenous gadolinium-based contrast-enhancement. CLINICAL RELEVANCE STATEMENT Dynamic intravenous contrast-enhanced MRI could visually evaluate the impaired drainage of the glymphatic-meningeal lymphatic system in patients with high-burden cerebral small-vessel disease and could help to explore a new therapeutic target. KEY POINTS • Signal intensity changes in regions representing the glymphatic-meningeal lymphatic system (GMLS) can reflect the drainage function based on contrast-enhanced 3D-FLAIR and 3D T1-weighted MRI. • Impaired drainage of the GMLS in patients with high-burden CSVD can be visually evaluated using dynamic intravenous contrast-enhanced MRI. • This direct, noninvasive technique could serve as a basis for further GMLS studies and could help to explore a new therapeutic target in CSVD patients.
Collapse
Affiliation(s)
- Miaoyi Zhang
- Department of Neurology, Huashan Hospital, Fudan University, No.12 Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Jie Tang
- Department of Neurology, Huashan Hospital, Fudan University, No.12 Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Ding Xia
- Department of Radiology, Huashan Hospital, Fudan University, No.12 Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Yang Xue
- Department of Neurology, Huashan Hospital, Fudan University, No.12 Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Xue Ren
- Department of Neurology, Huashan Hospital, Fudan University, No.12 Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Qi Huang
- Department of Neurology, Huashan Hospital, Fudan University, No.12 Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Langfeng Shi
- Department of Neurology, Huashan Hospital, Fudan University, No.12 Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Weijun Tang
- Department of Radiology, Huashan Hospital, Fudan University, No.12 Wulumuqi Zhong Road, Shanghai, 200040, China.
| | - Jianhui Fu
- Department of Neurology, Huashan Hospital, Fudan University, No.12 Wulumuqi Zhong Road, Shanghai, 200040, China.
| |
Collapse
|
30
|
Manukjan N, Majcher D, Leenders P, Caiment F, van Herwijnen M, Smeets HJ, Suidgeest E, van der Weerd L, Vanmierlo T, Jansen JFA, Backes WH, van Oostenbrugge RJ, Staals J, Fulton D, Ahmed Z, Blankesteijn WM, Foulquier S. Hypoxic oligodendrocyte precursor cell-derived VEGFA is associated with blood-brain barrier impairment. Acta Neuropathol Commun 2023; 11:128. [PMID: 37550790 PMCID: PMC10405482 DOI: 10.1186/s40478-023-01627-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/23/2023] [Indexed: 08/09/2023] Open
Abstract
Cerebral small vessel disease is characterised by decreased cerebral blood flow and blood-brain barrier impairments which play a key role in the development of white matter lesions. We hypothesised that cerebral hypoperfusion causes local hypoxia, affecting oligodendrocyte precursor cell-endothelial cell signalling leading to blood-brain barrier dysfunction as an early mechanism for the development of white matter lesions. Bilateral carotid artery stenosis was used as a mouse model for cerebral hypoperfusion. Pimonidazole, a hypoxic cell marker, was injected prior to humane sacrifice at day 7. Myelin content, vascular density, blood-brain barrier leakages, and hypoxic cell density were quantified. Primary mouse oligodendrocyte precursor cells were exposed to hypoxia and RNA sequencing was performed. Vegfa gene expression and protein secretion was examined in an oligodendrocyte precursor cell line exposed to hypoxia. Additionally, human blood plasma VEGFA levels were measured and correlated to blood-brain barrier permeability in normal-appearing white matter and white matter lesions of cerebral small vessel disease patients and controls. Cerebral blood flow was reduced in the stenosis mice, with an increase in hypoxic cell number and blood-brain barrier leakages in the cortical areas but no changes in myelin content or vascular density. Vegfa upregulation was identified in hypoxic oligodendrocyte precursor cells, which was mediated via Hif1α and Epas1. In humans, VEGFA plasma levels were increased in patients versus controls. VEGFA plasma levels were associated with increased blood-brain barrier permeability in normal appearing white matter of patients. Cerebral hypoperfusion mediates hypoxia induced VEGFA expression in oligodendrocyte precursor cells through Hif1α/Epas1 signalling. VEGFA could in turn increase BBB permeability. In humans, increased VEGFA plasma levels in cerebral small vessel disease patients were associated with increased blood-brain barrier permeability in the normal appearing white matter. Our results support a role of VEGFA expression in cerebral hypoperfusion as seen in cerebral small vessel disease.
Collapse
Affiliation(s)
- Narek Manukjan
- Department of Pharmacology and Toxicology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
| | - Daria Majcher
- Department of Pharmacology and Toxicology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Peter Leenders
- Department of Pharmacology and Toxicology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Florian Caiment
- Department of Toxicogenomics, GROW–School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Marcel van Herwijnen
- Department of Toxicogenomics, GROW–School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Hubert J. Smeets
- Department of Toxicogenomics, GROW–School for Oncology and Developmental Biology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Ernst Suidgeest
- C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, P.O. Box 9500, 2300 RA Leiden, the Netherlands
| | - Louise van der Weerd
- C.J. Gorter Center for High Field MRI, Department of Radiology, Leiden University Medical Center, P.O. Box 9500, 2300 RA Leiden, the Netherlands
- Department of Human Genetics, Leiden University Medical Center, P.O. Box 9500, 2300 RA Leiden, The Netherlands
| | - Tim Vanmierlo
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Neuroscience, Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium
- Department of Psychiatry and Neuropsychology, European Graduate School of Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Jacobus F. A. Jansen
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Walter H. Backes
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Robert J. van Oostenbrugge
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Neurology, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Julie Staals
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Neurology, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | - Daniel Fulton
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
| | - Zubair Ahmed
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
- Centre for Trauma Sciences Research, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
| | - W. Matthijs Blankesteijn
- Department of Pharmacology and Toxicology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Sébastien Foulquier
- Department of Pharmacology and Toxicology, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- CARIM - School for Cardiovascular Diseases, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- MHeNs—School for Mental Health and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Department of Neurology, Maastricht University Medical Center+, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| |
Collapse
|
31
|
Jia X, Li Y, Ying Y, Jia X, Tang W, Bian Y, Zhang J, Wang DJJ, Cheng X, Yang Q. Effect of corticosubcortical iron deposition on dysfunction in CADASIL is mediated by white matter microstructural damage. Neuroimage Clin 2023; 39:103485. [PMID: 37542975 PMCID: PMC10407949 DOI: 10.1016/j.nicl.2023.103485] [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: 03/30/2023] [Revised: 07/26/2023] [Accepted: 07/26/2023] [Indexed: 08/07/2023]
Abstract
Iron dysregulation may attenuate cognitive performance in patients with CADASIL. However, the underlying pathophysiological mechanisms remain incompletely understood. Whether white matter microstructural changes mediate these processes is largely unclear. In the present study, 30 cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) patients were confirmed via genetic analysis and 30 sex- and age-matched healthy controls underwent multimodal MRI examinations and neuropsychological assessments. Quantitative susceptibility mapping and peak width of skeletonized mean diffusivity (PSMD) were analyzed. Mediation effect analysis was performed to explore the interrelationship between iron deposition, white matter microstructural changes and cognitive deficits in CADASIL. Cognitive deterioration was most affected in memory and executive function, followed by attention and working memory in CADASIL. Excessive iron in the temporal-precuneus pathway and deep gray matter specific to CADASIL were identified. Mediation analysis further revealed that PSMD mediated the relationship between iron concentration and cognitive profile in CADASIL. The present findings provide a new perspective on iron deposition in the corticosubcortical circuit and its contribution to disease-related selective cognitive decline, in which iron concentration may affect cognition by white matter microstructural changes in CADASIL.
