1
|
Greenberg SM, Bax F, van Veluw SJ. Amyloid-related imaging abnormalities: manifestations, metrics and mechanisms. Nat Rev Neurol 2025; 21:193-203. [PMID: 39794509 DOI: 10.1038/s41582-024-01053-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2024] [Indexed: 01/13/2025]
Abstract
Three monoclonal antibodies directed against specific forms of the amyloid-β (Aβ) peptide have been granted accelerated or traditional approval by the FDA as treatments for Alzheimer disease, representing the first step towards bringing disease-modifying treatments for this disease into clinical practice. Here, we review the detection, underlying pathophysiological mechanisms and clinical implications of amyloid-related imaging abnormalities (ARIA), the most impactful adverse effect of anti-Aβ immunotherapy. ARIA appears as regions of oedema or effusions (ARIA-E) in brain parenchyma or sulci or as haemorrhagic lesions (ARIA-H) in the form of cerebral microbleeds, convexity subarachnoid haemorrhage, cortical superficial siderosis or intracerebral haemorrhage. Analysis of the radiographic appearance of ARIA, its clinical risk factors and underlying neuropathology, and results from animal models point to a central role for cerebral amyloid angiopathy - a condition characterized by cerebrovascular Aβ deposits - as a key component, either as a direct target for antibody-mediated inflammation or as recipient of Aβ mobilized from plaques in the Alzheimer brain parenchyma. The great majority of ARIA occurrences are associated with mild or no clinical symptoms. However, ~5% of all ARIA events are severe enough to result in hospitalization, permanent disability or death and thus raise challenging clinical questions regarding patient selection and use of concomitant agents. Therefore, identifying novel approaches to predicting, modelling, preventing and treating ARIA remains a key step towards allowing safe use of anti-Aβ immunotherapy for the world's rapidly ageing population.
Collapse
Affiliation(s)
- Steven M Greenberg
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Francesco Bax
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Clinical Neurology Unit, Department of Head, Neck and Neurosciences, Udine University Hospital, Udine, Italy
| | - Susanne J van Veluw
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
2
|
Iwaide S, Murakami T, Sedghi Masoud N, Kobayashi N, Fortin JS, Miyahara H, Higuchi K, Chambers JK. Classification of amyloidosis and protein misfolding disorders in animals 2024: A review on pathology and diagnosis. Vet Pathol 2025; 62:117-138. [PMID: 39389927 DOI: 10.1177/03009858241283750] [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: 10/12/2024]
Abstract
Amyloidosis is a group of diseases in which proteins become amyloid, an insoluble fibrillar aggregate, resulting in organ dysfunction. Amyloid deposition has been reported in various animal species. To diagnose and understand the pathogenesis of amyloidosis, it is important to identify the amyloid precursor protein involved in each disease. Although 42 amyloid precursor proteins have been reported in humans, little is known about amyloidosis in animals, except for a few well-described amyloid proteins, including amyloid A (AA), amyloid light chain (AL), amyloid β (Aβ), and islet amyloid polypeptide-derived amyloid. Recently, several types of novel amyloidosis have been identified in animals using immunohistochemistry and mass spectrometry-based proteomic analysis. Certain species are predisposed to specific types of amyloidosis, suggesting a genetic background for its pathogenesis. Age-related amyloidosis has also emerged due to the increased longevity of captive animals. In addition, experimental studies have shown that some amyloids may be transmissible. Accurate diagnosis and understanding of animal amyloidosis are necessary for appropriate therapeutic intervention and comparative pathological studies. This review provides an updated classification of animal amyloidosis, including associated protein misfolding disorders of the central nervous system, and the current understanding of their pathogenesis. Pathologic features are presented together with state-of-the-art diagnostic methods that can be applied for routine diagnosis and identification of novel amyloid proteins in animals.
Collapse
Affiliation(s)
- Susumu Iwaide
- Tokyo University of Agriculture and Technology, Fuchu-shi, Japan
| | - Tomoaki Murakami
- Tokyo University of Agriculture and Technology, Fuchu-shi, Japan
| | | | | | | | | | - Keiichi Higuchi
- Shinshu University, Matsumoto, Japan
- Meio University, Nago, Japan
| | | |
Collapse
|
3
|
Greenberg SM, Aparicio HJ, Furie KL, Goyal MS, Hinman JD, Kozberg M, Leonard A, Fisher MJ. Vascular Neurology Considerations for Antiamyloid Immunotherapy: A Science Advisory From the American Heart Association. Stroke 2025; 56:e30-e38. [PMID: 39660440 DOI: 10.1161/str.0000000000000480] [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: 12/12/2024]
Abstract
Antibodies directed at the amyloid-β peptide offer the prospect of disease-modifying therapy for early-stage Alzheimer disease but also carry the risk of brain edema or bleeding events, collectively designated amyloid-related imaging abnormalities. Introduction of the antiamyloid immunotherapies into practice is therefore likely to present a new set of questions for clinicians treating patients with cerebrovascular disease: Which manifestations of cerebrovascular disease should preclude, or permit, antibody treatment? Is it safe to prescribe amyloid immunotherapies to individuals who require antithrombotic treatment, or to administer thrombolysis to antibody-treated individuals with acute stroke? How should severe amyloid-related imaging abnormalities be managed? This science advisory summarizes the data and key considerations to guide these challenging decisions as the medical community collects further data and experience with these groundbreaking agents.
