1
|
Isik AT, Kaya D, Gokden M. Brain Banking in Dementia Studies. Methods Mol Biol 2024; 2785:287-295. [PMID: 38427200 DOI: 10.1007/978-1-0716-3774-6_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
It is now well-established practice in dementia that one clinical entity may be caused by various neurodegenerative disorders, each with different histopathological findings, whereas neuropathologically confirmed patients may have different, unusual, and atypical clinical manifestations.This inconsistency in dementia patients leads to neuropathological examination of cases, and neuropathological examination seems to be an inevitable part of dementia practice, at least until all clinical entities are properly identified for humans.Additionally, the development of disease-modifying therapies and confirmation of the actual accurate diagnosis of the neurodegenerative disease that the drug is thought to modify or act upon are of great importance for neuropathological evaluation in brain banks.Neuropathological processes coexisting among patients diagnosed with established clinical criteria or international guidelines have provided a new perspective in the context of drug development.Here, we review our routinely used methodology in the context of the brain banking process.
Collapse
Affiliation(s)
- Ahmet Turan Isik
- Unit for Aging Brain and Dementia, Department of Geriatric Medicine, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Derya Kaya
- Unit for Aging Brain and Dementia, Department of Geriatric Medicine, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Murat Gokden
- Division of Neuropathology, Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
2
|
Hainsworth AH, Markus HS, Schneider JA. Cerebral Small Vessel Disease, Hypertension, and Vascular Contributions to Cognitive Impairment and Dementia. Hypertension 2024; 81:75-86. [PMID: 38044814 PMCID: PMC10734789 DOI: 10.1161/hypertensionaha.123.19943] [Citation(s) in RCA: 54] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Hypertension-associated cerebral small vessel disease is a common finding in older people. Strongly associated with age and hypertension, small vessel disease is found at autopsy in over 50% of people aged ≥65 years, with a spectrum of clinical manifestations. It is the main cause of lacunar stroke and a major source of vascular contributions to cognitive impairment and dementia. The brain areas affected are subcortical and periventricular white matter and deep gray nuclei. Neuropathological sequelae are diffuse white matter lesions (seen as white matter hyperintensities on T2-weighted magnetic resonance imaging), small ischemic foci (lacunes or microinfarcts), and less commonly, subcortical microhemorrhages. The most common form of cerebral small vessel disease is concentric, fibrotic thickening of small penetrating arteries (up to 300 microns outer diameter) termed arteriolosclerosis. Less common forms are small artery atheroma and lipohyalinosis (the lesions described by C. Miller Fisher adjacent to lacunes). Other microvascular lesions that are not reviewed here include cerebral amyloid angiopathy and venous collagenosis. Here, we review the epidemiology, neuropathology, clinical management, genetics, preclinical models, and pathogenesis of hypertensive small vessel disease. Knowledge gaps include initiating factors, molecular pathogenesis, relationships between arterial pathology and tissue damage, possible reversibility, pharmacological targets, and molecular biomarkers. Progress is anticipated from multicell transcriptomic and proteomic profiling, novel experimental models and further target-finding and interventional clinical studies.
Collapse
Affiliation(s)
- Atticus H. Hainsworth
- Molecular and Clinical Sciences Research Institute, St George’s University of London, United Kingdom (A.H.H.)
- Department of Neurology, St George’s University Hospitals NHS Foundation Trust, London, United Kingdom (A.H.H.)
| | - Hugh S. Markus
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, United Kingdom (H.S.M.)
| | - Julie A. Schneider
- Rush Alzheimer’s Disease Center, Departments of Pathology and Neurological Sciences, Rush University Medical Center, Chicago, IL (J.A.S.)
| |
Collapse
|
3
|
Scheuermann BC, Parr SK, Schulze KM, Kunkel ON, Turpin VG, Liang J, Ade CJ. Associations of Cerebrovascular Regulation and Arterial Stiffness With Cerebral Small Vessel Disease: A Systematic Review and Meta-Analysis. J Am Heart Assoc 2023; 12:e032616. [PMID: 37930079 PMCID: PMC10727345 DOI: 10.1161/jaha.123.032616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 10/24/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND Cerebral small vessel disease (cSVD) is a major contributing factor to ischemic stroke and dementia. However, the vascular pathologies of cSVD remain inconclusive. The aim of this systematic review and meta-analysis was to characterize the associations between cSVD and cerebrovascular reactivity (CVR), cerebral autoregulation, and arterial stiffness (AS). METHODS AND RESULTS MEDLINE, Web of Science, and Embase were searched from inception to September 2023 for studies reporting CVR, cerebral autoregulation, or AS in relation to radiological markers of cSVD. Data were extracted in predefined tables, reviewed, and meta-analyses performed using inverse-variance random effects models to determine pooled odds ratios (ORs). A total of 1611 studies were identified; 142 were included in the systematic review, of which 60 had data available for meta-analyses. Systematic review revealed that CVR, cerebral autoregulation, and AS were consistently associated with cSVD (80.4%, 78.6%, and 85.4% of studies, respectively). Meta-analysis in 7 studies (536 participants, 32.9% women) revealed a borderline association between impaired CVR and cSVD (OR, 2.26 [95% CI, 0.99-5.14]; P=0.05). In 37 studies (27 952 participants, 53.0% women) increased AS, per SD, was associated with cSVD (OR, 1.24 [95% CI, 1.15-1.33]; P<0.01). Meta-regression adjusted for comorbidities accounted for one-third of the AS model variance (R2=29.4%, Pmoderators=0.02). Subgroup analysis of AS studies demonstrated an association with white matter hyperintensities (OR, 1.42 [95% CI, 1.18-1.70]; P<0.01). CONCLUSIONS The collective findings of the present systematic review and meta-analyses suggest an association between cSVD and impaired CVR and elevated AS. However, longitudinal investigations into vascular stiffness and regulatory function as possible risk factors for cSVD remain warranted.
Collapse
Affiliation(s)
| | - Shannon K. Parr
- Department of KinesiologyKansas State UniversityManhattanKSUSA
| | | | | | | | - Jia Liang
- Department of Biostatistics, St. Jude Children’s Research HospitalMemphisTNUSA
| | - Carl J. Ade
- Department of KinesiologyKansas State UniversityManhattanKSUSA
- Department of Physician’s Assistant Studies, Kansas State UniversityManhattanKSUSA
- Johnson Cancer Research CenterKansas State UniversityManhattanKSUSA
| |
Collapse
|
4
|
Bah TM, Siler DA, Ibrahim AH, Cetas JS, Alkayed NJ. Fluid dynamics in aging-related dementias. Neurobiol Dis 2023; 177:105986. [PMID: 36603747 DOI: 10.1016/j.nbd.2022.105986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/22/2022] [Accepted: 12/31/2022] [Indexed: 01/03/2023] Open
Abstract
Recent human and animal model experimental studies revealed novel pathways for fluid movement, immune cell trafficking and metabolic waste clearance in CNS. These studies raise the intriguing possibility that the newly discovered pathways, including the glymphatic system, lymphatic meningeal vessels and skull-brain communication channels, are impaired in aging and neurovascular and neurodegenerative diseases associated with dementia, including Alzheimer's disease (AD) and AD-related dementia. We provide an overview of the glymphatic and dural meningeal lymphatic systems, review current methods and approaches used to study glymphatic flow in humans and animals, and discuss current evidence and controversies related to its role in CNS flow homeostasis under physiological and pathophysiological conditions. Non-invasive imaging approaches are needed to fully understand the mechanisms and pathways driving fluid movement in CNS and their roles across lifespan including healthy aging and aging-related dementia.
Collapse
Affiliation(s)
- Thierno M Bah
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Dominic A Siler
- Department of Neurological Surgery, Oregon Health & Science University, Portland, OR, USA
| | - Aseel H Ibrahim
- Department of Neurosurgery, University of Arizona, Tucson, AZ, USA
| | - Justin S Cetas
- Department of Neurosurgery, University of Arizona, Tucson, AZ, USA
| | - Nabil J Alkayed
- Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA; Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
5
|
Moretti R, Caruso P. Small Vessel Disease: Ancient Description, Novel Biomarkers. Int J Mol Sci 2022; 23:3508. [PMID: 35408867 PMCID: PMC8998274 DOI: 10.3390/ijms23073508] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/20/2022] [Accepted: 03/21/2022] [Indexed: 12/22/2022] Open
Abstract
Small vessel disease (SVD) is one of the most frequent pathological conditions which lead to dementia. Biochemical and neuroimaging might help correctly identify the clinical diagnosis of this relevant brain disease. The microvascular alterations which underlie SVD have common origins, similar cognitive outcomes, and common vascular risk factors. Nevertheless, the arteriolosclerosis process, which underlines SVD development, is based on different mechanisms, not all completely understood, which start from a chronic hypoperfusion state and pass through a chronic brain inflammatory condition, inducing a significant endothelium activation and a consequent tissue remodeling action. In a recent review, we focused on the pathophysiology of SVD, which is complex, involving genetic conditions and different co-morbidities (i.e., diabetes, chronic hypoxia condition, and obesity). Currently, many points still remain unclear and discordant. In this paper, we wanted to focus on new biomarkers, which can be the expression of the endothelial dysfunction, or of the oxidative damage, which could be employed as markers of disease progression or for future targets of therapies. Therefore, we described the altered response to the endothelium-derived nitric oxide-vasodilators (ENOV), prostacyclin, C-reactive proteins, and endothelium-derived hyperpolarizing factors (EDHF). At the same time, due to the concomitant endothelial activation and chronic neuroinflammatory status, we described hypoxia-endothelial-related markers, such as HIF 1 alpha, VEGFR2, and neuroglobin, and MMPs. We also described blood-brain barrier disruption biomarkers and imaging techniques, which can also describe perivascular spaces enlargement and dysfunction. More studies should be necessary, in order to implement these results and give them a clinical benefit.
Collapse
Affiliation(s)
- Rita Moretti
- Neurology Clinic, Department of Medical, Surgical and Health Sciences, University of Trieste, 34127 Trieste, Italy;
| | | |
Collapse
|
6
|
Anad A, Barker MK, Katanga JA, Arfanakis K, Bridges LR, Esiri MM, Isaacs JD, Prpar Mihevc S, Pereira AC, Schneider JA, Hainsworth AH. Vasculocentric Axonal NfH in Small Vessel Disease. J Neuropathol Exp Neurol 2022; 81:182-192. [PMID: 35086142 PMCID: PMC8922195 DOI: 10.1093/jnen/nlab134] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Cerebral small vessel disease (SVD) causes lacunar stroke and vascular cognitive impairment in older people. The pathogenic pathways from vessel pathology to parenchymal damage in SVD are unknown. Neurofilaments are axonal structural proteins. Neurofilament-light (NfL) is an emerging biomarker for neurological disease. Here, we examined the high molecular weight form neurofilament-heavy (NfH) and quantified a characteristic pattern of peri-arterial (vasculocentric) NfH labeling. Subcortical frontal and parietal white matter from young adult controls, aged controls, and older people with SVD or severe Alzheimer disease (n = 52) was immunohistochemically labeled for hyperphosphorylated NfH (pNfH). The extent of pNfH immunolabeling and the degree of vasculocentric axonal pNfH were quantified. Axonal pNfH immunolabeling was sparse in young adults but a common finding in older persons (controls, SVD, or AD). Axonal pNfH was often markedly concentrated around small penetrating arteries. This vasculocentric feature was more common in older people with SVD than in those with severe AD (p = 0.004). We conclude that axonal pNfH is a feature of subcortical white matter in aged brains. Vasculocentric axonal pNfH is a novel parenchymal lesion that is co-located with SVD arteriopathy and could be a consequence of vessel pathology.
Collapse
Affiliation(s)
- Adam Anad
- From the Molecular and Clinical Sciences Research Institute, St George’s University of London, London, UK (AA, MKB, JAK, LRB, JDI, ACP, AHH)
| | - Miriam K Barker
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, Illinois, USA (KA, JAS)
| | - Jessica A Katanga
- From the Molecular and Clinical Sciences Research Institute, St George’s University of London, London, UK (AA, MKB, JAK, LRB, JDI, ACP, AHH)
| | - Konstantinos Arfanakis
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, Illinois, USA (KA, JAS)
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, Illinois, USA (KA)
| | - Leslie R Bridges
- From the Molecular and Clinical Sciences Research Institute, St George’s University of London, London, UK (AA, MKB, JAK, LRB, JDI, ACP, AHH)
- Department of Cellular Pathology, St George’s University Hospitals NHS Foundation Trust, London, UK (LRB)
| | - Margaret M Esiri
- Nuffield Department of Clinical Neurosciences, Oxford University, Oxford, UK (MME)
| | - Jeremy D Isaacs
- From the Molecular and Clinical Sciences Research Institute, St George’s University of London, London, UK (AA, MKB, JAK, LRB, JDI, ACP, AHH)
- Department of Neurology, St George’s University Hospitals NHS Foundation Trust, London, UK (JDI, ACP, AHH)
| | - Sonja Prpar Mihevc
- Institute for Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Ljubljana, Slovenia (SPM)
| | - Anthony C Pereira
- From the Molecular and Clinical Sciences Research Institute, St George’s University of London, London, UK (AA, MKB, JAK, LRB, JDI, ACP, AHH)
- Department of Neurology, St George’s University Hospitals NHS Foundation Trust, London, UK (JDI, ACP, AHH)
| | - Julie A Schneider
- From the Molecular and Clinical Sciences Research Institute, St George’s University of London, London, UK (AA, MKB, JAK, LRB, JDI, ACP, AHH)
| | - Atticus H Hainsworth
- From the Molecular and Clinical Sciences Research Institute, St George’s University of London, London, UK (AA, MKB, JAK, LRB, JDI, ACP, AHH)
- Department of Neurology, St George’s University Hospitals NHS Foundation Trust, London, UK (JDI, ACP, AHH)
| |
Collapse
|
7
|
McAleese KE, Miah M, Graham S, Hadfield GM, Walker L, Johnson M, Colloby SJ, Thomas AJ, DeCarli C, Koss D, Attems J. Frontal white matter lesions in Alzheimer's disease are associated with both small vessel disease and AD-associated cortical pathology. Acta Neuropathol 2021; 142:937-950. [PMID: 34608542 PMCID: PMC8568857 DOI: 10.1007/s00401-021-02376-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 09/26/2021] [Accepted: 09/27/2021] [Indexed: 12/22/2022]
Abstract
Cerebral white matter lesions (WML) encompass axonal loss and demyelination and are assumed to be associated with small vessel disease (SVD)-related ischaemia. However, our previous study in the parietal lobe white matter revealed that WML in Alzheimer's disease (AD) are linked with degenerative axonal loss secondary to the deposition of cortical AD pathology. Furthermore, neuroimaging data suggest that pathomechanisms for the development of WML differ between anterior and posterior lobes with AD-associated degenerative mechanism driving posterior white matter disruption, and both AD-associated degenerative and vascular mechanisms contributed to anterior matter disruption. In this pilot study, we used human post-mortem brain tissue to investigate the composition and aetiology of frontal WML from AD and non-demented controls to determine if frontal WML are SVD-associated and to reveal any regional differences in the pathogenesis of WML. Frontal WML tissue sections from 40 human post-mortem brains (AD, n = 19; controls, n = 21) were quantitatively assessed for demyelination, axonal loss, cortical hyperphosphorylated tau (HPτ) and amyloid-beta (Aβ) burden, and arteriolosclerosis as a measure of SVD. Biochemical assessment included Wallerian degeneration-associated protease calpain and the myelin-associated glycoprotein to proteolipid protein ratio as a measure of ante-mortem ischaemia. Arteriolosclerosis severity was found to be associated with and a significant predictor of frontal WML severity in both AD and non-demented controls. Interesting, frontal axonal loss was also associated with HPτ and calpain levels were associated with increasing Aβ burden in the AD group, suggestive of an additional degenerative influence. To conclude, this pilot data suggest that frontal WML in AD may result from both increased arteriolosclerosis and AD-associated degenerative changes. These preliminary findings in combination with previously published data tentatively indicate regional differences in the aetiology of WML in AD, which should be considered in the clinical diagnosis of dementia subtypes: posterior WML maybe associated with degenerative mechanisms secondary to AD pathology, while anterior WML could be associated with both SVD-associated and degenerative mechanisms.