Collapse
Affiliation(s)
- Xiuqin Jia
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China; Key Lab of Medical Engineering for Cardiovascular Disease, Ministry of Education, Beijing 100020, China
| | - Yingying Li
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Yunqing Ying
- Department of Neurology, National Center for Neurological Disorders, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xuejia Jia
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Weijun Tang
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yueyan Bian
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Jiajia Zhang
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Danny J J Wang
- Laboratory of FMRI Technology (LOFT), USC Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90007, United States
| | - Xin Cheng
- Department of Neurology, National Center for Neurological Disorders, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Qi Yang
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China; Key Lab of Medical Engineering for Cardiovascular Disease, Ministry of Education, Beijing 100020, China.
| |
Collapse
|
32
|
Yang YH, Li SS, Wang YC, Yu LL, Zhu HH, Wu JH, Yu WK, An L, Yuan WX, Ji Y, Xu YM, Gao Y, Li YS. Correlation between neutrophil gelatinase phase lipocalin and cerebral small vessel disease. Front Neurol 2023; 14:1177479. [PMID: 37521280 PMCID: PMC10375489 DOI: 10.3389/fneur.2023.1177479] [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: 03/01/2023] [Accepted: 06/16/2023] [Indexed: 08/01/2023] Open
Abstract
Background Cerebral small vessel disease (CSVD) is common in the elderly population. Neutrophil gelatinase-associated lipocalin (NGAL) is closely related to cardiovascular and cerebrovascular diseases. NGAL causes pathological changes, such as damage to the vascular endothelium, by causing inflammation, which results in other related diseases. The purpose of this study was to investigate whether serum NGAL levels could predict disease severity in patients with CSVD. Methods The patients with CSVD who visited the Department of Neurology at the First Affiliated Hospital of Zhengzhou University between January 2018 and June 2022 were prospectively included. The total CSVD burden score was calculated using whole-brain magnetic resonance imaging (MRI), and the patients were divided into a mild group (total CSVD burden score < 2 points) and a severe group (total CSVD burden score ≥ 2 points). Age, sex, height, smoking and alcohol consumption history, medical history, and serological results of patients were collected to perform the univariate analysis. Multivariate logistic regression was used to analyze the risk factors that affect CSVD severity. The multiple linear regression method was used to analyze which individual CSVD markers (periventricular white matter hyperintensities, deep white matter hyperintensities, lacune, and cerebral microbleed) play a role in the association between total CSVD burden score and NGAL. Results A total of 427 patients with CSVD (140 in the mild group and 287 in the severe group) were included in the study. A multivariate logistic regression analysis showed that the following factors were significantly associated with CSVD severity: male sex [odds ratio(OR), 1.912; 95% confidence interval (CI), 1.150-3.179], age (OR, 1.046; 95% CI, 1.022-1.070), history of cerebrovascular disease (OR, 3.050; 95% CI, 1.764-5.274), serum NGAL level (OR, 1.005; 95% CI, 1.002-1.008), and diabetes (OR, 2.593; 95% CI, 1.424-4.722). A multivariate linear regression shows that periventricular white matter hyperintensities and cerebral microbleed are associated with serum NGAL concentrations (P < 0.05). Conclusion Serum NGAL level is closely related to CSVD severity and is a risk factor for the burden of CSVD brain damage. Serum NGAL has high specificity in reflecting the severity of CSVD.
Collapse
|
33
|
Shao X, Zhao C, Shou Q, St Lawrence KS, Wang DJJ. Quantification of blood-brain barrier water exchange and permeability with multidelay diffusion-weighted pseudo-continuous arterial spin labeling. Magn Reson Med 2023; 89:1990-2004. [PMID: 36622951 PMCID: PMC10079266 DOI: 10.1002/mrm.29581] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/22/2022] [Accepted: 12/26/2022] [Indexed: 01/11/2023]
Abstract
PURPOSE To present a pulse sequence and mathematical models for quantification of blood-brain barrier water exchange and permeability. METHODS Motion-compensated diffusion-weighted (MCDW) gradient-and-spin echo (GRASE) pseudo-continuous arterial spin labeling (pCASL) sequence was proposed to acquire intravascular/extravascular perfusion signals from five postlabeling delays (PLDs, 1590-2790 ms). Experiments were performed on 11 healthy subjects at 3 T. A comprehensive set of perfusion and permeability parameters including cerebral blood flow (CBF), capillary transit time (τc ), and water exchange rate (kw ) were quantified, and permeability surface area product (PSw ), total extraction fraction (Ew ), and capillary volume (Vc ) were derived simultaneously by a three-compartment single-pass approximation (SPA) model on group-averaged data. With information (i.e., Vc and τc ) obtained from three-compartment SPA modeling, a simplified linear regression of logarithm (LRL) approach was proposed for individual kw quantification, and Ew and PSw can be estimated from long PLD (2490/2790 ms) signals. MCDW-pCASL was compared with a previously developed diffusion-prepared (DP) pCASL sequence, which calculates kw by a two-compartment SPA model from PLD = 1800 ms signals, to evaluate the improvements. RESULTS Using three-compartment SPA modeling, group-averaged CBF = 51.5/36.8 ml/100 g/min, kw = 126.3/106.7 min-1 , PSw = 151.6/93.8 ml/100 g/min, Ew = 94.7/92.2%, τc = 1409.2/1431.8 ms, and Vc = 1.2/0.9 ml/100 g in gray/white matter, respectively. Temporal SNR of MCDW-pCASL perfusion signals increased 3-fold, and individual kw maps calculated by the LRL method achieved higher spatial resolution (3.5 mm3 isotropic) as compared with DP pCASL (3.5 × 3.5 × 8 mm3 ). CONCLUSION MCDW-pCASL allows visualization of intravascular/extravascular ASL signals across multiple PLDs. The three-compartment SPA model provides a comprehensive measurement of blood-brain barrier water dynamics from group-averaged data, and a simplified LRL method was proposed for individual kw quantification.
Collapse
Affiliation(s)
- Xingfeng Shao
- Laboratory of FMRI Technology (LOFT), Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Chenyang Zhao
- Laboratory of FMRI Technology (LOFT), Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Qinyang Shou
- Laboratory of FMRI Technology (LOFT), Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Keith S St Lawrence
- Lawson Health Research Institute, London, Ontario, Canada
- Department of Medical Biophysics, Western University, London, Ontario, Canada
| | - Danny JJ Wang
- Laboratory of FMRI Technology (LOFT), Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
34
|
Elschot EP, Backes WH, de Jong JJA, Drenthen GS, Wong SM, Staals J, Postma AA, Rouhl RPW, van Oostenbrugge RJ, Jansen JFA. Assessment of the clinical feasibility of detecting subtle blood-brain barrier leakage in cerebral small vessel disease using dynamic susceptibility contrast MRI. Magn Reson Imaging 2023; 102:55-61. [PMID: 37137345 DOI: 10.1016/j.mri.2023.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 04/26/2023] [Accepted: 04/26/2023] [Indexed: 05/05/2023]
Abstract
PURPOSE Cerebral small vessel disease (cSVD) involves several pathologies affecting the small vessels, including blood-brain barrier (BBB) impairment. Dynamic susceptibility contrast (DSC) MRI is sensitive to both blood perfusion and BBB leakage, and correction methods may be crucial for obtaining reliable perfusion measures. These methods might also be applicable to detect BBB leakage itself. This study investigated to what extent DSC-MRI can measure subtle BBB leakage in a clinical feasibility setting. METHODS In vivo DCE and DSC data were collected from fifteen cSVD patients (71 (±10) years, 6F/9M) and twelve elderly controls (71 (±10) years, 4F/8M). DSC-derived leakage fractions were obtained using the Boxerman-Schmainda-Weisskoff method (K2). K2 was compared with the DCE-derived leakage rate Ki, obtained from Patlak analysis. Subsequently, differences were assessed between white matter hyperintensities (WMH), cortical gray matter (CGM), and normal-appearing white matter (NAWM). Additionally, computer simulations were performed to assess the sensitivity of DSC-MRI to BBB leakage. RESULTS K2 showed significant differences between tissue regions (P < 0.001 for CGM-NAWM and CGM-WMH, and P = 0.001 for NAWM-WMH). Conversely, according to the computer simulations the DSC sensitivity was insufficient to measure subtle BBB leakage, as the K2 values were below the derived limit of quantification (4∙10-3 min-1). As expected, Ki was elevated in the WMH compared to CGM and NAWM (P < 0.001). CONCLUSIONS Although clinical DSC-MRI seems capable to detect subtle BBB leakage differences between WMH and normal-appearing brain tissue it is not recommended. K2 as a direct measure for subtle BBB leakage remains ambiguous as its signal effects are due to mixed T1- and T2∗-weighting. Further research is warranted to better disentangle perfusion from leakage effects.