Collapse
|
4
|
Rasing I, Vlegels N, Schipper MR, Voigt S, Koemans EA, Kaushik K, van Dort R, van Harten TW, De Luca A, van Etten ES, van Zwet EW, van Buchem MA, Middelkoop HA, Biessels GJ, Terwindt GM, van Osch MJ, van Walderveen MA, Wermer MJ. Microstructural white matter damage on MRI is associated with disease severity in Dutch-type cerebral amyloid angiopathy. J Cereb Blood Flow Metab 2024; 44:1253-1261. [PMID: 38886875 PMCID: PMC11542140 DOI: 10.1177/0271678x241261771] [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: 12/18/2023] [Revised: 03/26/2024] [Accepted: 04/28/2024] [Indexed: 06/20/2024]
Abstract
Peak width of skeletonized mean diffusivity (PSMD) is an emerging diffusion-MRI based marker to study subtle early alterations to white matter microstructure. We assessed PSMD over the clinical continuum in Dutch-type hereditary CAA (D-CAA) and its association with other CAA-related MRI-markers and cognitive symptoms. We included (pre)symptomatic D-CAA mutation-carriers and calculated PSMD from diffusion-MRI data. Associations between PSMD-levels, cognitive performance and CAA-related MRI-markers were assessed with linear regression models. We included 59 participants (25/34 presymptomatic/symptomatic; mean age 39/58 y). PSMD-levels increased with disease severity and were higher in symptomatic D-CAA mutation-carriers (median [range] 4.90 [2.77-9.50]mm2/s × 10-4) compared with presymptomatic mutation-carriers (2.62 [1.96-3.43]mm2/s × 10-4) p = <0.001. PSMD was positively correlated with age, CAA-SVD burden on MRI (adj.B [confidence interval] = 0.42 [0.16-0.67], p = 0.002), with number of cerebral microbleeds (adj.B = 0.30 [0.08-0.53], p = 0.009), and with both deep (adj.B = 0.46 [0.22-0.69], p = <0.001) and periventricular (adj.B = 0.38 [0.13-0.62], p = 0.004) white matter hyperintensities. Increasing PSMD was associated with decreasing Trail Making Test (TMT)-A performance (B = -0.42 [-0.69-0.14], p = 0.04. In D-CAA mutation-carriers microstructural white matter damage is associated with disease phase, CAA burden on MRI and cognitive impairment as reflected by a decrease in information processing speed. PSMD, as a global measure of alterations to the white matter microstructure, may be a useful tool to monitor disease progression in CAA.
Collapse
Affiliation(s)
- Ingeborg Rasing
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Naomi Vlegels
- Department of Neurology and Neurosurgery, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Manon R Schipper
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Sabine Voigt
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Emma A Koemans
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Kanishk Kaushik
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Rosemarie van Dort
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Thijs W van Harten
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Alberto De Luca
- Department of Neurology and Neurosurgery, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Image Sciences Institute, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Ellis S van Etten
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Erik W van Zwet
- Department of Biostatistics, Leiden University Medical Center, Leiden, The Netherland
| | - Mark A van Buchem
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Huub Am Middelkoop
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
- Institute of Psychology, Health, Medical and Neuropsychology Unit, Leiden University, Leiden, The Netherlands
| | - Geert Jan Biessels
- Department of Neurology and Neurosurgery, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Gisela M Terwindt
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Matthias Jp van Osch
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Marieke Jh Wermer
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Neurology, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
5
|
Costa AS, Albrecht M, Reich A, Nikoubashman O, Schulz JB, Reetz K, Pinho J. Non-hemorrhagic imaging markers of cerebral amyloid angiopathy in memory clinic patients. Alzheimers Dement 2024; 20:4792-4802. [PMID: 38865440 PMCID: PMC11247708 DOI: 10.1002/alz.13920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/21/2024] [Accepted: 03/22/2024] [Indexed: 06/14/2024]
Abstract
INTRODUCTION The Boston criteria v2.0 for cerebral amyloid angiopathy (CAA) incorporated non-hemorrhagic imaging markers. Their prevalence and significance in patients with cognitive impairment remain uncertain. METHODS We studied 622 memory clinic patients with available magnetic resonance imaging (MRI) and cerebrospinal fluid (CSF) biomarkers. Two raters assessed non-hemorrhagic markers, and we explored their association with clinical characteristics through multivariate analyses. RESULTS Most patients had mild cognitive impairment; median age was 71 years and 50% were female. Using the v2.0 criteria, possible or probable CAA increased from 75 to 383 patients. Sixty-eight percent of the sample had non-hemorrhagic CAA markers, which were independently associated with age (odds ratio [OR] = 1.04, 95% confidence interval [CI] = 1.01-1.07), female sex (OR = 1.68, 95% CI = 1.11-2.54), and hemorrhagic CAA markers (OR = 2.11, 95% CI = 1.02-4.35). DISCUSSION Two-thirds of patients from a memory clinic cohort had non-hemorrhagic CAA markers, increasing the number of patients meeting the v2.0 CAA criteria. Longitudinal approaches should explore the implications of these markers, particularly the hemorrhagic risk in this population. HIGHLIGHTS The updated Boston criteria for cerebral amyloid angiopathy (CAA) now include non-hemorrhagic markers. The prevalence of non-hemorrhagic CAA markers in memory clinic patients is unknown. Two-thirds of patients in our memory clinic presented non-hemorrhagic CAA markers. The presence of these markers was associated with age, female sex, and hemorrhagic CAA markers. The hemorrhagic risk of patients presenting these type of markers remains unclear.
Collapse
Affiliation(s)
- Ana Sofia Costa
- Department of NeurologyUniversity Hospital RWTH AachenAachenGermany
- JARA Institute Molecular Neuroscience and Neuroimaging (INM‐11)Juelich Research Center GmbH and RWTH Aachen UniversityAachenGermany
| | - Milena Albrecht
- Department of NeurologyUniversity Hospital RWTH AachenAachenGermany
| | - Arno Reich
- Department of NeurologyUniversity Hospital RWTH AachenAachenGermany
| | - Omid Nikoubashman
- Department of Diagnostic and Interventional NeuroradiologyUniversity Hospital RWTH AachenAachenGermany
| | - Jörg B. Schulz
- Department of NeurologyUniversity Hospital RWTH AachenAachenGermany
- JARA Institute Molecular Neuroscience and Neuroimaging (INM‐11)Juelich Research Center GmbH and RWTH Aachen UniversityAachenGermany
| | - Kathrin Reetz
- Department of NeurologyUniversity Hospital RWTH AachenAachenGermany
- JARA Institute Molecular Neuroscience and Neuroimaging (INM‐11)Juelich Research Center GmbH and RWTH Aachen UniversityAachenGermany
| | - João Pinho
- Department of NeurologyUniversity Hospital RWTH AachenAachenGermany
| |
Collapse
|
6
|
