101
|
Vancheri F, Longo G, Vancheri S, Henein M. Coronary Microvascular Dysfunction. J Clin Med 2020; 9:E2880. [PMID: 32899944 PMCID: PMC7563453 DOI: 10.3390/jcm9092880] [Citation(s) in RCA: 174] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/02/2020] [Accepted: 09/02/2020] [Indexed: 01/09/2023] Open
Abstract
Many patients with chest pain undergoing coronary angiography do not show significant obstructive coronary lesions. A substantial proportion of these patients have abnormalities in the function and structure of coronary microcirculation due to endothelial and smooth muscle cell dysfunction. The coronary microcirculation has a fundamental role in the regulation of coronary blood flow in response to cardiac oxygen requirements. Impairment of this mechanism, defined as coronary microvascular dysfunction (CMD), carries an increased risk of adverse cardiovascular clinical outcomes. Coronary endothelial dysfunction accounts for approximately two-thirds of clinical conditions presenting with symptoms and signs of myocardial ischemia without obstructive coronary disease, termed "ischemia with non-obstructive coronary artery disease" (INOCA) and for a small proportion of "myocardial infarction with non-obstructive coronary artery disease" (MINOCA). More frequently, the clinical presentation of INOCA is microvascular angina due to CMD, while some patients present vasospastic angina due to epicardial spasm, and mixed epicardial and microvascular forms. CMD may be associated with focal and diffuse epicardial coronary atherosclerosis, which may reinforce each other. Both INOCA and MINOCA are more common in females. Clinical classification of CMD includes the association with conditions in which atherosclerosis has limited relevance, with non-obstructive atherosclerosis, and with obstructive atherosclerosis. Several studies already exist which support the evidence that CMD is part of systemic microvascular disease involving multiple organs, such as brain and kidney. Moreover, CMD is strongly associated with the development of heart failure with preserved ejection fraction (HFpEF), diabetes, hypertensive heart disease, and also chronic inflammatory and autoimmune diseases. Since coronary microcirculation is not visible on invasive angiography or computed tomographic coronary angiography (CTCA), the diagnosis of CMD is usually based on functional assessment of microcirculation, which can be performed by both invasive and non-invasive methods, including the assessment of delayed flow of contrast during angiography, measurement of coronary flow reserve (CFR) and index of microvascular resistance (IMR), evaluation of angina induced by intracoronary acetylcholine infusion, and assessment of myocardial perfusion by positron emission tomography (PET) and magnetic resonance (CMR).
Collapse
Affiliation(s)
- Federico Vancheri
- Department of Internal Medicine, S.Elia Hospital, 93100 Caltanissetta, Italy
| | - Giovanni Longo
- Cardiovascular and Interventional Department, S.Elia Hospital, 93100 Caltanissetta, Italy;
| | - Sergio Vancheri
- Radiology Department, I.R.C.C.S. Policlinico San Matteo, 27100 Pavia, Italy;
| | - Michael Henein
- Institute of Public Health and Clinical Medicine, Umea University, SE-90187 Umea, Sweden;
- Department of Fluid Mechanics, Brunel University, Middlesex, London UB8 3PH, UK
- Molecular and Nuclear Research Institute, St George’s University, London SW17 0RE, UK
| |
Collapse
|
102
|
Verheggen ICM, de Jong JJA, van Boxtel MPJ, Gronenschild EHBM, Palm WM, Postma AA, Jansen JFA, Verhey FRJ, Backes WH. Increase in blood-brain barrier leakage in healthy, older adults. GeroScience 2020; 42:1183-1193. [PMID: 32601792 PMCID: PMC7394987 DOI: 10.1007/s11357-020-00211-2] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/02/2020] [Indexed: 12/22/2022] Open
Abstract
Blood-brain barrier (BBB) breakdown can disrupt nutrient supply and waste removal, which affects neuronal functioning. Currently, dynamic contrast-enhanced (DCE) MRI is the preferred in-vivo method to quantify BBB leakage. Dedicated DCE MRI studies in normal aging individuals are lacking, which could hamper value estimation and interpretation of leakage rate in pathological conditions. Therefore, we applied DCE MRI to investigate the association between BBB disruption and age in a healthy sample. Fifty-seven cognitively and neurologically healthy, middle-aged to older participants (mean age: 66 years, range: 47-91 years) underwent MRI, including DCE MRI with intravenous injection of a gadolinium-based contrast agent. Pharmacokinetic modeling was applied to contrast concentration time-curves to estimate BBB leakage rate in each voxel. Subsequently, leakage rate was calculated in the white and gray matter, and primary (basic sensory and motor functions), secondary (association areas), and tertiary (higher-order cognition) brain regions. A difference in vulnerability to deterioration was expected between these regions, with especially tertiary regions being affected by age. Higher BBB leakage rate was significantly associated with older age in the white and gray matter, and also in tertiary, but not in primary or secondary brain regions. Even in healthy individuals, BBB disruption was stronger in older persons, which suggests BBB disruption is a normal physiologically aging phenomenon. Age-related increase in BBB disruption occurred especially in brain regions most vulnerable to age-related deterioration, which may indicate that BBB disruption is an underlying mechanism of normal age-related decline.Netherlands Trial Register number: NL6358, date of registration: 2017-03-24.
Collapse
Affiliation(s)
- Inge C M Verheggen
- Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.
- Alzheimer Center Limburg, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.
| | - Joost J A de Jong
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Martin P J van Boxtel
- Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- Alzheimer Center Limburg, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Ed H B M Gronenschild
- Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- Alzheimer Center Limburg, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Walter M Palm
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Alida A Postma
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Jacobus F A Jansen
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Frans R J Verhey
- Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- Alzheimer Center Limburg, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Walter H Backes
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
103
|
Kerkhofs D, van Hagen BT, Milanova IV, Schell KJ, van Essen H, Wijnands E, Goossens P, Blankesteijn WM, Unger T, Prickaerts J, Biessen EA, van Oostenbrugge RJ, Foulquier S. Pharmacological depletion of microglia and perivascular macrophages prevents Vascular Cognitive Impairment in Ang II-induced hypertension. Am J Cancer Res 2020; 10:9512-9527. [PMID: 32863942 PMCID: PMC7449902 DOI: 10.7150/thno.44394] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 06/12/2020] [Indexed: 12/19/2022] Open
Abstract
Rationale: Hypertension is a major risk factor for cerebral small vessel disease, the most prevalent cause of vascular cognitive impairment. As we have shown, hypertension induced by a prolonged Angiotensin II infusion is associated with increased permeability of the blood-brain barrier (BBB), chronic activation of microglia and myelin loss. In this study we therefore aim to determine the contribution of microglia to hypertension-induced cognitive impairment in an experimental hypertension model by a pharmacological depletion approach. Methods: For this study, adult Cx3Cr1gfp/wtxThy1yfp/0 reporter mice were infused for 12 weeks with Angiotensin II or saline and subgroups were treated with PLX5622, a highly selective CSF1R tyrosine kinase inhibitor. Systolic blood pressure (SBP) was measured via tail-cuff. Short- and long-term spatial memory was assessed during an Object Location task and a Morris Water Maze task (MWM). Microglia depletion efficacy was assessed by flow cytometry and immunohistochemistry. BBB leakages, microglia phenotype and myelin integrity were assessed by immunohistochemistry. Results: SBP, heart weight and carotid pulsatility were increased by Ang II and were not affected by PLX5622. Short-term memory was significantly impaired in Ang II hypertensive mice, and partly prevented in Ang II mice treated with PLX5622. Histological and flow cytometry analysis revealed almost complete ablation of microglia and a 60% depletion of brain resident perivascular macrophages upon CSF1R inhibition. Number and size of BBB leakages were increased in Ang II hypertensive mice, but not altered by PLX5622 treatment. Microglia acquired a pro-inflammatory phenotype at the site of BBB leakages in both Saline and Ang II mice and were successfully depleted by PLX5622. There was however no significant change in myelin integrity at the site of leakages. Conclusion: Our results show that depletion of microglia and PVMs, by CSF1R inhibition prevents short-term memory impairment in Ang II induced hypertensive mice. We suggest this beneficial effect is mediated by the major decrease of pro-inflammatory microglia within BBB leakages. This novel finding supports the critical role of brain immune cells in the pathogenesis of hypertension-related cognitive impairment. An adequate modulation of microglia /PVM density and phenotype may constitute a relevant approach to prevent and/or limit the progression of vascular cognitive impairment.
Collapse
|
104
|
Uchida Y, Kan H, Sakurai K, Arai N, Inui S, Kobayashi S, Kato D, Ueki Y, Matsukawa N. Iron leakage owing to blood–brain barrier disruption in small vessel disease CADASIL. Neurology 2020; 95:e1188-e1198. [DOI: 10.1212/wnl.0000000000010148] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 03/02/2020] [Indexed: 12/21/2022] Open
Abstract
ObjectiveTo assess the relationship among iron accumulation, blood–brain barrier (BBB) damage, and cognitive function in patients with cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL).MethodsWe enrolled 21 patients with NOTCH3 mutations and 21 age-matched healthy controls in this cross-sectional study. All participants underwent global physical and cognitive assessments and brain MRI using voxel-based quantitative susceptibility mapping (QSM; iron deposition measure) and dynamic contrast-enhanced MRI (BBB permeability measure). We compared behavioral and imaging data between the groups and analyzed the correlations in each group.ResultsAmong 21 NOTCH3 mutation carriers, 10 were symptomatic and 11 asymptomatic. Montreal Cognitive Assessment scores were significantly different among the groups (symptomatic < asymptomatic < control participants). Voxel-based QSM analysis revealed that the symptomatic group had higher QSM values than did the asymptomatic group in the putamen, caudate nucleus, temporal pole, and centrum semiovale. These QSM values were positively correlated with regional BBB permeabilities (putamen: r = 0.57, p = 0.006; caudate nucleus: r = 0.51, p = 0.019; temporal pole: r = 0.48, p = 0.030; centrum semiovale: r = 0.45, p = 0.044) and negatively correlated with Montreal Cognitive Assessment scores (caudate nucleus: r = −0.53, p = 0.012; temporal pole: r = −0.56, p = 0.008).ConclusionsThis study showed that cerebral iron burden was associated with regional BBB permeability and cognitive dysfunction in patients with CADASIL, highlighting the potential of these imaging techniques as auxiliary biomarkers to monitor the course of small vessel disease.
Collapse
|
105
|
Manukjan N, Ahmed Z, Fulton D, Blankesteijn WM, Foulquier S. A Systematic Review of WNT Signaling in Endothelial Cell Oligodendrocyte Interactions: Potential Relevance to Cerebral Small Vessel Disease. Cells 2020; 9:cells9061545. [PMID: 32630426 PMCID: PMC7349551 DOI: 10.3390/cells9061545] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 12/21/2022] Open
Abstract
Key pathological features of cerebral small vessel disease (cSVD) include impairment of the blood brain barrier (BBB) and the progression of white matter lesions (WMLs) amongst other structural lesions, leading to the clinical manifestations of cSVD. The function of endothelial cells (ECs) is of major importance to maintain a proper BBB. ECs interact with several cell types to provide structural and functional support to the brain. Oligodendrocytes (OLs) myelinate axons in the central nervous system and are crucial in sustaining the integrity of white matter. The interplay between ECs and OLs and their precursor cells (OPCs) has received limited attention yet seems of relevance for the study of BBB dysfunction and white matter injury in cSVD. Emerging evidence shows a crosstalk between ECs and OPCs/OLs, mediated by signaling through the Wingless and Int-1 (WNT)/β-catenin pathway. As the latter is involved in EC function (e.g., angiogenesis) and oligodendrogenesis, we reviewed the role of WNT/β-catenin signaling for both cell types and performed a systematic search to identify studies describing a WNT-mediated interplay between ECs and OPCs/OLs. Dysregulation of this interaction may limit remyelination of WMLs and render the BBB leaky, thereby initiating a vicious neuroinflammatory cycle. A better understanding of the role of this signaling pathway in EC-OL crosstalk is essential in understanding cSVD development.
Collapse
Affiliation(s)
- Narek Manukjan
- Department of Pharmacology and Toxicology, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands; or (W.M.B.)
- CARIM—School for Cardiovascular Diseases, Maastricht University Medical Center+, PO Box 616, 6200 MD Maastricht, The Netherlands
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK; (Z.A.); (D.F.)
| | - Zubair Ahmed
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK; (Z.A.); (D.F.)
| | - Daniel Fulton
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, UK; (Z.A.); (D.F.)
| | - W. Matthijs Blankesteijn
- Department of Pharmacology and Toxicology, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands; or (W.M.B.)
- CARIM—School for Cardiovascular Diseases, Maastricht University Medical Center+, PO Box 616, 6200 MD Maastricht, The Netherlands
| | - Sébastien Foulquier
- Department of Pharmacology and Toxicology, Maastricht University, PO Box 616, 6200 MD Maastricht, The Netherlands; or (W.M.B.)
- CARIM—School for Cardiovascular Diseases, Maastricht University Medical Center+, PO Box 616, 6200 MD Maastricht, The Netherlands
- Department of Neurology, MHeNs—School for Mental Health and Neuroscience, Maastricht University Medical Center+, PO Box 616, 6200 MD Maastricht, The Netherlands
- Correspondence: ; Tel.: +31-43-3881409
| |
Collapse
|
106
|
The role of semaphorins in small vessels of the eye and brain. Pharmacol Res 2020; 160:105044. [PMID: 32590102 DOI: 10.1016/j.phrs.2020.105044] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 06/19/2020] [Accepted: 06/19/2020] [Indexed: 12/20/2022]
Abstract
Small vessel diseases, such as ischemic retinopathy and cerebral small vessel disease (CSVD), are increasingly recognized in patients with diabetes, dementia and cerebrovascular disease. The mechanisms of small vessel diseases are poorly understood, but the latest studies suggest a role for semaphorins. Initially identified as axon guidance cues, semaphorins are mainly studied in neuronal morphogenesis, neural circuit assembly, and synapse assembly and refinement. In recent years, semaphorins have been found to play important roles in regulating vascular growth and development and in many pathophysiological processes, including atherosclerosis, angiogenesis after stroke and retinopathy. Growing evidence indicates that semaphorins affect the occurrence, perfusion and regression of both the macrovasculature and microvasculature by regulating the proliferation, apoptosis, migration, barrier function and inflammatory response of endothelial cells, vascular smooth muscle cells (VSMCs) and pericytes. In this review, we concentrate on the regulatory effects of semaphorins on the cell components of the vessel wall and their potential roles in microvascular diseases, especially in the retina and cerebral small vessel. Finally, we discuss potential molecular approaches in targeting semaphorins as therapies for microvascular disorders in the eye and brain.