Collapse
Affiliation(s)
- Elles P Elschot
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P. Debyelaan 25, Maastricht, the Netherlands; School for Mental Health and Neuroscience (MHeNs), Maastricht University, Minderbroedersberg 4-6, Maastricht, the Netherlands
| | - Walter H Backes
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P. Debyelaan 25, Maastricht, the Netherlands; School for Mental Health and Neuroscience (MHeNs), Maastricht University, Minderbroedersberg 4-6, Maastricht, the Netherlands; School for Cardiovascular Diseases (CARIM), Maastricht University, Minderbroedersberg 4-6, Maastricht, the Netherlands
| | - Joost J A de Jong
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P. Debyelaan 25, Maastricht, the Netherlands; School for Mental Health and Neuroscience (MHeNs), Maastricht University, Minderbroedersberg 4-6, Maastricht, the Netherlands
| | - Gerhard S Drenthen
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P. Debyelaan 25, Maastricht, the Netherlands; School for Mental Health and Neuroscience (MHeNs), Maastricht University, Minderbroedersberg 4-6, Maastricht, the Netherlands
| | - Sau May Wong
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P. Debyelaan 25, Maastricht, the Netherlands
| | - Julie Staals
- Department of Neurology, Maastricht University Medical Center+, P. Debyelaan 25, Maastricht, the Netherlands; School for Cardiovascular Diseases (CARIM), Maastricht University, Minderbroedersberg 4-6, Maastricht, the Netherlands
| | - Alida A Postma
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P. Debyelaan 25, Maastricht, the Netherlands; School for Mental Health and Neuroscience (MHeNs), Maastricht University, Minderbroedersberg 4-6, Maastricht, the Netherlands
| | - Rob P W Rouhl
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Minderbroedersberg 4-6, Maastricht, the Netherlands; Department of Neurology, Maastricht University Medical Center+, P. Debyelaan 25, Maastricht, the Netherlands; Academic Center for Epileptology Kempenhaeghe/MUMC+, Heeze and Maastricht, the Netherlands
| | - Robert J van Oostenbrugge
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Minderbroedersberg 4-6, Maastricht, the Netherlands; Department of Neurology, Maastricht University Medical Center+, P. Debyelaan 25, Maastricht, the Netherlands; School for Cardiovascular Diseases (CARIM), Maastricht University, Minderbroedersberg 4-6, Maastricht, the Netherlands
| | - Jacobus F A Jansen
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P. Debyelaan 25, Maastricht, the Netherlands; School for Mental Health and Neuroscience (MHeNs), Maastricht University, Minderbroedersberg 4-6, Maastricht, the Netherlands; Department of Electrical Engineering, Eindhoven University of Technology, De Rondom 70, Eindhoven, the Netherlands; Academic Center for Epileptology Kempenhaeghe/MUMC+, Heeze and Maastricht, the Netherlands.
| |
Collapse
|
35
|
Wang H, Chai C, Wu G, Li J, Zhao C, Fu D, Zhang S, Wang H, Wang B, Zhu J, Shen W, Xia S. Cerebral blood flow regulates iron overload in the cerebral nuclei of hemodialysis patients with anemia. J Cereb Blood Flow Metab 2023; 43:749-762. [PMID: 36545834 PMCID: PMC10108183 DOI: 10.1177/0271678x221147363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 09/16/2022] [Accepted: 10/09/2022] [Indexed: 12/24/2022]
Abstract
Hemodialysis patients exhibit anemia-related cerebral hyperperfusion and iron deposition (ID). However, the mechanisms underlying the pathology of cerebral ID are not clear. We investigated the role of cerebral blood flow (CBF) in the pathophysiology of cerebral ID in hemodialysis patients with anemia. This study recruited 33 hemodialysis patients with anemia and thirty-three healthy controls (HCs). All the subjects underwent quantitative susceptibility mapping (QSM) and arterial spin labeling (ASL) to measure ID and CBF in the cerebral nuclei. Furthermore, we evaluated lacunar infarction (LI), cerebral microbleeds, and total white matter hyperintensity volume (TWMHV). Hemodialysis patients with anemia showed significantly higher ID and CBF in some nuclei compared to the HCs after adjusting for age, sex, and total intracranial volume (TIV) [P < 0.05, false discovery rate (FDR) corrected]. CBF showed a positive correlation with ID in both patients and HCs after adjustments for age, gender, and TIV (P < 0.05, FDR corrected). Serum phosphorus, calcium, TWMHV, hypertension, and dialysis duration were independently associated with ID (P < 0.05). Hemoglobin, serum phosphorus, and LI were independently associated with CBF (P < 0.05). Mediation analysis demonstrated that CBF mediated the effects between hemoglobin and ID. Our study demonstrated that CBF mediated aberrant cerebral ID in hemodialysis patients with anemia.
Collapse
Affiliation(s)
- Huiying Wang
- The School of Medicine, Nankai
University, Tianjin, China
| | - Chao Chai
- Department of Radiology, Tianjin
First Central Hospital, School of Medicine, Nankai University, Tianjin,
China
- Imaging Medicine Institute of
Tianjin, Tianjin, China
| | - Gemuer Wu
- The School of Medicine, Nankai
University, Tianjin, China
| | - Jinping Li
- Department of Hemodialysis, Tianjin
First Central Hospital, School of Medicine, Nankai University, Tianjin,
China
| | - Chenxi Zhao
- Department of Radiology, First
Central Clinical College, Tianjin Medical University, Tianjin, China
| | - Dingwei Fu
- Department of Radiology, First
Central Clinical College, Tianjin Medical University, Tianjin, China
| | | | - Huapeng Wang
- Department of Radiology, First
Central Clinical College, Tianjin Medical University, Tianjin, China
| | - Beini Wang
- Department of Radiology, First
Central Clinical College, Tianjin Medical University, Tianjin, China
| | - Jinxia Zhu
- MR Collaboration, Siemens
Healthcare, Northeast Asia, Beijing, China
| | - Wen Shen
- Department of Radiology, Tianjin
First Central Hospital, School of Medicine, Nankai University, Tianjin,
China
- Imaging Medicine Institute of
Tianjin, Tianjin, China
| | - Shuang Xia
- Department of Radiology, Tianjin
First Central Hospital, School of Medicine, Nankai University, Tianjin,
China
- Imaging Medicine Institute of
Tianjin, Tianjin, China
| |
Collapse
|
36
|
Xu J, Su Y, Fu J, Shen Y, Dong Q, Cheng X. Glymphatic pathway in sporadic cerebral small vessel diseases: From bench to bedside. Ageing Res Rev 2023; 86:101885. [PMID: 36801378 DOI: 10.1016/j.arr.2023.101885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023]
Abstract
Cerebral small vessel diseases (CSVD) consist of a group of diseases with high heterogeneity induced by pathologies of intracranial small blood vessels. Endothelium dysfunction, bloodbrain barrier leakage and the inflammatory response are traditionally considered to participate in the pathogenesis of CSVD. However, these features cannot fully explain the complex syndrome and related neuroimaging characteristics. In recent years, the glymphatic pathway has been discovered to play a pivotal role in clearing perivascular fluid and metabolic solutes, which has provided novel insights into neurological disorders. Researchers have also explored the potential role of perivascular clearance dysfunction in CSVD. In this review, we presented a brief overview of CSVD and the glymphatic pathway. In addition, we elucidated CSVD pathogenesis from the perspective of glymphatic failure, including basic animal models and clinical neuroimaging markers. Finally, we proposed forthcoming clinical applications targeting the glymphatic pathway, hoping to provide novel ideas on promising therapies and preventions of CSVD.