Rasing I, Voigt S, Koemans EA, de Kort AM, van Harten TW, van Etten ES, van Zwet EW, Stoops E, Francois C, Kuiperij HB, Klijn CJM, Schreuder FHBM, van der Weerd L, van Osch MJP, van Walderveen MAA, Verbeek MM, Terwindt GM, Wermer MJH. Serum and cerebrospinal fluid neurofilament light chain and glial fibrillary acid protein levels in early and advanced stages of cerebral amyloid Angiopathy. Alzheimers Res Ther 2024; 16:86. [PMID: 38654326 PMCID: PMC11036675 DOI: 10.1186/s13195-024-01457-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 04/12/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND Neurofilament light chain (NFL) is a biomarker for neuroaxonal damage and glial fibrillary acidic protein (GFAP) for reactive astrocytosis. Both processes occur in cerebral amyloid angiopathy (CAA), but studies investigating the potential of NFL and GFAP as markers for CAA are lacking. We aimed to investigate NFL and GFAP as biomarkers for neuroaxonal damage and astrocytosis in CAA. METHODS For this cross-sectional study serum and cerebrospinal fluid (CSF) samples were collected between 2010 and 2020 from controls, (pre)symptomatic Dutch-type hereditary (D-CAA) mutation-carriers and participants with sporadic CAA (sCAA) from two prospective CAA studies at two University hospitals in the Netherlands. NFL and GFAP levels were measured with Simoa-assays. The association between NFL and GFAP levels and age, cognitive performance (MoCA), CAA-related MRI markers (CAA-CSVD-burden) and Aβ40 and Aβ42 levels in CSF were assessed with linear regression adjusted for confounders. The control group was divided in age < 55 and ≥55 years to match the specific groups. RESULTS We included 187 participants: 28 presymptomatic D-CAA mutation-carriers (mean age 40 years), 29 symptomatic D-CAA participants (mean age 58 years), 59 sCAA participants (mean age 72 years), 33 controls < 55 years (mean age 42 years) and 38 controls ≥ 55 years (mean age 65 years). In presymptomatic D-CAA, only GFAP in CSF (7.7*103pg/mL vs. 4.4*103pg/mL in controls; P<.001) was increased compared to controls. In symptomatic D-CAA, both serum (NFL:26.2pg/mL vs. 12.5pg/mL; P=0.008, GFAP:130.8pg/mL vs. 123.4pg/mL; P=0.027) and CSF (NFL:16.8*102pg/mL vs. 7.8*102pg/mL; P=0.01 and GFAP:11.4*103pg/mL vs. 7.5*103pg/mL; P<.001) levels were higher than in controls and serum levels (NFL:26.2pg/mL vs. 6.7pg/mL; P=0.05 and GFAP:130.8pg/mL vs. 66.0pg/mL; P=0.004) were higher than in pre-symptomatic D-CAA. In sCAA, only NFL levels were increased compared to controls in both serum (25.6pg/mL vs. 12.5pg/mL; P=0.005) and CSF (20.0*102pg/mL vs 7.8*102pg/mL; P=0.008). All levels correlated with age. Serum NFL correlated with MoCA (P=0.008) and CAA-CSVD score (P<.001). NFL and GFAP in CSF correlated with Aβ42 levels (P=0.01/0.02). CONCLUSIONS GFAP level in CSF is an early biomarker for CAA and is increased years before symptom onset. NFL and GFAP levels in serum and CSF are biomarkers for advanced CAA.
Collapse
Affiliation(s)
- Ingeborg Rasing
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Sabine Voigt
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Emma A Koemans
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Anna M de Kort
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Thijs W van Harten
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ellis S van Etten
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Erik W van Zwet
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - H Bea Kuiperij
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Catharina J M Klijn
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Floris H B M Schreuder
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Louise van der Weerd
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Marcel M Verbeek
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gisela M Terwindt
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marieke J H Wermer
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Neurology, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
7
|
Liu YT, Lei CY, Zhong LM. Research Advancements on the Correlation Between Spontaneous Intracerebral Hemorrhage of Different Etiologies and Imaging Markers of Cerebral Small Vessel Disease. Neuropsychiatr Dis Treat 2024; 20:307-316. [PMID: 38405425 PMCID: PMC10893791 DOI: 10.2147/ndt.s442334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 01/24/2024] [Indexed: 02/27/2024] Open
Abstract
Objective The purpose of this review is to identify the correlation between ICH and CSVD imaging markers under SMASH-U classification by searching and analyzing a large number of literatures in recent years, laying a theoretical foundation for future clinical research. At the same time, by collecting clinical data to evaluate patient prognosis, analyzing whether there are differences or supplements between clinical trial conclusions and previous theories, and ultimately guiding clinical diagnosis and treatment through the analysis of imaging biomarkers. Methods In this review, by searching CNKI, Web of Science, PubMed, FMRS and other databases, the use of "spontaneous intracerebral hemorrhage", "hypertensive hemorrhagic cerebral small vessel disease", "cerebral small vessel disease imaging", "Based cerebral small vessel diseases", "SMASH the -u classification" and their Chinese equivalents for the main search term. We focused on reading and analyzing hundreds of relevant literatures in the last decade from August 2011 to April 2020, and also included some earlier literatures with conceptual data sources. After screening and ranking the degree of relevance to this study, sixty of them were cited for analysis and elaboration. Results In patients with ICH, the number of cerebral microbleeds in lobes, basal ganglia, and the deep brain is positively correlated with ICH volume and independently correlated with neurological functional outcomes; white matter hyperintensity severity is positively correlated with ICH recurrence risk; multiple lacunar infarction independently predict the risk of ICH; severe brain atrophy is an independent risk factor for a poor prognosis in the long term in patients diagnosed with ICH; and the number of enlarged perivascular spaces is correlated with ICH recurrence. However, small subcortical infarct and ICH are the subject of few studies. Higher CSVD scores are independently associated with functional outcomes at 90 days in patients diagnosed with ICH.
Collapse
Affiliation(s)
- Yu-Tong Liu
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, People’s Republic of China
| | - Chun-Yan Lei
- Department of Neurology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, People’s Republic of China
| | - Lian-Mei Zhong
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
8
|
Bangad A, Abbasi M, Payabvash S, de Havenon A. Imaging of Amyloid-beta-related Arteritis. Neuroimaging Clin N Am 2024; 34:167-173. [PMID: 37951701 DOI: 10.1016/j.nic.2023.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Cerebral amyloid angiopathy (CAA) is a cerebrovascular disorder marked by the accumulation of amyloid-beta peptide (Aβ) within the leptomeninges and smaller blood vessels of the brain. CAA can be both noninflammatory and inflammatory, and the inflammatory version includes Aβ-related angiitis (ABRA). ABRA is a vasculitis of the central nervous system related to an inflammatory response to Aβ in the vascular walls, which necessitates differentiating ABRA from noninflammatory CAA, as ABRA may require immunosuppressive treatment. MR imaging is typically the most effective imaging modality of choice to screen for these conditions, and they should be obtained at varying time points to track disease progression.