Collapse
Affiliation(s)
- Kirsty E McAleese
- Translation and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK.
| | - Mohi Miah
- Translation and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Sophie Graham
- Translation and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Georgina M Hadfield
- Translation and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Lauren Walker
- Translation and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Mary Johnson
- Translation and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Sean J Colloby
- Translation and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Alan J Thomas
- Translation and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Charles DeCarli
- Department of Neurology, University of California, Davis, CA, USA
| | - David Koss
- Translation and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Johannes Attems
- Translation and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| |
Collapse
|
8
|
Bir SC, Khan MW, Javalkar V, Toledo EG, Kelley RE. Emerging Concepts in Vascular Dementia: A Review. J Stroke Cerebrovasc Dis 2021; 30:105864. [PMID: 34062312 DOI: 10.1016/j.jstrokecerebrovasdis.2021.105864] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/29/2021] [Accepted: 04/28/2021] [Indexed: 01/18/2023] Open
Abstract
OBJECTIVE Vascular dementia (VaD) is the second most common cause of dementia and a major health concern worldwide. A comprehensive review on VaD is warranted for better understanding and guidance for the practitioner. We provide an updated overview of the epidemiology, pathophysiological mechanisms, neuroimaging patterns as well as current diagnostic and therapeutic approaches. MATERIALS AND METHODS A narrative review of current literature in VaD was performed based on publications from the database of PubMed, Scopus and Google Scholar up to January, 2021. RESULTS VaD can be the result of ischemic or hemorrhagic tissue injury in a particular region of the brain which translates into clinically significant cognitive impairment. For example, a cerebral infarct in the speech area of the dominant hemisphere would translate into clinically significant impairment as would involvement of projection pathways such as the arcuate fasciculus. Specific involvement of the angular gyrus of the dominant hemisphere, with resultant Gerstman's syndrome, could have a pronounced effect on functional ability despite being termed a "minor stroke". Small vessel cerebrovascular disease can have a cumulate effect on cognitive function over time. It is unfortunately well recognized that "good" functional recovery in acute ischemic or haemorrhagic stroke, including subarachnoid haemorrhage, does not necessarily translate into good cognitive recovery. The victim may often be left unable to have gainful employment, drive a car safely or handle their affairs independently. CONCLUSIONS This review should serve as a compendium of updated information on VaD and provide guidance in terms of newer diagnostic and potential therapeutic approaches.
Collapse
Affiliation(s)
- Shyamal C Bir
- Department of Neurology Ocshner/LSU Health Sciences Center-Sheveport, Shreveport, LA, USA
| | - Muhammad W Khan
- Department of Neurology Ocshner/LSU Health Sciences Center-Sheveport, Shreveport, LA, USA
| | - Vijayakumar Javalkar
- Department of Neurology Ocshner/LSU Health Sciences Center-Sheveport, Shreveport, LA, USA
| | | | - Roger E Kelley
- Department of Neurology Ocshner/LSU Health Sciences Center-Sheveport, Shreveport, LA, USA.
| |
Collapse
|
9
|
Frantellizzi V, Pani A, Ricci M, Locuratolo N, Fattapposta F, De Vincentis G. Neuroimaging in Vascular Cognitive Impairment and Dementia: A Systematic Review. J Alzheimers Dis 2021; 73:1279-1294. [PMID: 31929166 DOI: 10.3233/jad-191046] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cerebrovascular diseases are well established causes of cognitive impairment. Different etiologic entities, such as vascular dementia (VaD), vascular cognitive impairment, subcortical (ischemic) VaD, and vascular cognitive disorder, are included in the umbrella definition of vascular cognitive impairment and dementia (VCID). Because of the variability of VCID clinical presentation, there is no agreement on criteria defining the neuropathological threshold of this disorder. In fact, VCID is characterized by cerebral hemodynamic alteration which ranges from decreased cerebral blood flow to small vessels disease and involves a multifactorial process that leads to demyelination and gliosis, including blood-brain barrier disruption, hypoxia, and hypoperfusion, oxidative stress, neuroinflammation and alteration on neurovascular unit coupling, cerebral microbleeds, or superficial siderosis. Numerous criteria for the definition of VaD have been described: the National Institute of Neurological Disorders and Stroke Association Internationale pour Recherche'-et-l'Enseignement en Neurosciences criteria, the State of California Alzheimer's Disease Diagnostic and Treatment Centers criteria, DSM-V criteria, the Diagnostic Criteria for Vascular Cognitive Disorders (a VASCOG Statement), and Vascular Impairment of Cognition Classification Consensus Study. Neuroimaging is fundamental for definition and diagnosis of VCID and should be used to assess the extent, location, and type of vascular lesions. MRI is the most sensible technique, especially if used according to standardized protocols, even if CT plays an important role in several conditions. Functional neuroimaging, in particular functional MRI and PET, may facilitate differential diagnosis among different forms of dementia. This systematic review aims to explore the state of the art and future perspective of non-invasive diagnostics of VCID.
Collapse
Affiliation(s)
| | - Arianna Pani
- Clinical Pharmacology and Toxicology, University of Milan "Statale", Italy
| | - Maria Ricci
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, Rome, Italy
| | | | | | - Giuseppe De Vincentis
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
10
|
Lee H, Xu F, Liu X, Koundal S, Zhu X, Davis J, Yanez D, Schrader J, Stanisavljevic A, Rothman DL, Wardlaw J, Van Nostrand WE, Benveniste H. Diffuse white matter loss in a transgenic rat model of cerebral amyloid angiopathy. J Cereb Blood Flow Metab 2021; 41:1103-1118. [PMID: 32791876 PMCID: PMC8054716 DOI: 10.1177/0271678x20944226] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Diffuse white matter (WM) disease is highly prevalent in elderly with cerebral small vessel disease (cSVD). In humans, cSVD such as cerebral amyloid angiopathy (CAA) often coexists with Alzheimer's disease imposing a significant impediment for characterizing their distinct effects on WM. Here we studied the burden of age-related CAA pathology on WM disease in a novel transgenic rat model of CAA type 1 (rTg-DI). A cohort of rTg-DI and wild-type rats was scanned longitudinally using MRI for characterization of morphometry, cerebral microbleeds (CMB) and WM integrity. In rTg-DI rats, a distinct pattern of WM loss was observed at 9 M and 11 M. MRI also revealed manifestation of small CMB in thalamus at 6 M, which preceded WM loss and progressively enlarged until the moribund disease stage. Histology revealed myelin loss in the corpus callosum and thalamic CMB in all rTg-DI rats, the latter of which manifested in close proximity to occluded and calcified microvessels. The quantitation of CAA load in rTg-DI rats revealed that the most extensive microvascular Aβ deposition occurred in the thalamus. For the first time using in vivo MRI, we show that CAA type 1 pathology alone is associated with a distinct pattern of WM loss.
Collapse
Affiliation(s)
- Hedok Lee
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Feng Xu
- George and Anne Ryan Institute for Neuroscience and the Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, RI, USA
| | - Xiaodan Liu
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Sunil Koundal
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Xiaoyue Zhu
- George and Anne Ryan Institute for Neuroscience and the Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, RI, USA
| | - Judianne Davis
- George and Anne Ryan Institute for Neuroscience and the Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, RI, USA
| | - David Yanez
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA
| | - Joseph Schrader
- George and Anne Ryan Institute for Neuroscience and the Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, RI, USA
| | - Aleksandra Stanisavljevic
- George and Anne Ryan Institute for Neuroscience and the Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, RI, USA
| | - Douglas L Rothman
- Departments of Radiology and Biomedical Imaging, Yale School of Medicine New Haven, CT, USA.,Department of Biomedical Engineering, Yale School of Medicine New Haven, CT, USA
| | - Joanna Wardlaw
- Brain Research Imaging Centre, Centre for Clinical Brain Sciences, Dementia Research Institute at the University of Edinburgh, Edinburgh, UK
| | - William E Van Nostrand
- George and Anne Ryan Institute for Neuroscience and the Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, RI, USA
| | - Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, CT, USA.,Department of Biomedical Engineering, Yale School of Medicine New Haven, CT, USA
| |
Collapse
|
11
|
Li M, Li Y, Zuo L, Hu W, Jiang T. Increase of blood-brain barrier leakage is related to cognitive decline in vascular mild cognitive impairment. BMC Neurol 2021; 21:159. [PMID: 33858381 PMCID: PMC8048027 DOI: 10.1186/s12883-021-02189-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 04/08/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Blood-brain barrier (BBB) breakdown, as an early biomarker for vascular mild cognitive impairment (vMCI), has only been validated by a few studies. The aim of this study was to investigate whether compromised BBB integrity is involved in vMCI patients, and detect the relationship between BBB breakdown and cognitive function. BBB leakage in vMCI was explored, and the relationship between BBB leakage and cognitive function was discussed in this study. METHODS This is a cross-sectional study involving 26 vMCI patients and 21 sex- and age-matched healthy controls. Dynamic contrast-enhanced-magnetic resonance imaging was performed for all participants, to determine BBB leakage. Leakage volume, leakage rate, and fractional blood plasma volume (Vp) in the grey and white matter were evaluated. Neuropsychological tests were used to determine cognitive function. Leakage rate, leakage volume, and Vp in different brain locations, including deep grey matter, cortical grey matter, white matter hyperintensity, and normal-appearing white matter were compared between the two groups. RESULTS Multivariable linear regression analyses revealed that in all regions of interest, the leakage rate was significantly higher in vMCI patients relative to controls. Leakage volume in normal-appearing white matter and white matter hyperintensity were significantly higher, while Vp in normal-appearing white matter, deep grey matter, and cortical grey matter were significantly lower in vMCI patients. Moreover, Montreal Cognitive Assessment scores decreased with the increase of leakage rate in white matter hyperintensity. CONCLUSION Increased BBB permeability was detected in vMCI patients and was related to cognitive decline, which suggested that BBB breakdown might be involved in cognitive dysfunction pathogenesis.
Collapse
Affiliation(s)
- Man Li
- Radiology Department, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, South Gongti Road, Chaoyang District, Beijing, 100020, P.R. China.
| | - Yue Li
- Neurology Department, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, South Gongti Road, Chaoyang District, Beijing, P.R. China
| | - Long Zuo
- Radiology Department, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, South Gongti Road, Chaoyang District, Beijing, 100020, P.R. China
| | - Wenli Hu
- Neurology Department, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, South Gongti Road, Chaoyang District, Beijing, P.R. China
| | - Tao Jiang
- Radiology Department, Beijing Chao-Yang Hospital, Capital Medical University, No. 8, South Gongti Road, Chaoyang District, Beijing, 100020, P.R. China
| |
Collapse
|
12
|
Moretti R, Giuffré M, Caruso P, Gazzin S, Tiribelli C. Homocysteine in Neurology: A Possible Contributing Factor to Small Vessel Disease. Int J Mol Sci 2021; 22:2051. [PMID: 33669577 PMCID: PMC7922986 DOI: 10.3390/ijms22042051] [Citation(s) in RCA: 16] [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: 01/31/2021] [Revised: 02/14/2021] [Accepted: 02/15/2021] [Indexed: 12/19/2022] Open
Abstract
Homocysteine (Hcy) is a sulfur-containing amino acid generated during methionine metabolism, accumulation of which may be caused by genetic defects or the deficit of vitamin B12 and folate. A serum level greater than 15 micro-mols/L is defined as hyperhomocysteinemia (HHcy). Hcy has many roles, the most important being the active participation in the transmethylation reactions, fundamental for the brain. Many studies focused on the role of homocysteine accumulation in vascular or degenerative neurological diseases, but the results are still undefined. More is known in cardiovascular disease. HHcy is a determinant for the development and progression of inflammation, atherosclerotic plaque formation, endothelium, arteriolar damage, smooth muscle cell proliferation, and altered-oxidative stress response. Conversely, few studies focused on the relationship between HHcy and small vessel disease (SVD), despite the evidence that mice with HHcy showed a significant end-feet disruption of astrocytes with a diffuse SVD. A severe reduction of vascular aquaporin-4-water channels, lower levels of high-functioning potassium channels, and higher metalloproteinases are also observed. HHcy modulates the N-homocysteinylation process, promoting a pro-coagulative state and damage of the cellular protein integrity. This altered process could be directly involved in the altered endothelium activation, typical of SVD and protein quality, inhibiting the ubiquitin-proteasome system control. HHcy also promotes a constant enhancement of microglia activation, inducing the sustained pro-inflammatory status observed in SVD. This review article addresses the possible role of HHcy in small-vessel disease and understands its pathogenic impact.