Collapse
Affiliation(s)
- Jiajie Xu
- Department of Neurology, National Center for Neurological Disorders, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Ya Su
- Department of Neurology, National Center for Neurological Disorders, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiayu Fu
- Department of Neurology, National Center for Neurological Disorders, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yong Shen
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC and Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qiang Dong
- Department of Neurology, National Center for Neurological Disorders, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China; State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Xin Cheng
- Department of Neurology, National Center for Neurological Disorders, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
37
|
Lee RL, Funk KE. Imaging blood–brain barrier disruption in neuroinflammation and Alzheimer’s disease. Front Aging Neurosci 2023; 15:1144036. [PMID: 37009464 PMCID: PMC10063921 DOI: 10.3389/fnagi.2023.1144036] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/28/2023] [Indexed: 03/19/2023] Open
Abstract
The blood–brain barrier (BBB) is the neurovascular structure that regulates the passage of cells and molecules to and from the central nervous system (CNS). Alzheimer’s disease (AD) is a neurodegenerative disorder that is associated with gradual breakdown of the BBB, permitting entry of plasma-derived neurotoxins, inflammatory cells, and microbial pathogens into the CNS. BBB permeability can be visualized directly in AD patients using imaging technologies including dynamic contrast-enhanced and arterial spin labeling magnetic resonance imaging, and recent studies employing these techniques have shown that subtle changes in BBB stability occur prior to deposition of the pathological hallmarks of AD, senile plaques, and neurofibrillary tangles. These studies suggest that BBB disruption may be useful as an early diagnostic marker; however, AD is also accompanied by neuroinflammation, which can complicate these analyses. This review will outline the structural and functional changes to the BBB that occur during AD pathogenesis and highlight current imaging technologies that can detect these subtle changes. Advancing these technologies will improve both the diagnosis and treatment of AD and other neurodegenerative diseases.
Collapse
|
38
|
Lapucci C, Tazza F, Rebella S, Boffa G, Sbragia E, Bruschi N, Mancuso E, Mavilio N, Signori A, Roccatagliata L, Cellerino M, Schiavi S, Inglese M. Central vein sign and diffusion MRI differentiate microstructural features within white matter lesions of multiple sclerosis patients with comorbidities. Front Neurol 2023; 14:1084661. [PMID: 36970546 PMCID: PMC10030505 DOI: 10.3389/fneur.2023.1084661] [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: 10/30/2022] [Accepted: 01/30/2023] [Indexed: 03/29/2023] Open
Abstract
Introduction The Central Vein Sign (CVS) has been suggested as a potential biomarker to improve diagnostic specificity in multiple sclerosis (MS). Nevertheless, the impact of comorbidities on CVS performance has been poorly investigated so far. Despite the similar features shared by MS, migraine and Small Vessel Disease (SVD) at T2-weighted conventional MRI sequences, ex-vivo studies demonstrated their heterogeneous histopathological substrates. If in MS, inflammation, primitive demyelination and axonal loss coexist, in SVD demyelination is secondary to ischemic microangiopathy, while the contemporary presence of inflammatory and ischemic processes has been suggested in migraine. The aims of this study were to investigate the impact of comorbidities (risk factors for SVD and migraine) on the global and subregional assessment of the CVS in a large cohort of MS patients and to apply the Spherical Mean Technique (SMT) diffusion model to evaluate whether perivenular and non-perivenular lesions show distinctive microstructural features. Methods 120 MS patients stratified into 4 Age Groups performed 3T brain MRI. WM lesions were classified in "perivenular" and "non-perivenular" by visual inspection of FLAIR* images; mean values of SMT metrics, indirect estimators of inflammation, demyelination and fiber disruption (EXTRAMD: extraneurite mean diffusivity, EXTRATRANS: extraneurite transverse diffusivity and INTRA: intraneurite signal fraction, respectively) were extracted. Results Of the 5303 lesions selected for the CVS assessment, 68.7% were perivenular. Significant differences were found between perivenular and non-perivenular lesion volume in the whole brain (p < 0.001) and between perivenular and non-perivenular lesion volume and number in all the four subregions (p < 0.001 for all). The percentage of perivenular lesions decreased from youngest to oldest patients (79.7%-57.7%), with the deep/subcortical WM of oldest patients as the only subregion where the number of non-perivenular was higher than the number of perivenular lesions. Older age and migraine were independent predictors of a higher percentage of non-perivenular lesions (p < 0.001 and p = 0.013 respectively). Whole brain perivenular lesions showed higher inflammation, demyelination and fiber disruption than non perivenular lesions (p = 0.001, p = 0.001 and p = 0.02 for EXTRAMD, EXTRATRANS and INTRA respectively). Similar findings were found in the deep/subcortical WM (p = 0.001 for all). Compared to non-perivenular lesions, (i) perivenular lesions located in periventricular areas showed a more severe fiber disruption (p = 0.001), (ii) perivenular lesions located in juxtacortical and infratentorial regions exhibited a higher degree of inflammation (p = 0.01 and p = 0.05 respectively) and (iii) perivenular lesions located in infratentorial areas showed a higher degree of demyelination (p = 0.04). Discussion Age and migraine have a relevant impact in reducing the percentage of perivenular lesions, particularly in the deep/subcortical WM. SMT may differentiate perivenular lesions, characterized by higher inflammation, demyelination and fiber disruption, from non perivenular lesions, where these pathological processes seemed to be less pronounced. The development of new non-perivenular lesions, especially in the deep/subcortical WM of older patients, should be considered a "red flag" for a different -other than MS- pathophysiology.
Collapse
Affiliation(s)
- Caterina Lapucci
- HNSR, IRRCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Francesco Tazza
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | | | - Giacomo Boffa
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Elvira Sbragia
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Nicolò Bruschi
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Elisabetta Mancuso
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Nicola Mavilio
- Department of Neuroradiology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Alessio Signori
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | - Luca Roccatagliata
- Department of Neuroradiology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | - Maria Cellerino
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Simona Schiavi
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Matilde Inglese
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino IRCCS, Genoa, Italy
| |
Collapse
|
39
|
He Y, Li Z, Shi X, Ding J, Wang X. Roles of NG2 Glia in Cerebral Small Vessel Disease. Neurosci Bull 2023; 39:519-530. [PMID: 36401147 PMCID: PMC10043141 DOI: 10.1007/s12264-022-00976-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 09/02/2022] [Indexed: 11/19/2022] Open
Abstract
Cerebral small vessel disease (CSVD) is one of the most prevalent pathologic processes affecting 5% of people over 50 years of age and contributing to 45% of dementia cases. Increasing evidence has demonstrated the pathological roles of chronic hypoperfusion, impaired cerebral vascular reactivity, and leakage of the blood-brain barrier in CSVD. However, the pathogenesis of CSVD remains elusive thus far, and no radical treatment has been developed. NG2 glia, also known as oligodendrocyte precursor cells, are the fourth type of glial cell in addition to astrocytes, microglia, and oligodendrocytes in the mammalian central nervous system. Many novel functions for NG2 glia in physiological and pathological states have recently been revealed. In this review, we discuss the role of NG2 glia in CSVD and the underlying mechanisms.