Collapse
Affiliation(s)
- Aaron Bangad
- Department of Neurology, Yale University, New Haven, CT, USA
| | - Mehdi Abbasi
- Department of Neurology, Yale University, New Haven, CT, USA
| | - Sam Payabvash
- Center for Brain and Mind Health, Yale University, New Haven, CT, USA
| | - Adam de Havenon
- Department of Neurology, Yale University, New Haven, CT, USA; Center for Brain and Mind Health, Yale University, New Haven, CT, USA.
| |
Collapse
|
9
|
Wang S, Wang J, Guo J, Dove A, Xu H, Qi X, Xu W. Association of Kidney Function With Dementia and Structural Brain Differences: A Large Population-Based Cohort Study. J Gerontol A Biol Sci Med Sci 2024; 79:glad192. [PMID: 37578935 PMCID: PMC10733178 DOI: 10.1093/gerona/glad192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Indexed: 08/16/2023] Open
Abstract
BACKGROUND The association between kidney function and dementia risk and the mechanisms underlying this relationship remain unclear. METHODS Within the UK Biobank, 191 970 dementia-free participants aged ≥60 (mean age: 64.1 ± 2.9 years) were followed for 16 years to detect incident dementia. Serum creatinine and Cystatin C were measured at baseline to calculate estimated glomerular filtration rate (eGFR, mL/min/1.73 m2). Kidney function was categorized as normal (eGFR ≥ 90), mildly impaired (60 ≤ eGFR < 90), or moderately to severely impaired (eGFR < 60). Dementia was assessed based on self-reported medical history and medical records. During the follow-up, a subsample of 12 637 participants underwent brain MRI scans. Volumes of total brain, gray matter, white matter, hippocampus, and white matter hyperintensities were assessed. RESULTS Over the follow-up, 5 327 (2.8%) participants developed dementia. Compared to normal kidney function, there was an increased risk of dementia with moderate to severely impaired kidney function (hazard ratio = 1.53, 95% confidence interval [CI]: 1.32-1.76) but not mildly impaired kidney function. In Laplace regression, dementia onset among people with moderate to severely impaired kidney function occurred 1.53 (95% CI: 0.98-2.08) years earlier than those with normal kidney function. Moderate to severely impaired kidney function was related to significantly lower gray matter volume (β = -0.11, 95% CI: -0.19 to -0.03), but not to other brain magnetic resonance imaging measures. CONCLUSIONS Impaired kidney function is associated with about 50% increased risk of dementia and anticipates dementia onset by more than 1.5 years. Brain neurodegeneration may underlie the kidney function-dementia association.
Collapse
Affiliation(s)
- Shuqi Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Jiao Wang
- Department of Epidemiology, College of Preventive Medicine, the Army Medical University (Third Military Medical University), Chongqing, China
| | - Jie Guo
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Abigail Dove
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Hong Xu
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Xiuying Qi
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Weili Xu
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
10
|
Theodorou A, Palaiodimou L, Papagiannopoulou G, Kargiotis O, Psychogios K, Safouris A, Bakola E, Chondrogianni M, Kotsali-Peteinelli V, Melanis K, Tsibonakis A, Andreadou E, Vasilopoulou S, Lachanis S, Velonakis G, Tzavellas E, Tzartos JS, Voumvourakis K, Paraskevas GP, Tsivgoulis G. Clinical Characteristics, Neuroimaging Markers, and Outcomes in Patients with Cerebral Amyloid Angiopathy: A Prospective Cohort Study. J Clin Med 2023; 12:5591. [PMID: 37685658 PMCID: PMC10488273 DOI: 10.3390/jcm12175591] [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: 07/04/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Background and purpose: Sporadic cerebral amyloid angiopathy (CAA) is a small vessel disease, resulting from progressive amyloid-β deposition in the media/adventitia of cortical and leptomeningeal arterioles. We sought to assess the prevalence of baseline characteristics, clinical and radiological findings, as well as outcomes among patients with CAA, in the largest study to date conducted in Greece. Methods: Sixty-eight patients fulfilling the Boston Criteria v1.5 for probable/possible CAA were enrolled and followed for at least twelve months. Magnetic Resonance Imaging was used to assess specific neuroimaging markers. Data regarding cerebrospinal fluid biomarker profile and Apolipoprotein-E genotype were collected. Multiple logistic regression analyses were performed to identify predictors of clinical phenotypes. Cox-proportional hazard regression models were used to calculate associations with the risk of recurrent intracerebral hemorrhage (ICH). Results: Focal neurological deficits (75%), cognitive decline (57%), and transient focal neurological episodes (TFNEs; 21%) were the most common clinical manifestations. Hemorrhagic lesions, including lobar cerebral microbleeds (CMBs; 93%), cortical superficial siderosis (cSS; 48%), and lobar ICH (43%) were the most prevalent neuroimaging findings. cSS was independently associated with the likelihood of TFNEs at presentation (OR: 4.504, 95%CI:1.258-19.088), while multiple (>10) lobar CMBs were independently associated with cognitive decline at presentation (OR:5.418, 95%CI:1.316-28.497). cSS emerged as the only risk factor of recurrent ICH (HR:4.238, 95%CI:1.509-11.900) during a median follow-up of 20 months. Conclusions: cSS was independently associated with TFNEs at presentation and ICH recurrence at follow-up, while a higher burden of lobar CMBs with cognitive decline at baseline. These findings highlight the prognostic value of neuroimaging markers, which may influence clinical decision-making.
Collapse
Affiliation(s)
- Aikaterini Theodorou
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.T.); (L.P.); (G.P.); (E.B.); (M.C.); (V.K.-P.); (K.M.); (A.T.); (J.S.T.); (K.V.); (G.P.P.)
| | - Lina Palaiodimou
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.T.); (L.P.); (G.P.); (E.B.); (M.C.); (V.K.-P.); (K.M.); (A.T.); (J.S.T.); (K.V.); (G.P.P.)
| | - Georgia Papagiannopoulou
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.T.); (L.P.); (G.P.); (E.B.); (M.C.); (V.K.-P.); (K.M.); (A.T.); (J.S.T.); (K.V.); (G.P.P.)
| | - Odysseas Kargiotis
- Stroke Unit, Metropolitan Hospital, 18547 Piraeus, Greece; (O.K.); (K.P.); (A.S.)
| | - Klearchos Psychogios
- Stroke Unit, Metropolitan Hospital, 18547 Piraeus, Greece; (O.K.); (K.P.); (A.S.)
| | - Apostolos Safouris
- Stroke Unit, Metropolitan Hospital, 18547 Piraeus, Greece; (O.K.); (K.P.); (A.S.)