Collapse
|
107
|
Yu C, Lu W, Qiu J, Wang F, Li J, Wang L. Alterations of the Whole Cerebral Blood Flow in Patients With Different Total Cerebral Small Vessel Disease Burden. Front Aging Neurosci 2020; 12:175. [PMID: 32655393 PMCID: PMC7324936 DOI: 10.3389/fnagi.2020.00175] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 05/19/2020] [Indexed: 12/18/2022] Open
Abstract
Background Cerebral small vessel disease (CSVD) is a common age-related vascular disease of the brain associated with slowly accumulating tissue damage. At present, total CSVD burden score is a commonly used method to evaluate the severity of the disease. Purpose To observe whether global and regional cerebral perfusion is related to total CSVD score and to explore global and regional cerebral blood flow (CBF) changes in patients with different degrees of CSVD. Methods We collected 130 subjects with different total burden score of CSVD (0 point: 33 subjects, 1 point: 39 subjects, 2 points: 24 subjects, 3 points: 24 subjects, 4 points: 10 subjects). Total CSVD burden score was evaluated by clinically routine sequences (T2WI, T2-FLAIR, T1WI, DWI, and SWAN sequence). Global and regional CBF were calculated and correlation analysis was used to investigate the relationship between total CSVD score and CBF of the whole brain and several brain regions. Results The analysis results showed that there was a negative correlation between total CSVD burden score and global CBF (r = −0.33, p = 0.001). Total CSVD burden score also had moderately negative correlations with CBF of almost all the brain regions. Conclusion CSVD is a disease that affects the whole brain. With the increase of total CSVD burden score, the global and regional CBF decreased. The CSVD total burden score could be used to evaluate the overall condition of brain perfusion.
Collapse
Affiliation(s)
- Chunyan Yu
- Department of Radiology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, China
| | - Weizhao Lu
- Department of Radiology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, China
| | - Jianfeng Qiu
- Department of Radiology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, China
| | - Feng Wang
- The Second Affiliated Hospital of Shandong First Medical University, Tai'an, China
| | | | - Liru Wang
- Department of Radiology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, China
| |
Collapse
|
108
|
Abrahamson EE, Ikonomovic MD. Brain injury-induced dysfunction of the blood brain barrier as a risk for dementia. Exp Neurol 2020; 328:113257. [PMID: 32092298 DOI: 10.1016/j.expneurol.2020.113257] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/31/2020] [Accepted: 02/20/2020] [Indexed: 02/06/2023]
Abstract
The blood-brain barrier (BBB) is a complex and dynamic physiological interface between brain parenchyma and cerebral vasculature. It is composed of closely interacting cells and signaling molecules that regulate movement of solutes, ions, nutrients, macromolecules, and immune cells into the brain and removal of products of normal and abnormal brain cell metabolism. Dysfunction of multiple components of the BBB occurs in aging, inflammatory diseases, traumatic brain injury (TBI, severe or mild repetitive), and in chronic degenerative dementing disorders for which aging, inflammation, and TBI are considered risk factors. BBB permeability changes after TBI result in leakage of serum proteins, influx of immune cells, perivascular inflammation, as well as impairment of efflux transporter systems and accumulation of aggregation-prone molecules involved in hallmark pathologies of neurodegenerative diseases with dementia. In addition, cerebral vascular dysfunction with persistent alterations in cerebral blood flow and neurovascular coupling contribute to brain ischemia, neuronal degeneration, and synaptic dysfunction. While the idea of TBI as a risk factor for dementia is supported by many shared pathological features, it remains a hypothesis that needs further testing in experimental models and in human studies. The current review focusses on pathological mechanisms shared between TBI and neurodegenerative disorders characterized by accumulation of pathological protein aggregates, such as Alzheimer's disease and chronic traumatic encephalopathy. We discuss critical knowledge gaps in the field that need to be explored to clarify the relationship between TBI and risk for dementia and emphasize the need for longitudinal in vivo studies using imaging and biomarkers of BBB dysfunction in people with single or multiple TBI.
Collapse
Affiliation(s)
- Eric E Abrahamson
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Milos D Ikonomovic
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, University of Pittsburgh, Pittsburgh, PA, United States; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, United States; Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
109
|
Decreased visible deep medullary veins is a novel imaging marker for cerebral small vessel disease. Neurol Sci 2020; 41:1497-1506. [PMID: 31955350 DOI: 10.1007/s10072-019-04203-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 12/16/2019] [Indexed: 12/15/2022]
Abstract
PURPOSE Visibility of deep medullary veins (DMVs) seen at SWI is predictive of poor prognosis in ischemic stroke. Few attentions have been paid to DMVs in atherosclerotic cerebral small vessel disease (aCSVD) which is attributed to long-term imbalanced microhemodynamics. We conducted this retrospective study to explore the association between DMVs profiles and aCSVD risk factors, neuroimaging markers. METHODS Two hundred and two patients identified as aCSVD from January 2017 to March 2019 were included in the study. Their demographic, clinical, laboratory, and neuroimaging data were reviewed. The quantity and morphology of DMVs were assessed with a 5-grade (range 0~4) visual rating scale. Total CSVD burden was calculated with an ordinal "SVD score" (range 0~4). Spearman rank correlation and multivariable logistic regression analysis were performed to determine the association between DMV scale and CSVD markers. RESULTS DMV scale showed strong positive correlation with CSVD burden (rs = 0.629, P < 0.001). Age (OR 1.078, 95% CI 1.015-1.145, P = 0.015) and hypertension (OR 2.629, 95% CI 1.024-6.749, P = 0.045) were two demographic risk factors for high DMV scale. Among CSVD neuroimaging markers, periventricular WMH (OR 2.925, 95% CI 1.464-5.845, P = 0.002), deep WMH (OR 2.872, 95% CI 1.174-7.022, P = 0.021), lacunae (OR 1.961, 95% CI 1.181-3.254, P = 0.009), and cerebral atrophy (OR 2.046, 95% CI 1.079-3.880, P = 0.028) were associated with high DMV scale after adjusting for clinical and metabolic confounders. CONCLUSION Multifactorial association between DMV scale and epidemiological, radiological contributors of aCSVD suggests DMV's involved pathomechanism may participate in aCSVD development. Attach importance to DMV radiological profile in aCSVD will provide more neuroimaging information for diagnosis and prognosis.
Collapse
|
110
|
Li CC, Chen WX, Wang J, Xia M, Jia ZC, Guo C, Tang XQ, Li MX, Yin Y, Liu X, Feng H. Nicotinamide riboside rescues angiotensin II-induced cerebral small vessel disease in mice. CNS Neurosci Ther 2020; 26:438-447. [PMID: 31943833 PMCID: PMC7080427 DOI: 10.1111/cns.13276] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/08/2019] [Accepted: 11/23/2019] [Indexed: 12/20/2022] Open
Abstract
Aims Hypertension is a leading cause of cerebral small vessel disease (CSVD). Currently, treatments for CSVD are limited. Nicotinamide riboside (NR) can protect against vascular injury and cognitive impairment in neurodegenerative diseases. In this study, the protective effects of NR against angiotensin ‐ (Ang ‐)–induced CSVD were evaluated. Methods To explore the effects of NR in CSVD, C57BL/6 mice were infused with Ang ‐, and NR was added to the food of the mice for 28 days. Then, short‐term memory, blood‐brain barrier (BBB) integrity, and endothelial function were detected. Arteriole injury and glial activation were also evaluated. Results Our data showed that mice infused with Ang ‐ exhibited decreased short‐term memory function and BBB leakage due to decreased claudin‐5 expression and increased caveolae‐mediated endocytosis after 28 days. Furthermore, Ang ‐ decreased the expression of α‐smooth muscle actin (α‐SMA) and increased the expression of proliferating cell nuclear antigen (PCNA) in arterioles and decreased the expression of neurofilament 200 (NF200) and myelin basic protein (MBP) in the white matter. These CSVD‐related damages induced by Ang ‐ were inhibited by NR administration. Moreover, NR administration significantly reduced glial activation around the vessels. Conclusion Our results indicated that NR administration alleviated Ang ‐–induced CSVD by protecting BBB integrity, vascular remodeling, neuroinflammation, and white matter injury (WMI)–associated cognitive impairment.
Collapse
Affiliation(s)
- Cheng-Cheng Li
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Wei-Xiang Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jie Wang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Min Xia
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zheng-Cai Jia
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Chao Guo
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiao-Qin Tang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Ming-Xi Li
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yi Yin
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xin Liu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Department of Neurosurgery Southwest Hospital, Collaborative Innovation Center for Brain Science, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
111
|
Moon WJ. Alzheimer Dementia and Microvascular Pathology: Blood-Brain Barrier Permeability Imaging. JOURNAL OF THE KOREAN SOCIETY OF RADIOLOGY 2020; 81:488-500. [PMID: 36238621 PMCID: PMC9431923 DOI: 10.3348/jksr.2020.81.3.488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/14/2020] [Accepted: 04/17/2020] [Indexed: 11/15/2022]
Affiliation(s)
- Won-Jin Moon
- Department of Radiology, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Korea
| |
Collapse
|
112
|
Marini S, Anderson CD, Rosand J. Genetics of Cerebral Small Vessel Disease. Stroke 2020; 51:12-20. [PMID: 31752611 PMCID: PMC7337039 DOI: 10.1161/strokeaha.119.024151] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 08/20/2019] [Indexed: 02/07/2023]
Affiliation(s)
- Sandro Marini
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
- Henry and Allison McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| | - Christopher D. Anderson
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
- Henry and Allison McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| | - Jonathan Rosand
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
- Henry and Allison McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
113
|
Eide PK, Hansson HA. Blood-brain barrier leakage of blood proteins in idiopathic normal pressure hydrocephalus. Brain Res 2020; 1727:146547. [DOI: 10.1016/j.brainres.2019.146547] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 10/14/2019] [Accepted: 11/07/2019] [Indexed: 01/05/2023]
|
114
|
Freeze WM, Jacobs HI, de Jong JJ, Verheggen IC, Gronenschild EH, Palm WM, Hoff EI, Wardlaw JM, Jansen JF, Verhey FR, Backes WH. White matter hyperintensities mediate the association between blood-brain barrier leakage and information processing speed. Neurobiol Aging 2020; 85:113-122. [DOI: 10.1016/j.neurobiolaging.2019.09.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 08/27/2019] [Accepted: 09/22/2019] [Indexed: 12/22/2022]
|
115
|
Chokesuwattanaskul A, Lertjitbanjong P, Thongprayoon C, Bathini T, Sharma K, Mao MA, Cheungpasitporn W, Chokesuwattanaskul R. Impact of obstructive sleep apnea on silent cerebral small vessel disease: a systematic review and meta-analysis. Sleep Med 2019; 68:80-88. [PMID: 32028230 DOI: 10.1016/j.sleep.2019.11.1262] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/28/2019] [Accepted: 11/27/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND Cerebral small vessel disease (CSVD) is a well-known cause of vascular dementia, a leading medical morbidity in the aging population. Obstructive sleep apnea (OSA) has been validated as a cardiovascular risk factor. However, the relationship between these two clinical syndromes is not well established. We aimed to assess the association between OSA and CSVD. METHODS Databases were searched from inception through May 2019. Studies that reported incidence or odd ratios of CSVD in patients with OSA were included. Effect estimates from the individual studies were extracted and combined using random-effect, generic inverse variance method of DerSimonian and Laird. RESULTS A total of 14 observational studies comprising of 4335 patients were included into the analysis. Compared to patients without OSA, patients with OSA were significantly associated with CSVD magnetic resonance imaging (MRI) findings of white matter hyperintensity (WMH) and asymptomatic lacunar infarction (ALI) with a pooled OR of 2.31 (95% confidence interval [CI], 1.46-3.66, I2 = 79%) and 1.78 (95% CI, 1.06-3.01, I2 = 41%), respectively. However, there was no significant association between OSA and findings of cerebral microbleeds (CMBs), with a pooled odds ratio (OR) of 2.15 (95% CI, 0.64-7.29, I2 = 55%). CONCLUSIONS Our study demonstrated the association between OSA and CSVD MRI findings of white matter hyperintensity (WMH) and asymptomatic lacunar infarction (ALI) when compared to patients without OSA. The absence of an association of CMBs findings with OSA could be due either by a lower sensitivity of neuroimaging techniques utilized to detect CMBs or a potentially different pathogenesis of CMBs.
Collapse
Affiliation(s)
- Anthipa Chokesuwattanaskul
- Division of Neurology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand; King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand.
| | | | | | - Tarun Bathini
- Department of Internal Medicine, University of Arizona, Tucson, AZ, USA
| | - Konika Sharma
- Department of Internal Medicine, Bassett Medical Center, Cooperstown, NY, USA
| | - Michael A Mao
- Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, 32224, USA
| | | | - Ronpichai Chokesuwattanaskul
- King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand; Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| |
Collapse
|
116
|
Zhang CE, Staals J, van Oostenbrugge RJ, Vink H. Uncoupling of Microvascular Blood Flow and Capillary Density in Vascular Cognitive Impairment. Front Neurol 2019; 10:1268. [PMID: 31849826 PMCID: PMC6901497 DOI: 10.3389/fneur.2019.01268] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 11/15/2019] [Indexed: 12/14/2022] Open
Abstract
Cerebral small vessel disease (cSVD) plays an important role in dementia and is a major cause for vascular cognitive impairment (VCI). Recent studies hypothesized that capillary dysfunction including reduction of capillary patency, rather than a flow-limiting pathology is crucial in cSVD. As cSVD is considered a systemic microvascular disease, we examined sublingual microvascular blood flow and capillary density in patients with VCI and controls. Fifteen patients with VCI due to cSVD and 15 controls underwent intravital microscopy of the sublingual microvessels. Microvascular blood flow and capillary density in high and low flow areas were determined for each participant. Flow-density coupling was examined by determining the ratio of density changes to flow changes, and the ratio of feed vessel red blood cell (RBC) velocity to capillary RBC velocity. These were compared between VCI and controls. In healthy controls, capillary density increased proportionally with feed vessel blood flow increase. In patients with VCI, no increase of capillary density was observed. Moreover, increase of feed vessel RBC velocity led to significant increase of capillary RBC velocity in VCI, whereas in controls, the capillary RBC increased only slightly. Flow-density coupling differed significantly between VCI and controls, also after correcting for age and hypertension. Our findings suggest uncoupling of microvascular blood flow and capillary density in patients with VCI. This uncoupling may impair oxygen and nutrients exchange when blood flow increases in response to increased metabolic demand, ultimately leading to tissue damage.