Collapse
Affiliation(s)
- Yixi He
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Zhenghao Li
- Institute of Neuroscience, MOE Key Laboratory of Molecular Neurobiology, NMU, Shanghai, 200433, China
| | - Xiaoyu Shi
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Jing Ding
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| | - Xin Wang
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
40
|
Yang Y, Knol MJ, Wang R, Mishra A, Liu D, Luciano M, Teumer A, Armstrong N, Bis JC, Jhun MA, Li S, Adams HHH, Aziz NA, Bastin ME, Bourgey M, Brody JA, Frenzel S, Gottesman RF, Hosten N, Hou L, Kardia SLR, Lohner V, Marquis P, Maniega SM, Satizabal CL, Sorond FA, Valdés Hernández MC, van Duijn CM, Vernooij MW, Wittfeld K, Yang Q, Zhao W, Boerwinkle E, Levy D, Deary IJ, Jiang J, Mather KA, Mosley TH, Psaty BM, Sachdev PS, Smith JA, Sotoodehnia N, DeCarli CS, Breteler MMB, Ikram MA, Grabe HJ, Wardlaw J, Longstreth WT, Launer LJ, Seshadri S, Debette S, Fornage M. Epigenetic and integrative cross-omics analyses of cerebral white matter hyperintensities on MRI. Brain 2023; 146:492-506. [PMID: 35943854 PMCID: PMC9924914 DOI: 10.1093/brain/awac290] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 06/23/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Cerebral white matter hyperintensities on MRI are markers of cerebral small vessel disease, a major risk factor for dementia and stroke. Despite the successful identification of multiple genetic variants associated with this highly heritable condition, its genetic architecture remains incompletely understood. More specifically, the role of DNA methylation has received little attention. We investigated the association between white matter hyperintensity burden and DNA methylation in blood at ∼450 000 cytosine-phosphate-guanine (CpG) sites in 9732 middle-aged to older adults from 14 community-based studies. Single CpG and region-based association analyses were carried out. Functional annotation and integrative cross-omics analyses were performed to identify novel genes underlying the relationship between DNA methylation and white matter hyperintensities. We identified 12 single CpG and 46 region-based DNA methylation associations with white matter hyperintensity burden. Our top discovery single CpG, cg24202936 (P = 7.6 × 10-8), was associated with F2 expression in blood (P = 6.4 × 10-5) and co-localized with FOLH1 expression in brain (posterior probability = 0.75). Our top differentially methylated regions were in PRMT1 and in CCDC144NL-AS1, which were also represented in single CpG associations (cg17417856 and cg06809326, respectively). Through Mendelian randomization analyses cg06809326 was putatively associated with white matter hyperintensity burden (P = 0.03) and expression of CCDC144NL-AS1 possibly mediated this association. Differentially methylated region analysis, joint epigenetic association analysis and multi-omics co-localization analysis consistently identified a role of DNA methylation near SH3PXD2A, a locus previously identified in genome-wide association studies of white matter hyperintensities. Gene set enrichment analyses revealed functions of the identified DNA methylation loci in the blood-brain barrier and in the immune response. Integrative cross-omics analysis identified 19 key regulatory genes in two networks related to extracellular matrix organization, and lipid and lipoprotein metabolism. A drug-repositioning analysis indicated antihyperlipidaemic agents, more specifically peroxisome proliferator-activated receptor-alpha, as possible target drugs for white matter hyperintensities. Our epigenome-wide association study and integrative cross-omics analyses implicate novel genes influencing white matter hyperintensity burden, which converged on pathways related to the immune response and to a compromised blood-brain barrier possibly due to disrupted cell-cell and cell-extracellular matrix interactions. The results also suggest that antihyperlipidaemic therapy may contribute to lowering risk for white matter hyperintensities possibly through protection against blood-brain barrier disruption.
Collapse
Affiliation(s)
- Yunju Yang
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science at Houston, Houston, TX 77030, USA
| | - Maria J Knol
- Department of Epidemiology, Erasmus MC University Medical Center, 3015 GD, Rotterdam, The Netherlands
| | - Ruiqi Wang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Aniket Mishra
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, Team VINTAGE, UMR 1219, F-33000 Bordeaux, France
| | - Dan Liu
- Population Health Sciences, German Centre for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Michelle Luciano
- Department of Psychology, University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald 17475, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, Greifswald 17475, Germany
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Bialystok, Bialystok, 15-269, Poland
| | - Nicola Armstrong
- Mathematics and Statistics, Curtin University, 6845 Perth, Australia
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA 02115, USA
| | - Min A Jhun
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48104, USA
| | - Shuo Li
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Hieab H H Adams
- Department of Epidemiology, Erasmus MC University Medical Center, 3015 GD, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, 3015 GD, Rotterdam, The Netherlands
| | - Nasir Ahmad Aziz
- Population Health Sciences, German Centre for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
- Department of Neurology, Faculty of Medicine, University of Bonn, 53127 Bonn, Germany
| | - Mark E Bastin
- Centre for Clinical Brain Sciences, Department of Neuroimaging Sciences, University of Edinburgh, Edinburgh, EH8 9AB, UK
| | - Mathieu Bourgey
- Canadian Centre for Computational Genomics, McGill University, Montréal, Quebec, Canada H3A 0G1
- Department for Human Genetics, McGill University Genome Centre, McGill University, Montréal, Quebec, Canada H3A 0G1
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA 02115, USA
| | - Stefan Frenzel
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald 17475, Germany
| | - Rebecca F Gottesman
- Stroke Branch, National Institutes of Neurological Disorders and Stroke, Bethesda, MD 20814, USA
| | - Norbert Hosten
- Department of Radiology and Neuroradiology, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Lifang Hou
- Department of Preventive Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48104, USA
| | - Valerie Lohner
- Population Health Sciences, German Centre for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Pascale Marquis
- Canadian Centre for Computational Genomics, McGill University, Montréal, Quebec, Canada H3A 0G1
- Department for Human Genetics, McGill University Genome Centre, McGill University, Montréal, Quebec, Canada H3A 0G1
| | - Susana Muñoz Maniega
- Centre for Clinical Brain Sciences, Department of Neuroimaging Sciences, University of Edinburgh, Edinburgh, EH8 9AB, UK
| | - Claudia L Satizabal
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases and Department of Population Health Sciences, UT Health San Antonio, San Antonio, TX 78229, USA
- The Framingham Heart Study, Framingham, MA 01701, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 02115, USA
| | - Farzaneh A Sorond
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Maria C Valdés Hernández
- Centre for Clinical Brain Sciences, Department of Neuroimaging Sciences, University of Edinburgh, Edinburgh, EH8 9AB, UK
| | - Cornelia M van Duijn
- Department of Epidemiology, Erasmus MC University Medical Center, 3015 GD, Rotterdam, The Netherlands
- Nuffield Department of Population Health, Oxford University, Oxford, OX3 7LF, UK
| | - Meike W Vernooij
- Department of Epidemiology, Erasmus MC University Medical Center, 3015 GD, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center, 3015 GD, Rotterdam, The Netherlands
| | - Katharina Wittfeld
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald 17475, Germany
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, 17475 Rostock, Germany
| | - Qiong Yang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
- The Framingham Heart Study, Framingham, MA 01701, USA
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48104, USA
| | - Eric Boerwinkle
- Human Genetics Center, School of Public Health, University of Texas Health Science at Houston, Houston, TX 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Daniel Levy
- The Framingham Heart Study, Framingham, MA 01701, USA
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20814, USA
| | - Ian J Deary
- Department of Psychology, University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Jiyang Jiang
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW 2052, Australia
| | - Karen A Mather
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW 2052, Australia
- Neuroscience Research Australia, Sydney, NSW 2031, Australia
| | - Thomas H Mosley
- The Memory Impairment Neurodegenerative Dementia (MIND) Research Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA 02115, USA
- Department of Epidemiology, University of Washington, Seattle, WA 98104, USA
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW 2052, Australia
- Neuropsychiatric Institute, The Prince of Wales Hospital, University of New South Wales, Randwick, NSW 2031, Australia