Collapse
Affiliation(s)
- Rita Moretti
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy; (M.G.); (P.C.)
| | - Mauro Giuffré
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy; (M.G.); (P.C.)
| | - Paola Caruso
- Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy; (M.G.); (P.C.)
| | - Silvia Gazzin
- Italian Liver Foundation, AREA SCIENCE PARK, 34149 Trieste, Italy; (S.G.); (C.T.)
| | - Claudio Tiribelli
- Italian Liver Foundation, AREA SCIENCE PARK, 34149 Trieste, Italy; (S.G.); (C.T.)
| |
Collapse
|
13
|
Blevins BL, Vinters HV, Love S, Wilcock DM, Grinberg LT, Schneider JA, Kalaria RN, Katsumata Y, Gold BT, Wang DJJ, Ma SJ, Shade LMP, Fardo DW, Hartz AMS, Jicha GA, Nelson KB, Magaki SD, Schmitt FA, Teylan MA, Ighodaro ET, Phe P, Abner EL, Cykowski MD, Van Eldik LJ, Nelson PT. Brain arteriolosclerosis. Acta Neuropathol 2021; 141:1-24. [PMID: 33098484 PMCID: PMC8503820 DOI: 10.1007/s00401-020-02235-6] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/04/2020] [Accepted: 10/05/2020] [Indexed: 12/14/2022]
Abstract
Brain arteriolosclerosis (B-ASC), characterized by pathologic arteriolar wall thickening, is a common finding at autopsy in aged persons and is associated with cognitive impairment. Hypertension and diabetes are widely recognized as risk factors for B-ASC. Recent research indicates other and more complex risk factors and pathogenetic mechanisms. Here, we describe aspects of the unique architecture of brain arterioles, histomorphologic features of B-ASC, relevant neuroimaging findings, epidemiology and association with aging, established genetic risk factors, and the co-occurrence of B-ASC with other neuropathologic conditions such as Alzheimer's disease and limbic-predominant age-related TDP-43 encephalopathy (LATE). There may also be complex physiologic interactions between metabolic syndrome (e.g., hypertension and inflammation) and brain arteriolar pathology. Although there is no universally applied diagnostic methodology, several classification schemes and neuroimaging techniques are used to diagnose and categorize cerebral small vessel disease pathologies that include B-ASC, microinfarcts, microbleeds, lacunar infarcts, and cerebral amyloid angiopathy (CAA). In clinical-pathologic studies that factored in comorbid diseases, B-ASC was independently associated with impairments of global cognition, episodic memory, working memory, and perceptual speed, and has been linked to autonomic dysfunction and motor symptoms including parkinsonism. We conclude by discussing critical knowledge gaps related to B-ASC and suggest that there are probably subcategories of B-ASC that differ in pathogenesis. Observed in over 80% of autopsied individuals beyond 80 years of age, B-ASC is a complex and under-studied contributor to neurologic disability.
Collapse
Affiliation(s)
- Brittney L Blevins
- Department of Neuroscience, University Kentucky, Lexington, KY, 40536, USA
| | - Harry V Vinters
- Department of Pathology and Laboratory Medicine, David Geffen SOM at UCLA and Ronald Reagan UCLA Medical Center, Los Angeles, CA, 90095-1732, USA
| | - Seth Love
- University of Bristol and Southmead Hospital, Bristol, BS10 5NB, UK
| | - Donna M Wilcock
- Sanders-Brown Center on Aging, Department of Neuroscience, University Kentucky, Lexington, KY, 40536, USA
| | - Lea T Grinberg
- Department of Neurology and Pathology, UCSF, San Francisco, CA, USA
- Global Brain Health Institute, UCSF, San Francisco, CA, USA
- LIM-22, Department of Pathology, University of Sao Paulo Medical School, São Paulo, Brazil
| | - Julie A Schneider
- Departments of Neurology and Pathology, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Rajesh N Kalaria
- Translational and Clinical Research Institute, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, NE4 5PL, UK
| | - Yuriko Katsumata
- Sanders-Brown Center on Aging, Department of Biostatistics, University Kentucky, Lexington, KY, 40536, USA
| | - Brian T Gold
- Sanders-Brown Center on Aging, Department of Neuroscience, University Kentucky, Lexington, KY, 40536, USA
| | - Danny J J Wang
- Laboratory of FMRI Technology (LOFT), USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Samantha J Ma
- Laboratory of FMRI Technology (LOFT), USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Lincoln M P Shade
- Sanders-Brown Center on Aging, Department of Biostatistics, University Kentucky, Lexington, KY, 40536, USA
| | - David W Fardo
- Sanders-Brown Center on Aging, Department of Biostatistics, University Kentucky, Lexington, KY, 40536, USA
| | - Anika M S Hartz
- Sanders-Brown Center on Aging, Department of Pharmacology and Nutritional Sciences, University Kentucky, Lexington, KY, 40536, USA
| | - Gregory A Jicha
- Sanders-Brown Center on Aging, Department of Neurology, University Kentucky, Lexington, KY, 40536, USA
| | | | - Shino D Magaki
- Department of Pathology and Laboratory Medicine, David Geffen SOM at UCLA and Ronald Reagan UCLA Medical Center, Los Angeles, CA, 90095-1732, USA
| | - Frederick A Schmitt
- Sanders-Brown Center on Aging, Department of Neurology, University Kentucky, Lexington, KY, 40536, USA
| | - Merilee A Teylan
- Department of Epidemiology, University Washington, Seattle, WA, 98105, USA
| | | | - Panhavuth Phe
- Sanders-Brown Center on Aging, University Kentucky, Lexington, KY, 40536, USA
| | - Erin L Abner
- Sanders-Brown Center on Aging, Department of Epidemiology, University Kentucky, Lexington, KY, 40536, USA
| | - Matthew D Cykowski
- Departments of Pathology and Genomic Medicine and Neurology, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Linda J Van Eldik
- Sanders-Brown Center on Aging, Department of Neuroscience, University Kentucky, Lexington, KY, 40536, USA
| | - Peter T Nelson
- Sanders-Brown Center on Aging, Department of Pathology, University of Kentucky, Lexington, KY, 40536, USA.
- Rm 311 Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone Avenue, Lexington, KY, 40536, USA.
| |
Collapse
|
14
|
Nie S, Shen C, Guo Y, Hou X, Hong Y, Xu S, Lv R, Liu X. Preliminary Findings on Visual Event-Related Potential P3 in Asymptomatic Patients with Cerebral Small Vessel Disease. Neuropsychiatr Dis Treat 2021; 17:3379-3394. [PMID: 34848959 PMCID: PMC8626861 DOI: 10.2147/ndt.s338717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/09/2021] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Cerebral small vessel disease is the primary cause of cognitive impairment. Therefore, early recognition is of great significance. Some studies have shown that asymptomatic cerebral small vessel disease (aCSVD) patients have abnormal neurocognitive function, but this is not readily apparent at the initial stage. The objective of this paper was to assess visual spatial attention by event-related potential (ERP) examination and to analyze the relationship between ERP data and clinical characteristics in patients with aCSVD. METHODS We selected 25 aCSVD patients and enrolled 23 age-matched normal subjects as the control group. We measured the latency and amplitude of original/corresponding differential ERP components using the modified visual oddball paradigm, which included a standard stimulus, target stimulus, and new stimulus. Additionally, we selected aberrant ERP components to study the correlations between the ERP data and clinical characteristics of the patients with aCSVD. RESULTS We found not only lower amplitude but also significantly longer P3 latency in the aCSVD patients. The above results were further verified by analyzing the different components (target minus standard and novel minus standard) of P3. Furthermore, abnormal ERPs in the aCSVD patients were closely related to the changes observed with imaging. CONCLUSION It was demonstrated that the speed and capability of processing visual spatial information was impaired in aCSVD patients compared with healthy controls. Thus, ERP examination could detect the presence of attentional deficits and might become a rapid and sensitive method for the early diagnosis of aCSVD. However, its availability needs further investigation.
Collapse
Affiliation(s)
- Shanjing Nie
- Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China.,Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China.,Anti-Aging Monitoring Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China
| | - Chao Shen
- Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China.,Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China
| | - Yunliang Guo
- Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China.,Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China.,Anti-Aging Monitoring Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China
| | - Xunyao Hou
- Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China.,Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China.,Anti-Aging Monitoring Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China
| | - Yan Hong
- Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China.,Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China.,Anti-Aging Monitoring Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China
| | - Song Xu
- Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China.,Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China.,Anti-Aging Monitoring Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China
| | - Renjun Lv
- Shandong Provincial Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Xueping Liu
- Department of Geriatrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China.,Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China.,Anti-Aging Monitoring Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China
| |
Collapse
|
15
|
Abstract
Cerebrovascular disease is a significant cause of cognitive impairment leading to a reduction or loss of functioning, including social and occupational. The connection cause-effect between cerebrovascular disease and cerebral infarction was originally theorized by the studies from Newcastle-Upon-Tyne, England, in the 1960s, where vascular dementia (VaD) was defined as a disease originated from several infarctions that overcome a determined threshold. It differs from Alzheimer's disease (AD), although there are various overlaps in risk factors, symptomatology, the similarity of vascular lesions, and treatment benefits. Nevertheless, AD is one-half of all cases of dementia. Cognitive impairment and dementia (VCID) has recently been proposed to include different entities such as VaD, Vascular cognitive impairment, subcortical (ischemic) VaD, and vascular cognitive disorders. VaD is the most common cause of dementia after AD. Neuroimaging is an essential part of the workup of patients with cognitive decline and in those with suspected VCID it should be used to assess the extent, location, and type of vascular lesions. Computed tomography (CT) or structural magnetic resonance imaging (MRI) are usually used for the diagnosis of vascular diseases of the brain. However, images obtained from new hybrid devices could help the neurologist in the differential diagnosis between various neuropathological entities related to VCID. Single-photon emission computed tomography (SPECT) combined with CT or MRI and positron emission tomography (PET) combined with CT or MRI represent the future of neuroimaging tools as morphological and functional data can be provided simultaneously. New prospects have been developed such as hybrid PET/SPECT/CT, a high-performance prototype able to produce high-quality images but for now suitable only for small animals. Nowadays, PET/CT and PET/MRI are good performance and high-quality instruments, even if the magnetic field of MRI represents a limitation that affects the PET electronics and positron detection ability. SPECT/MRI delineates as a potential and tempting device. It could give us both functional and anatomical details, with the advantage of lack of extra ionizing radiation and high soft-tissue contrast, important features, and considerable auxiliary for differential diagnosis in the variegate word of vascular cognitive impairment. The aim of this review is to summarize the newest viewpoints in hybrid imaging in the diagnosis of VaD and to highlight pros and cons of each methodic.
Collapse
Affiliation(s)
| | - Miriam Conte
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, Rome, Italy
| | - Giuseppe De Vincentis
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
16
|
Moroni F, Ammirati E, Hainsworth AH, Camici PG. Association of White Matter Hyperintensities and Cardiovascular Disease: The Importance of Microcirculatory Disease. Circ Cardiovasc Imaging 2020; 13:e010460. [PMID: 33232175 DOI: 10.1161/circimaging.120.010460] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Cardiac and cerebrovascular diseases are currently the leading causes of mortality and disability worldwide. Both the heart and brain display similar vascular anatomy, with large conduit arteries running on the surface of the organ providing tissue perfusion through an intricate network of penetrating small vessels. Both organs rely on fine tuning of local blood flow to match metabolic demand. Blood flow regulation requires adequate functioning of the microcirculation in both organs, with loss of microvascular function, termed small vessel disease (SVD) underlying different potential clinical manifestations. SVD in the heart, known as coronary microvascular dysfunction, can cause chronic or acute myocardial ischemia and may lead to development of heart failure. In the brain, cerebral SVD can cause an acute stroke syndrome known as lacunar stroke or more subtle pathological alterations of the brain parenchyma, which may eventually lead to neurological deficits or cognitive decline in the long term. Coronary microcirculation cannot be visualized in vivo in humans, and functional information can be deduced by measuring the coronary flow reserve. The diagnosis of cerebral SVD is largely based on brain magnetic resonance imaging, with white matter hyperintensities, microbleeds, and brain atrophy reflecting key structural changes. There is evidence that such structural changes reflect underlying cerebral SVD. Here, we review interactions between SVD and cardiovascular risk factors, and we discuss the evidence linking cerebral SVD with large vessel atheroma, atrial fibrillation, heart failure, and heart valve disease.
Collapse
Affiliation(s)
- Francesco Moroni
- Cardiothoracic and Vascular Department, Vita-Salute University and San Raffaele Hospital, Milan, Italy (F.M., P.G.C.)
| | - Enrico Ammirati
- De Gasperis Cardio Center, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy (E.A.)
| | - Atticus H Hainsworth
- Molecular and Clinical Sciences Research Institute, St George's, University of London, United Kingdom (A.H.H.)
- Department of Neurology, St George's University Hospitals NHS Foundation Trust, London, United Kingdom (A.H.H.)
| | - Paolo G Camici
- Cardiothoracic and Vascular Department, Vita-Salute University and San Raffaele Hospital, Milan, Italy (F.M., P.G.C.)
| |
Collapse
|
17
|
Moretti R, Caruso P. Small Vessel Disease-Related Dementia: An Invalid Neurovascular Coupling? Int J Mol Sci 2020; 21:1095. [PMID: 32046035 PMCID: PMC7036993 DOI: 10.3390/ijms21031095] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 02/04/2020] [Accepted: 02/04/2020] [Indexed: 12/18/2022] Open
Abstract
The arteriosclerosis-dependent alteration of brain perfusion is one of the major determinants in small vessel disease, since small vessels have a pivotal role in the brain's autoregulation. Nevertheless, as far as we know, endothelium distress can potentiate the flow dysregulation and lead to subcortical vascular dementia that is related to small vessel disease (SVD), also being defined as subcortical vascular dementia (sVAD), as well as microglia activation, chronic hypoxia and hypoperfusion, vessel-tone dysregulation, altered astrocytes, and pericytes functioning blood-brain barrier disruption. The molecular basis of this pathology remains controversial. The apparent consequence (or a first event, too) is the macroscopic alteration of the neurovascular coupling. Here, we examined the possible mechanisms that lead a healthy aging process towards subcortical dementia. We remarked that SVD and white matter abnormalities related to age could be accelerated and potentiated by different vascular risk factors. Vascular function changes can be heavily influenced by genetic and epigenetic factors, which are, to the best of our knowledge, mostly unknown. Metabolic demands, active neurovascular coupling, correct glymphatic process, and adequate oxidative and inflammatory responses could be bulwarks in defense of the correct aging process; their impairments lead to a potentially catastrophic and non-reversible condition.