Collapse
Affiliation(s)
- Chenxing Eleana Zhang
- Department of Neurology, Maastricht University Medical Center, Maastricht, Netherlands.,CARIM and MHeNs, Maastricht University Medical Center, Maastricht, Netherlands
| | - Julie Staals
- Department of Neurology, Maastricht University Medical Center, Maastricht, Netherlands.,CARIM and MHeNs, Maastricht University Medical Center, Maastricht, Netherlands
| | - Robert Jan van Oostenbrugge
- Department of Neurology, Maastricht University Medical Center, Maastricht, Netherlands.,CARIM and MHeNs, Maastricht University Medical Center, Maastricht, Netherlands
| | - Hans Vink
- CARIM and MHeNs, Maastricht University Medical Center, Maastricht, Netherlands.,Department of Physiology, Maastricht University Medical Center, Maastricht, Netherlands
| |
Collapse
|
117
|
Weaver NA, Zhao L, Biesbroek JM, Kuijf HJ, Aben HP, Bae HJ, Caballero MÁ, Chappell FM, Chen CP, Dichgans M, Duering M, Georgakis MK, van der Giessen RS, Gyanwali B, Hamilton OK, Hilal S, vom Hofe EM, de Kort PL, Koudstaal PJ, Lam BY, Lim JS, Makin SD, Mok VC, Shi L, Valdés Hernández MC, Venketasubramanian N, Wardlaw JM, Wollenweber FA, Wong A, Xin X, Biessels GJ. The Meta VCI Map consortium for meta-analyses on strategic lesion locations for vascular cognitive impairment using lesion-symptom mapping: Design and multicenter pilot study. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2019; 11:310-326. [PMID: 31011619 PMCID: PMC6465616 DOI: 10.1016/j.dadm.2019.02.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION The Meta VCI Map consortium performs meta-analyses on strategic lesion locations for vascular cognitive impairment using lesion-symptom mapping. Integration of data from different cohorts will increase sample sizes, to improve brain lesion coverage and support comprehensive lesion-symptom mapping studies. METHODS Cohorts with available imaging on white matter hyperintensities or infarcts and cognitive testing were invited. We performed a pilot study to test the feasibility of multicenter data processing and analysis and determine the benefits to lesion coverage. RESULTS Forty-seven groups have joined Meta VCI Map (stroke n = 7800 patients; memory clinic n = 4900; population-based n = 14,400). The pilot study (six ischemic stroke cohorts, n = 878) demonstrated feasibility of multicenter data integration (computed tomography/magnetic resonance imaging) and achieved marked improvement of lesion coverage. DISCUSSION Meta VCI Map will provide new insights into the relevance of vascular lesion location for cognitive dysfunction. After the successful pilot study, further projects are being prepared. Other investigators are welcome to join.
Collapse
Affiliation(s)
- Nick A. Weaver
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Lei Zhao
- BrainNow Medical Technology Limited, Hong Kong Science and Technology Park, Shatin, Hong Kong SAR, China
| | - J. Matthijs Biesbroek
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Hugo J. Kuijf
- Image Sciences Institute, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Hugo P. Aben
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Neurology, Elisabeth Tweesteden Hospital, Tilburg, the Netherlands
| | - Hee-Joon Bae
- Department of Neurology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Miguel Á.A. Caballero
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Francesca M. Chappell
- Neuroimaging Sciences, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, United Kingdom
| | - Christopher P.L.H. Chen
- Department of Pharmacology, National University of Singapore, Singapore, Singapore
- Memory, Aging and Cognition Center, National University Health System, Singapore, Singapore
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Marco Duering
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Marios K. Georgakis
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | | | - Bibek Gyanwali
- Department of Pharmacology, National University of Singapore, Singapore, Singapore
- Memory, Aging and Cognition Center, National University Health System, Singapore, Singapore
| | - Olivia K.L. Hamilton
- Neuroimaging Sciences, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, United Kingdom
| | - Saima Hilal
- Department of Pharmacology, National University of Singapore, Singapore, Singapore
- Memory, Aging and Cognition Center, National University Health System, Singapore, Singapore
- Departments of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, the Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Elise M. vom Hofe
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Paul L.M. de Kort
- Department of Neurology, Elisabeth Tweesteden Hospital, Tilburg, the Netherlands
| | - Peter J. Koudstaal
- Department of Neurology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Bonnie Y.K. Lam
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Gerald Choa Neuroscience Centre, Therese Pei Fong Chow Research Centre for Prevention of Dementia, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Jae-Sung Lim
- Department of Neurology, Hallym University Sacred Heart Hospital, Anyang, Republic of Korea
| | - Stephen D.J. Makin
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Vincent C.T. Mok
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Gerald Choa Neuroscience Centre, Therese Pei Fong Chow Research Centre for Prevention of Dementia, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Lin Shi
- BrainNow Medical Technology Limited, Hong Kong Science and Technology Park, Shatin, Hong Kong SAR, China
- Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Maria C. Valdés Hernández
- Neuroimaging Sciences, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, United Kingdom
| | | | - Joanna M. Wardlaw
- Neuroimaging Sciences, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, United Kingdom
| | - Frank A. Wollenweber
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Adrian Wong
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Gerald Choa Neuroscience Centre, Therese Pei Fong Chow Research Centre for Prevention of Dementia, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Xu Xin
- Department of Pharmacology, National University of Singapore, Singapore, Singapore
- Memory, Aging and Cognition Center, National University Health System, Singapore, Singapore
| | - Geert Jan Biessels
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
118
|
Smith EE, Biessels GJ, De Guio F, de Leeuw FE, Duchesne S, Düring M, Frayne R, Ikram MA, Jouvent E, MacIntosh BJ, Thrippleton MJ, Vernooij MW, Adams H, Backes WH, Ballerini L, Black SE, Chen C, Corriveau R, DeCarli C, Greenberg SM, Gurol ME, Ingrisch M, Job D, Lam BY, Launer LJ, Linn J, McCreary CR, Mok VC, Pantoni L, Pike GB, Ramirez J, Reijmer YD, Romero JR, Ropele S, Rost NS, Sachdev PS, Scott CJ, Seshadri S, Sharma M, Sourbron S, Steketee RM, Swartz RH, van Oostenbrugge R, van Osch M, van Rooden S, Viswanathan A, Werring D, Dichgans M, Wardlaw JM. Harmonizing brain magnetic resonance imaging methods for vascular contributions to neurodegeneration. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2019; 11:191-204. [PMID: 30859119 PMCID: PMC6396326 DOI: 10.1016/j.dadm.2019.01.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Many consequences of cerebrovascular disease are identifiable by magnetic resonance imaging (MRI), but variation in methods limits multicenter studies and pooling of data. The European Union Joint Program on Neurodegenerative Diseases (EU JPND) funded the HARmoNizing Brain Imaging MEthodS for VaScular Contributions to Neurodegeneration (HARNESS) initiative, with a focus on cerebral small vessel disease. METHODS Surveys, teleconferences, and an in-person workshop were used to identify gaps in knowledge and to develop tools for harmonizing imaging and analysis. RESULTS A framework for neuroimaging biomarker development was developed based on validating repeatability and reproducibility, biological principles, and feasibility of implementation. The status of current MRI biomarkers was reviewed. A website was created at www.harness-neuroimaging.org with acquisition protocols, a software database, rating scales and case report forms, and a deidentified MRI repository. CONCLUSIONS The HARNESS initiative provides resources to reduce variability in measurement in MRI studies of cerebral small vessel disease.
Collapse
Affiliation(s)
- Eric E. Smith
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Department of Radiology, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Alberta, Canada
| | - Geert Jan Biessels
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands
| | - François De Guio
- Department of Neurology, Lariboisière Hospital, University Paris Diderot, Paris, France
| | - Frank Erik de Leeuw
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Donders Center for Medical Neuroscience, Radboud University Medical Center, Nijmegen, Netherlands
| | - Simon Duchesne
- CERVO Research Center, Quebec Mental Health Institute, Québec, Canada
- Radiology Department, Université Laval, Québec, Canada
| | - Marco Düring
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-Universität LMU, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE, Munich), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Richard Frayne
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Department of Radiology, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Alberta, Canada
- Seaman Family MR Centre, Foothills Medical Centre, Calgary, Alberta, Canada
| | - M. Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Eric Jouvent
- Department of Neurology, Lariboisière Hospital, University Paris Diderot, Paris, France
| | - Bradley J. MacIntosh
- Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Department of Medical Biophysics, Sunnybrook Research Institute, University of Toronto, Ontario, Canada
| | - Michael J. Thrippleton
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Meike W. Vernooij
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Hieab Adams
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Walter H. Backes
- Department of Radiology & Nuclear Medicine, School for Mental Health & Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Lucia Ballerini
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Sandra E. Black
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
- Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Toronto, Ontario, Canada
- Department of Medicine (Neurology), Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Christopher Chen
- Memory Aging and Cognition Centre, Department of Pharmacology, National University of Singapore, Singapore
| | - Rod Corriveau
- National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Charles DeCarli
- Department of Neurology and Center for Neuroscience, University of California at Davis, Davis, CA, USA
| | - Steven M. Greenberg
- J. Philip Kistler Stroke Research Center, Stroke Service and Memory Disorders Unit, Massachusetts General Hospital, Boston, MA, USA
| | - M. Edip Gurol
- J. Philip Kistler Stroke Research Center, Stroke Service and Memory Disorders Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Michael Ingrisch
- Department of Radiology, Ludwig-Maximilians-University Hospital Munich, Munich, Germany
| | - Dominic Job
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Bonnie Y.K. Lam
- Therese Pei Fong Chow Research Centre for Prevention of Dementia, Gerald Choa Neuroscience Centre, Lui Che Woo Institute of Innovative Medicine, Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Lenore J. Launer
- National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Jennifer Linn
- Institute of Neuroradiology, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Cheryl R. McCreary
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Department of Radiology, University of Calgary, Calgary, Alberta, Canada
- Seaman Family MR Centre, Foothills Medical Centre, Calgary, Alberta, Canada
| | - Vincent C.T. Mok
- Therese Pei Fong Chow Research Centre for Prevention of Dementia, Gerald Choa Neuroscience Centre, Lui Che Woo Institute of Innovative Medicine, Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Leonardo Pantoni
- Luigi Sacco Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - G. Bruce Pike
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
- Department of Radiology, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Alberta, Canada
| | - Joel Ramirez
- Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Department of Medical Biophysics, Sunnybrook Research Institute, University of Toronto, Ontario, Canada
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Yael D. Reijmer
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands
| | - Jose Rafael Romero
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Framingham Heart Study, Framingham, MA, USA
| | - Stefan Ropele
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Natalia S. Rost
- J. Philip Kistler Stroke Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Perminder S. Sachdev
- Centre for Healthy Brain Ageing, University of New South Wales, Sydney, Australia
| | - Christopher J.M. Scott
- Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Department of Medical Biophysics, Sunnybrook Research Institute, University of Toronto, Ontario, Canada
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| | - Mukul Sharma
- Population Health Research Institute, Hamilton, Ontario, Canada
- Department of Medicine (Neurology) McMaster University, Hamilton, Ontario, Canada
| | - Steven Sourbron
- Imaging Biomarkers Group, Department of Biomedical Imaging Sciences, University of Leeds, Leeds, UK
| | - Rebecca M.E. Steketee
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Richard H. Swartz
- Department of Medicine (Neurology), University of Toronto, Toronto, Canada
- Hurvitz Brain Sciences Program, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - Robert van Oostenbrugge
- Department of Neurology, School for Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Matthias van Osch
- C.J. Gorter Center for high field MRI, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Sanneke van Rooden
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Anand Viswanathan
- J. Philip Kistler Stroke Research Center, Stroke Service and Memory Disorders Unit, Massachusetts General Hospital, Boston, MA, USA
| | - David Werring
- University College London Queen Square institute of Neurology, London, UK
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilians-Universität LMU, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE, Munich), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Joanna M. Wardlaw
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
119
|
Mustapha M, Nassir CMNCM, Aminuddin N, Safri AA, Ghazali MM. Cerebral Small Vessel Disease (CSVD) - Lessons From the Animal Models. Front Physiol 2019; 10:1317. [PMID: 31708793 PMCID: PMC6822570 DOI: 10.3389/fphys.2019.01317] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 09/30/2019] [Indexed: 12/28/2022] Open
Abstract
Cerebral small vessel disease (CSVD) refers to a spectrum of clinical and imaging findings resulting from pathological processes of various etiologies affecting cerebral arterioles, perforating arteries, capillaries, and venules. Unlike large vessels, it is a challenge to visualize small vessels in vivo, hence the difficulty to directly monitor the natural progression of the disease. CSVD might progress for many years during the early stage of the disease as it remains asymptomatic. Prevalent among elderly individuals, CSVD has been alarmingly reported as an important precursor of full-blown stroke and vascular dementia. Growing evidence has also shown a significant association between CSVD's radiological manifestation with dementia and Alzheimer's disease (AD) pathology. Although it remains contentious as to whether CSVD is a cause or sequelae of AD, it is not far-fetched to posit that effective therapeutic measures of CSVD would mitigate the overall burden of dementia. Nevertheless, the unifying theory on the pathomechanism of the disease remains elusive, hence the lack of effective therapeutic approaches. Thus, this chapter consolidates the contemporary insights from numerous experimental animal models of CSVD, to date: from the available experimental animal models of CSVD and its translational research value; the pathomechanical aspects of the disease; relevant aspects on systems biology; opportunities for early disease biomarkers; and finally, converging approaches for future therapeutic directions of CSVD.
Collapse
Affiliation(s)
- Muzaimi Mustapha
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | | | - Niferiti Aminuddin
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
- Department of Basic Medical Sciences, Kulliyyah of Pharmacy, International Islamic University Malaysia, Kuantan, Malaysia
| | - Amanina Ahmad Safri
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Mazira Mohamad Ghazali
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| |
Collapse
|
120
|
Schreiber S, Wilisch-Neumann A, Schreiber F, Assmann A, Scheumann V, Perosa V, Jandke S, Mawrin C, Carare RO, Werring DJ. Invited Review: The spectrum of age-related small vessel diseases: potential overlap and interactions of amyloid and nonamyloid vasculopathies. Neuropathol Appl Neurobiol 2019; 46:219-239. [PMID: 31386773 DOI: 10.1111/nan.12576] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 07/10/2019] [Accepted: 07/11/2019] [Indexed: 12/12/2022]
Abstract
Deep perforator arteriopathy (DPA) and cerebral amyloid angiopathy (CAA) are the commonest known cerebral small vessel diseases (CSVD), which cause ischaemic stroke, intracebral haemorrhage (ICH) and vascular cognitive impairment (VCI). While thus far mainly considered as separate entities, we here propose that DPA and CAA share similarities, overlap and interact, so that 'pure' DPA or CAA are extremes along a continuum of age-related small vessel pathologies. We suggest blood-brain barrier (BBB) breakdown, endothelial damage and impaired perivascular β-amyloid (Aβ) drainage are hallmark common mechanisms connecting DPA and CAA. We also suggest a need for new biomarkers (e.g. high-resolution imaging) to deepen understanding of the complex relationships between DPA and CAA.