| | - Eleni Bakola
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.T.); (L.P.); (G.P.); (E.B.); (M.C.); (V.K.-P.); (K.M.); (A.T.); (J.S.T.); (K.V.); (G.P.P.)
| | - Maria Chondrogianni
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.T.); (L.P.); (G.P.); (E.B.); (M.C.); (V.K.-P.); (K.M.); (A.T.); (J.S.T.); (K.V.); (G.P.P.)
| | - Vasiliki Kotsali-Peteinelli
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.T.); (L.P.); (G.P.); (E.B.); (M.C.); (V.K.-P.); (K.M.); (A.T.); (J.S.T.); (K.V.); (G.P.P.)
| | - Konstantinos Melanis
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.T.); (L.P.); (G.P.); (E.B.); (M.C.); (V.K.-P.); (K.M.); (A.T.); (J.S.T.); (K.V.); (G.P.P.)
| | - Athanasios Tsibonakis
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.T.); (L.P.); (G.P.); (E.B.); (M.C.); (V.K.-P.); (K.M.); (A.T.); (J.S.T.); (K.V.); (G.P.P.)
| | - Elissavet Andreadou
- First Department of Neurology, “Eginition” Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (E.A.); (S.V.)
| | - Sofia Vasilopoulou
- First Department of Neurology, “Eginition” Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (E.A.); (S.V.)
| | - Stefanos Lachanis
- Iatropolis Magnetic Resonance Diagnostic Centre, 15231 Athens, Greece;
| | - Georgios Velonakis
- Second Department of Radiology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Elias Tzavellas
- First Department of Psychiatry, “Aiginition” Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | - John S. Tzartos
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.T.); (L.P.); (G.P.); (E.B.); (M.C.); (V.K.-P.); (K.M.); (A.T.); (J.S.T.); (K.V.); (G.P.P.)
| | - Konstantinos Voumvourakis
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.T.); (L.P.); (G.P.); (E.B.); (M.C.); (V.K.-P.); (K.M.); (A.T.); (J.S.T.); (K.V.); (G.P.P.)
| | - Georgios P. Paraskevas
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.T.); (L.P.); (G.P.); (E.B.); (M.C.); (V.K.-P.); (K.M.); (A.T.); (J.S.T.); (K.V.); (G.P.P.)
| | - Georgios Tsivgoulis
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.T.); (L.P.); (G.P.); (E.B.); (M.C.); (V.K.-P.); (K.M.); (A.T.); (J.S.T.); (K.V.); (G.P.P.)
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
11
|
Pansieri J, Hadley G, Lockhart A, Pisa M, DeLuca GC. Regional contribution of vascular dysfunction in white matter dementia: clinical and neuropathological insights. Front Neurol 2023; 14:1199491. [PMID: 37396778 PMCID: PMC10313211 DOI: 10.3389/fneur.2023.1199491] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 05/25/2023] [Indexed: 07/04/2023] Open
Abstract
The maintenance of adequate blood supply and vascular integrity is fundamental to ensure cerebral function. A wide range of studies report vascular dysfunction in white matter dementias, a group of cerebral disorders characterized by substantial white matter damage in the brain leading to cognitive impairment. Despite recent advances in imaging, the contribution of vascular-specific regional alterations in white matter dementia has been not extensively reviewed. First, we present an overview of the main components of the vascular system involved in the maintenance of brain function, modulation of cerebral blood flow and integrity of the blood-brain barrier in the healthy brain and during aging. Second, we review the regional contribution of cerebral blood flow and blood-brain barrier disturbances in the pathogenesis of three distinct conditions: the archetypal white matter predominant neurocognitive dementia that is vascular dementia, a neuroinflammatory predominant disease (multiple sclerosis) and a neurodegenerative predominant disease (Alzheimer's). Finally, we then examine the shared landscape of vascular dysfunction in white matter dementia. By emphasizing the involvement of vascular dysfunction in the white matter, we put forward a hypothetical map of vascular dysfunction during disease-specific progression to guide future research aimed to improve diagnostics and facilitate the development of tailored therapies.
Collapse
|
12
|
Maitre M, Jeltsch-David H, Okechukwu NG, Klein C, Patte-Mensah C, Mensah-Nyagan AG. Myelin in Alzheimer's disease: culprit or bystander? Acta Neuropathol Commun 2023; 11:56. [PMID: 37004127 PMCID: PMC10067200 DOI: 10.1186/s40478-023-01554-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/20/2023] [Indexed: 04/03/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder with neuronal and synaptic losses due to the accumulation of toxic amyloid β (Αβ) peptide oligomers, plaques, and tangles containing tau (tubulin-associated unit) protein. While familial AD is caused by specific mutations, the sporadic disease is more common and appears to result from a complex chronic brain neuroinflammation with mitochondriopathies, inducing free radicals' accumulation. In aged brain, mutations in DNA and several unfolded proteins participate in a chronic amyloidosis response with a toxic effect on myelin sheath and axons, leading to cognitive deficits and dementia. Αβ peptides are the most frequent form of toxic amyloid oligomers. Accumulations of misfolded proteins during several years alters different metabolic mechanisms, induce chronic inflammatory and immune responses with toxic consequences on neuronal cells. Myelin composition and architecture may appear to be an early target for the toxic activity of Aβ peptides and others hydrophobic misfolded proteins. In this work, we describe the possible role of early myelin alterations in the genesis of neuronal alterations and the onset of symptomatology. We propose that some pathophysiological and clinical forms of the disease may arise from structural and metabolic disorders in the processes of myelination/demyelination of brain regions where the accumulation of non-functional toxic proteins is important. In these forms, the primacy of the deleterious role of amyloid peptides would be a matter of questioning and the initiating role of neuropathology would be primarily the fact of dysmyelination.