Collapse
Affiliation(s)
- Rita Moretti
- Neurology Clinic, Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy;
| | | |
Collapse
|
18
|
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
|
19
|
Moretti R, Peinkhofer C. B Vitamins and Fatty Acids: What Do They Share with Small Vessel Disease-Related Dementia? Int J Mol Sci 2019; 20:5797. [PMID: 31752183 PMCID: PMC6888477 DOI: 10.3390/ijms20225797] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 10/21/2019] [Accepted: 11/12/2019] [Indexed: 12/12/2022] Open
Abstract
Many studies have been written on vitamin supplementation, fatty acid, and dementia, but results are still under debate, and no definite conclusion has yet been drawn. Nevertheless, a significant amount of lab evidence confirms that vitamins of the B group are tightly related to gene control for endothelium protection, act as antioxidants, play a co-enzymatic role in the most critical biochemical reactions inside the brain, and cooperate with many other elements, such as choline, for the synthesis of polyunsaturated phosphatidylcholine, through S-adenosyl-methionine (SAM) methyl donation. B-vitamins have anti-inflammatory properties and act in protective roles against neurodegenerative mechanisms, for example, through modulation of the glutamate currents and a reduction of the calcium currents. In addition, they also have extraordinary antioxidant properties. However, laboratory data are far from clinical practice. Many studies have tried to apply these results in everyday clinical activity, but results have been discouraging and far from a possible resolution of the associated mysteries, like those represented by Alzheimer's disease (AD) or small vessel disease dementia. Above all, two significant problems emerge from the research: No consensus exists on general diagnostic criteria-MCI or AD? Which diagnostic criteria should be applied for small vessel disease-related dementia? In addition, no general schema exists for determining a possible correct time of implementation to have effective results. Here we present an up-to-date review of the literature on such topics, shedding some light on the possible interaction of vitamins and phosphatidylcholine, and their role in brain metabolism and catabolism. Further studies should take into account all of these questions, with well-designed and world-homogeneous trials.
Collapse
Affiliation(s)
- Rita Moretti
- Neurology Clinic, Department of Medical, Surgical and Health Sciences, University of Trieste, 34149 Trieste, Italy;
| | | |
Collapse
|
20
|
Norton EJ, Bridges LR, Kenyon LC, Esiri MM, Bennett DC, Hainsworth AH. Cell Senescence and Cerebral Small Vessel Disease in the Brains of People Aged 80 Years and Older. J Neuropathol Exp Neurol 2019; 78:1066-1072. [PMID: 31553444 DOI: 10.1093/jnen/nlz088] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 07/21/2019] [Indexed: 12/17/2022] Open
Abstract
Cerebral small vessel disease (cSVD) in penetrating arteries is a major cause of age-related morbidity. Cellular senescence is a molecular process targeted by novel senolytic drugs. We quantified senescence in penetrating arteries and tested whether myocyte senescence was associated with cSVD. We immunolabeled subcortical white matter of older persons (age 80-96 years, n = 60) with minimal AD, using antibodies to 2 established senescence markers (H3K9me3, γH2AX) and a myocyte marker (hSMM). Within the walls of penetrating arteries (20-300 µm), we quantified senescence-associated heterochromatic foci (SAHF)-positive nuclei, cell density (nuclei/µm2), and sclerotic index (SI). Senescent-appearing mural cells were present in small arteries of all cases. cSVD cases exhibited a lower proportion of senescent-appearing cells and lower area fraction (AF%) of SAHF-positive nuclei compared to controls (p = 0.014, 0.016, respectively). cSVD severity and SI both correlated negatively with AF% (p = 0.013, 0.002, respectively). Mural cell density was lower (p < 0.001) and SI higher (p < 0.001) in cSVD, relative to controls. In conclusion, senescent myocyte-like cells were universal in penetrating arteries of an AD-free cohort aged 80 years and older. Senescent-appearing nuclei were more common in persons aged 80 years and older without cSVD compared to cSVD cases, indicating caution in senolytic drug prescribing. Myocyte senescence and cSVD may represent alternative vessel fates in the aging human brain.
Collapse
Affiliation(s)
- Emma J Norton
- Molecular and Clinical Sciences Research Institute, St George's, University of London, London, UK; Department of Cellular Pathology, St George's University Hospitals NHS Foundation Trust, London, UK; Department of Pathology, Anatomy & Cell Biology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania; Department of Neuropathology, Oxford-Radcliffe NHS Trust, Oxford, UK; Department of Clinical Neurology, Oxford University, John Radcliffe Hospital, Oxford, UK; and Department of Neurology, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Leslie R Bridges
- Molecular and Clinical Sciences Research Institute, St George's, University of London, London, UK; Department of Cellular Pathology, St George's University Hospitals NHS Foundation Trust, London, UK; Department of Pathology, Anatomy & Cell Biology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania; Department of Neuropathology, Oxford-Radcliffe NHS Trust, Oxford, UK; Department of Clinical Neurology, Oxford University, John Radcliffe Hospital, Oxford, UK; and Department of Neurology, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Lawrence C Kenyon
- Molecular and Clinical Sciences Research Institute, St George's, University of London, London, UK; Department of Cellular Pathology, St George's University Hospitals NHS Foundation Trust, London, UK; Department of Pathology, Anatomy & Cell Biology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania; Department of Neuropathology, Oxford-Radcliffe NHS Trust, Oxford, UK; Department of Clinical Neurology, Oxford University, John Radcliffe Hospital, Oxford, UK; and Department of Neurology, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Margaret M Esiri
- Molecular and Clinical Sciences Research Institute, St George's, University of London, London, UK; Department of Cellular Pathology, St George's University Hospitals NHS Foundation Trust, London, UK; Department of Pathology, Anatomy & Cell Biology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania; Department of Neuropathology, Oxford-Radcliffe NHS Trust, Oxford, UK; Department of Clinical Neurology, Oxford University, John Radcliffe Hospital, Oxford, UK; and Department of Neurology, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Dorothy C Bennett
- Molecular and Clinical Sciences Research Institute, St George's, University of London, London, UK; Department of Cellular Pathology, St George's University Hospitals NHS Foundation Trust, London, UK; Department of Pathology, Anatomy & Cell Biology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania; Department of Neuropathology, Oxford-Radcliffe NHS Trust, Oxford, UK; Department of Clinical Neurology, Oxford University, John Radcliffe Hospital, Oxford, UK; and Department of Neurology, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Atticus H Hainsworth
- Molecular and Clinical Sciences Research Institute, St George's, University of London, London, UK; Department of Cellular Pathology, St George's University Hospitals NHS Foundation Trust, London, UK; Department of Pathology, Anatomy & Cell Biology, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania; Department of Neuropathology, Oxford-Radcliffe NHS Trust, Oxford, UK; Department of Clinical Neurology, Oxford University, John Radcliffe Hospital, Oxford, UK; and Department of Neurology, St George's University Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
21
|
Becker CE, Quinn TJ, Williams A. Association Between Endothelial Cell Stabilizing Medication and Small Vessel Disease Stroke: A Case-Control Study. Front Neurol 2019; 10:1029. [PMID: 31608006 PMCID: PMC6773869 DOI: 10.3389/fneur.2019.01029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 09/10/2019] [Indexed: 11/23/2022] Open
Abstract
Increasing evidence suggests a role for endothelial cell (EC) dysfunction in pathogenesis of cerebral small vessel disease. Commonly used medications including certain antihypertensives and statins have EC-stabilizing effects. We used individual patient data from completed acute stroke trials to assess whether prior exposure to EC-stabilizing medications was associated with lacunar stroke, using lacunar stroke as a clinical proxy for cerebral small vessel disease. Across 12,002 patients with relevant data, 2,855 (24%) had a lacunar stroke presentation. Univariable analyses suggested potential confounding from vascular diseases treated with EC-stabilizing medications. Initial multivariable logistic regression gave conflicting results when describing the independent association of exposure to EC-stabilizing medication and lacunar stroke in the complete population (O.R. 0.87, 95% C.I.: 0.77– 0.98) and limited to those taking any antihypertensive (O.R. 1.51, 95% C.I.: 1.21–1.88). Re-running the analyses including statins in the EC-stabilizing category suggested a beneficial effect of EC-stabilizing medication exposure on lacunar stroke incidence (O.R. 0.83, 95% C.I.: 0.73–0.93). These results align with recent pre-clinical data and would support interventional trials of EC-stabilizing medication for preventing cerebral small vessel disease. Our results also suggest that analyses of EC-stabilizing interventions need to adjust for potential endothelial effects of other co-prescribed medication.
Collapse
Affiliation(s)
- Charlotte Elisabeth Becker
- Centre for Regenerative Medicine, UK Dementia Research Institute, University of Edinburgh, Edinburgh, United Kingdom.,School of Medicine and Health Sciences, University of Oldenburg, Oldenburg, Germany
| | - Terence J Quinn
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Anna Williams
- Centre for Regenerative Medicine, UK Dementia Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
22
|
Caruso P, Signori R, Moretti R. Small vessel disease to subcortical dementia: a dynamic model, which interfaces aging, cholinergic dysregulation and the neurovascular unit. Vasc Health Risk Manag 2019; 15:259-281. [PMID: 31496716 PMCID: PMC6689673 DOI: 10.2147/vhrm.s190470] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 01/14/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Small vessels have the pivotal role for the brain's autoregulation. The arteriosclerosis-dependent alteration of the brain perfusion is one of the major determinants in small vessel disease. Endothelium distress can potentiate the flow dysregulation and lead to subcortical vascular dementia (sVAD). sVAD increases morbidity and disability. Epidemiological studies have shown that sVAD shares with cerebrovascular disease most of the common risk factors. The molecular basis of this pathology remains controversial. PURPOSE To detect the possible mechanisms between small vessel disease and sVAD, giving a broad vision on the topic, including pathological aspects, clinical and laboratory findings, metabolic process and cholinergic dysfunction. METHODS We searched MEDLINE using different search terms ("vascular dementia", "subcortical vascular dementia", "small vessel disease", "cholinergic afferents", etc). Publications were selected from the past 20 years. Searches were extended to Embase, Cochrane Library, and LILIACS databases. All searches were done from January 1, 1998 up to January 31, 2018. RESULTS A total of 560 studies showed up, and appropriate studies were included. Associations between traditional vascular risk factors have been isolated. We remarked that SVD and white matter abnormalities are seen frequently with aging and also that vascular and endothelium changes are related with age; the changes can be accelerated by different vascular risk factors. Vascular function changes can be heavily influenced by genetic and epigenetic factors. CONCLUSION Small vessel disease and the related dementia are two pathologies that deserve attention for their relevance and impact in clinical practice. Hypertension might be a historical problem for SVD and SVAD, but low pressure might be even more dangerous; CBF regional selective decrease seems to be a critical factor for small vessel disease-related dementia. In those patients, endothelium damage is a super-imposed condition. Several issues are still debatable, and more research is needed.
Collapse
Affiliation(s)
- Paola Caruso
- Department of Medical, Surgical and Health Sciences, Neurology Clinic, University of Trieste, Trieste, Italy
| | - Riccardo Signori
- Department of Medical, Surgical and Health Sciences, Neurology Clinic, University of Trieste, Trieste, Italy
| | - Rita Moretti
- Department of Medical, Surgical and Health Sciences, Neurology Clinic, University of Trieste, Trieste, Italy
| |
Collapse
|
23
|
Scott TM, Bhadelia RA, Qiu WQ, Folstein MF, Rosenberg IH. Small Vessel Cerebrovascular Pathology Identified by Magnetic Resonance Imaging Is Prevalent in Alzheimer's Disease and Mild Cognitive Impairment: A Potential Target for Intervention. J Alzheimers Dis 2019; 65:293-302. [PMID: 30040728 DOI: 10.3233/jad-180366] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND There is evidence that Alzheimer's disease (AD) has significant cerebrovascular etiopathogenesis. Understanding potentially modifiable risk factors for vascular disease can help design long-term intervention strategies for controlling or preventing cognitive dysfunction attributable to cerebrovascular disease. OBJECTIVE To evaluate the presence and severity of markers of cerebrovascular pathology, its relationship to diagnostic categories of dementia, including AD, and association with the metabolic biomarker homocysteine. METHODS In a cross-sectional observational study, 340 community-dwelling elders received a clinical evaluation including brain MRI and neuropsychological tests. Dementia and mild cognitive impairment (MCI) were diagnosed by consensus committee. Fasting total plasma homocysteine was measured. Statistical analyses were adjusted for demographics and cerebrovascular risk factors. RESULTS Nearly 25% of those diagnosed with AD had small vessel infarcts (SVI). Periventricular white matter hyperintensity (pvWMHI) was prevalent in participants with AD (61%) or MCI (amnesic 61% and non-amnesic 54%, respectively). Participants with SVI and/or pvWMHI also had greater brain atrophy. Homocysteine concentrations were higher in individuals with cerebrovascular findings than in those without. In individuals with cerebrovascular disease, homocysteine was inversely related to executive function (p = 0.022) and directly related to degree of brain atrophy (p = 0.009). CONCLUSIONS We demonstrated a significant prevalence of small vessel markers of cerebrovascular pathology in individuals diagnosed with AD, with a significant concurrence between cerebrovascular disease and brain and ventricular atrophy. While current research on AD has focused on amyloid-βpeptide deposition, tau-pathology, and microglial activation and inflammation, greater attention to the cerebrovascular contribution to this neurodegenerative disease presents an additional target for therapeutic prevention and intervention.
Collapse
Affiliation(s)
- Tammy M Scott
- Friedman School of Nutrition Science and Policy, Boston, MA, USA.,Tufts University School of Medicine, Boston, MA, USA
| | | | - Wei Qiao Qiu
- Boston University School of Medicine, Boston, MA, USA
| | | | - Irwin H Rosenberg
- Friedman School of Nutrition Science and Policy, Boston, MA, USA.,Tufts University School of Medicine, Boston, MA, USA.,USDA Jean Mayer Human Nutrition Research Center on Aging, Boston, MA, USA
| |
Collapse
|
24
|
Abstract
Proper functioning of the brain is dependent on integrity of the cerebral vasculature. During ageing, a number of factors including aortic or arterial stiffness, autonomic dysregulation, neurovascular uncoupling and blood-brain barrier (BBB) damage will define the dynamics of brain blood flow and local perfusion. The nature and extent of ageing-related cerebrovascular changes, the degree of involvement of the heart and extracranial vessels and the consequent location of tissue pathology may vary considerably. Atheromatous disease retarding flow is a common vascular insult, which increases exponentially with increasing age. Arteriolosclerosis characterized as a prominent feature of small vessel disease is one of the first changes to occur during the natural history of cerebrovascular pathology. At the capillary level, the cerebral endothelium, which forms the BBB undergoes changes including reduced cytoplasm, fewer mitochondria, loss of tight junctions and thickened basement membranes with collagenosis. Astrocyte end-feet protecting the BBB retract as part of the clasmatodendrotic response whereas pericyte coverage is altered. The consequences of these microvascular changes are lacunar infarcts, cortical and subcortical microinfarcts, microbleeds and diffuse white matter disease, which involves myelin loss and axonal abnormalities. The deeper structures are particularly vulnerable because of the relatively reduced density of the microvascular network formed by perforating and penetrating end arteries. Ultimately, the integrity of both the neurovascular and gliovascular units is compromised such that there is an overall synergistic effect reflecting on ageing associated cerebral perfusion and permeability. More than one protagonist appears to be involved in ageing-related cognitive dysfunction characteristically associated with the neurocognitive disorders.