Collapse
Affiliation(s)
- S Schreiber
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany.,German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, Magdeburg, Germany.,Center for behavioral brain sciences (CBBS), Magdeburg, Germany
| | - A Wilisch-Neumann
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany.,German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, Magdeburg, Germany
| | - F Schreiber
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany.,German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, Magdeburg, Germany
| | - A Assmann
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany.,German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, Magdeburg, Germany
| | - V Scheumann
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany
| | - V Perosa
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany.,German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, Magdeburg, Germany
| | - S Jandke
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany.,German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, Magdeburg, Germany
| | - C Mawrin
- Department of Neuropathology, Otto-von-Guericke University, Magdeburg, Germany
| | - R O Carare
- Faculty of Medicine, University of Southampton, Southampton, UK
| | - D J Werring
- Stroke Research Centre, Department of Brain Repair & Rehabilitation, UCL Institute of Neurology, The National Hospital for Neurology and Neurosurgery, London, UK
| |
Collapse
|
121
|
Wong SM, Backes WH, Drenthen GS, Zhang CE, Voorter PHM, Staals J, van Oostenbrugge RJ, Jansen JFA. Spectral Diffusion Analysis of Intravoxel Incoherent Motion MRI in Cerebral Small Vessel Disease. J Magn Reson Imaging 2019; 51:1170-1180. [PMID: 31486211 PMCID: PMC7078988 DOI: 10.1002/jmri.26920] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 08/15/2019] [Accepted: 08/15/2019] [Indexed: 01/22/2023] Open
Abstract
Background Cerebral intravoxel incoherent motion (IVIM) imaging assumes two components. However, more compartments are likely present in pathologic tissue. We hypothesized that spectral analysis using a nonnegative least‐squares (NNLS) approach can detect an additional, intermediate diffusion component, distinct from the parenchymal and microvascular components, in lesion‐prone regions. Purpose To investigate the presence of this intermediate diffusion component and its relation with cerebral small vessel disease (cSVD)‐related lesions. Study Type Prospective cross‐sectional study. Population Patients with cSVD (n = 69, median age 69.8) and controls (n = 39, median age 68.9). Field Strength/Sequence Whole‐brain inversion recovery IVIM acquisition at 3.0T. Assessment Enlarged perivascular spaces (PVS) were rated by three raters. White matter hyperintensities (WMH) were identified on a fluid attenuated inversion recovery (FLAIR) image using a semiautomated algorithm. Statistical Tests Relations between IVIM measures and cSVD‐related lesions were studied using the Spearman's rank order correlation. Results NNLS yielded diffusion spectra from which the intermediate volume fraction fint was apparent between parenchymal diffusion and microvasular pseudodiffusion. WMH volume and the extent of MRI‐visible enlarged PVS in the basal ganglia (BG) and centrum semiovale (CSO) were correlated with fint in the WMHs, BG, and CSO, respectively. fint was 4.2 ± 1.7%, 7.0 ± 4.1% and 13.6 ± 7.7% in BG and 3.9 ± 1.3%, 4.4 ± 1.4% and 4.5 ± 1.2% in CSO for the groups with low, moderate, and high number of enlarged PVS, respectively, and increased with the extent of enlarged PVS (BG: r = 0.49, P < 0.01; CSO: r = 0.23, P = 0.02). fint in the WMHs was 27.1 ± 13.1%, and increased with the WMH volume (r = 0.57, P < 0.01). Data Conclusion We revealed the presence of an intermediate diffusion component in lesion‐prone regions of cSVD and demonstrated its relation with enlarged PVS and WMHs. In tissue with these lesions, tissue degeneration or perivascular edema can lead to more freely diffusing interstitial fluid contributing to fint. Level of Evidence: 2 Technical Efficacy: Stage 2 J. Magn. Reson. Imaging 2020;51:1170–1180.
Collapse
Affiliation(s)
- Sau May Wong
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands.,Department of Neurology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Walter H Backes
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands.,Department of Neurology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Gerhard S Drenthen
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands.,Department of Neurology, Maastricht University Medical Centre, Maastricht, the Netherlands.,Department of School for Mental Health and Neuroscience (MHeNs), Maastricht University Medical Centre, Maastricht, the Netherlands
| | - C Eleana Zhang
- Department of Neurology, Maastricht University Medical Centre, Maastricht, the Netherlands.,Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, the Netherlands.,Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Paulien H M Voorter
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands.,Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Julie Staals
- Department of Neurology, Maastricht University Medical Centre, Maastricht, the Netherlands.,Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Robert J van Oostenbrugge
- Department of Neurology, Maastricht University Medical Centre, Maastricht, the Netherlands.,Department of School for Mental Health and Neuroscience (MHeNs), Maastricht University Medical Centre, Maastricht, the Netherlands.,Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Jacobus F A Jansen
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands.,Department of Neurology, Maastricht University Medical Centre, Maastricht, the Netherlands.,Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| |
Collapse
|
122
|
Blood-brain barrier leakage in relation to white matter hyperintensity volume and cognition in small vessel disease and normal aging. Brain Imaging Behav 2019; 13:389-395. [PMID: 29572621 PMCID: PMC6486901 DOI: 10.1007/s11682-018-9855-7] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Blood–brain barrier (BBB) leakage increases with age and is involved in the pathophysiology of cerebral small vessel disease (cSVD). We examined the relationship between BBB leakage and white matter hyperintensity (WMH) volume and cognition, in cSVD patients and healthy controls. Seventy-seven patients with clinically overt cSVD and thirty-nine age matched healthy controls underwent dynamic contract-enhanced and structural brain MRI and neuropsychological assessment. We quantified BBB leakage volume and rate in normal appearing white matter (NAWM), WMH and cortical grey matter (CGM). Larger leakage volume and lower leakage rate in WMH were associated with larger WMH volume in cSVD but not in controls. Higher leakage rate in NAWM was associated with lower scores on executive function and information processing speed in healthy controls, whereas no relation with cognition was found in cSVD patients. Our findings support the involvement of BBB leakage in cSVD and aging. They also suggest that the mechanism of cognitive dysfunction in cSVD is more complex and multifactorial in cSVD compared with normal aging.
Collapse
|
123
|
Cannistraro RJ, Badi M, Eidelman BH, Dickson DW, Middlebrooks EH, Meschia JF. CNS small vessel disease: A clinical review. Neurology 2019; 92:1146-1156. [PMID: 31142635 PMCID: PMC6598791 DOI: 10.1212/wnl.0000000000007654] [Citation(s) in RCA: 378] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 03/13/2019] [Indexed: 11/15/2022] Open
Abstract
CNS small vessel disease (CSVD) causes 25% of strokes and contributes to 45% of dementia cases. Prevalence increases with age, affecting about 5% of people aged 50 years to almost 100% of people older than 90 years. Known causes and risk factors include age, hypertension, branch atheromatous disease, cerebral amyloid angiopathy, radiation exposure, immune-mediated vasculitides, certain infections, and several genetic diseases. CSVD can be asymptomatic; however, depending on location, lesions can cause mild cognitive dysfunction, dementia, mood disorders, motor and gait dysfunction, and urinary incontinence. CSVD is diagnosed on the basis of brain imaging biomarkers, including recent small subcortical infarcts, white matter hyperintensities, lacunes, cerebral microbleeds, enlarged perivascular spaces, and cerebral atrophy. Advanced imaging modalities can detect signs of disease even earlier than current standard imaging techniques. Diffusion tensor imaging can identify altered white matter connectivity, and blood oxygenation level-dependent imaging can identify decreased vascular reactivity. Pathogenesis is thought to begin with an etiologically specific insult, with or without genetic predisposition, which results in dysfunction of the neurovascular unit. Uncertainties regarding pathogenesis have delayed development of effective treatment. The most widely accepted approach to treatment is to intensively control well-established vascular risk factors, of which hypertension is the most important. With better understanding of pathogenesis, specific therapies may emerge. Early identification of pathologic characteristics with advanced imaging provides an opportunity to forestall progression before emergence of symptoms.
Collapse
Affiliation(s)
- Rocco J Cannistraro
- From the Department of Neurology (R.J.C., M.B., B.H.E., J.F.M.), Department of Laboratory Medicine and Pathology (D.W.D.), Department of Neuroscience (D.W.D.), and Department of Radiology (E.H.M.), Mayo Clinic, Jacksonville, FL
| | - Mohammed Badi
- From the Department of Neurology (R.J.C., M.B., B.H.E., J.F.M.), Department of Laboratory Medicine and Pathology (D.W.D.), Department of Neuroscience (D.W.D.), and Department of Radiology (E.H.M.), Mayo Clinic, Jacksonville, FL
| | - Benjamin H Eidelman
- From the Department of Neurology (R.J.C., M.B., B.H.E., J.F.M.), Department of Laboratory Medicine and Pathology (D.W.D.), Department of Neuroscience (D.W.D.), and Department of Radiology (E.H.M.), Mayo Clinic, Jacksonville, FL
| | - Dennis W Dickson
- From the Department of Neurology (R.J.C., M.B., B.H.E., J.F.M.), Department of Laboratory Medicine and Pathology (D.W.D.), Department of Neuroscience (D.W.D.), and Department of Radiology (E.H.M.), Mayo Clinic, Jacksonville, FL
| | - Erik H Middlebrooks
- From the Department of Neurology (R.J.C., M.B., B.H.E., J.F.M.), Department of Laboratory Medicine and Pathology (D.W.D.), Department of Neuroscience (D.W.D.), and Department of Radiology (E.H.M.), Mayo Clinic, Jacksonville, FL
| | - James F Meschia
- From the Department of Neurology (R.J.C., M.B., B.H.E., J.F.M.), Department of Laboratory Medicine and Pathology (D.W.D.), Department of Neuroscience (D.W.D.), and Department of Radiology (E.H.M.), Mayo Clinic, Jacksonville, FL.
| |
Collapse
|
124
|
Li Y, Li M, Yang L, Qin W, Yang S, Yuan J, Jiang T, Hu W. The relationship between blood-brain barrier permeability and enlarged perivascular spaces: a cross-sectional study. Clin Interv Aging 2019; 14:871-878. [PMID: 31190773 PMCID: PMC6519012 DOI: 10.2147/cia.s204269] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 04/15/2019] [Indexed: 12/01/2022] Open
Abstract
Purpose: Enlarged perivascular spaces (EPVS) have been widely considered as a feature of cerebral small vessel disease (cSVD) but the pathogenesis of EPVS remains unclear. Compromised blood–brain barrier (BBB) integrity may play a role since previous studies have shown that BBB breakdown is a critical contributor to the pathogenesis of other cSVD markers. This study aimed to investigate the association of EPVS in the centrum semiovale (CSO) and basal ganglia (BG) with BBB permeability. Patients and methods: Consecutive participants free of symptomatic stroke history presented for physical examination were enrolled in this cross-sectional study. CSO- and BG-EPVS on T2-weighted (T2-W) magnetic resonance imaging (MRI) were rated using a five-point validated scale. Dynamic contrast-enhanced (DCE)-MRI and Patlak pharmacokinetic model were applied to quantify BBB permeability in the CSO and BG. Results: A total of 109 participants aged 49–90 years (mean age of 69.85 years) were enrolled. The proportions of participants presenting high-grade (>10) EPVS in the CSO and BG were 50.5% and 44.0%, respectively. After adjustments for potential confounders by logistic regression, leakage rate and fractional blood plasma volume were correlated with the severity of BG-EPVS (OR: 5.33; 95%CI: 1.95–14.60 and OR: 0.93; 95%CI: 0.87–0.99). Conclusion: Our study demonstrates that BG-EPVS are associated with compromised BBB integrity, supporting the hypothesis that the BBB dysfunction may be involved in the pathogenesis of BG-EPVS. EPVS in the CSO and BG may have distinct pathophysiology.
Collapse
Affiliation(s)
- Yue Li
- Department of Neurology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Man Li
- Department of Radiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Lei Yang
- Department of Neurology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Wei Qin
- Department of Neurology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Shuna Yang
- Department of Neurology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Junliang Yuan
- Department of Neurology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Tao Jiang
- Department of Radiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Wenli Hu
- Department of Neurology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
125
|
Thrippleton MJ, Backes WH, Sourbron S, Ingrisch M, van Osch MJP, Dichgans M, Fazekas F, Ropele S, Frayne R, van Oostenbrugge RJ, Smith EE, Wardlaw JM. Quantifying blood-brain barrier leakage in small vessel disease: Review and consensus recommendations. Alzheimers Dement 2019; 15:840-858. [PMID: 31031101 PMCID: PMC6565805 DOI: 10.1016/j.jalz.2019.01.013] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/22/2018] [Accepted: 01/18/2019] [Indexed: 12/12/2022]
Abstract
Cerebral small vessel disease (cSVD) comprises pathological processes of the small vessels in the brain that may manifest clinically as stroke, cognitive impairment, dementia, or gait disturbance. It is generally accepted that endothelial dysfunction, including blood-brain barrier (BBB) failure, is pivotal in the pathophysiology. Recent years have seen increasing use of imaging, primarily dynamic contrast-enhanced magnetic resonance imaging, to assess BBB leakage, but there is considerable variability in the approaches and findings reported in the literature. Although dynamic contrast-enhanced magnetic resonance imaging is well established, challenges emerge in cSVD because of the subtle nature of BBB impairment. The purpose of this work, authored by members of the HARNESS Initiative, is to provide an in-depth review and position statement on magnetic resonance imaging measurement of subtle BBB leakage in clinical research studies, with aspects requiring further research identified. We further aim to provide information and consensus recommendations for new investigators wishing to study BBB failure in cSVD and dementia.