Collapse
Affiliation(s)
- Michel Maitre
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM U1119, Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, Strasbourg, 67000, France.
| | - Hélène Jeltsch-David
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM U1119, Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, Strasbourg, 67000, France
- Biotechnologie et signalisation cellulaire, UMR 7242 CNRS, Université de Strasbourg, 300 Boulevard Sébastien Brant CS 10413, Illkirch cedex, 67412, France
| | - Nwife Getrude Okechukwu
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM U1119, Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, Strasbourg, 67000, France
| | - Christian Klein
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM U1119, Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, Strasbourg, 67000, France
| | - Christine Patte-Mensah
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM U1119, Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, Strasbourg, 67000, France
| | - Ayikoe-Guy Mensah-Nyagan
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM U1119, Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, Strasbourg, 67000, France
| |
Collapse
|
13
|
Lu J, Huang R, Peng Y, Zhang J, Liang K, Wang Y, Feng Y, Wang Z. Mendelian Randomization Analyses Accounting for Causal Effect of COVID-19 on Brain Imaging-Derived Phenotypes. J Alzheimers Dis 2023; 96:1059-1070. [PMID: 37955088 DOI: 10.3233/jad-230626] [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: 11/14/2023]
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) has been a major challenge to global health and a financial burden. Little is known regarding the possible causal effects of COVID-19 on the macro- and micro-structures of the human brain. OBJECTIVE To determine the causal links between susceptibility, hospitalization, and the severity of COVID-19 and brain imaging-derived phenotypes (IDPs). METHODS Mendelian randomization (MR) analyses were performed to investigate the causal effect of three COVID-19 exposures (SARS-CoV-2 infection, hospitalized COVID-19, and critical COVID-19) on brain structure employing summary datasets of genome-wide association studies. RESULTS In terms of cortical phenotypes, hospitalization due to COVID-19 was associated with a global decrease in the surface area (SA) of the cortex structure (β= -624.77, 95% CI: -1227.88 to -21.66, p = 0.042). At the regional level, SARS-CoV-2 infection was found to have a nominally causal effect on the thickness (TH) of the postcentral region (β= -0.004, 95% CI: -0.007 to -0.001, p = 0.01), as well as eight other IDPs. Hospitalized COVID-19 has a nominally causal relationship with TH of postcentral (β= -0.004, 95% CI: -0.007 to -0.001, p = 0.01) and other 6 IDPs. The nominally causal effects of critical COVID-19 on TH of medial orbitofrontal (β=0.004, 95% CI: 0.001to 0.007, p = 0.004) and other 7 IDPs were revealed. CONCLUSIONS Our study provides compelling genetic evidence supporting causal relationships between three COVID-19 traits and brain IDPs. This discovery holds promise for enhancing predictions and interventions in brain imaging.
Collapse
Affiliation(s)
- Jiajie Lu
- Institute of Neuroscience, Department of Neurosurgery, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Clinical Medicine, The Second Clinical School of Guangzhou Medical University, Guangzhou, China
| | - Rihong Huang
- Institute of Neuroscience, Department of Neurosurgery, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Clinical Medicine, The Second Clinical School of Guangzhou Medical University, Guangzhou, China
| | - Yuecheng Peng
- Institute of Neuroscience, Department of Neurosurgery, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Clinical Medicine, The Second Clinical School of Guangzhou Medical University, Guangzhou, China
| | - Jinming Zhang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kairong Liang
- Institute of Neuroscience, Department of Neurosurgery, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yezhong Wang
- Institute of Neuroscience, Department of Neurosurgery, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yi Feng
- Department of Thoracic Surgery and Oncology, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Institute of Respiratory Health, Guangzhou, China
| | - Zhaotao Wang
- Institute of Neuroscience, Department of Neurosurgery, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Department of Clinical Medicine, The Second Clinical School of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
14
|
Chen CH, Khnaijer MK, Beaudin AE, McCreary CR, Gee M, Saad F, Frayne R, Ismail Z, Pike GB, Camicioli R, Smith EE. Subcortical volumes in cerebral amyloid angiopathy compared with Alzheimer's disease and controls. Front Neurosci 2023; 17:1139196. [PMID: 37139517 PMCID: PMC10149850 DOI: 10.3389/fnins.2023.1139196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 03/28/2023] [Indexed: 05/05/2023] Open
Abstract
Background Previous reports have suggested that patients with cerebral amyloid angiopathy (CAA) may harbor smaller white matter, basal ganglia, and cerebellar volumes compared to age-matched healthy controls (HC) or patients with Alzheimer's disease (AD). We investigated whether CAA is associated with subcortical atrophy. Methods The study was based on the multi-site Functional Assessment of Vascular Reactivity cohort and included 78 probable CAA (diagnosed according to the Boston criteria v2.0), 33 AD, and 70 HC. Cerebral and cerebellar volumes were extracted from brain 3D T1-weighted MRI using FreeSurfer (v6.0). Subcortical volumes, including total white matter, thalamus, basal ganglia, and cerebellum were reported as proportion (%) of estimated total intracranial volume. White matter integrity was quantified by the peak width of skeletonized mean diffusivity. Results Participants in the CAA group were older (74.0 ± 7.0, female 44%) than the AD (69.7 ± 7.5, female 42%) and HC (68.8 ± 7.8, female 69%) groups. CAA participants had the highest white matter hyperintensity volume and worse white matter integrity of the three groups. After adjusting for age, sex, and study site, CAA participants had smaller putamen volumes (mean differences, -0.024% of intracranial volume; 95% confidence intervals, -0.041% to -0.006%; p = 0.005) than the HCs but not AD participants (-0.003%; -0.024 to 0.018%; p = 0.94). Other subcortical volumes including subcortical white matter, thalamus, caudate, globus pallidus, cerebellar cortex or cerebellar white matter were comparable between all three groups. Conclusion In contrast to prior studies, we did not find substantial atrophy of subcortical volumes in CAA compared to AD or HCs, except for the putamen. Differences between studies may reflect heterogeneity in CAA presenting syndromes or severity.