Collapse
|
25
|
Vasita E, Yasmeen S, Andoh J, Bridges LR, Kruuse C, Pauls MMH, Pereira AC, Hainsworth AH. The cGMP-Degrading Enzyme Phosphodiesterase-5 (PDE5) in Cerebral Small Arteries of Older People. J Neuropathol Exp Neurol 2019; 78:191-194. [PMID: 30590671 DOI: 10.1093/jnen/nly117] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2024] Open
Abstract
Cerebral small vessel disease in deep penetrating arteries is a major cause of lacunar infarcts, white matter lesions and vascular cognitive impairment. Local cerebral blood flow in these small vessels is controlled by endothelial-derived nitric oxide, which exerts a primary vasodilator stimulus on vascular myocytes, via cytoplasmic cyclic GMP. Here, we investigated whether the cGMP-degrading enzyme phosphodiesterase-5 (PDE5) is present in small penetrating arteries in the deep subcortical white matter of older people. Frontal cortical tissue blocks were examined from donated brains of older people (n = 42, 24 male: 18 female, median age 81, range: 59-100 years). PDE5, detected by immunohistochemical labeling, was graded as absent, sparse, or abundant in vascular cells within small arteries in subcortical white matter (vessel outer diameter: 20-100 µm). PDE5 labeling within arterial myocytes was detected in all cases. Degree of PDE5 expression (absent, sparse, or abundant) was not associated with age or with neuropathological diagnosis of small vessel disease. In conclusion, PDE5 is present in vascular myocytes within small penetrating arteries in older people. This is a potential molecular target for pharmacological interventions.
Collapse
Affiliation(s)
- Ekta Vasita
- Molecular and Clinical Sciences Research Institute, St. Georges University of London, London, United Kingdom
| | - Saiqa Yasmeen
- Department of Neurology, Herlev Hospital, Herlev Ringvej 75, 2730 Herlev, Denmark
| | - Joycelyn Andoh
- Molecular and Clinical Sciences Research Institute, St. Georges University of London, London, United Kingdom
| | - Leslie R Bridges
- Department of Cellular Pathology, St. George's University Hospitals NHS Foundation Trust, Blackshaw Road, SW17 0QT, London, United Kingdom
| | - Christina Kruuse
- Department of Neurology, Herlev Hospital, Herlev Ringvej 75, 2730 Herlev, Denmark
| | - Mathilde M H Pauls
- Molecular and Clinical Sciences Research Institute, St. Georges University of London, London, United Kingdom
| | - Anthony C Pereira
- Department of Neurology, St. George's University Hospitals NHS Foundation Trust, London, Blackshaw Road, SW17 0QT, United Kingdom
| | - Atticus H Hainsworth
- Molecular and Clinical Sciences Research Institute, St. Georges University of London, London, United Kingdom
| |
Collapse
|
26
|
Kelly SC, McKay EC, Beck JS, Collier TJ, Dorrance AM, Counts SE. Locus Coeruleus Degeneration Induces Forebrain Vascular Pathology in a Transgenic Rat Model of Alzheimer's Disease. J Alzheimers Dis 2019; 70:371-388. [PMID: 31177220 PMCID: PMC6929678 DOI: 10.3233/jad-190090] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Noradrenergic locus coeruleus (LC) neuron loss is a significant feature of mild cognitive impairment and Alzheimer's disease (AD). The LC is the primary source of norepinephrine in the forebrain, where it modulates attention and memory in vulnerable cognitive regions such as prefrontal cortex (PFC) and hippocampus. Furthermore, LC-mediated norepinephrine signaling is thought to play a role in blood-brain barrier (BBB) maintenance and neurovascular coupling, suggesting that LC degeneration may impact the high comorbidity of cerebrovascular disease and AD. However, the extent to which LC projection system degeneration influences vascular pathology is not fully understood. To address this question in vivo, we stereotactically lesioned LC projection neurons innervating the PFC of six-month-old Tg344-19 AD rats using the noradrenergic immunotoxin, dopamine-β-hydroxylase IgG-saporin (DBH-sap), or an untargeted control IgG-saporin (IgG-sap). DBH-sap-lesioned animals performed significantly worse than IgG-sap animals on the Barnes maze task in measures of both spatial and working memory. DBH-sap-lesioned rats also displayed increased amyloid and inflammation pathology compared to IgG-sap controls. However, we also discovered prominent parenchymal albumin extravasation with DBH-sap lesions indicative of BBB breakdown. Moreover, microvessel wall-to-lumen ratios were increased in the PFC of DBH-sap compared to IgG-sap rats, suggesting that LC deafferentation results in vascular remodeling. Finally, we noted an early emergence of amyloid angiopathy in the DBH-sap-lesioned Tg344-19 AD rats. Taken together, these data indicate that LC projection system degeneration is a nexus lesion that compromises both vascular and neuronal function in cognitive brain areas during the prodromal stages of AD.
Collapse
Affiliation(s)
- Sarah C. Kelly
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Cell and Molecular Biology Program, Michigan State University, East Lansing, MI, USA
| | - Erin C. McKay
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - John S. Beck
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Timothy J. Collier
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Cell and Molecular Biology Program, Michigan State University, East Lansing, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
| | - Anne M. Dorrance
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Scott E. Counts
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI, USA
- Cell and Molecular Biology Program, Michigan State University, East Lansing, MI, USA
- Neuroscience Program, Michigan State University, East Lansing, MI, USA
- Department of Family Medicine, Michigan State University, Grand Rapids, MI, USA
- Hauenstein Neurosciences Center, Mercy Health Saint Mary’s Hospital, Grand Rapids, MI, USA
- Michigan Alzheimer’s Disease Core Center, Ann Arbor, MI, USA
| |
Collapse
|
27
|
Humphreys CA, Jansen MA, Muñoz Maniega S, González-Castro V, Pernet C, Deary IJ, Al-Shahi Salman R, Wardlaw JM, Smith C. A protocol for precise comparisons of small vessel disease lesions between ex vivo magnetic resonance imaging and histopathology. Int J Stroke 2018; 14:310-320. [PMID: 30196792 PMCID: PMC6604680 DOI: 10.1177/1747493018799962] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Rationale Neuroimaging and clinical studies have defined human sporadic cerebral small vessel disease but the pathophysiology remains relatively poorly understood. To develop effective therapies and preventative strategies, we must better understand the heterogeneity and development of small vessel disease at a cellular level. Hypothesis Small vessel disease lesions as seen on neuroimaging have specific neuropathological correlates. Methods and design Standard histological samples are taken from strategic areas of the brain typically affected by small vessel disease, in cases with a range of disease from mild to severe and controls. Tissue is formalin fixed, scanned using 7-tesla magnetic resonance imaging and processed for histology. Histological slides are digitalized then registered with the corresponding magnetic resonance image. Small vessel disease burden is assessed and lesions are precisely identified on the ex vivo imaging and microscopy independently then compared. The tissue can be interrogated using multiple magnetic resonance sequences and histological methods targeting the gliovascular unit. Study outcomes The primary outcome is identifying and defining the cellular characteristics of small vessel disease lesions compared to imaging. Secondary outcomes are related to obtaining information about abnormalities of protein expression in the gliovascular unit, defining groups of small vessel disease severity in our cohorts for future analysis and developing a reliable, reproducible protocol for accurate radiological–histological lesion comparison, which can be applied to other neurological diseases in the future. Discussion Comprehensive, precise pathological–radiological–clinical correlations in small vessel disease will provide greater insight into associations and pathophysiology underlying magnetic resonance imaging findings in normal- and abnormal-appearing tissue, ex vivo and in vivo.
Collapse
Affiliation(s)
- Catherine A Humphreys
- 1 Academic Neuropathology, University of Edinburgh, Edinburgh, UK.,2 Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Maurits A Jansen
- 2 Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Susana Muñoz Maniega
- 2 Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.,3 Neuroimaging Sciences, University of Edinburgh, Edinburgh, UK
| | - Víctor González-Castro
- 4 Department of Electrical, Systems and Automatic Engineering, Universidad de León, León, Spain
| | - Cyril Pernet
- 2 Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.,3 Neuroimaging Sciences, University of Edinburgh, Edinburgh, UK
| | - Ian J Deary
- 2 Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.,5 Department of Psychology, University of Edinburgh, Edinburgh, UK
| | | | - Joanna M Wardlaw
- 2 Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.,3 Neuroimaging Sciences, University of Edinburgh, Edinburgh, UK.,6 UK Dementia Research Institute at the University of Edinburgh, Edinburgh, UK
| | - Colin Smith
- 1 Academic Neuropathology, University of Edinburgh, Edinburgh, UK.,2 Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
28
|
Li H, Guo J, Wang A, Zhang D, Luo Y, Wang W, Li X, Tang Z, Guo X. Assessment of risk factors for cerebrovascular disease among the elderly in Beijing: A 23-year community-based prospective study in China. Arch Gerontol Geriatr 2018; 79:39-44. [PMID: 30096587 DOI: 10.1016/j.archger.2018.07.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 07/12/2018] [Accepted: 07/27/2018] [Indexed: 11/18/2022]
Abstract
INTRODUCTION There are few studies on how lifestyle factors and mental conditions modulate the cerebrovascular diseases (CBVD) mortality risk are rare in the Asian elderly. AIM To comprehensively assess the impact of lifestyle factors and mental conditions on the mortality risk of CBVD among the Chinese older adults. MATERIAL AND METHODS This community-based prospective cohort study was based on the Beijing Longitudinal Study of Aging. We included 2101 participants aged ≥55 years who were interviewed in August 1992 and followed until December 2015. Baseline sociodemographic variables, lifestyle behaviors, and medical conditions were collected using a standard questionnaire. In addition, biochemical parameters, the Activities of Daily Living (ADL) scale, Center for Epidemiological Studies Depression (CES-D) scale, and Mini-Mental State Examination (MMSE) were performed. Hazard ratio (HR) and 95% confidence intervals (CI) was estimated from the competing risk model. RESULTS During the follow-up period, 576 (27.42%) CBVD events were documented. Multivariable analysis showed that hypertension (HR = 2.331, 95% CI = 1.652-3.288,P < 0.001), depression (HR=2.331, 95% CI=1.652-3.288, P < 0.001), cognitive impairment (HR=1.382, 95% CI=1.132-1.689, P < 0.001), and coronary heart diseases (HR=1.360, 95% CI=1.095-1.689, P = 0.005) were independently associated with CBVD, while body mass index, fasting blood glucose, triglycerides, drinking, and smoking were not associated with CBVD (all P > 0.05). CONCLUSIONS Males were at higher risk of CBVD than females. Age, gender, hypertension, cognitive impairment, and depression were associated with CBVD among the elderly in Beijing, China.
Collapse
Affiliation(s)
- Haibin Li
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, China
| | - Jin Guo
- Greenwood Medical Company, 300 Highway Burwood, Melbourne, Victoria, Australia
| | - Anxin Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, China
| | - Deqiang Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, China
| | - Yanxia Luo
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, China
| | - Wei Wang
- Global Health and Genomics, School of Medical Sciences and Health, Edith Cowan University, Perth, Western Australia, Australia
| | - Xia Li
- Department of Mathematics and Statistics, La Trobe University, Victoria, Australia
| | - Zhe Tang
- Beijing Geriatric Healthcare Center, Xuan Wu Hospital, Capital Medical University, Beijing, China.
| | - Xiuhua Guo
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China; Beijing Municipal Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, China.
| |
Collapse
|
29
|
Patel T, Brookes KJ, Turton J, Chaudhury S, Guetta-Baranes T, Guerreiro R, Bras J, Hernandez D, Singleton A, Francis PT, Hardy J, Morgan K. Whole-exome sequencing of the BDR cohort: evidence to support the role of the PILRA gene in Alzheimer's disease. Neuropathol Appl Neurobiol 2018; 44:506-521. [PMID: 29181857 PMCID: PMC6005734 DOI: 10.1111/nan.12452] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 11/22/2017] [Indexed: 02/03/2023]
Abstract
AIM Late-onset Alzheimer's disease (LOAD) accounts for 95% of all Alzheimer's cases and is genetically complex in nature. Overlapping clinical and neuropathological features between AD, FTD and Parkinson's disease highlight the potential role of genetic pleiotropy across diseases. Recent genome-wide association studies (GWASs) have uncovered 20 new loci for AD risk; however, these exhibit small effect sizes. Using NGS, here we perform association analyses using exome-wide and candidate-gene-driven approaches. METHODS Whole-exome sequencing was performed on 132 AD cases and 53 control samples. Exome-wide single-variant association and gene burden tests were performed for 76 640 nonsingleton variants. Samples were also screened for known causative mutations in familial genes in AD and other dementias. Single-variant association and burden analysis was also carried out on variants in known AD and other neurological dementia genes. RESULTS Tentative single-variant and burden associations were seen in several genes with kinase and protease activity. Exome-wide burden analysis also revealed significant burden of variants in PILRA (P = 3.4 × 10-5 ), which has previously been linked to AD via GWAS, hit ZCWPW1. Screening for causative mutations in familial AD and other dementia genes revealed no pathogenic variants. Variants identified in ABCA7, SLC24A4, CD33 and LRRK2 were nominally associated with disease (P < 0.05) but did not withstand correction for multiple testing. APOE (P = 0.02) and CLU (P = 0.04) variants showed significant burden on AD. CONCLUSIONS In addition, polygenic risk scores (PRS) were able to distinguish between cases and controls with 83.8% accuracy using 3268 variants, sex, age at death and APOE ε4 and ε2 status as predictors.