Collapse
Affiliation(s)
- Michael J Thrippleton
- Centre for Clinical Brain Science, University of Edinburgh, Edinburgh, UK; Dementia Research Institute, University of Edinburgh, Edinburgh, UK; Edinburgh Imaging, University of Edinburgh, Edinburgh, UK.
| | - Walter H Backes
- Department of Radiology & Nuclear Medicine, School for Mental Health and Neuroscience, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Steven Sourbron
- Leeds Imaging Biomarkers group, Department of Biomedical Imaging Sciences, University of Leeds, Leeds, UK
| | - Michael Ingrisch
- Department of Radiology, Ludwig-Maximilians-University Hospital Munich, Munich, Germany
| | - Matthias J P van Osch
- Department of Radiology, C. J. Gorter Center for High Field MRI, Leiden University Medical Center, Leiden, The Netherlands
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-University München & Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Franz Fazekas
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Stefan Ropele
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Richard Frayne
- Department of Radiology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Seaman Family MR Research Centre, Foothills Medical Centre, Calgary, Alberta, Canada
| | - Robert J van Oostenbrugge
- Department of Neurology, School for Mental Health and Neuroscience, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Eric E Smith
- Department of Radiology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada; Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Joanna M Wardlaw
- Centre for Clinical Brain Science, University of Edinburgh, Edinburgh, UK; Dementia Research Institute, University of Edinburgh, Edinburgh, UK; Edinburgh Imaging, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
126
|
Wong SM, Jansen JFA, Zhang CE, Hoff EI, Staals J, van Oostenbrugge RJ, Backes WH. Blood-brain barrier impairment and hypoperfusion are linked in cerebral small vessel disease. Neurology 2019; 92:e1669-e1677. [PMID: 30867275 DOI: 10.1212/wnl.0000000000007263] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 11/28/2018] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE To investigate the link between blood-brain-barrier (BBB) permeability and cerebral blood flow (CBF) and the relation with white matter hyperintensities (WMH) in cerebral small vessel disease (cSVD). METHODS Twenty-seven patients with cSVD received dynamic susceptibility contrast and dynamic contrast-enhanced MRI to determine CBF and BBB permeability (expressed as leakage rate and volume), respectively. Structural MRI were segmented into normal-appearing white matter (NAWM) and WMH, for which a perilesional zone was defined. In these regions, we investigated the BBB permeability, CBF, and their relation using Pearson correlation r. RESULTS We found a decrease in CBF of 2.2 mL/min/100 g (p < 0.01) and an increase in leakage volume of 0.7% (p < 0.01) per mm closer to the WMH in the perilesional zones. Lower CBF values correlated with higher leakage measures in the NAWM and WMH (-0.53 < r < -0.40, p < 0.05). This relation was also observed in the perilesional zones, which became stronger in the proximity of WMH (p = 0.03). CONCLUSION BBB impairment and hypoperfusion appear in the WMH and NAWM, which increase in the proximity of the WMH, and are linked. Both BBB and CBF are regulated in the neurovascular unit (NVU) and the observed link might be due to the physiologic regulation mechanism of the NVU. This link may suggest an early overall deterioration of this unit.
Collapse
Affiliation(s)
- Sau May Wong
- From the Departments of Radiology and Nuclear Medicine (S.M.W., J.F.A.J., W.H.B.), Neurology (C.E.Z., J.S., R.J.v.O.), School for Mental Health and Neuroscience (MHeNs) (S.M.W., J.F.A.J., C.E.Z., R.J.v.O., W.H.B.), and Cardiovascular Research Institute Maastricht (CARIM) (C.E.Z., J.S., R.J.v.O.), Maastricht University Medical Centre; and the Department of Neurology (E.I.H.), Zuyderland Medical Centre, Heerlen, the Netherlands
| | - Jacobus F A Jansen
- From the Departments of Radiology and Nuclear Medicine (S.M.W., J.F.A.J., W.H.B.), Neurology (C.E.Z., J.S., R.J.v.O.), School for Mental Health and Neuroscience (MHeNs) (S.M.W., J.F.A.J., C.E.Z., R.J.v.O., W.H.B.), and Cardiovascular Research Institute Maastricht (CARIM) (C.E.Z., J.S., R.J.v.O.), Maastricht University Medical Centre; and the Department of Neurology (E.I.H.), Zuyderland Medical Centre, Heerlen, the Netherlands
| | - C Eleana Zhang
- From the Departments of Radiology and Nuclear Medicine (S.M.W., J.F.A.J., W.H.B.), Neurology (C.E.Z., J.S., R.J.v.O.), School for Mental Health and Neuroscience (MHeNs) (S.M.W., J.F.A.J., C.E.Z., R.J.v.O., W.H.B.), and Cardiovascular Research Institute Maastricht (CARIM) (C.E.Z., J.S., R.J.v.O.), Maastricht University Medical Centre; and the Department of Neurology (E.I.H.), Zuyderland Medical Centre, Heerlen, the Netherlands
| | - Erik I Hoff
- From the Departments of Radiology and Nuclear Medicine (S.M.W., J.F.A.J., W.H.B.), Neurology (C.E.Z., J.S., R.J.v.O.), School for Mental Health and Neuroscience (MHeNs) (S.M.W., J.F.A.J., C.E.Z., R.J.v.O., W.H.B.), and Cardiovascular Research Institute Maastricht (CARIM) (C.E.Z., J.S., R.J.v.O.), Maastricht University Medical Centre; and the Department of Neurology (E.I.H.), Zuyderland Medical Centre, Heerlen, the Netherlands
| | - Julie Staals
- From the Departments of Radiology and Nuclear Medicine (S.M.W., J.F.A.J., W.H.B.), Neurology (C.E.Z., J.S., R.J.v.O.), School for Mental Health and Neuroscience (MHeNs) (S.M.W., J.F.A.J., C.E.Z., R.J.v.O., W.H.B.), and Cardiovascular Research Institute Maastricht (CARIM) (C.E.Z., J.S., R.J.v.O.), Maastricht University Medical Centre; and the Department of Neurology (E.I.H.), Zuyderland Medical Centre, Heerlen, the Netherlands
| | - Robert J van Oostenbrugge
- From the Departments of Radiology and Nuclear Medicine (S.M.W., J.F.A.J., W.H.B.), Neurology (C.E.Z., J.S., R.J.v.O.), School for Mental Health and Neuroscience (MHeNs) (S.M.W., J.F.A.J., C.E.Z., R.J.v.O., W.H.B.), and Cardiovascular Research Institute Maastricht (CARIM) (C.E.Z., J.S., R.J.v.O.), Maastricht University Medical Centre; and the Department of Neurology (E.I.H.), Zuyderland Medical Centre, Heerlen, the Netherlands
| | - Walter H Backes
- From the Departments of Radiology and Nuclear Medicine (S.M.W., J.F.A.J., W.H.B.), Neurology (C.E.Z., J.S., R.J.v.O.), School for Mental Health and Neuroscience (MHeNs) (S.M.W., J.F.A.J., C.E.Z., R.J.v.O., W.H.B.), and Cardiovascular Research Institute Maastricht (CARIM) (C.E.Z., J.S., R.J.v.O.), Maastricht University Medical Centre; and the Department of Neurology (E.I.H.), Zuyderland Medical Centre, Heerlen, the Netherlands.
| |
Collapse
|
127
|
Förster A, Al-Zghloul M, Wenz H, Böhme J, Groden C, Alonso A. Gadolinium Leakage in Ocular Structures Is Common in Lacunar Infarction. Stroke 2019; 50:193-195. [PMID: 30580698 DOI: 10.1161/strokeaha.118.023573] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background and Purpose- We investigated the frequency and pattern of blood-brain barrier, as well as blood-retina barrier, impairment in acute lacunar infarction as demonstrated by hyperintense acute reperfusion marker and gadolinium leakage in ocular structures (GLOS), respectively, on fluid-attenuated inversion recovery images. Methods- Acute lacunar infarction patients who underwent repeated magnetic resonance imaging after intravenous contrast agent administration were identified and the presence of GLOS in the anterior chamber and vitreous body and hyperintense acute reperfusion marker noted on fluid-attenuated inversion recovery. Results- Overall, 24 acute lacunar infarction patients (median age 64.5 years; interquartile range, 54-78 years) were included. On contrast-enhanced fluid-attenuated inversion recovery, GLOS was observed in 11 (45.8%) patients: in 4 (16.7%) in the anterior chamber only and in 7 (29.2%) in the anterior chamber and vitreous body. In all patients, GLOS was bilateral and symmetrical. In patients with GLOS in the anterior chamber only, the time between initial and follow-up magnetic resonance imaging was significantly shorter (7.5 [interquartile range, 4.25-11.5] hours) compared with patients with GLOS in the anterior chamber and vitreous body (28 [interquartile range, 10-43] hours; P=0.047). Hyperintense acute reperfusion marker could not be demonstrated in any of the patients. Conclusions- In acute lacunar infarction patients, unlike hyperintense acute reperfusion marker, GLOS is a frequent finding and shows a similar temporal evolution like in larger ischemic stroke.
Collapse
Affiliation(s)
- Alex Förster
- From the Department of Neuroradiology (A.F., M.A.-Z., H.W., J.B., C.G.), Universitätsmedizin Mannheim, University of Heidelberg, Mannheim, Germany
| | - Mansour Al-Zghloul
- From the Department of Neuroradiology (A.F., M.A.-Z., H.W., J.B., C.G.), Universitätsmedizin Mannheim, University of Heidelberg, Mannheim, Germany
| | - Holger Wenz
- From the Department of Neuroradiology (A.F., M.A.-Z., H.W., J.B., C.G.), Universitätsmedizin Mannheim, University of Heidelberg, Mannheim, Germany
| | - Johannes Böhme
- From the Department of Neuroradiology (A.F., M.A.-Z., H.W., J.B., C.G.), Universitätsmedizin Mannheim, University of Heidelberg, Mannheim, Germany
| | - Christoph Groden
- From the Department of Neuroradiology (A.F., M.A.-Z., H.W., J.B., C.G.), Universitätsmedizin Mannheim, University of Heidelberg, Mannheim, Germany
| | - Angelika Alonso
- Department of Neurology (A.A.), Universitätsmedizin Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
128
|
Cipolla MJ, Liebeskind DS, Chan SL. The importance of comorbidities in ischemic stroke: Impact of hypertension on the cerebral circulation. J Cereb Blood Flow Metab 2018; 38:2129-2149. [PMID: 30198826 PMCID: PMC6282213 DOI: 10.1177/0271678x18800589] [Citation(s) in RCA: 187] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Comorbidities are a hallmark of stroke that both increase the incidence of stroke and worsen outcome. Hypertension is prevalent in the stroke population and the most important modifiable risk factor for stroke. Hypertensive disorders promote stroke through increased shear stress, endothelial dysfunction, and large artery stiffness that transmits pulsatile flow to the cerebral microcirculation. Hypertension also promotes cerebral small vessel disease through several mechanisms, including hypoperfusion, diminished autoregulatory capacity and localized increase in blood-brain barrier permeability. Preeclampsia, a hypertensive disorder of pregnancy, also increases the risk of stroke 4-5-fold compared to normal pregnancy that predisposes women to early-onset cognitive impairment. In this review, we highlight how comorbidities and concomitant disorders are not only risk factors for ischemic stroke, but alter the response to acute ischemia. We focus on hypertension as a comorbidity and its effects on the cerebral circulation that alters the pathophysiology of ischemic stroke and should be considered in guiding future therapeutic strategies.
Collapse
Affiliation(s)
- Marilyn J Cipolla
- 1 Department of Neurological Sciences, University of Vermont Larner College of Medicine, Burlington, VT, USA
| | - David S Liebeskind
- 2 Neurovascular Imaging Research Core and Stroke Center, Department of Neurology, University of California at Los Angeles, Los Angeles, CA, USA
| | - Siu-Lung Chan
- 1 Department of Neurological Sciences, University of Vermont Larner College of Medicine, Burlington, VT, USA
| |
Collapse
|
129
|
Elahi FM, Casaletto KB, Altendahl M, Staffaroni AM, Fletcher E, Filshtein TJ, Glymour MM, Miller BL, Hinman JD, DeCarli C, Goetzl EJ, Kramer JH. "Liquid Biopsy" of White Matter Hyperintensity in Functionally Normal Elders. Front Aging Neurosci 2018; 10:343. [PMID: 30483114 PMCID: PMC6244607 DOI: 10.3389/fnagi.2018.00343] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 10/11/2018] [Indexed: 12/14/2022] Open
Abstract
Background and Objective: In the aging brain, increased blood-brain barrier (BBB) leakage and white matter hyperintensity (WMH) on MRI are frequently presumed secondary to cerebral small vessel disease (cSVD) or endotheliopathy. We investigate this association in vivo by quantifying protein cargo from endothelial-derived exosomes (EDE), and comparing levels between two groups of functionally normal elders with and without WMH. In addition, we study associations of EDE proteins with upstream and downstream factors, such as inflammation and neurodegenerative changes, respectively. Methods: Twenty six neurologically normal older adults completed general health questionnaires, neuropsychological and physical examinations, and brain MRI. WMH was visually graded with modified Fazekas score of 2 or greater used to classify 11 subjects as cases, and 15 without WMH as controls. Plasma total exosomes were precipitated and EDEs enriched by sequential immuno-precipitations. In addition, we quantified three inflammatory cytokines from plasma and imaging variables on MRI. Group means were compared, the discriminant functions of biomarkers calculated, and the association of EDE biomarkers with plasma inflammatory markers, cognition, and imaging outcomes assessed via regression modeling. Results: Plasma levels of EDE cargo proteins GLUT1, LAT1, P-GP, and NOSTRIN were significantly higher in subjects with WMH in comparison to those without. In contrast, EDE levels of the marker with low expression in brain (VCAM1) were equal between groups. The effect sizes for each of the brain-expressed cargo proteins (GLUT1, LAT1, and P-GP) were such that age-adjusted logistic regressions revealed areas under the curve (AUC) with range of 0.82–0.89, differentiating subjects with WMH from those without. VCAM1 poorly discriminated between groups (AUC:0.55). Higher levels of all brain-expressed EDE proteins were also associated with lower cognitive function, unrelated to burden of WMH. Levels of LAT1 and P-GP were significantly inversely associated with global gray matter volumes, and EDE GLUT1, LAT-1, and P-GP concentrations were significantly associated with systemic IL-6 levels. Conclusion: In a case control study of clinically normal adults with and without WMH, concentrations of EDE proteins were significantly higher in subjects with WMH in comparison to controls. This work is a first step toward in vivo dissection of molecular changes in endothelia of functionally normal subjects with radiographic evidence of age-associated white matter disease.
Collapse
Affiliation(s)
- Fanny M Elahi
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - Kaitlin B Casaletto
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - Marie Altendahl
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - Adam M Staffaroni
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - Evan Fletcher
- Department of Neurology, University of California, Davis, Davis, CA, United States
| | - Teresa J Filshtein
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, United States
| | - Maria M Glymour
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, United States
| | - Bruce L Miller
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| | - Jason D Hinman
- Department of Neurology, University of California, Los Angeles, Los Angeles, CA, United States
| | - Charles DeCarli
- Department of Neurology, University of California, Davis, Davis, CA, United States
| | - Edward J Goetzl
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States.,Jewish Home of San Francisco, San Francisco, CA, United States
| | - Joel H Kramer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
130
|
Kim YK, Nam KI, Song J. The Glymphatic System in Diabetes-Induced Dementia. Front Neurol 2018; 9:867. [PMID: 30429819 PMCID: PMC6220044 DOI: 10.3389/fneur.2018.00867] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 09/26/2018] [Indexed: 12/14/2022] Open
Abstract
The glymphatic system has emerged as an important player in central nervous system (CNS) diseases, by regulating the vasculature impairment, effectively controlling the clearance of toxic peptides, modulating activity of astrocytes, and being involved in the circulation of neurotransmitters in the brain. Recently, several studies have indicated decreased activity of the glymphatic pathway under diabetes conditions such as in insulin resistance and hyperglycemia. Furthermore, diabetes leads to the disruption of the blood-brain barrier and decrease of apolipoprotein E (APOE) expression and the secretion of norepinephrine in the brain, involving the impairment of the glymphatic pathway and ultimately resulting in cognitive decline. Considering the increased prevalence of diabetes-induced dementia worldwide, the relationship between the glymphatic pathway and diabetes-induced dementia should be investigated and the mechanisms underlying their relationship should be discussed to promote the development of an effective therapeutic approach in the near future. Here, we have reviewed recent evidence for the relationship between glymphatic pathway dysfunction and diabetes. We highlight that the enhancement of the glymphatic system function during sleep may be beneficial to the attenuation of neuropathology in diabetes-induced dementia. Moreover, we suggest that improving glymphatic system activity may be a potential therapeutic strategy for the prevention of diabetes-induced dementia.