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48104, USA
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA 02115, USA
| | - Charles S DeCarli
- Department of Neurology and Center for Neuroscience, University of California at Davis, Sacramento, CA 95816, USA
| | - Monique M B Breteler
- Population Health Sciences, German Centre for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
- Institute for Medical Biometry, Informatics and Epidemiology (IMBIE), Faculty of Medicine, University of Bonn, 53127 Bonn, Germany
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, 3015 GD, Rotterdam, The Netherlands
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald 17475, Germany
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, 17475 Rostock, Germany
| | - Joanna Wardlaw
- Centre for Clinical Brain Sciences, Department of Neuroimaging Sciences, University of Edinburgh, Edinburgh, EH8 9AB, UK
| | - W T Longstreth
- Department of Epidemiology, University of Washington, Seattle, WA 98104, USA
- Department of Neurology, University of Washington, Seattle, WA 98104, USA
| | - Lenore J Launer
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Bethesda, MD 20814, USA
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases and Department of Population Health Sciences, UT Health San Antonio, San Antonio, TX 78229, USA
- The Framingham Heart Study, Framingham, MA 01701, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA 02115, USA
| | - Stephanie Debette
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, Team VINTAGE, UMR 1219, F-33000 Bordeaux, France
- Department of Neurology, Boston University School of Medicine, Boston, MA 02115, USA
- CHU de Bordeaux, Department of Neurology, F-33000 Bordeaux, France
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science at Houston, Houston, TX 77030, USA
- Human Genetics Center, School of Public Health, University of Texas Health Science at Houston, Houston, TX 77030, USA
| |
Collapse
|
41
|
Uchida Y, Kan H, Sakurai K, Oishi K, Matsukawa N. Contributions of blood-brain barrier imaging to neurovascular unit pathophysiology of Alzheimer's disease and related dementias. Front Aging Neurosci 2023; 15:1111448. [PMID: 36861122 PMCID: PMC9969807 DOI: 10.3389/fnagi.2023.1111448] [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: 11/29/2022] [Accepted: 01/26/2023] [Indexed: 02/11/2023] Open
Abstract
The blood-brain barrier (BBB) plays important roles in the maintenance of brain homeostasis. Its main role includes three kinds of functions: (1) to protect the central nervous system from blood-borne toxins and pathogens; (2) to regulate the exchange of substances between the brain parenchyma and capillaries; and (3) to clear metabolic waste and other neurotoxic compounds from the central nervous system into meningeal lymphatics and systemic circulation. Physiologically, the BBB belongs to the glymphatic system and the intramural periarterial drainage pathway, both of which are involved in clearing interstitial solutes such as β-amyloid proteins. Thus, the BBB is believed to contribute to preventing the onset and progression for Alzheimer's disease. Measurements of BBB function are essential toward a better understanding of Alzheimer's pathophysiology to establish novel imaging biomarkers and open new avenues of interventions for Alzheimer's disease and related dementias. The visualization techniques for capillary, cerebrospinal, and interstitial fluid dynamics around the neurovascular unit in living human brains have been enthusiastically developed. The purpose of this review is to summarize recent BBB imaging developments using advanced magnetic resonance imaging technologies in relation to Alzheimer's disease and related dementias. First, we give an overview of the relationship between Alzheimer's pathophysiology and BBB dysfunction. Second, we provide a brief description about the principles of non-contrast agent-based and contrast agent-based BBB imaging methodologies. Third, we summarize previous studies that have reported the findings of each BBB imaging method in individuals with the Alzheimer's disease continuum. Fourth, we introduce a wide range of Alzheimer's pathophysiology in relation to BBB imaging technologies to advance our understanding of the fluid dynamics around the BBB in both clinical and preclinical settings. Finally, we discuss the challenges of BBB imaging techniques and suggest future directions toward clinically useful imaging biomarkers for Alzheimer's disease and related dementias.
Collapse
Affiliation(s)
- Yuto Uchida
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States,*Correspondence: Yuto Uchida, ; Noriyuki Matsukawa,
| | - Hirohito Kan
- Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Keita Sakurai
- Department of Radiology, National Center for Geriatrics and Gerontology, Ōbu, Aichi, Japan
| | - Kenichi Oishi
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Noriyuki Matsukawa
- Department of Neurology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan,*Correspondence: Yuto Uchida, ; Noriyuki Matsukawa,
| |
Collapse
|
42
|
Jia X, Ling C, Li Y, Zhang J, Li Z, Jia X, Wang DJJ, Zhang Z, Yuan Y, Yang Q. Sex differences in frontotemporal atrophy in CADASIL revealed by 7-Tesla MRI. Neuroimage Clin 2023; 37:103298. [PMID: 36577270 DOI: 10.1016/j.nicl.2022.103298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022]
Abstract
Brain damage caused by small vessel disease (SVD) differs between males and females. We aimed to examine the pure sex-specific neuroanatomical mechanisms of SVD adjusted for voxel-based expected effects of age and sex on healthy brain volume. Thirty-one female and 32 male genetic SVD (cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy, CADASIL) patients and 55 sex- and age-matched healthy controls (HCs) underwent 7-Tesla MRI examinations. Voxel-based W-score maps were calculated from volumes and deformations of brain tissues, controlling for the expected effects of age and sex in HCs. Significant cognitive declines in working memory and executive function were identified in male CADASIL patients compared to female patients. Greater gray matter (GM) atrophy was found in the bilateral orbitofrontal cortex (OFC), left anterior cingulate cortex (ACC), left entorhinal cortex (EC), and right temporooccipital cortex in male CADASIL patients than in females. Working memory was associated with volumes in the right OFC specific to female CADASIL patients, whereas visuospatial ability was associated with the right hOcl (primary visual area, BA 17) volume specific to males. The current findings indicate that sex affects the pathogenesis of CADASIL, ranging from differences in neuroanatomy to those in behavioral performance, which may facilitate the development of more effective sex-specific therapeutic strategies for CADASIL and SVD.
Collapse
Affiliation(s)
- Xiuqin Jia
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China; Key Lab of Medical Engineering for Cardiovascular Disease, Ministry of Education, China
| | - Chen Ling
- Department of Neurology, Peking University First Hospital, Beijing 100034, China
| | - Yingying Li
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Jinyuan Zhang
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing 100101, China
| | - Zhixin Li
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing 100101, China
| | - Xuejia Jia
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Danny J J Wang
- Lab of FMRI Technology (LOFT), Mark & Mary Stevens Neuroimaging and Informatics Institute Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| | - Zihao Zhang
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Beijing 100101, China; Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China.
| | - Yun Yuan
- Department of Neurology, Peking University First Hospital, Beijing 100034, China.
| | - Qi Yang
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China; Key Lab of Medical Engineering for Cardiovascular Disease, Ministry of Education, China; Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, China.
| |
Collapse
|
43
|
Huang P, Zhang M. Magnetic Resonance Imaging Studies of Neurodegenerative Disease: From Methods to Translational Research. Neurosci Bull 2023; 39:99-112. [PMID: 35771383 PMCID: PMC9849544 DOI: 10.1007/s12264-022-00905-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 05/07/2022] [Indexed: 01/22/2023] Open
Abstract
Neurodegenerative diseases (NDs) have become a significant threat to an aging human society. Numerous studies have been conducted in the past decades to clarify their pathologic mechanisms and search for reliable biomarkers. Magnetic resonance imaging (MRI) is a powerful tool for investigating structural and functional brain alterations in NDs. With the advantages of being non-invasive and non-radioactive, it has been frequently used in both animal research and large-scale clinical investigations. MRI may serve as a bridge connecting micro- and macro-level analysis and promoting bench-to-bed translational research. Nevertheless, due to the abundance and complexity of MRI techniques, exploiting their potential is not always straightforward. This review aims to briefly introduce research progress in clinical imaging studies and discuss possible strategies for applying MRI in translational ND research.