Collapse
Affiliation(s)
- Chih-Hao Chen
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Mary Klir Khnaijer
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - Andrew E. Beaudin
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Cheryl R. McCreary
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Department of Radiology, University of Calgary, Calgary, AB, Canada
| | - Myrlene Gee
- Division of Neurology, Department of Medicine and Neurosciences and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Feryal Saad
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - Richard Frayne
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Department of Radiology, University of Calgary, Calgary, AB, Canada
| | - Zahinoor Ismail
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Department of Psychiatry, University of Calgary, Calgary, AB, Canada
| | - G. Bruce Pike
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Department of Radiology, University of Calgary, Calgary, AB, Canada
| | - Richard Camicioli
- Division of Neurology, Department of Medicine and Neurosciences and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Eric E. Smith
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Department of Radiology, University of Calgary, Calgary, AB, Canada
- *Correspondence: Eric E. Smith,
| |
Collapse
|
15
|
Han X, Zhang J, Chen S, Yu W, Zhou Y, Gu X. Mapping the current trends and hotspots of vascular cognitive impairment from 2000-2021: A bibliometric analysis. CNS Neurosci Ther 2022; 29:771-782. [PMID: 36415118 PMCID: PMC9928552 DOI: 10.1111/cns.14026] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/03/2022] [Accepted: 10/30/2022] [Indexed: 11/24/2022] Open
Abstract
AIMS To visualize the trends and hotspots in the scientific research related to vascular cognitive impairment (VCI) quantitatively and qualitatively. METHODS Cross-sectional bibliometric analysis of publications that related to VCI was conducted. Publications were found by searching in the Web of Science Core Collection database (WoSCC) - Edition: Science Citation Index Expanded (SCI-Expanded) from January 2000 to December 2021. Publication type was restricted to article and review in the English language. The downloaded data were screened and analyzed in January 2022. RESULTS In total, 16,264 publications were identified, with a steady increase in annual publications. The United States was the leading country in VCI research regarding publication numbers and national influence. National Institute of Aging had the highest influence among all the institutes in the field of VCI. Philip Scheltens was the most active author. The top five active authors' publications focused on pathobiology, neuroimaging standards, risk factors, prevention, and the standard diagnosis of vascular dementia (VaD). A co-cited publication clustering resulted in 19 main clusters, and the prevention, blood-brain barrier, cholesterol, cerebral amyloid angiopathy, and VaD were the top 5 clusters. Moreover, burst keywords detection revealed that the "small vessel disease" is the current hotspot in the field of VCI. CONCLUSIONS This bibliometric analysis mapped the overall research structure of VCI and analyzed the current research trends and hotspots for future studies orientation. Neuroimaging, risk factors detection, and pathobiology are the current trends in VCI research. Small vessel disease and its mechanisms are the current hotspots of VCI research.
Collapse
Affiliation(s)
- Xu Han
- Department of Radiology, Department of Anesthesiology, Renji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jian Zhang
- Department of Radiology, Department of Anesthesiology, Renji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Sifan Chen
- Department of Radiology, Department of Anesthesiology, Renji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Weifeng Yu
- Department of Radiology, Department of Anesthesiology, Renji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yan Zhou
- Department of Radiology, Department of Anesthesiology, Renji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiyao Gu
- Department of Radiology, Department of Anesthesiology, Renji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
16
|
Che R, Zhang M, Sun H, Ma J, Hu W, Liu X, Ji X. Long-term outcome of cerebral amyloid angiopathy-related hemorrhage. CNS Neurosci Ther 2022; 28:1829-1837. [PMID: 35975394 PMCID: PMC9532921 DOI: 10.1111/cns.13922] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 11/30/2022] Open
Abstract
OBJECT The long-term functional outcome of cerebral amyloid angiopathy-related hemorrhage (CAAH) patients is unclear. We sought to assess the long-term functional outcome of CAAH and determine the prognostic factors associated with unfavorable outcomes. METHODS We enrolled consecutive CAAH patients from 2014 to 2020 in this observational study. Baseline characteristics and clinical outcomes were presented. Multivariable logistic regression analysis was performed to identify the prognostic factors associated with long-term outcome. RESULTS Among the 141 CAAH patients, 76 (53.9%) achieved favorable outcomes and 28 (19.9%) of them died at 1-year follow-up. For the longer-term follow-up with a median observation time of 19.0 (interquartile range, 12.0-26.5) months, 71 (50.4%) patients obtained favorable outcomes while 33 (23.4%) died. GCS on admission (OR, 0.109; 95% CI, 0.021-0.556; p = 0.008), recurrence of ICH (OR, 2923.687; 95% CI, 6.282-1360730.14; p = 0.011), WML grade 3-4 (OR, 31.007; 95% CI, 1.041-923.573; p = 0.047), severe central atrophy (OR, 4220.303; 95% CI, 9.135-1949674.84; p = 0.008) assessed by CT was identified as independent predictors for long-term outcome. INTERPRETATION Nearly 50% of CAAH patients achieved favorable outcomes at long-term follow-up. GCS, recurrence of ICH, WML grade and cerebral atrophy were identified as independent prognostic factors of long-term outcome.
Collapse
Affiliation(s)
- Ruiwen Che
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, China
| | - Mengke Zhang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Hailiang Sun
- Department of Neurosurgery, Beijing Fengtai You'anmen Hospital, Beijing, China
| | - Jin Ma
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wenbo Hu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xin Liu
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Xunming Ji
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, China
- Department of Neurosurgery, Xuan Wu Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Brain Disorders, Beijing, China
- Capital Medical University, Beijing, China
| |
Collapse
|
17
|
Pamiparib Induces Neurodevelopmental Defects and Cerebral Haemorrhage in Zebrafish Embryos via Inhibiting Notch Signalling. Mol Neurobiol 2022; 59:6652-6665. [PMID: 35982279 DOI: 10.1007/s12035-022-02988-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 08/06/2022] [Indexed: 10/15/2022]
Abstract
Pamiparib is a poly ADP-ribose polymerase (PARP) inhibitor used in clinical studies, which can penetrate the blood-brain barrier efficiently. At present, there are few studies on its effect on vertebrate neurodevelopment. In this study, we exposed zebrafish embryos to 1, 2 and 3 µM of Pamiparib from 6 to 72 h post-fertilisation (hpf). Results showed that pamiparib can specifically induce cerebral haemorrhage, brain atrophy and movement disorders in fish larvae. In addition, pamiparib exposure leads to downregulation of acetylcholinesterase (AChE) and adenosine triphosphate (ATPase) activities, and upregulation of oxidative stress which then leads to apoptosis and disrupts the gene expression involved in the neurodevelopment, neurotransmitter pathways and Parkinson's disease (PD) like symptoms. Meanwhile, astaxanthin can partially rescue neurodevelopmental defects by downregulating oxidative stress. After exposure to pamiparib, the Notch signalling is downregulated, and the use of an activator of Notch signalling can partially rescue neurodevelopmental toxicity. Therefore, our research indicates that pamiparib may induce zebrafish neurotoxicity by downregulating Notch signalling and provides a reference for the potential neurotoxicity of pamiparib during embryonic development.