Collapse
Affiliation(s)
- T Patel
- Human Genetics Group, University of Nottingham, Nottingham, UK
| | - K J Brookes
- Human Genetics Group, University of Nottingham, Nottingham, UK
| | - J Turton
- Human Genetics Group, University of Nottingham, Nottingham, UK
| | - S Chaudhury
- Human Genetics Group, University of Nottingham, Nottingham, UK
| | | | - R Guerreiro
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute at UCL (UK DRI), London, UK
- Department of Medical Sciences, Institute of Biomedicine-iBiMED, University of Aveiro, Aveiro, Portugal
| | - J Bras
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute at UCL (UK DRI), London, UK
- Department of Medical Sciences, Institute of Biomedicine-iBiMED, University of Aveiro, Aveiro, Portugal
| | - D Hernandez
- Laboratory of Neurogenetics, National Institute of Aging, National Institute of Health, Bethesda, MD, USA
| | - A Singleton
- Laboratory of Neurogenetics, National Institute of Aging, National Institute of Health, Bethesda, MD, USA
| | - P T Francis
- Brains for Dementia Research Resource, Wolfson Centre for Age Related Diseases, King's College London, London, UK
| | - J Hardy
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute at UCL (UK DRI), London, UK
| | - K Morgan
- Human Genetics Group, University of Nottingham, Nottingham, UK
| |
Collapse
|
30
|
Paradise MB, Shepherd CE, Wen W, Sachdev PS. Neuroimaging and neuropathology indices of cerebrovascular disease burden. Neurology 2018; 91:310-320. [DOI: 10.1212/wnl.0000000000005997] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 05/17/2018] [Indexed: 11/15/2022] Open
Abstract
ObjectiveTo systematically review the literature on the use of both neuroimaging and neuropathologic indices of cerebrovascular disease (CVD) burden, as estimation of this burden could have multiple benefits in the diagnosis and prognosis of cognitive impairment and dementia.MethodsMEDLINE and EMBASE databases were searched (inception to June 2017) to obtain and then systematically review all pertinent neuroimaging and neuropathology studies, where an index of CVD was developed or tested.ResultsTwenty-five neuroimaging articles were obtained, which included 4 unique indices. These utilized a limited range of CVD markers from mainly structural MRI, most commonly white matter hyperintensities (WMH), cerebral microbleeds, and dilated perivascular spaces. Weighting of the constituent markers was often coarse. There were 7 unique neuropathology indices, which were heterogeneous in their regions sampled and lesions examined.ConclusionThere is increasing interest in indices of total CVD burden that incorporate multiple lesions, as traditional individual markers of CVD such as WMH only provide limited information. Neuropathologic indices are needed to validate neuroimaging findings. The studies clearly demonstrated proof of concept that information from multiple imaging measures of CVD provide more information, including a stronger association with cognitive impairment and dementia, than that provided by a single measure. There has been limited exploration of the psychometric properties of published indices and no comparison between indices. Further development of indices is recommended, including the use of data from diffusion tensor and perfusion imaging.
Collapse
|
31
|
Rajani RM, Quick S, Ruigrok SR, Graham D, Harris SE, Verhaaren BFJ, Fornage M, Seshadri S, Atanur SS, Dominiczak AF, Smith C, Wardlaw JM, Williams A. Reversal of endothelial dysfunction reduces white matter vulnerability in cerebral small vessel disease in rats. Sci Transl Med 2018; 10:10/448/eaam9507. [DOI: 10.1126/scitranslmed.aam9507] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 01/31/2018] [Accepted: 06/08/2018] [Indexed: 12/23/2022]
|
32
|
Wallin A, Román GC, Esiri M, Kettunen P, Svensson J, Paraskevas GP, Kapaki E. Update on Vascular Cognitive Impairment Associated with Subcortical Small-Vessel Disease. J Alzheimers Dis 2018; 62:1417-1441. [PMID: 29562536 PMCID: PMC5870030 DOI: 10.3233/jad-170803] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2017] [Indexed: 02/06/2023]
Abstract
Subcortical small-vessel disease (SSVD) is a disorder well characterized from the clinical, imaging, and neuropathological viewpoints. SSVD is considered the most prevalent ischemic brain disorder, increasing in frequency with age. Vascular risk factors include hypertension, diabetes, hyperlipidemia, elevated homocysteine, and obstructive sleep apnea. Ischemic white matter lesions are the hallmark of SSVD; other pathological lesions include arteriolosclerosis, dilatation of perivascular spaces, venous collagenosis, cerebral amyloid angiopathy, microbleeds, microinfarcts, lacunes, and large infarcts. The pathogenesis of SSVD is incompletely understood but includes endothelial changes and blood-brain barrier alterations involving metalloproteinases, vascular endothelial growth factors, angiotensin II, mindin/spondin, and the mammalian target of rapamycin pathway. Metabolic and genetic conditions may also play a role but hitherto there are few conclusive studies. Clinical diagnosis of SSVD includes early executive dysfunction manifested by impaired capacity to use complex information, to formulate strategies, and to exercise self-control. In comparison with Alzheimer's disease (AD), patients with SSVD show less pronounced episodic memory deficits. Brain imaging has advanced substantially the diagnostic tools for SSVD. With the exception of cortical microinfarcts, all other lesions are well visualized with MRI. Diagnostic biomarkers that separate AD from SSVD include reduction of cerebrospinal fluid amyloid-β (Aβ)42 and of the ratio Aβ42/Aβ40 often with increased total tau levels. However, better markers of small-vessel function of intracerebral blood vessels are needed. The treatment of SSVD remains unsatisfactory other than control of vascular risk factors. There is an urgent need of finding targets to slow down and potentially halt the progression of this prevalent, but often unrecognized, disorder.
Collapse
Affiliation(s)
- Anders Wallin
- Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Gothenburg Sweden and Memory Clinic at Department of Neuropsychiatry, Sahlgrenska University, Hospital, Gothenburg, Sweden
| | - Gustavo C. Román
- Department of Neurology, Methodist Neurological Institute, Houston, TX, USA
- Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Margaret Esiri
- Neuropathology Department, West Wing, John Radcliffe Hospital, Oxford, UK
| | - Petronella Kettunen
- Institute of Neuroscience and Physiology at Sahlgrenska Academy, University of Gothenburg, Gothenburg Sweden and Memory Clinic at Department of Neuropsychiatry, Sahlgrenska University, Hospital, Gothenburg, Sweden
- Nuffield Department of Clinical Neurosciences, University of Oxford, West Wing, John Radcliffe Hospital, Oxford, UK
| | - Johan Svensson
- Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - George P. Paraskevas
- 1st Department of Neurology, Neurochemistry Unit, National and Kapodistrian University of Athens, Athens, Greece
| | - Elisabeth Kapaki
- 1st Department of Neurology, Neurochemistry Unit, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
33
|
Kalaria RN. The pathology and pathophysiology of vascular dementia. Neuropharmacology 2017; 134:226-239. [PMID: 29273521 DOI: 10.1016/j.neuropharm.2017.12.030] [Citation(s) in RCA: 211] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 12/13/2017] [Accepted: 12/18/2017] [Indexed: 02/07/2023]
Abstract
Vascular dementia (VaD) is widely recognised as the second most common type of dementia. Consensus and accurate diagnosis of clinically suspected VaD relies on wide-ranging clinical, neuropsychological and neuroimaging measures in life but more importantly pathological confirmation. Factors defining subtypes of VaD include the nature and extent of vascular pathologies, degree of involvement of extra and intracranial vessels and the anatomical location of tissue changes as well as time after the initial vascular event. Atherosclerotic and cardioembolic diseases combined appear the most common subtypes of vascular brain injury. In recent years, cerebral small vessel disease (SVD) has gained prominence worldwide as an important substrate of cognitive impairment. SVD is characterised by arteriolosclerosis, lacunar infarcts and cortical and subcortical microinfarcts and diffuse white matter changes, which involve myelin loss and axonal abnormalities. Global brain atrophy and focal degeneration of the cerebrum including medial temporal lobe atrophy are also features of VaD similar to Alzheimer's disease. Hereditary arteriopathies have provided insights into the mechanisms of dementia particularly how arteriolosclerosis, a major contributor of SVD promotes cognitive impairment. Recently developed and validated neuropathology guidelines indicated that the best predictors of vascular cognitive impairment were small or lacunar infarcts, microinfarcts, perivascular space dilation, myelin loss, arteriolosclerosis and leptomeningeal cerebral amyloid angiopathy. While these substrates do not suggest high specificity, VaD is likely defined by key neuronal and dendro-synaptic changes resulting in executive dysfunction and related cognitive deficits. Greater understanding of the molecular pathology is needed to clearly define microvascular disease and vascular substrates of dementia. This article is part of the Special Issue entitled 'Cerebral Ischemia'.
Collapse
Affiliation(s)
- Raj N Kalaria
- Institute of Neuroscience, Newcastle University, Campus for Ageing & Vitality, Newcastle Upon Tyne NE4 5PL, United Kingdom.
| |
Collapse
|
34
|
Zhang XJ, Sun TC, Liu ZW, Wang FJ, Wang YD, Liu J. Effects of Tianmagouteng particles on brain cognitive function in spontaneously hypertensive rats with hyperactivity of liver-yang: A [F-18] FDG micro-PET imaging study. Biomed Pharmacother 2017; 95:1838-1843. [PMID: 28968928 DOI: 10.1016/j.biopha.2017.08.100] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 08/11/2017] [Accepted: 08/23/2017] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE To collect visualized proof of Tianmagouteng particles (TMGTP) in alleviating cognitive dysfunction and to explore its effects on brain activity in spontaneously hypertensive rats (SHRs) with hyperactivity of liver-yang (Gan Yang Shang Kang, GYSK). METHODS Sixteen SHRs were randomized into treatment group and non-treatment. The SHR with GYSK was induced by gavaging aconite decoction (10mL/kg at 0.2g/mL). After the SHR models were prepared, the rats in the treatment group were administered TMGTP (10mL/kg) once a day for 14days.The rats in the non-treatment group or normal rats (control group) received an equivalent volume of saline. Morris water maze test was conducted before and after the treatment to observe cognitive function. Fluorine 18-deoxy glucose [F-18]FDG micro-PET brain imaging scans was performed after treatment. Data were analyzed with two-sample t-test (P<0. 001) using SPM2 image analysis software. RESULTS Compared with the non-treatment group, the escape latency significantly decreased but the frequency of entrance into the target zone significantly increased in the treatment group. Consistent with the alteration of cognitive functions, TMGTP induced strong brain activity in the following sites: right dorsolateral nucleus and ventrolateral nucleus of thalamus, amygdala, left met thalamus, cerebellum leaflets, original crack, front cone crack, loop-shaped leaflets; but deactivation of right medial frontal gyrus, bilateral corpus callosum, hippocampus, and left dentate gyrus. CONCLUSION TMGTP could alleviate cognitive dysfunction in SHRs with GYSK, which was possibly by inducing alteration of glucose metabolism in different brain regions with corresponding functions.
Collapse
Affiliation(s)
- Xiu-Jing Zhang
- Department of Cardiology, Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Tian-Cai Sun
- Department of Emergency, Chinese Medicine Hospital of Zhengzhou, Zhengzhou, Henan, China
| | - Zi-Wang Liu
- Department of Cardiology, Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China.
| | - Feng-Jiao Wang
- Department of Digestive Disease, Beijing Fengtai Hospital of Integrated Chinese and Western Medicine, Beijing, China
| | - Yong-De Wang
- Department of Cardiology, Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Liu
- Department of Cardiology, Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
35
|
McAleese KE, Walker L, Graham S, Moya ELJ, Johnson M, Erskine D, Colloby SJ, Dey M, Martin-Ruiz C, Taylor JP, Thomas AJ, McKeith IG, De Carli C, Attems J. Parietal white matter lesions in Alzheimer's disease are associated with cortical neurodegenerative pathology, but not with small vessel disease. Acta Neuropathol 2017. [PMID: 28638989 PMCID: PMC5563333 DOI: 10.1007/s00401-017-1738-2] [Citation(s) in RCA: 188] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cerebral white matter lesions (WML) encompass axonal loss and demyelination, and the pathogenesis is assumed to be small vessel disease (SVD)-related ischemia. However, WML may also result from the activation of Wallerian degeneration as a consequence of cortical Alzheimer's disease (AD) pathology, i.e. hyperphosphorylated tau (HPτ) and amyloid-beta (Aβ) deposition. WML seen in AD have a posterior predominance compared to non-demented individuals but it is unclear whether the pathological and molecular signatures of WML differ between these two groups. We investigated differences in the composition and aetiology of parietal WML from AD and non-demented controls. Parietal WML tissue from 55 human post-mortem brains (AD, n = 27; non-demented controls, n = 28) were quantitatively assessed for axonal loss and demyelination, as well as for cortical HPτ and Aβ burden and SVD. Biochemical assessment included Wallerian degeneration protease calpain and the myelin-associated glycoprotein (MAG) to proteolipid protein (PLP) ratio (MAG:PLP) as a measure of hypoperfusion. WML severity was associated with both axonal loss and demyelination in AD, but only with demyelination in controls. Calpain was significantly increased in WML tissue in AD, whereas MAG:PLP was significantly reduced in controls. Calpain levels were associated with increasing amounts of cortical AD-pathology but not SVD. We conclude that parietal WML seen in AD differ in their pathological composition and aetiology compared to WML seen in aged controls: WML seen in AD may be associated with Wallerian degeneration that is triggered by cortical AD-pathology, whereas WML in aged controls are due to ischaemia. Hence, parietal WML as seen on MRI should not invariably be interpreted as a surrogate biomarker for SVD as they may be indicative of cortical AD-pathology, and therefore, AD should also be considered as the main underlying cause for cognitive impairment in cases with parietal WML.
Collapse
|
36
|
Solfrizzi V, Custodero C, Lozupone M, Imbimbo BP, Valiani V, Agosti P, Schilardi A, D’Introno A, La Montagna M, Calvani M, Guerra V, Sardone R, Abbrescia DI, Bellomo A, Greco A, Daniele A, Seripa D, Logroscino G, Sabbá C, Panza F. Relationships of Dietary Patterns, Foods, and Micro- and Macronutrients with Alzheimer’s Disease and Late-Life Cognitive Disorders: A Systematic Review. J Alzheimers Dis 2017; 59:815-849. [DOI: 10.3233/jad-170248] [Citation(s) in RCA: 179] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Vincenzo Solfrizzi
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari Aldo Moro, Bari, Italy
| | - Carlo Custodero
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari Aldo Moro, Bari, Italy
| | - Madia Lozupone
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Bruno P. Imbimbo
- Department of Research and Development, Chiesi Farmaceutici, Parma, Italy
| | - Vincenzo Valiani
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari Aldo Moro, Bari, Italy
| | - Pasquale Agosti
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari Aldo Moro, Bari, Italy
| | - Andrea Schilardi
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari Aldo Moro, Bari, Italy
| | - Alessia D’Introno
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari Aldo Moro, Bari, Italy
| | - Maddalena La Montagna
- Department of Clinical and Experimental Medicine, Psychiatric Unit, University of Foggia, Foggia, Italy
| | - Mariapaola Calvani
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Vito Guerra
- National Institute for Digestive Diseases, IRCCS “Saverio de Bellis”, Castellana, Bari, Italy
| | - Rodolfo Sardone
- National Institute for Digestive Diseases, IRCCS “Saverio de Bellis”, Castellana, Bari, Italy
| | - Daniela I. Abbrescia
- National Institute for Digestive Diseases, IRCCS “Saverio de Bellis”, Castellana, Bari, Italy
| | - Antonello Bellomo
- Department of Clinical and Experimental Medicine, Psychiatric Unit, University of Foggia, Foggia, Italy
| | - Antonio Greco
- Geriatric Unit and Laboratory of Gerontology and Geriatrics, Department of Medical Sciences, IRCCS “Casa Sollievo della Sofferenza”, San Giovanni Rotondo, Foggia, Italy
| | - Antonio Daniele
- Institute of Neurology, Catholic University of Sacred Heart, Rome, Italy
| | - Davide Seripa
- Geriatric Unit and Laboratory of Gerontology and Geriatrics, Department of Medical Sciences, IRCCS “Casa Sollievo della Sofferenza”, San Giovanni Rotondo, Foggia, Italy
| | - Giancarlo Logroscino
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
- Department of Clinical Research in Neurology, University of Bari Aldo Moro, “Pia Fondazione Cardinale G. Panico”, Tricase, Lecce, Italy
| | - Carlo Sabbá
- Geriatric Medicine-Memory Unit and Rare Disease Centre, University of Bari Aldo Moro, Bari, Italy
| | - Francesco Panza
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
- Geriatric Unit and Laboratory of Gerontology and Geriatrics, Department of Medical Sciences, IRCCS “Casa Sollievo della Sofferenza”, San Giovanni Rotondo, Foggia, Italy
- Department of Clinical Research in Neurology, University of Bari Aldo Moro, “Pia Fondazione Cardinale G. Panico”, Tricase, Lecce, Italy
| |
Collapse
|
37
|
Endothelial cell-oligodendrocyte interactions in small vessel disease and aging. Clin Sci (Lond) 2017; 131:369-379. [PMID: 28202749 PMCID: PMC5310718 DOI: 10.1042/cs20160618] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/28/2016] [Accepted: 12/14/2016] [Indexed: 12/11/2022]
Abstract
Cerebral small vessel disease (SVD) is a prevalent, neurological disease that significantly increases the risk of stroke and dementia. The main pathological changes are vascular, in the form of lipohyalinosis and arteriosclerosis, and in the white matter (WM), in the form of WM lesions. Despite this, it is unclear to what extent the key cell types involved–the endothelial cells (ECs) of the vasculature and the oligodendrocytes of the WM–interact. Here, we describe the work that has so far been carried out suggesting an interaction between ECs and oligodendrocytes in SVD. As these interactions have been studied in more detail in other disease states and in development, we explore these systems and discuss the role these mechanisms may play in SVD.