Collapse
Affiliation(s)
- Peiyu Huang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009 China
| | - Minming Zhang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009 China
| |
Collapse
|
44
|
Yu WK, Wang YC, Gao Y, Shi CH, Fan Y, Yu LL, Zhao ZC, Li SS, Xu YM, Li YS. Genetic analysis of the ATP11B gene in Chinese Han population with cerebral small vessel disease. BMC Genomics 2022; 23:822. [PMID: 36510145 PMCID: PMC9746074 DOI: 10.1186/s12864-022-09051-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 11/25/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND A loss-of-function mutation in ATPase phospholipid transporting 11-B (putative) (ATP11B) gene causing cerebral small vessel disease (SVD) in vivo, and a single intronic nucleotide polymorphism in ATP11B: rs148771930 that was associated with white matter hyperintensities burden in European patients with SVD, was recently identified. Our results suggest that ATP11B may not play an essential role in SVD in the Chinese population. RESULTS We performed target region sequencing including ATP11B gene in 182 patients with sporadic SVD, and identified five rare variants and two novel variants of ATP11B. A case-control study was then performed in 524 patients and matched 550 controls to investigate the relationship between ATP11B and sporadic SVD in the Chinese Han population. Although none of these variants were significantly associated with SVD in our samples, it is important to mention that we identified a novel variant, p. G238W, which was predicted to be pathogenic in silico. This variant was present in our cohort of patients with an extremely low frequency and was absent in the controls. CONCLUSION Our results suggest that ATP11B may not play an essential role in SVD in the Chinese population.
Collapse
Affiliation(s)
- Wen-Kai Yu
- grid.207374.50000 0001 2189 3846Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan China
| | - Yun-Chao Wang
- grid.207374.50000 0001 2189 3846Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan China
| | - Yuan Gao
- grid.207374.50000 0001 2189 3846Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan China
| | - Chang-He Shi
- grid.207374.50000 0001 2189 3846Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan China
| | - Yu Fan
- grid.207374.50000 0001 2189 3846Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan China
| | - Lu-Lu Yu
- grid.207374.50000 0001 2189 3846Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan China
| | - Zi-Chen Zhao
- grid.207374.50000 0001 2189 3846Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan China
| | - Shan-Shan Li
- grid.207374.50000 0001 2189 3846Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan China
| | - Yu-Ming Xu
- grid.207374.50000 0001 2189 3846Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan China ,National Health Commission Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, Zhengzhou, Henan China
| | - Yu-Sheng Li
- grid.207374.50000 0001 2189 3846Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan China
| |
Collapse
|
45
|
Wang RZ, Huang SY, Li HQ, Yang YX, Chen SD, Yu JT. Genetic determinants of circulating metabolites and the risk of stroke and its subtypes. Eur J Neurol 2022; 29:3711-3719. [PMID: 36086915 DOI: 10.1111/ene.15549] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/30/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND PURPOSE Circulating metabolites have been implicated in stroke pathogenesis, but their genetic determinants are understudied. Using a Mendelian randomization approach, our aim was to provide evidence for the relationship of circulating metabolites and the risk of stroke and its subtypes. METHODS Genetic instruments of 102 circulating metabolites were obtained from a genome-wide association study, including 24,925 European individuals. Stroke was extracted from the MEGASTROKE dataset (67,162 cases; 454,450 controls) and a lacunar stroke dataset (7338 cases; 254,798 controls). The magnetic resonance imaging markers of cerebral small vessel disease and microstructural injury were evaluated by a genome-wide association study of white matter hyperintensities (N = 18,381), fractional anisotropy (N = 17,663), mean diffusivity (N = 17,467) and brain microbleeds (N = 25,862). The inverse-variance weighted method Mendelian randomization was used as the primary analytical method, and directional pleiotropy and heterogeneity were examined in sensitivity analyses. RESULTS A genetic predisposition to a higher level of cholesterol in small and low-density lipoprotein (LDL) was associated with risk of stroke (odds ratio [OR] 1.14, 95% confidence interval [CI] 1.08-1.21, p = 5.98 × 10-7 ), especially for large-artery atherosclerotic stroke (OR 1.34, 95% CI 1.19-1.52, p = 1.90 × 10-6 ). Total lipids in LDL particles were also associated with risk of stroke. A genetically determined higher cholesterol level in high-density lipoprotein (HDL-C) was associated with risk of intracerebral haemorrhage (OR 1.74, 95% CI 1.23-2.45, p = 1.66 × 10-3 ). No statistically significant association was found between genetic predisposition to circulating metabolites and magnetic resonance imaging markers of cerebral small vessel disease and microstructural injury. CONCLUSIONS Genetically determined levels of lipids in small LDL were associated with the risk of stroke, suggesting that a therapeutic strategy targeting small LDL levels may be crucial for stroke prevention. HDL-C was positively associated with the risk of intracerebral haemorrhage.
Collapse
Affiliation(s)
- Rong-Ze Wang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shu-Yi Huang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hong-Qi Li
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yu-Xiang Yang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shi-Dong Chen
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
46
|
Wang S, Zhang F, Huang P, Hong H, Jiaerken Y, Yu X, Zhang R, Zeng Q, Zhang Y, Kikinis R, Rathi Y, Makris N, Lou M, Pasternak O, Zhang M, O'Donnell LJ. Superficial white matter microstructure affects processing speed in cerebral small vessel disease. Hum Brain Mapp 2022; 43:5310-5325. [PMID: 35822593 PMCID: PMC9812245 DOI: 10.1002/hbm.26004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 06/10/2022] [Accepted: 06/15/2022] [Indexed: 01/15/2023] Open
Abstract
White matter hyperintensities (WMH) are a typical feature of cerebral small vessel disease (CSVD), which contributes to about 50% of dementias worldwide. Microstructural alterations in deep white matter (DWM) have been widely examined in CSVD. However, little is known about abnormalities in superficial white matter (SWM) and their relevance for processing speed, the main cognitive deficit in CSVD. In 141 CSVD patients, processing speed was assessed using Trail Making Test Part A. White matter abnormalities were assessed by WMH burden (volume on T2-FLAIR) and diffusion MRI measures. SWM imaging measures had a large contribution to processing speed, despite a relatively low SWM WMH burden. Across all imaging measures, SWM free water (FW) had the strongest association with processing speed, followed by SWM mean diffusivity (MD). SWM FW was the only marker to significantly increase between two subgroups with the lowest WMH burdens. When comparing two subgroups with the highest WMH burdens, the involvement of WMH in the SWM was accompanied by significant differences in processing speed and white matter microstructure. Mediation analysis revealed that SWM FW fully mediated the association between WMH volume and processing speed, while no mediation effect of MD or DWM FW was observed. Overall, results suggest that the SWM has an important contribution to processing speed, while SWM FW is a sensitive imaging marker associated with cognition in CSVD. This study extends the current understanding of CSVD-related dysfunction and suggests that the SWM, as an understudied region, can be a potential target for monitoring pathophysiological processes.