Collapse
Affiliation(s)
- Young-Kook Kim
- Department of Biochemistry, Chonnam National University Medical School, Gwangju, South Korea.,Department of Biomedical Sciences, Center for Creative Biomedical Scientists, Chonnam National University, Gwangju, South Korea
| | - Kwang Il Nam
- Department of Anatomy, Chonnam National University Medical School, Gwangju, South Korea
| | - Juhyun Song
- Department of Biomedical Sciences, Center for Creative Biomedical Scientists, Chonnam National University, Gwangju, South Korea.,Department of Anatomy, Chonnam National University Medical School, Gwangju, South Korea
| |
Collapse
|
131
|
Hamanaka G, Ohtomo R, Takase H, Lok J, Arai K. White-matter repair: Interaction between oligodendrocytes and the neurovascular unit. Brain Circ 2018; 4:118-123. [PMID: 30450418 PMCID: PMC6187946 DOI: 10.4103/bc.bc_15_18] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 09/06/2018] [Accepted: 09/10/2018] [Indexed: 02/06/2023] Open
Abstract
There are currently no adequate treatments for white-matter injury, which often follows central nervous system maladies and their accompanying neurodegenerative processes. Indeed, the white matter is compromised by the deterioration of the blood–brain barrier and the demyelination of neuronal axons. Key repairs to the white matter are mediated by oligodendrocyte lineage cells after damaging events. Oligodendrocytes are supported by other cells in the neurovascular unit and these cells collaborate in processes such as angiogenesis, neurogenesis, and oligodendrogenesis. Understanding the various interactions between these cells and oligodendrocytes will be imperative for developing reparative therapies for impaired white matter. This minireview will discuss how oligodendrocytes and oligodendrocyte lineage cells mend damage to the white matter and restore brain function ensuing neural injury.
Collapse
Affiliation(s)
- Gen Hamanaka
- Department of Radiology and Neurology, Neuroprotection Research Laboratory, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Ryo Ohtomo
- Department of Radiology and Neurology, Neuroprotection Research Laboratory, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Hajime Takase
- Department of Radiology and Neurology, Neuroprotection Research Laboratory, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Josephine Lok
- Department of Radiology and Neurology, Neuroprotection Research Laboratory, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Ken Arai
- Department of Radiology and Neurology, Neuroprotection Research Laboratory, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| |
Collapse
|
132
|
Cuadrado-Godia E, Dwivedi P, Sharma S, Ois Santiago A, Roquer Gonzalez J, Balcells M, Laird J, Turk M, Suri HS, Nicolaides A, Saba L, Khanna NN, Suri JS. Cerebral Small Vessel Disease: A Review Focusing on Pathophysiology, Biomarkers, and Machine Learning Strategies. J Stroke 2018; 20:302-320. [PMID: 30309226 PMCID: PMC6186915 DOI: 10.5853/jos.2017.02922] [Citation(s) in RCA: 181] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 04/02/2018] [Indexed: 12/15/2022] Open
Abstract
Cerebral small vessel disease (cSVD) has a crucial role in lacunar stroke and brain hemorrhages and is a leading cause of cognitive decline and functional loss in elderly patients. Based on underlying pathophysiology, cSVD can be subdivided into amyloidal and non-amyloidal subtypes. Genetic factors of cSVD play a pivotal role in terms of unraveling molecular mechanism. An important pathophysiological mechanism of cSVD is blood-brain barrier leakage and endothelium dysfunction which gives a clue in identification of the disease through circulating biological markers. Detection of cSVD is routinely carried out by key neuroimaging markers including white matter hyperintensities, lacunes, small subcortical infarcts, perivascular spaces, cerebral microbleeds, and brain atrophy. Application of neural networking, machine learning and deep learning in image processing have increased significantly for correct severity of cSVD. A linkage between cSVD and other neurological disorder, such as Alzheimer's and Parkinson's disease and non-cerebral disease, has also been investigated recently. This review draws a broad picture of cSVD, aiming to inculcate new insights into its pathogenesis and biomarkers. It also focuses on the role of deep machine strategies and other dimensions of cSVD by linking it with several cerebral and non-cerebral diseases as well as recent advances in the field to achieve sensitive detection, effective prevention and disease management.
Collapse
Affiliation(s)
- Elisa Cuadrado-Godia
- Department of Neurology, Hospital del Mar Medical Research Institute, Barcelona, Spain
| | | | - Sanjiv Sharma
- Department of Computer Science & Engineering and Information Technology, Madhav Institute of Technology and Science, Gwalior, India
| | - Angel Ois Santiago
- Department of Neurology, Hospital del Mar Medical Research Institute, Barcelona, Spain
| | - Jaume Roquer Gonzalez
- Department of Neurology, Hospital del Mar Medical Research Institute, Barcelona, Spain
| | - Mercedes Balcells
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Biological Engineering, IQS School of Engineering, Barcelona, Spain
| | - John Laird
- Department of Cardiology, St. Helena Hospital, St. Helena, CA, USA
| | - Monika Turk
- Deparment of Neurology, University Medical Centre Maribor, Maribor, Slovenia
| | | | | | - Luca Saba
- Department of Radiology, Azienda Ospedaliero Universitaria, Cagliari, Italy
| | | | - Jasjit S Suri
- Stroke Monitoring Division, AtheroPoint, Roseville, CA, USA
| |
Collapse
|
133
|
Abstract
Cerebral small vessel disease (CSVD) is composed of several diseases affecting the small arteries, arterioles, venules, and capillaries of the brain, and refers to several pathological processes and etiologies. Neuroimaging features of CSVD include recent small subcortical infarcts, lacunes, white matter hyperintensities, perivascular spaces, microbleeds, and brain atrophy. The main clinical manifestations of CSVD include stroke, cognitive decline, dementia, psychiatric disorders, abnormal gait, and urinary incontinence. Currently, there are no specific preventive or therapeutic measures to improve this condition. In this review, we will discuss the pathophysiology, clinical aspects, neuroimaging, progress of research to treat and prevent CSVD and current treatment of this disease.
Collapse
Affiliation(s)
- Qian Li
- 1 Department of Pediatrics, The Third Affiliated Hospital & Field Surgery Institution, Army Medical University, Chongqing, China.,Both the authors contributed equally as co-authors
| | - Yang Yang
- 2 Department of Neurology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.,Both the authors contributed equally as co-authors
| | - Cesar Reis
- 3 Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Tao Tao
- 2 Department of Neurology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Wanwei Li
- 1 Department of Pediatrics, The Third Affiliated Hospital & Field Surgery Institution, Army Medical University, Chongqing, China
| | - Xiaogang Li
- 2 Department of Neurology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - John H Zhang
- 3 Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, USA.,4 Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| |
Collapse
|
134
|
Hamanaka G, Ohtomo R, Takase H, Lok J, Arai K. Role of oligodendrocyte-neurovascular unit in white matter repair. Neurosci Lett 2018; 684:175-180. [PMID: 30006018 DOI: 10.1016/j.neulet.2018.07.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 07/09/2018] [Indexed: 12/20/2022]
Abstract
White matter injury caused by acute or chronic neuropathologies is a major characteristic of many CNS diseases, and an effective treatment is still out of our reach. White matter damage is associated with the collapse of the axon-myelin complex and with blood-brain barrier (BBB) breakdown, which results in disruption of white matter function. While white matter damage cannot completely resolve spontaneously, some compensative responses may occur after the injury. Oligodendrocyte lineage cells perform critical functions in repairing damaged white matter. In this mini-review, we will focus on the reparative actions of the oligodendrocytes, and highlight the important role of oligodendrocyte lineage cells in brain recovery after injury.
Collapse
Affiliation(s)
- Gen Hamanaka
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Ryo Ohtomo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Hajime Takase
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Josephine Lok
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA.
| |
Collapse
|
135
|
Cox SR, Allerhand M, Ritchie SJ, Muñoz Maniega S, Valdés Hernández M, Harris SE, Dickie DA, Anblagan D, Aribisala BS, Morris Z, Sherwood R, Abbott NJ, Starr JM, Bastin ME, Wardlaw JM, Deary IJ. Longitudinal serum S100β and brain aging in the Lothian Birth Cohort 1936. Neurobiol Aging 2018; 69:274-282. [PMID: 29933100 PMCID: PMC6075468 DOI: 10.1016/j.neurobiolaging.2018.05.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 05/22/2018] [Accepted: 05/23/2018] [Indexed: 12/22/2022]
Abstract
Elevated serum and cerebrospinal fluid concentrations of S100β, a protein predominantly found in glia, are associated with intracranial injury and neurodegeneration, although concentrations are also influenced by several other factors. The longitudinal association between serum S100β concentrations and brain health in nonpathological aging is unknown. In a large group (baseline N = 593; longitudinal N = 414) of community-dwelling older adults at ages 73 and 76 years, we examined cross-sectional and parallel longitudinal changes between serum S100β and brain MRI parameters: white matter hyperintensities, perivascular space visibility, white matter fractional anisotropy and mean diffusivity (MD), global atrophy, and gray matter volume. Using bivariate change score structural equation models, correcting for age, sex, diabetes, and hypertension, higher S100β was cross-sectionally associated with poorer general fractional anisotropy (r = -0.150, p = 0.001), which was strongest in the anterior thalamic (r = -0.155, p < 0.001) and cingulum bundles (r = -0.111, p = 0.005), and survived false discovery rate correction. Longitudinally, there were no significant associations between changes in brain imaging parameters and S100β after false discovery rate correction. These data provide some weak evidence that S100β may be an informative biomarker of brain white matter aging.
Collapse
Affiliation(s)
- Simon R Cox
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, Scotland, UK; Department of Psychology, University of Edinburgh, Edinburgh, Scotland, UK.
| | - Mike Allerhand
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, Scotland, UK; Department of Psychology, University of Edinburgh, Edinburgh, Scotland, UK
| | - Stuart J Ritchie
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, Scotland, UK; Department of Psychology, University of Edinburgh, Edinburgh, Scotland, UK
| | - Susana Muñoz Maniega
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, Scotland, UK; Department of Neuroimaging Sciences, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK; UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
| | - Maria Valdés Hernández
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, Scotland, UK; Department of Neuroimaging Sciences, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK; UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
| | - Sarah E Harris
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, Scotland, UK; Medical Genetics Section, University of Edinburgh Centre for Genomic and Experimental Medicine and MRC Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, UK
| | - David Alexander Dickie
- Institute of Cardiovascular and Medical Sciences College of Medical, Veterinary & Life Sciences University of Glasgow, UK
| | - Devasuda Anblagan
- Department of Neuroimaging Sciences, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| | - Benjamin S Aribisala
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, Scotland, UK; Department of Neuroimaging Sciences, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK; Department of Computer Science, Lagos State University, Lagos, Nigeria
| | - Zoe Morris
- Department of Neuroimaging Sciences, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK; UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
| | - Roy Sherwood
- Department of Clinical Biochemistry, King's College Hospital NHS Foundation Trust, London, UK
| | - N Joan Abbott
- Institute of Pharmaceutical Science, King's College London, London, UK
| | - John M Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, Scotland, UK; Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, Scotland, UK
| | - Mark E Bastin
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, Scotland, UK; Department of Neuroimaging Sciences, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| | - Joanna M Wardlaw
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, Scotland, UK; Department of Neuroimaging Sciences, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK; UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, Scotland, UK; Department of Psychology, University of Edinburgh, Edinburgh, Scotland, UK
| |
Collapse
|
136
|
Brown R, Benveniste H, Black SE, Charpak S, Dichgans M, Joutel A, Nedergaard M, Smith KJ, Zlokovic BV, Wardlaw JM. Understanding the role of the perivascular space in cerebral small vessel disease. Cardiovasc Res 2018; 114:1462-1473. [PMID: 29726891 PMCID: PMC6455920 DOI: 10.1093/cvr/cvy113] [Citation(s) in RCA: 223] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/18/2018] [Accepted: 05/02/2018] [Indexed: 12/17/2022] Open
Abstract
Small vessel diseases (SVDs) are a group of disorders that result from pathological alteration of the small blood vessels in the brain, including the small arteries, capillaries and veins. Of the 35-36 million people that are estimated to suffer from dementia worldwide, up to 65% have an SVD component. Furthermore, SVD causes 20-25% of strokes, worsens outcome after stroke and is a leading cause of disability, cognitive impairment and poor mobility. Yet the underlying cause(s) of SVD are not fully understood. Magnetic resonance imaging has confirmed enlarged perivascular spaces (PVS) as a hallmark feature of SVD. In healthy tissue, these spaces are proposed to form part of a complex brain fluid drainage system which supports interstitial fluid exchange and may also facilitate clearance of waste products from the brain. The pathophysiological signature of PVS and what this infers about their function and interaction with cerebral microcirculation, plus subsequent downstream effects on lesion development in the brain has not been established. Here we discuss the potential of enlarged PVS to be a unique biomarker for SVD and related brain disorders with a vascular component. We propose that widening of PVS suggests presence of peri-vascular cell debris and other waste products that form part of a vicious cycle involving impaired cerebrovascular reactivity, blood-brain barrier dysfunction, perivascular inflammation and ultimately impaired clearance of waste proteins from the interstitial fluid space, leading to accumulation of toxins, hypoxia, and tissue damage. Here, we outline current knowledge, questions and hypotheses regarding understanding the brain fluid dynamics underpinning dementia and stroke through the common denominator of SVD.