Collapse
|
38
|
Geraldes R, Esiri MM, DeLuca GC, Palace J. Age-related small vessel disease: a potential contributor to neurodegeneration in multiple sclerosis. Brain Pathol 2017; 27:707-722. [PMID: 27864848 DOI: 10.1111/bpa.12460] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Accepted: 11/17/2016] [Indexed: 12/18/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disorder of the central nervous system wherein, after an initial phase of transient neurological defects, slow neurological deterioration due to progressive neuronal loss ensues. Age is a major determinant of MS progression onset and disability. Over the past years, several mechanisms have been proposed to explain the key drivers of neurodegeneration and disability accumulation in MS. However, the effect of commonly encountered age-related cerebral vessel disease, namely small vessel disease (SVD), has been largely neglected and constitutes the aim of this review. SVD shares some features with MS, that is, white matter demyelination and brain atrophy, and has been shown to contribute to the neuronal damage seen in vascular cognitive impairment. Several lines of evidence suggest that an interaction between MS and SVD may influence MS-related neurodegeneration. SVD may contribute to hypoperfusion, reduced vascular reactivity and tissue hypoxia, features seen in MS. Venule and endothelium abnormalities have been documented in MS but the role of arterioles and of other neurovascular unit structures, such as the pericyte, has not been explored. Vascular risk factors (VRF) have recently been associated with faster progression in MS, though the mechanisms are unclear since very few studies have addressed the impact of VRF and SVD on MS imaging and pathology outcomes. Therapeutic agents targeting the microvasculature and the neurovascular unit may impact both SVD and MS and may benefit patients with dual pathology.
Collapse
Affiliation(s)
- Ruth Geraldes
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, UK
| | - Margaret M Esiri
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, UK
| | - Gabriele C DeLuca
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, UK
| | - Jacqueline Palace
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, Oxford, UK
| |
Collapse
|
39
|
The Vascular Impairment of Cognition Classification Consensus Study. Alzheimers Dement 2016; 13:624-633. [DOI: 10.1016/j.jalz.2016.10.007] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 08/31/2016] [Accepted: 10/22/2016] [Indexed: 01/03/2023]
|
40
|
Wardlaw JM, Makin SJ, Valdés Hernández MC, Armitage PA, Heye AK, Chappell FM, Muñoz‐Maniega S, Sakka E, Shuler K, Dennis MS, Thrippleton MJ. Blood‐brain barrier failure as a core mechanism in cerebral small vessel disease and dementia: evidence from a cohort study. Alzheimers Dement 2016. [PMCID: PMC5472180 DOI: 10.1016/j.jalz.2016.09.006] [Citation(s) in RCA: 174] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Introduction Methods Results Discussion In 201 patients with non-disabling stroke and white matter hyperintensities (WMH), we measured blood-brain barrier (BBB) leakage with contrast-enhanced MRI. BBB leakage was higher in WMH than in normal appearing white matter. BBB leakage increased in both WMH and normal appearing white matter with the burden of small vessel disease, hypertension, and age. BBB leakage predicted cognitive decline one year later. The leakage pattern supported the hypothesis that BBB leak was pathogenic of diffuse brain damage in cerebral small vessel disease.
Collapse
Affiliation(s)
- Joanna M. Wardlaw
- Centre for Clinical Brain Sciences University of Edinburgh Edinburgh UK
| | - Stephen J. Makin
- Centre for Clinical Brain Sciences University of Edinburgh Edinburgh UK
| | | | - Paul A. Armitage
- Academic Unit of Radiology, Department of Cardiovascular Science University of Sheffield, Royal Hallamshire Hospital Sheffield UK
| | - Anna K. Heye
- Centre for Clinical Brain Sciences University of Edinburgh Edinburgh UK
| | | | | | - Eleni Sakka
- Centre for Clinical Brain Sciences University of Edinburgh Edinburgh UK
| | - Kirsten Shuler
- Centre for Clinical Brain Sciences University of Edinburgh Edinburgh UK
| | - Martin S. Dennis
- Centre for Clinical Brain Sciences University of Edinburgh Edinburgh UK
| | | |
Collapse
|
41
|
Charidimou A, Pantoni L, Love S. The concept of sporadic cerebral small vessel disease: A road map on key definitions and current concepts. Int J Stroke 2016; 11:6-18. [PMID: 26763016 DOI: 10.1177/1747493015607485] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Sporadic cerebral small vessel disease is considered to be among the most common known neuropathological processes and has an important role in stroke, cognitive impairment, and functional loss in elderly persons. The term is now commonly used to describe a range of neuroimaging, neuropathological, and associated clinical features, the pathogenesis of which is largely unclear but that are thought to arise from disease affecting the perforating cerebral arterioles, capillaries, and venules. Modern neuroimaging has revolutionized our understanding of the consequences of small vessels disease on the brain parenchyma, even though small arteries, arterioles, capillaries, and venules are difficult to be directly visualized with current techniques used in clinical practice. In this short review, we focus on histopathological and neuroimaging perspectives, basic definitions, and recent advances in the field.
Collapse
Affiliation(s)
- Andreas Charidimou
- Hemorrhagic Stroke Research Program, Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, USA UCL Institute of Neurology and The National Hospital for Neurology and Neurosurgery, Queen Square, London WC1N 3BG, UK
| | - Leonardo Pantoni
- NEUROFARBA Department, University of Florence and Stroke Unit and Neurology, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | - Seth Love
- Dementia Research Group, Institute of Clinical Neurosciences, University of Bristol, Learning & Research Level 2, Southmead Hospital, Bristol, UK
| |
Collapse
|
42
|
Skrobot OA, Attems J, Esiri M, Hortobágyi T, Ironside JW, Kalaria RN, King A, Lammie GA, Mann D, Neal J, Ben-Shlomo Y, Kehoe PG, Love S. Vascular cognitive impairment neuropathology guidelines (VCING): the contribution of cerebrovascular pathology to cognitive impairment. Brain 2016; 139:2957-2969. [DOI: 10.1093/brain/aww214] [Citation(s) in RCA: 171] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Accepted: 07/03/2016] [Indexed: 01/01/2023] Open
|
43
|
McAleese KE, Alafuzoff I, Charidimou A, De Reuck J, Grinberg LT, Hainsworth AH, Hortobagyi T, Ince P, Jellinger K, Gao J, Kalaria RN, Kovacs GG, Kövari E, Love S, Popovic M, Skrobot O, Taipa R, Thal DR, Werring D, Wharton SB, Attems J. Post-mortem assessment in vascular dementia: advances and aspirations. BMC Med 2016; 14:129. [PMID: 27600683 PMCID: PMC5011905 DOI: 10.1186/s12916-016-0676-5] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 08/19/2016] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Cerebrovascular lesions are a frequent finding in the elderly population. However, the impact of these lesions on cognitive performance, the prevalence of vascular dementia, and the pathophysiology behind characteristic in vivo imaging findings are subject to controversy. Moreover, there are no standardised criteria for the neuropathological assessment of cerebrovascular disease or its related lesions in human post-mortem brains, and conventional histological techniques may indeed be insufficient to fully reflect the consequences of cerebrovascular disease. DISCUSSION Here, we review and discuss both the neuropathological and in vivo imaging characteristics of cerebrovascular disease, prevalence rates of vascular dementia, and clinico-pathological correlations. We also discuss the frequent comorbidity of cerebrovascular pathology and Alzheimer's disease pathology, as well as the difficult and controversial issue of clinically differentiating between Alzheimer's disease, vascular dementia and mixed Alzheimer's disease/vascular dementia. Finally, we consider additional novel approaches to complement and enhance current post-mortem assessment of cerebral human tissue. CONCLUSION Elucidation of the pathophysiology of cerebrovascular disease, clarification of characteristic findings of in vivo imaging and knowledge about the impact of combined pathologies are needed to improve the diagnostic accuracy of clinical diagnoses.
Collapse
Affiliation(s)
- Kirsty E McAleese
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Irina Alafuzoff
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Andreas Charidimou
- Hemorrhagic Stroke Research Program, Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | | | - Lea T Grinberg
- Departments of neurology and Pathology, University of California, San Francisco, USA.,Department of Pathology - LIM-22, University of Sao Paulo Medical School, São Paulo, Brazil
| | - Atticus H Hainsworth
- Institute of Cardiovascular and Cell Sciences, St George's University of London, London, UK
| | - Tibor Hortobagyi
- Department of Neuropathology, University of Debrecen, Debrecen, Hungary
| | - Paul Ince
- Sheffield Institute for Translational Neuroscience, Sheffield, UK
| | | | - Jing Gao
- Neurological Department, Peking Union Medical College Hospital, Beijing, China
| | - Raj N Kalaria
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Gabor G Kovacs
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Enikö Kövari
- Department of Mental Health and Psychiatry, University of Geneva, Geneva, Switzerland
| | - Seth Love
- Clincial Neurosciences, University of Bristol, Bristol, UK
| | - Mara Popovic
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Olivia Skrobot
- Clincial Neurosciences, University of Bristol, Bristol, UK
| | - Ricardo Taipa
- Unit of Neuropathology, Centro Hospitalar do Porto, University of Porto, Porto, Portugal
| | - Dietmar R Thal
- Department of Neuroscience, KU-Leuven and Department of Pathology, UZ-Leuven, Leuven, Belgium
| | - David Werring
- Institute of Neurology, University College London, London, UK
| | | | - Johannes Attems
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
44
|
Docherty AB, Shenkin SD. Cognitive decline after surgery and anaesthesia: correlation does not mean causation. Anaesthesia 2016; 71:1131-5. [PMID: 27538826 DOI: 10.1111/anae.13592] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- A B Docherty
- Department of Anaesthesia and Critical Care, University of Edinburgh, Edinburgh, UK.
| | - S D Shenkin
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK.,Department of Geriatric Medicine, NHS Lothian, Edinburgh, UK
| |
Collapse
|
45
|
Charidimou A, Martinez-Ramirez S, Reijmer YD, Oliveira-Filho J, Lauer A, Roongpiboonsopit D, Frosch M, Vashkevich A, Ayres A, Rosand J, Gurol ME, Greenberg SM, Viswanathan A. Total Magnetic Resonance Imaging Burden of Small Vessel Disease in Cerebral Amyloid Angiopathy: An Imaging-Pathologic Study of Concept Validation. JAMA Neurol 2016; 73:994-1001. [PMID: 27366898 PMCID: PMC5283697 DOI: 10.1001/jamaneurol.2016.0832] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
IMPORTANCE Cerebral amyloid angiopathy (CAA) is characteristically associated with magnetic resonance imaging (MRI) biomarkers of small vessel brain injury, including strictly lobar cerebral microbleeds, cortical superficial siderosis, centrum semiovale perivascular spaces, and white matter hyperintensities. Although these neuroimaging markers reflect distinct pathophysiologic aspects in CAA, no studies to date have combined these structural imaging features to gauge total brain small vessel disease burden in CAA. OBJECTIVES To investigate whether a composite score can be developed to capture the total brain MRI burden of small vessel disease in CAA and to explore whether this score contributes independent and complementary information about CAA severity, defined as intracerebral hemorrhage during life or bleeding-related neuropathologic changes. DESIGN, SETTING, AND PARTICIPANTS This retrospective, cross-sectional study examined a single-center neuropathologic CAA cohort of eligible patients from the Massachusetts General Hospital from January 1, 1997, through December 31, 2012. Data analysis was performed from January 2, 2015, to January 9, 2016. Patients with pathologic evidence of CAA (ie, any presence of CAA from routinely collected brain biopsy specimen, biopsy specimen at hematoma evacuation, or autopsy) and available brain MRI sequences of adequate quality, including T2-weighted, T2*-weighted gradient-recalled echo, and/or susceptibility-weighted imaging and fluid-attenuated inversion recovery sequences, were considered for the study. MAIN OUTCOMES AND MEASURES Brain MRIs were rated for lobar cerebral microbleeds, cortical superficial siderosis, centrum semiovale perivascular spaces, and white matter hyperintensities. All 4 MRI lesions were incorporated into a prespecified ordinal total small vessel disease score, ranging from 0 to 6 points. Associations with severity of CAA-associated vasculopathic changes (fibrinoid necrosis and concentric splitting of the wall), clinical presentation, number of intracerebral hemorrhages, and other imaging markers not included in the score were explored using logistic and ordinal regression. RESULTS In total, 105 patients with pathologically defined CAA were included: 52 with autopsies, 22 with brain biopsy specimens, and 31 with pathologic samples from hematoma evacuations. The mean (range) age of the patients was 73 (71-74) years, and 55 (52.4%) were women. In multivariable ordinal regression analysis, severity of CAA-associated vasculopathic changes (odds ratio, 2.40; 95% CI, 1.06-5.45; P = .04) and CAA presentation with symptomatic intracerebral hemorrhage (odds ratio, 2.23; 95% CI, 1.07-4.64; P = .03) were independently associated with the total MRI small vessel disease score. The score was associated with small, acute, diffusion-weighted imaging lesions and posterior white matter hyperintensities in adjusted analyses. CONCLUSIONS AND RELEVANCE This study provides evidence of concept validity of a total MRI small vessel disease score in CAA. After further validation, this approach can be potentially used in prospective clinical studies.