Collapse
Affiliation(s)
- Shuyue Wang
- Department of Radiologythe Second Affiliated Hospital of Zhejiang University School of MedicineChina
- Brigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Fan Zhang
- Brigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Peiyu Huang
- Department of Radiologythe Second Affiliated Hospital of Zhejiang University School of MedicineChina
| | - Hui Hong
- Department of Radiologythe Second Affiliated Hospital of Zhejiang University School of MedicineChina
| | - Yeerfan Jiaerken
- Department of Radiologythe Second Affiliated Hospital of Zhejiang University School of MedicineChina
| | - Xinfeng Yu
- Department of Radiologythe Second Affiliated Hospital of Zhejiang University School of MedicineChina
| | - Ruiting Zhang
- Department of Radiologythe Second Affiliated Hospital of Zhejiang University School of MedicineChina
| | - Qingze Zeng
- Department of Radiologythe Second Affiliated Hospital of Zhejiang University School of MedicineChina
| | - Yao Zhang
- Department of Radiologythe Second Affiliated Hospital of Zhejiang University School of MedicineChina
| | - Ron Kikinis
- Brigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Yogesh Rathi
- Brigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Nikos Makris
- Brigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Center for Morphometric AnalysisMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Min Lou
- Department of Neurologythe Second Affiliated Hospital of Zhejiang University School of MedicineChina
| | - Ofer Pasternak
- Brigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Minming Zhang
- Department of Radiologythe Second Affiliated Hospital of Zhejiang University School of MedicineChina
| | | |
Collapse
|
47
|
Liu X, Sun P, Yang J, Fan Y. Biomarkers involved in the pathogenesis of cerebral small-vessel disease. Front Neurol 2022; 13:969185. [PMID: 36119691 PMCID: PMC9475115 DOI: 10.3389/fneur.2022.969185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
Cerebral small-vessel disease (CSVD) has been found to have a strong association with vascular cognitive impairment (VCI) and functional loss in elderly patients. At present, the diagnosis of CSVD mainly relies on brain neuroimaging markers, but they cannot fully reflect the overall picture of the disease. Currently, some biomarkers were found to be related to CSVD, but the underlying mechanisms remain unclear. We aimed to systematically review and summarize studies on the progress of biomarkers related to the pathogenesis of CSVD, which is mainly the relationship between these indicators and neuroimaging markers of CSVD. Concerning the pathophysiological mechanism of CSVD, the biomarkers of CSVD have been described as several categories related to sporadic and genetic factors. Monitoring of biomarkers might contribute to the early diagnosis and progression prediction of CSVD, thus providing ideas for better diagnosis and treatment of CSVD.
Collapse
|
48
|
Gao Y, Li D, Lin J, Thomas AM, Miao J, Chen D, Li S, Chu C. Cerebral small vessel disease: Pathological mechanisms and potential therapeutic targets. Front Aging Neurosci 2022; 14:961661. [PMID: 36034144 PMCID: PMC9412755 DOI: 10.3389/fnagi.2022.961661] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
Cerebral small vessel disease (CSVD) represents a diverse cluster of cerebrovascular diseases primarily affecting small arteries, capillaries, arterioles and venules. The diagnosis of CSVD relies on the identification of small subcortical infarcts, lacunes, white matter hyperintensities, perivascular spaces, and microbleeds using neuroimaging. CSVD is observed in 25% of strokes worldwide and is the most common pathology of cognitive decline and dementia in the elderly. Still, due to the poor understanding of pathophysiology in CSVD, there is not an effective preventative or therapeutic approach for CSVD. The most widely accepted approach to CSVD treatment is to mitigate vascular risk factors and adopt a healthier lifestyle. Thus, a deeper understanding of pathogenesis may foster more specific therapies. Here, we review the underlying mechanisms of pathological characteristics in CSVD development, with a focus on endothelial dysfunction, blood-brain barrier impairment and white matter change. We also describe inflammation in CSVD, whose role in contributing to CSVD pathology is gaining interest. Finally, we update the current treatments and preventative measures of CSVD, as well as discuss potential targets and novel strategies for CSVD treatment.
Collapse
Affiliation(s)
- Yue Gao
- Department of Neurointervention and Neurological Intensive Care, Dalian Municipal Central Hospital, Dalian, China
| | - Di Li
- Department of Neurointervention and Neurological Intensive Care, Dalian Municipal Central Hospital, Dalian, China
| | - Jianwen Lin
- Department of Neurology, Dalian Municipal Central Hospital, Dalian, China
| | - Aline M. Thomas
- F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institution, Baltimore, MD, United States
| | - Jianyu Miao
- Department of Neurology, Dalian Municipal Central Hospital, Dalian, China
| | - Dong Chen
- Department of Neurosurgery, Dalian Municipal Central Hospital, Dalian, China
| | - Shen Li
- Department of Neurology and Psychiatry, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Chengyan Chu
- Department of Neurology, Dalian Municipal Central Hospital, Dalian, China
- *Correspondence: Chengyan Chu,
| |
Collapse
|
49
|
Meng F, Yang Y, Jin G. Research Progress on MRI for White Matter Hyperintensity of Presumed Vascular Origin and Cognitive Impairment. Front Neurol 2022; 13:865920. [PMID: 35873763 PMCID: PMC9301233 DOI: 10.3389/fneur.2022.865920] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 06/14/2022] [Indexed: 11/17/2022] Open
Abstract
White matter hyperintensity of presumed vascular origin (WMH) is a common medical imaging manifestation in the brains of middle-aged and elderly individuals. WMH can lead to cognitive decline and an increased risk of cognitive impairment and dementia. However, the pathogenesis of cognitive impairment in patients with WMH remains unclear. WMH increases the risk of cognitive impairment, the nature and severity of which depend on lesion volume and location and the patient's cognitive reserve. Abnormal changes in microstructure, cerebral blood flow, metabolites, and resting brain function are observed in patients with WMH with cognitive impairment. Magnetic resonance imaging (MRI) is an indispensable tool for detecting WMH, and novel MRI techniques have emerged as the key approaches for exploring WMH and cognitive impairment. This article provides an overview of the association between WMH and cognitive impairment and the application of dynamic contrast-enhanced MRI, structural MRI, diffusion tensor imaging, 3D-arterial spin labeling, intravoxel incoherent motion, magnetic resonance spectroscopy, and resting-state functional MRI for examining WMH and cognitive impairment.
Collapse
Affiliation(s)
- Fanhua Meng
- North China University of Science and Technology, Tangshan, China
| | - Ying Yang
- Department of Radiology, China Emergency General Hospital, Beijing, China
| | - Guangwei Jin
- Department of Radiology, China Emergency General Hospital, Beijing, China
- *Correspondence: Guangwei Jin
| |
Collapse
|
50
|
Libecap TJ, Zachariou V, Bauer CE, Wilcock DM, Jicha GA, Raslau FD, Gold BT. Enlarged Perivascular Spaces Are Negatively Associated With Montreal Cognitive Assessment Scores in Older Adults. Front Neurol 2022; 13:888511. [PMID: 35847209 PMCID: PMC9283758 DOI: 10.3389/fneur.2022.888511] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 06/06/2022] [Indexed: 11/30/2022] Open
Abstract
Emerging evidence suggests that enlarged perivascular spaces (ePVS) may be a clinically significant neuroimaging marker of global cognitive function related to cerebral small vessel disease (cSVD). We tested this possibility by assessing the relationship between ePVS and both a standardized measure of global cognitive function, the Montreal Cognitive Assessment (MoCA), and an established marker of cSVD, white matter hyperintensity volume (WMH) volume. One hundred and eleven community-dwelling older adults (56-86) underwent neuroimaging and MoCA testing. Quantification of region-specific ePVS burden was performed using a previously validated visual rating method and WMH volumes were computed using the standard ADNI pipeline. Separate linear regression models were run with ePVS as a predictor of MoCA scores and whole brain WMH volume. Results indicated a negative association between MoCA scores and both total ePVS counts (P ≤ 0.001) and centrum semiovale ePVS counts (P ≤ 0.001), after controlling for other relevant cSVD variables. Further, WMH volumes were positively associated with total ePVS (P = 0.010), basal ganglia ePVS (P ≤ 0.001), and centrum semiovale ePVS (P = 0.027). Our results suggest that ePVS burden, particularly in the centrum semiovale, may be a clinically significant neuroimaging marker of global cognitive dysfunction related to cSVD.
Collapse
Affiliation(s)
- Timothy J. Libecap
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Valentinos Zachariou
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Christopher E. Bauer
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Donna M. Wilcock
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Gregory A. Jicha
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, United States
- Department of Neurology, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Flavius D. Raslau
- Department of Radiology, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Brian T. Gold
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, United States
- Magnetic Resonance Imaging and Spectroscopy Center, College of Medicine, University of Kentucky, Lexington, KY, United States
| |
Collapse
|