Collapse
Affiliation(s)
- Rosalind Brown
- Centre for Clinical Brain Sciences, The University of Edinburgh, Chancellor's Building, Edinburgh, UK
| | - Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, USA
| | - Sandra E Black
- LC Campbell Cognitive Neurology Research Unit, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
- Department of Medicine (Neurology), Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
- Hurvitz Brain Sciences Program, Sunnybrook Health Sciences Center, University of Toronto, Toronto, Canada
- Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - Serge Charpak
- INSERM U1128, Laboratory of Neurophysiology and New Microscopies, Université Paris Descartes, Paris, France
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE, Munich), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Anne Joutel
- Genetics and Pathogenesis of Cerebrovascular Diseases, INSERM, Université Paris Diderot-Paris 7, Paris, France
- DHU NeuroVasc, Sorbonne Paris Cité, Paris, France
| | - Maiken Nedergaard
- Section for Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
- Division of Glia Disease and Therapeutics, Center for Translational Neuromedicine, University of Rochester Medical School, Rochester, USA
| | - Kenneth J Smith
- Department of Neuroinflammation, UCL Institute of Neurology, London, UK
| | - Berislav V Zlokovic
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Joanna M Wardlaw
- Centre for Clinical Brain Sciences, Chancellor's Building, Edinburgh, UK
- UK Dementia Research Institute at The University of Edinburgh, Chancellor's Building, Edinburgh, UK
- Row Fogo Centre for Research into Ageing and the Brain, The University of Edinburgh, Chancellor's Building, Edinburgh, UK
| |
Collapse
|
137
|
Yoo RE, Choi SH, Oh BM, Do Shin S, Lee EJ, Shin DJ, Jo SW, Kang KM, Yun TJ, Kim JH, Sohn CH. Quantitative dynamic contrast-enhanced MR imaging shows widespread blood-brain barrier disruption in mild traumatic brain injury patients with post-concussion syndrome. Eur Radiol 2018; 29:1308-1317. [DOI: 10.1007/s00330-018-5656-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 07/01/2018] [Accepted: 07/04/2018] [Indexed: 12/27/2022]
|
138
|
New insights into cerebral small vessel disease and vascular cognitive impairment from MRI. Curr Opin Neurol 2018; 31:36-43. [PMID: 29084064 DOI: 10.1097/wco.0000000000000513] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW We review recent MRI research that addresses two important challenges in cerebral small vessel disease (SVD) research: early diagnosis, and linking SVD with cognitive impairment. First, we review studies of MRI measurements of blood flow and blood-brain barrier integrity. Second, we review MRI studies identifying neuroimaging correlates of SVD-related cognitive dysfunction, focusing on brain connectivity and white matter microarchitecture. This research is placed in context through discussion of recent recommendations for management of incidentally discovered SVD, and neuroimaging biomarker use in clinical trials. RECENT FINDINGS Cerebral perfusion, cerebrovascular reactivity (CVR), blood-brain barrier permeability, and white matter microarchitecture are measurable using MRI, and are altered in SVD. Lower cerebral blood flow predicts a higher future risk for dementia, whereas decreased CVR occurs at early stages of SVD and is associated with future white matter hyperintensity growth. Two new approaches to analyzing diffusion tensor imaging (DTI) data in SVD patients have emerged: graph theory-based analysis of networks of DTI connectivity between cortical nodes, and analysis of histograms of mean diffusivity of the hemispheric white matter. SUMMARY New, advanced quantitative neuroimaging techniques are not ready for routine radiological practice but are already being employed as monitoring biomarkers in the newest generation of trials for SVD.
Collapse
|
139
|
Duering M, Finsterwalder S, Baykara E, Tuladhar AM, Gesierich B, Konieczny MJ, Malik R, Franzmeier N, Ewers M, Jouvent E, Biessels GJ, Schmidt R, de Leeuw FE, Pasternak O, Dichgans M. Free water determines diffusion alterations and clinical status in cerebral small vessel disease. Alzheimers Dement 2018; 14:764-774. [PMID: 29406155 PMCID: PMC5994358 DOI: 10.1016/j.jalz.2017.12.007] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 12/16/2017] [Accepted: 12/19/2017] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Diffusion tensor imaging detects early tissue alterations in Alzheimer's disease and cerebral small vessel disease (SVD). However, the origin of diffusion alterations in SVD is largely unknown. METHODS To gain further insight, we applied free water (FW) imaging to patients with genetically defined SVD (Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy [CADASIL], n = 57), sporadic SVD (n = 444), and healthy controls (n = 28). We modeled freely diffusing water in the extracellular space (FW) and measures reflecting fiber structure (tissue compartment). We tested associations between these measures and clinical status (processing speed and disability). RESULTS Diffusion alterations in SVD were mostly driven by increased FW and less by tissue compartment alterations. Among imaging markers, FW showed the strongest association with clinical status (R2 up to 34%, P < .0001). Findings were consistent across patients with CADASIL and sporadic SVD. DISCUSSION Diffusion alterations and clinical status in SVD are largely determined by extracellular fluid increase rather than alterations of white matter fiber organization.
Collapse
Affiliation(s)
- Marco Duering
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany.
| | - Sofia Finsterwalder
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Ebru Baykara
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Anil Man Tuladhar
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Benno Gesierich
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Marek J Konieczny
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Rainer Malik
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Michael Ewers
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Eric Jouvent
- Univ Paris Diderot, DHU NeuroVasc Sorbonne Paris Cité, UMR-S 1161 INSERM, Paris, France; Department of Neurology, Assistance publique - hôpitaux de Paris (AP-HP), Lariboisière Hospital, Paris, France
| | - Geert Jan Biessels
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Reinhold Schmidt
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Frank-Erik de Leeuw
- Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Ofer Pasternak
- Departments of Psychiatry and Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| |
Collapse
|
140
|
Raja R, Rosenberg GA, Caprihan A. MRI measurements of Blood-Brain Barrier function in dementia: A review of recent studies. Neuropharmacology 2018; 134:259-271. [PMID: 29107626 PMCID: PMC6044415 DOI: 10.1016/j.neuropharm.2017.10.034] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 10/24/2017] [Accepted: 10/26/2017] [Indexed: 12/26/2022]
Abstract
Blood-brain barrier (BBB) separates the systemic circulation and the brain, regulating transport of most molecules to protect the brain microenvironment. Multiple structural and functional components preserve the integrity of the BBB. Several imaging modalities are available to study disruption of the BBB. However, the subtle changes in BBB leakage that occurs in vascular cognitive impairment and Alzheimer's disease have been less well studied. Dynamic contrast enhanced magnetic resonance imaging (DCE-MRI) is the most widely adopted non-invasive imaging technique for evaluating BBB breakdown. It is used as a significant marker for a wide variety of diseases with large permeability leaks, such as brain tumors and multiple sclerosis, to more subtle disruption in chronic vascular disease and dementia. DCE-MRI analysis of BBB includes both model-free parameters and quantitative parameters using pharmacokinetic modelling. We review MRI studies of BBB breakdown in dementia. The challenges in measuring subtle BBB changes and the state of the art techniques are initially examined. Subsequently, a systematic review comparing methodologies from recent in-vivo MRI studies is presented. Various factors related to subtle BBB permeability measurement such as DCE-MRI acquisition parameters, arterial input assessment, T1 mapping and data analysis methods are reviewed with the focus on finding the optimal technique. Finally, the reported BBB permeability values in dementia are compared across different studies and across various brain regions. We conclude that reliable measurement of low-level BBB permeability across sites remains a difficult problem and a standardization of the methodology for both data acquisition and quantitative analysis is required. This article is part of the Special Issue entitled 'Cerebral Ischemia'.
Collapse
Affiliation(s)
| | - Gary A Rosenberg
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | | |
Collapse
|
141
|
Li Y, Li M, Zuo L, Shi Q, Qin W, Yang L, Jiang T, Hu W. Compromised Blood-Brain Barrier Integrity Is Associated With Total Magnetic Resonance Imaging Burden of Cerebral Small Vessel Disease. Front Neurol 2018; 9:221. [PMID: 29681883 PMCID: PMC5897516 DOI: 10.3389/fneur.2018.00221] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 03/21/2018] [Indexed: 11/17/2022] Open
Abstract
Objective Several studies have demonstrated that compromised blood–brain barrier (BBB) integrity may play a pivotal role in the pathogenesis of individual cerebral small vessel disease (cSVD) markers, but the association between BBB permeability and total magnetic resonance imaging (MRI) cSVD burden remains unclear. This study aimed to investigate the relationship between BBB permeability and total MRI cSVD burden. Methods Consecutive participants without symptomatic stroke history presented for physical examination were enrolled in this cross-sectional study. The presence of lacunes, white matter hyperintensities (WMH), cerebral microbleeds, and enlarged perivascular spaces was recorded in an ordinal score (range 0–4). We used dynamic contrast-enhanced-MRI and Patlak pharmacokinetic model to quantify BBB permeability in the normal-appearing white matter (NAWM), WMH, cortical gray matter (CGM), and deep gray matter (DGM). Results All 99 participants averaged 70.33 years old (49–90 years). Multivariable linear regression analyses adjusted for age, sex, and vascular risk factors showed that leakage rate and area under the leakage curve in the NAWM, WMH, CGM, and DGM were positively associated with total MRI cSVD burden (all P < 0.01). Moreover, fractional blood plasma volumes in the NAWM, CGM, and DGM were negatively associated with total MRI cSVD burden (all P < 0.05). Conclusion This study verified that compromised BBB integrity is associated with total MRI cSVD burden, suggesting that BBB dysfunction may be a critical contributor to the pathogenesis of cSVD. Longitudinal studies are required to determine whether there is a causal relationship between BBB permeability and total MRI cSVD burden.
Collapse
Affiliation(s)
- Yue Li
- Department of Neurology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Man Li
- Department of Radiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Long Zuo
- Department of Radiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Qinglei Shi
- Diagnosis Imaging, Siemens Healthcare Ltd., Beijing, China
| | - Wei Qin
- Department of Neurology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Lei Yang
- Department of Neurology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Tao Jiang
- Department of Radiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Wenli Hu
- Department of Neurology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
142
|
Progress in brain barriers and brain fluid research in 2017. Fluids Barriers CNS 2018; 15:6. [PMID: 29391031 PMCID: PMC5796342 DOI: 10.1186/s12987-018-0091-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 01/22/2018] [Indexed: 12/11/2022] Open
Abstract
The past year, 2017, has seen many important papers published in the fields covered by Fluids and Barriers of the CNS. This article from the Editors highlights some.
Collapse
|
143
|
Fu Y, Yan Y. Emerging Role of Immunity in Cerebral Small Vessel Disease. Front Immunol 2018; 9:67. [PMID: 29422904 PMCID: PMC5788893 DOI: 10.3389/fimmu.2018.00067] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 01/10/2018] [Indexed: 01/06/2023] Open
Abstract
Cerebral small vessel disease (CSVD) is one of the main causes of vascular dementia in older individuals. Apart from risk containment, efforts to prevent or treat CSVD are ineffective due to the unknown pathogenesis of the disease. CSVD, a subtype of stroke, is characterized by recurrent strokes and neurodegeneration. Blood-brain barrier (BBB) impairment, chronic inflammatory responses, and leukocyte infiltration are classical pathological features of CSVD. Understanding how BBB disruption instigates inflammatory and degenerative processes may be informative for CSVD therapy. Antigens derived from the brain are found in the peripheral blood of lacunar stroke patients, and antibodies and sensitized T cells against brain antigens are also detected in patients with leukoaraiosis. These findings suggest that antigen-specific immune responses could occur in CSVD. This review describes the neurovascular unit features of CSVD, the immune responses to specific neuronal and glial processes that may be involved in a distinct mechanism of CSVD, and the current evidence of the association between mechanisms of inflammation and interventions in CSVD. We suggest that autoimmune activity should be assessed in future studies; this knowledge would benefit the development of effective therapeutic interventions in CSVD.
Collapse
Affiliation(s)
- Ying Fu
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yaping Yan
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| |
Collapse
|
144
|
Chen Y, Yan S, Xu M, Zhong G, Liebeskind DS, Lou M. More extensive white matter hyperintensity is linked with higher risk of remote intracerebral hemorrhage after intravenous thrombolysis. Eur J Neurol 2017; 25:380-e15. [PMID: 29115734 DOI: 10.1111/ene.13517] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 11/02/2017] [Indexed: 11/27/2022]
Affiliation(s)
- Y. Chen
- Department of Neurology The Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou China
| | - S. Yan
- Department of Neurology The Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou China
| | - M. Xu
- Department of Neurology The Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou China
| | - G. Zhong
- Department of Neurology The Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou China
| | | | - M. Lou
- Department of Neurology The Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou China
| |
Collapse
|
145
|
Staffaroni AM, Elahi FM, McDermott D, Marton K, Karageorgiou E, Sacco S, Paoletti M, Caverzasi E, Hess CP, Rosen HJ, Geschwind MD. Neuroimaging in Dementia. Semin Neurol 2017; 37:510-537. [PMID: 29207412 PMCID: PMC5823524 DOI: 10.1055/s-0037-1608808] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Although the diagnosis of dementia still is primarily based on clinical criteria, neuroimaging is playing an increasingly important role. This is in large part due to advances in techniques that can assist with discriminating between different syndromes. Magnetic resonance imaging remains at the core of differential diagnosis, with specific patterns of cortical and subcortical changes having diagnostic significance. Recent developments in molecular PET imaging techniques have opened the door for not only antemortem but early, even preclinical, diagnosis of underlying pathology. This is vital, as treatment trials are underway for pharmacological agents with specific molecular targets, and numerous failed trials suggest that earlier treatment is needed. This article provides an overview of classic neuroimaging findings as well as new and cutting-edge research techniques that assist with clinical diagnosis of a range of dementia syndromes, with an emphasis on studies using pathologically proven cases.
Collapse
Affiliation(s)
- Adam M. Staffaroni
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California
| | - Fanny M. Elahi
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California
| | - Dana McDermott
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California
| | - Kacey Marton
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California
| | - Elissaios Karageorgiou
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California
- Neurological Institute of Athens, Athens, Greece
| | - Simone Sacco
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California
- Institute of Radiology, Department of Clinical Surgical Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Matteo Paoletti
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California
- Institute of Radiology, Department of Clinical Surgical Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Eduardo Caverzasi
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Christopher P. Hess
- Division of Neuroradiology, Department of Radiology, University of California, San Francisco (UCSF), California
| | - Howard J. Rosen
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California
| | - Michael D. Geschwind
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California
| |
Collapse
|
146
|
Hainsworth AH, Minett T, Andoh J, Forster G, Bhide I, Barrick TR, Elderfield K, Jeevahan J, Markus HS, Bridges LR. Neuropathology of White Matter Lesions, Blood-Brain Barrier Dysfunction, and Dementia. Stroke 2017; 48:2799-2804. [PMID: 28855392 DOI: 10.1161/strokeaha.117.018101] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 07/28/2017] [Accepted: 08/01/2017] [Indexed: 01/17/2023]
Abstract
BACKGROUND AND PURPOSE We tested whether blood-brain barrier dysfunction in subcortical white matter is associated with white matter abnormalities or risk of clinical dementia in older people (n=126; mean age 86.4, SD: 7.7 years) in the MRC CFAS (Medical Research Council Cognitive Function and Ageing Study). METHODS Using digital pathology, we quantified blood-brain barrier dysfunction (defined by immunohistochemical labeling for the plasma marker fibrinogen). This was assessed within subcortical white matter tissue samples harvested from postmortem T2 magnetic resonance imaging (MRI)-detected white matter hyperintensities, from normal-appearing white matter (distant from coexistent MRI-defined hyperintensities), and from equivalent areas in MRI normal brains. Histopathologic lesions were defined using a marker for phagocytic microglia (CD68, clone PGM1). RESULTS Extent of fibrinogen labeling was not significantly associated with white matter abnormalities defined either by MRI (odds ratio, 0.90; 95% confidence interval, 0.79-1.03; P=0.130) or by histopathology (odds ratio, 0.93; 95% confidence interval, 0.77-1.12; P=0.452). Among participants with normal MRI (no detectable white matter hyperintensities), increased fibrinogen was significantly related to decreased risk of clinical dementia (odds ratio, 0.74; 95% confidence interval, 0.58-0.94; P=0.013). Among participants with histological lesions, increased fibrinogen was related to increased risk of dementia (odds ratio, 2.26; 95% confidence interval, 1.25-4.08; P=0.007). CONCLUSIONS Our data suggest that some degree of blood-brain barrier dysfunction is common in older people and that this may be related to clinical dementia risk, additional to standard MRI biomarkers.