Collapse
Affiliation(s)
- Andreas Charidimou
- Hemorrhagic Stroke Research Program, Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Sergi Martinez-Ramirez
- Hemorrhagic Stroke Research Program, Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Yael D. Reijmer
- Hemorrhagic Stroke Research Program, Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Jamary Oliveira-Filho
- Hemorrhagic Stroke Research Program, Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Arne Lauer
- Hemorrhagic Stroke Research Program, Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Duangnapa Roongpiboonsopit
- Hemorrhagic Stroke Research Program, Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Matthew Frosch
- C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Anastasia Vashkevich
- Hemorrhagic Stroke Research Program, Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Alison Ayres
- Hemorrhagic Stroke Research Program, Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Jonathan Rosand
- Hemorrhagic Stroke Research Program, Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
- Division of Neurocritical Care and Emergency Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
- Center for Human Genetic Research, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Mahmut Edip Gurol
- Hemorrhagic Stroke Research Program, Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Steven M. Greenberg
- Hemorrhagic Stroke Research Program, Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| | - Anand Viswanathan
- Hemorrhagic Stroke Research Program, Department of Neurology, Massachusetts General Hospital Stroke Research Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
46
|
Nelson PT, Trojanowski JQ, Abner EL, Al-Janabi OM, Jicha GA, Schmitt FA, Smith CD, Fardo DW, Wang WX, Kryscio RJ, Neltner JH, Kukull WA, Cykowski MD, Van Eldik LJ, Ighodaro ET. "New Old Pathologies": AD, PART, and Cerebral Age-Related TDP-43 With Sclerosis (CARTS). J Neuropathol Exp Neurol 2016; 75:482-98. [PMID: 27209644 PMCID: PMC6366658 DOI: 10.1093/jnen/nlw033] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Indexed: 12/12/2022] Open
Abstract
The pathology-based classification of Alzheimer's disease (AD) and other neurodegenerative diseases is a work in progress that is important for both clinicians and basic scientists. Analyses of large autopsy series, biomarker studies, and genomics analyses have provided important insights about AD and shed light on previously unrecognized conditions, enabling a deeper understanding of neurodegenerative diseases in general. After demonstrating the importance of correct disease classification for AD and primary age-related tauopathy, we emphasize the public health impact of an underappreciated AD "mimic," which has been termed "hippocampal sclerosis of aging" or "hippocampal sclerosis dementia." This pathology affects >20% of individuals older than 85 years and is strongly associated with cognitive impairment. In this review, we provide an overview of current hypotheses about how genetic risk factors (GRN, TMEM106B, ABCC9, and KCNMB2), and other pathogenetic influences contribute to TDP-43 pathology and hippocampal sclerosis. Because hippocampal sclerosis of aging affects the "oldest-old" with arteriolosclerosis and TDP-43 pathologies that extend well beyond the hippocampus, more appropriate terminology for this disease is required. We recommend "cerebral age-related TDP-43 and sclerosis" (CARTS). A detailed case report is presented, which includes neuroimaging and longitudinal neurocognitive data. Finally, we suggest a neuropathology-based diagnostic rubric for CARTS.
Collapse
Affiliation(s)
- Peter T Nelson
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC).
| | - John Q Trojanowski
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC)
| | - Erin L Abner
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC)
| | - Omar M Al-Janabi
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC)
| | - Gregory A Jicha
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC)
| | - Frederick A Schmitt
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC)
| | - Charles D Smith
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC)
| | - David W Fardo
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC)
| | - Wang-Xia Wang
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC)
| | - Richard J Kryscio
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC)
| | - Janna H Neltner
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC)
| | - Walter A Kukull
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC)
| | - Matthew D Cykowski
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC)
| | - Linda J Van Eldik
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC)
| | - Eseosa T Ighodaro
- From the Department of Pathology, Division of Neuropathology (PTN, JHN), Department of Neurology (GAJ, FAS, CDS), Department of Statistics (DWF, RJK), Department of Anatomy and Neurobiology (PTN, JHN, LJVE, ETI), Department of Epidemiology (ELA), and Sanders-Brown Center on Aging (PTN, ELA, OMA-J, GAJ, FAS, CDS, DWF, WXW, RJK, LJVE, ETI), University of Kentucky, Lexington, Kentucky; Department of Pathology & Laboratory Medicine and Center for Neurodegenerative Disease Research, University of Pennsylvania, Philadelphia, Pennsylvannia (JQT); Department of Epidemiology, University of Washington, Seattle, Washington (WAK); and Department of Pathology, Houston Methodist Hospital, Houston, Texas (MDC)
| |
Collapse
|
47
|
Neuropathological diagnosis of vascular cognitive impairment and vascular dementia with implications for Alzheimer's disease. Acta Neuropathol 2016; 131:659-85. [PMID: 27062261 PMCID: PMC4835512 DOI: 10.1007/s00401-016-1571-z] [Citation(s) in RCA: 274] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 03/23/2016] [Accepted: 03/24/2016] [Indexed: 12/16/2022]
Abstract
Vascular dementia (VaD) is recognised as a neurocognitive disorder, which is explained by numerous vascular causes in the general absence of other pathologies. The heterogeneity of cerebrovascular disease makes it challenging to elucidate the neuropathological substrates and mechanisms of VaD as well as vascular cognitive impairment (VCI). Consensus and accurate diagnosis of VaD relies on wide-ranging clinical, neuropsychometric and neuroimaging measures with subsequent pathological confirmation. Pathological diagnosis of suspected clinical VaD requires adequate postmortem brain sampling and rigorous assessment methods to identify important substrates. Factors that define the subtypes of VaD include the nature and extent of vascular pathologies, degree of involvement of extra and intracranial vessels and the anatomical location of tissue changes. Atherosclerotic and cardioembolic diseases appear the most common substrates of vascular brain injury or infarction. Small vessel disease characterised by arteriolosclerosis and lacunar infarcts also causes cortical and subcortical microinfarcts, which appear to be the most robust substrates of cognitive impairment. Diffuse WM changes with loss of myelin and axonal abnormalities are common to almost all subtypes of VaD. Medial temporal lobe and hippocampal atrophy accompanied by variable hippocampal sclerosis are also features of VaD as they are of Alzheimer’s disease. Recent observations suggest that there is a vascular basis for neuronal atrophy in both the temporal and frontal lobes in VaD that is entirely independent of any Alzheimer pathology. Further knowledge on specific neuronal and dendro-synaptic changes in key regions resulting in executive dysfunction and other cognitive deficits, which define VCI and VaD, needs to be gathered. Hereditary arteriopathies such as cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy or CADASIL have provided insights into the mechanisms of dementia associated with cerebral small vessel disease. Greater understanding of the neurochemical and molecular investigations is needed to better define microvascular disease and vascular substrates of dementia. The investigation of relevant animal models would be valuable in exploring the pathogenesis as well as prevention of the vascular causes of cognitive impairment.
Collapse
|
48
|
Chen A, Akinyemi RO, Hase Y, Firbank MJ, Ndung'u MN, Foster V, Craggs LJL, Washida K, Okamoto Y, Thomas AJ, Polvikoski TM, Allan LM, Oakley AE, O'Brien JT, Horsburgh K, Ihara M, Kalaria RN. Frontal white matter hyperintensities, clasmatodendrosis and gliovascular abnormalities in ageing and post-stroke dementia. Brain 2015; 139:242-58. [PMID: 26667280 PMCID: PMC4905522 DOI: 10.1093/brain/awv328] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 09/29/2015] [Indexed: 01/16/2023] Open
Abstract
White matter hyperintensities as seen on brain T
2
-weighted magnetic resonance imaging are associated with varying degrees of cognitive dysfunction in stroke, cerebral small vessel disease and dementia. The pathophysiological mechanisms within the white matter accounting for cognitive dysfunction remain unclear. With the hypothesis that gliovascular interactions are impaired in subjects with high burdens of white matter hyperintensities, we performed clinicopathological studies in post-stroke survivors, who had exhibited greater frontal white matter hyperintensities volumes that predicted shorter time to dementia onset. Histopathological methods were used to identify substrates in the white matter that would distinguish post-stroke demented from post-stroke non-demented subjects. We focused on the reactive cell marker glial fibrillary acidic protein (GFAP) to study the incidence and location of clasmatodendrosis, a morphological attribute of irreversibly injured astrocytes. In contrast to normal appearing GFAP+ astrocytes, clasmatodendrocytes were swollen and had vacuolated cell bodies. Other markers such as aldehyde dehydrogenase 1 family, member L1 (ALDH1L1) showed cytoplasmic disintegration of the astrocytes. Total GFAP+ cells in both the frontal and temporal white matter were not greater in post-stroke demented versus post-stroke non-demented subjects. However, the percentage of clasmatodendrocytes was increased by >2-fold in subjects with post-stroke demented compared to post-stroke non-demented subjects (
P =
0.026) and by 11-fold in older controls versus young controls (
P <
0.023) in the frontal white matter. High ratios of clasmotodendrocytes to total astrocytes in the frontal white matter were consistent with lower Mini-Mental State Examination and the revised Cambridge Cognition Examination scores in post-stroke demented subjects. Double immunofluorescent staining showed aberrant co-localization of aquaporin 4 (AQP4) in retracted GFAP+ astrocytes with disrupted end-feet juxtaposed to microvessels. To explore whether this was associated with the disrupted gliovascular interactions or blood–brain barrier damage, we assessed the co-localization of GFAP and AQP4 immunoreactivities in post-mortem brains from adult baboons with cerebral hypoperfusive injury, induced by occlusion of three major vessels supplying blood to the brain. Analysis of the frontal white matter in perfused brains from the animals surviving 1–28 days after occlusion revealed that the highest intensity of fibrinogen immunoreactivity was at 14 days. At this survival time point, we also noted strikingly similar redistribution of AQP4 and GFAP+ astrocytes transformed into clasmatodendrocytes. Our findings suggest novel associations between irreversible astrocyte injury and disruption of gliovascular interactions at the blood–brain barrier in the frontal white matter and cognitive impairment in elderly post-stroke survivors. We propose that clasmatodendrosis is another pathological substrate, linked to white matter hyperintensities and frontal white matter changes, which may contribute to post-stroke or small vessel disease dementia.
Collapse
Affiliation(s)
- Aiqing Chen
- 1 Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle Upon Tyne, UK
| | - Rufus O Akinyemi
- 1 Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle Upon Tyne, UK
| | - Yoshiki Hase
- 1 Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle Upon Tyne, UK
| | - Michael J Firbank
- 1 Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle Upon Tyne, UK
| | | | - Vincent Foster
- 1 Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle Upon Tyne, UK
| | - Lucy J L Craggs
- 1 Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle Upon Tyne, UK
| | - Kazuo Washida
- 1 Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle Upon Tyne, UK
| | - Yoko Okamoto
- 1 Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle Upon Tyne, UK
| | - Alan J Thomas
- 1 Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle Upon Tyne, UK
| | - Tuomo M Polvikoski
- 1 Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle Upon Tyne, UK
| | - Louise M Allan
- 1 Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle Upon Tyne, UK
| | - Arthur E Oakley
- 1 Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle Upon Tyne, UK
| | - John T O'Brien
- 1 Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle Upon Tyne, UK
| | - Karen Horsburgh
- 3 Centre for Neuroregeneration, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Masafumi Ihara
- 4 Department of Stroke and Cerebrovascular Diseases, National Cerebral and Cardiovascular Center, 5-7-1 Fujishiro-dai, Suita, Osaka 565-8565, Japan
| | - Raj N Kalaria
- 1 Neurovascular Research Group, Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle Upon Tyne, UK
| |
Collapse
|
49
|
Hainsworth AH, Yeo NE, Weekman EM, Wilcock DM. Homocysteine, hyperhomocysteinemia and vascular contributions to cognitive impairment and dementia (VCID). Biochim Biophys Acta Mol Basis Dis 2015; 1862:1008-17. [PMID: 26689889 DOI: 10.1016/j.bbadis.2015.11.015] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 11/27/2015] [Accepted: 11/29/2015] [Indexed: 11/29/2022]
Abstract
Homocysteine is produced physiologically in all cells, and is present in plasma of healthy individuals (plasma [HCy]: 3-10μM). While rare genetic mutations (CBS, MTHFR) cause severe hyperhomocysteinemia ([HCy]: 100-200μM), mild-moderate hyperhomocysteinemia ([HCy]: 10-100μM) is common in older people, and is an independent risk factor for stroke and cognitive impairment. As B-vitamin supplementation (B6, B12 and folate) has well-validated homocysteine-lowering efficacy, this may be a readily-modifiable risk factor in vascular contributions to cognitive impairment and dementia (VCID). Here we review the biochemical and cellular actions of HCy related to VCID. Neuronal actions of HCy were at concentrations above the clinically-relevant range. Effects of HCy <100μM were primarily vascular, including myocyte proliferation, vessel wall fibrosis, impaired nitric oxide signalling, superoxide generation and pro-coagulant actions. HCy-lowering clinical trials relevant to VCID are discussed. Extensive clinical and preclinical data support HCy as a mediator for VCID. In our view further trials of combined B-vitamin supplementation are called for, incorporating lessons from previous trials and from recent experimental work. To maximise likelihood of treatment effect, a future trial should: supply a high-dose, combination supplement (B6, B12 and folate); target the at-risk age range; and target cohorts with low baseline B-vitamin status. This article is part of a Special Issue entitled: Vascular Contributions to Cognitive Impairment and Dementia edited by M. Paul Murphy, Roderick A. Corriveau and Donna M. Wilcock.
Collapse
Affiliation(s)
- Atticus H Hainsworth
- Cardiovascular and Cell Sciences Research Centre, St Georges University of London, London SW17 0RE, UK.
| | - Natalie E Yeo
- Cardiovascular and Cell Sciences Research Centre, St Georges University of London, London SW17 0RE, UK
| | - Erica M Weekman
- Sanders-Brown Center on Aging, University of Kentucky, Lexington KY 40536, USA
| | - Donna M Wilcock
- Sanders-Brown Center on Aging, University of Kentucky, Lexington KY 40536, USA.
| |
Collapse
|
50
|
Tight junction disruption of blood–brain barrier in white matter lesions in chronic hypertensive rats. Neuroreport 2015; 26:1039-43. [DOI: 10.1097/wnr.0000000000000464] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|