Collapse
Affiliation(s)
- Atticus H Hainsworth
- From the Cell Biology and Genetics Research Centre (A.H.H., J.A., I.B., L.R.B.) and Neuroscience Research Centre (A.H.H., J.A., I.B., T.R.B., L.R.B.), Molecular and Clinical Sciences Research Institute, St George's University of London, United Kingdom; Department of Neurology (A.H.H.) and Department of Cellular Pathology (K.E., J.J., L.R.B.), St George's University Hospitals NHS Foundation Trust, London, United Kingdom; Department of Public Health and Primary Care (T.M.), Department of Radiology (T.M.), and Stroke Research Group, Department of Clinical Neurosciences (H.S.M.), University of Cambridge, United Kingdom; and The Sheffield Institute for Translational Neuroscience, University of Sheffield, United Kingdom (G.F.).
| | - Thais Minett
- From the Cell Biology and Genetics Research Centre (A.H.H., J.A., I.B., L.R.B.) and Neuroscience Research Centre (A.H.H., J.A., I.B., T.R.B., L.R.B.), Molecular and Clinical Sciences Research Institute, St George's University of London, United Kingdom; Department of Neurology (A.H.H.) and Department of Cellular Pathology (K.E., J.J., L.R.B.), St George's University Hospitals NHS Foundation Trust, London, United Kingdom; Department of Public Health and Primary Care (T.M.), Department of Radiology (T.M.), and Stroke Research Group, Department of Clinical Neurosciences (H.S.M.), University of Cambridge, United Kingdom; and The Sheffield Institute for Translational Neuroscience, University of Sheffield, United Kingdom (G.F.)
| | - Joycelyn Andoh
- From the Cell Biology and Genetics Research Centre (A.H.H., J.A., I.B., L.R.B.) and Neuroscience Research Centre (A.H.H., J.A., I.B., T.R.B., L.R.B.), Molecular and Clinical Sciences Research Institute, St George's University of London, United Kingdom; Department of Neurology (A.H.H.) and Department of Cellular Pathology (K.E., J.J., L.R.B.), St George's University Hospitals NHS Foundation Trust, London, United Kingdom; Department of Public Health and Primary Care (T.M.), Department of Radiology (T.M.), and Stroke Research Group, Department of Clinical Neurosciences (H.S.M.), University of Cambridge, United Kingdom; and The Sheffield Institute for Translational Neuroscience, University of Sheffield, United Kingdom (G.F.)
| | - Gillian Forster
- From the Cell Biology and Genetics Research Centre (A.H.H., J.A., I.B., L.R.B.) and Neuroscience Research Centre (A.H.H., J.A., I.B., T.R.B., L.R.B.), Molecular and Clinical Sciences Research Institute, St George's University of London, United Kingdom; Department of Neurology (A.H.H.) and Department of Cellular Pathology (K.E., J.J., L.R.B.), St George's University Hospitals NHS Foundation Trust, London, United Kingdom; Department of Public Health and Primary Care (T.M.), Department of Radiology (T.M.), and Stroke Research Group, Department of Clinical Neurosciences (H.S.M.), University of Cambridge, United Kingdom; and The Sheffield Institute for Translational Neuroscience, University of Sheffield, United Kingdom (G.F.)
| | - Ishaan Bhide
- From the Cell Biology and Genetics Research Centre (A.H.H., J.A., I.B., L.R.B.) and Neuroscience Research Centre (A.H.H., J.A., I.B., T.R.B., L.R.B.), Molecular and Clinical Sciences Research Institute, St George's University of London, United Kingdom; Department of Neurology (A.H.H.) and Department of Cellular Pathology (K.E., J.J., L.R.B.), St George's University Hospitals NHS Foundation Trust, London, United Kingdom; Department of Public Health and Primary Care (T.M.), Department of Radiology (T.M.), and Stroke Research Group, Department of Clinical Neurosciences (H.S.M.), University of Cambridge, United Kingdom; and The Sheffield Institute for Translational Neuroscience, University of Sheffield, United Kingdom (G.F.)
| | - Thomas R Barrick
- From the Cell Biology and Genetics Research Centre (A.H.H., J.A., I.B., L.R.B.) and Neuroscience Research Centre (A.H.H., J.A., I.B., T.R.B., L.R.B.), Molecular and Clinical Sciences Research Institute, St George's University of London, United Kingdom; Department of Neurology (A.H.H.) and Department of Cellular Pathology (K.E., J.J., L.R.B.), St George's University Hospitals NHS Foundation Trust, London, United Kingdom; Department of Public Health and Primary Care (T.M.), Department of Radiology (T.M.), and Stroke Research Group, Department of Clinical Neurosciences (H.S.M.), University of Cambridge, United Kingdom; and The Sheffield Institute for Translational Neuroscience, University of Sheffield, United Kingdom (G.F.)
| | - Kay Elderfield
- From the Cell Biology and Genetics Research Centre (A.H.H., J.A., I.B., L.R.B.) and Neuroscience Research Centre (A.H.H., J.A., I.B., T.R.B., L.R.B.), Molecular and Clinical Sciences Research Institute, St George's University of London, United Kingdom; Department of Neurology (A.H.H.) and Department of Cellular Pathology (K.E., J.J., L.R.B.), St George's University Hospitals NHS Foundation Trust, London, United Kingdom; Department of Public Health and Primary Care (T.M.), Department of Radiology (T.M.), and Stroke Research Group, Department of Clinical Neurosciences (H.S.M.), University of Cambridge, United Kingdom; and The Sheffield Institute for Translational Neuroscience, University of Sheffield, United Kingdom (G.F.)
| | - Jamuna Jeevahan
- From the Cell Biology and Genetics Research Centre (A.H.H., J.A., I.B., L.R.B.) and Neuroscience Research Centre (A.H.H., J.A., I.B., T.R.B., L.R.B.), Molecular and Clinical Sciences Research Institute, St George's University of London, United Kingdom; Department of Neurology (A.H.H.) and Department of Cellular Pathology (K.E., J.J., L.R.B.), St George's University Hospitals NHS Foundation Trust, London, United Kingdom; Department of Public Health and Primary Care (T.M.), Department of Radiology (T.M.), and Stroke Research Group, Department of Clinical Neurosciences (H.S.M.), University of Cambridge, United Kingdom; and The Sheffield Institute for Translational Neuroscience, University of Sheffield, United Kingdom (G.F.)
| | - Hugh S Markus
- From the Cell Biology and Genetics Research Centre (A.H.H., J.A., I.B., L.R.B.) and Neuroscience Research Centre (A.H.H., J.A., I.B., T.R.B., L.R.B.), Molecular and Clinical Sciences Research Institute, St George's University of London, United Kingdom; Department of Neurology (A.H.H.) and Department of Cellular Pathology (K.E., J.J., L.R.B.), St George's University Hospitals NHS Foundation Trust, London, United Kingdom; Department of Public Health and Primary Care (T.M.), Department of Radiology (T.M.), and Stroke Research Group, Department of Clinical Neurosciences (H.S.M.), University of Cambridge, United Kingdom; and The Sheffield Institute for Translational Neuroscience, University of Sheffield, United Kingdom (G.F.)
| | - Leslie R Bridges
- From the Cell Biology and Genetics Research Centre (A.H.H., J.A., I.B., L.R.B.) and Neuroscience Research Centre (A.H.H., J.A., I.B., T.R.B., L.R.B.), Molecular and Clinical Sciences Research Institute, St George's University of London, United Kingdom; Department of Neurology (A.H.H.) and Department of Cellular Pathology (K.E., J.J., L.R.B.), St George's University Hospitals NHS Foundation Trust, London, United Kingdom; Department of Public Health and Primary Care (T.M.), Department of Radiology (T.M.), and Stroke Research Group, Department of Clinical Neurosciences (H.S.M.), University of Cambridge, United Kingdom; and The Sheffield Institute for Translational Neuroscience, University of Sheffield, United Kingdom (G.F.)
| |
Collapse
|
147
|
Hypercholesterolemia induced cerebral small vessel disease. PLoS One 2017; 12:e0182822. [PMID: 28796818 PMCID: PMC5552130 DOI: 10.1371/journal.pone.0182822] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 07/25/2017] [Indexed: 01/01/2023] Open
Abstract
Background While hypercholesterolemia plays a causative role for the development of ischemic stroke in large vessels, its significance for cerebral small vessel disease (CSVD) remains unclear. We thus aimed to understand the detailed relationship between hypercholesterolemia and CSVD using the well described Ldlr-/- mouse model. Methods We used Ldlr-/- mice (n = 16) and wild-type (WT) mice (n = 15) at the age of 6 and 12 months. Ldlr-/- mice develop high plasma cholesterol levels following a high fat diet. We analyzed cerebral capillaries and arterioles for intravascular erythrocyte accumulations, thrombotic vessel occlusions, blood-brain barrier (BBB) dysfunction and microbleeds. Results We found a significant increase in the number of erythrocyte stases in 6 months old Ldlr-/- mice compared to all other groups (P < 0.05). Ldlr-/- animals aged 12 months showed the highest number of thrombotic occlusions while in WT animals hardly any occlusions could be observed (P < 0.001). Compared to WT mice, Ldlr-/- mice did not display significant gray matter BBB breakdown. Microhemorrhages were observed in one Ldlr-/- mouse that was 6 months old. Results did not differ when considering subcortical and cortical regions. Conclusions In Ldlr-/- mice, hypercholesterolemia is related to a thrombotic CSVD phenotype, which is different from hypertension-related CSVD that associates with a hemorrhagic CSVD phenotype. Our data demonstrate a relationship between hypercholesterolemia and the development of CSVD. Ldlr-/- mice appear to be an adequate animal model for research into CSVD.
Collapse
|
148
|
Li Y, Li M, Zhang X, Shi Q, Yang S, Fan H, Qin W, Yang L, Yuan J, Jiang T, Hu W. Higher blood-brain barrier permeability is associated with higher white matter hyperintensities burden. J Neurol 2017; 264:1474-1481. [PMID: 28653212 DOI: 10.1007/s00415-017-8550-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 06/15/2017] [Accepted: 06/16/2017] [Indexed: 01/11/2023]
Abstract
The pathogenesis of white matter hyperintensities (WMH) is incompletely understood but blood-brain barrier (BBB) dysfunction may play a key role. This study aimed to investigate the relationship between BBB permeability and the severity of WMH burden. Consecutive participants without symptomatic stroke history presented for physical examination were recruited in this cross-sectional study and divided into three WMH burden groups according to total Fazekas scores. They received dynamic contrast-enhanced-magnetic resonance imaging to measure BBB permeability, and received Mini-Mental State Examination (MMSE) and Montreal Cognitive Assessment (MoCA). A total of 102 participants aged 49-90 years (mean age of 69.82 years) were enrolled (36 with low WMH burden, 35 with medium WMH burden, and 31 with high WMH burden). Multivariable linear regression analyses revealed that participants with higher WMH burden had significantly higher BBB leakage rate and area under the leakage curve in normal-appearing white matter, WMH, cortical gray matter, and deep gray matter (DGM) after adjustment for age, sex, and vascular risk factors. Scores on MMSE and MoCA decreased with increasing leakage rate in WMH and DGM after adjustment for age, sex, WMH burden, and education years. We found that higher BBB permeability is associated with higher WMH burden and cognitive decline. The compromised BBB integrity may be a critical contributor to the pathogenesis of WMH and part of a series of pathological processes that finally lead to cognitive impairment.
Collapse
Affiliation(s)
- Yue Li
- Department of Neurology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, South Gongti Road, Beijing, 100020, People's Republic of China
| | - Man Li
- Department of Radiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Xiaoyu Zhang
- Department of Neurology, Qianfoshan Hospital, Shandong University, Beijing, People's Republic of China
| | - Qinglei Shi
- Diagnosis Imaging, Siemens Healthcare Ltd., Beijing, People's Republic of China
| | - Shuna Yang
- Department of Neurology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, South Gongti Road, Beijing, 100020, People's Republic of China
| | - Huimin Fan
- Department of Neurology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, South Gongti Road, Beijing, 100020, People's Republic of China
| | - Wei Qin
- Department of Neurology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, South Gongti Road, Beijing, 100020, People's Republic of China
| | - Lei Yang
- Department of Neurology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, South Gongti Road, Beijing, 100020, People's Republic of China
| | - Junliang Yuan
- Department of Neurology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, South Gongti Road, Beijing, 100020, People's Republic of China
| | - Tao Jiang
- Department of Radiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Wenli Hu
- Department of Neurology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, South Gongti Road, Beijing, 100020, People's Republic of China.
| |
Collapse
|
149
|
Hainsworth AH, Fisher MJ. A dysfunctional blood-brain barrier and cerebral small vessel disease. Neurology 2016; 88:420-421. [PMID: 28031393 DOI: 10.1212/wnl.0000000000003561] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- Atticus H Hainsworth
- From the Molecular and Clinical Sciences Research Institute (A.H.H.), St Georges University of London; Neurology (A.H.H.), St George's University Hospitals NHS Foundation Trust, London, UK; and Departments of Neurology, Anatomy & Neurobiology, and Pathology & Laboratory Medicine (M.J.F.), University of California, Irvine.
| | - Mark J Fisher
- From the Molecular and Clinical Sciences Research Institute (A.H.H.), St Georges University of London; Neurology (A.H.H.), St George's University Hospitals NHS Foundation Trust, London, UK; and Departments of Neurology, Anatomy & Neurobiology, and Pathology & Laboratory Medicine (M.J.F.), University of California, Irvine.
| |
Collapse
|