Collapse
|
18
|
Gokcal E, Horn MJ, Becker JA, Das AS, Schwab K, Biffi A, Rost N, Rosand J, Viswanathan A, Polimeni JR, Johnson KA, Greenberg SM, Gurol ME. Effect of vascular amyloid on white matter disease is mediated by vascular dysfunction in cerebral amyloid angiopathy. J Cereb Blood Flow Metab 2022; 42:1272-1281. [PMID: 35086372 PMCID: PMC9207495 DOI: 10.1177/0271678x221076571] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
We postulated that vascular dysfunction mediates the relationship between amyloid load and white matter hyperintensities (WMH) in cerebral amyloid angiopathy (CAA). Thirty-eight cognitively healthy patients with CAA (mean age 70 ± 7.1) were evaluated. WMH was quantified and expressed as percent of total intracranial volume (pWMH) using structural MRI. Mean global cortical Distribution Volume Ratio representing Pittsburgh Compound B (PiB) uptake (PiB-DVR) was calculated from PET scans. Time-to-peak [TTP] of blood oxygen level-dependent response to visual stimulation was used as an fMRI measure of vascular dysfunction. Higher PiB-DVR correlated with prolonged TTP (r = 0.373, p = 0.021) and higher pWMH (r = 0.337, p = 0.039). Prolonged TTP also correlated with higher pWMH (r = 0.485, p = 0.002). In a multivariate linear regression model, TTP remained independently associated with pWMH (p = 0.006) while PiB-DVR did not (p = 0.225). In a bootstrapping model, TTP had a significant indirect effect (ab = 0.97, 95% CI: 0.137-2.461), supporting that the association between PiB-DVR and pWMH is mediated by TTP response. There was no longer a direct effect independent of the hypothesized pathway. Our study suggests that the effect of vascular amyloid load on white matter disease is mediated by vascular dysfunction in CAA. Amyloid lowering strategies might prevent pathophysiological processes leading to vascular dysfunction, therefore limiting ischemic brain injury.
Collapse
Affiliation(s)
- Elif Gokcal
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mitchell J Horn
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - J Alex Becker
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Alvin S Das
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kristin Schwab
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Alessandro Biffi
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Natalia Rost
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jonathan Rosand
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Anand Viswanathan
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Keith A Johnson
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Steven M Greenberg
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - M Edip Gurol
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
19
|
Shaikh I, Beaulieu C, Gee M, McCreary CR, Beaudin AE, Valdés-Cabrera D, Smith EE, Camicioli R. Diffusion tensor tractography of the fornix in cerebral amyloid angiopathy, mild cognitive impairment and Alzheimer's disease. Neuroimage Clin 2022; 34:103002. [PMID: 35413649 PMCID: PMC9010796 DOI: 10.1016/j.nicl.2022.103002] [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] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 03/26/2022] [Accepted: 04/02/2022] [Indexed: 11/16/2022]
Abstract
The fornix was delineated with deterministic tractography from diffusion tensor images (DTI). Fornix diffusion changes were found in the fornix in CAA, AD and MCI compared to controls. Higher fornix diffusivity correlated with smaller hippocampal volume and larger ventricles. Fornix diffusion measures correlated with cognitive measures in the combined disease groups.
Purpose Cerebral amyloid angiopathy (CAA) is a common neuropathological finding and clinical entity that occurs independently and with co-existent Alzheimer’s disease (AD) and small vessel disease. We compared diffusion tensor imaging (DTI) metrics of the fornix, the primary efferent tract of the hippocampus between CAA, AD and Mild Cognitive Impairment (MCI) and healthy controls. Methods Sixty-eight healthy controls, 32 CAA, 21 AD, and 26 MCI patients were recruited at two centers. Diffusion tensor images were acquired at 3 T with high spatial resolution and fluid-attenuated inversion recovery (FLAIR) to suppress cerebrospinal fluid (CSF) and minimize partial volume effects on the fornix. The fornix was delineated with deterministic tractography to yield mean diffusivity (MD), axial diffusivity (AXD), radial diffusivity (RD), fractional anisotropy (FA) and tract volume. Volumetric measurements of the hippocampus, thalamus, and lateral ventricles were obtained using T1-weighted MRI. Results Diffusivity (MD, AXD, and RD) of the fornix was highest in AD followed by CAA compared to controls; the MCI group was not significantly different from controls. FA was similar between groups. Fornix tract volume was ∼ 30% lower for all three patient groups compared to controls, but not significantly different between the patient groups. Thalamic and hippocampal volumes were preserved in CAA, but lower in AD and MCI compared to controls. Lateral ventricular volumes were increased in CAA, AD and MCI. Global cognition, memory, and executive function all correlated negatively with fornix diffusivity across the combined clinical group. Conclusion There were significant diffusion changes of the fornix in CAA, AD and MCI compared to controls, despite relatively intact thalamic and hippocampal volumes in CAA, suggesting the mechanisms for fornix diffusion abnormalities may differ in CAA compared to AD and MCI.
Collapse
Affiliation(s)
- Ibrahim Shaikh
- Department of Medicine, Division of Neurology and Neuroscience and Mental Health Institute (NMHI), University of Alberta, Edmonton, AB, Canada; Department of Biomedical Engineering, University of Alberta, Edmonton, AB, Canada
| | - Christian Beaulieu
- Department of Biomedical Engineering, University of Alberta, Edmonton, AB, Canada
| | - Myrlene Gee
- Department of Medicine, Division of Neurology and Neuroscience and Mental Health Institute (NMHI), University of Alberta, Edmonton, AB, Canada
| | - Cheryl R McCreary
- Department of Radiology, University of Calgary, Calgary, AB, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada; Seaman Family MR Research Centre, Foothills Medical Centre, Alberta Health Services, Calgary, AB, Canada
| | - Andrew E Beaudin
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada; Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - Diana Valdés-Cabrera
- Department of Biomedical Engineering, University of Alberta, Edmonton, AB, Canada
| | - Eric E Smith
- Department of Radiology, University of Calgary, Calgary, AB, Canada; Seaman Family MR Research Centre, Foothills Medical Centre, Alberta Health Services, Calgary, AB, Canada
| | - Richard Camicioli
- Department of Medicine, Division of Neurology and Neuroscience and Mental Health Institute (NMHI), University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
20
|
Horn MJ, Gokcal E, Becker AJ, Das AS, Warren AD, Schwab K, Goldstein JN, Biffi A, Rosand J, Polimeni JR, Viswanathan A, Greenberg SM, Gurol ME. Cerebellar atrophy and its implications on gait in cerebral amyloid angiopathy. J Neurol Neurosurg Psychiatry 2022; 93:jnnp-2021-328553. [PMID: 35534189 PMCID: PMC10936558 DOI: 10.1136/jnnp-2021-328553] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 04/06/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVE Recent data suggest that cerebral amyloid angiopathy (CAA) causes haemorrhagic lesions in cerebellar cortex as well as subcortical cerebral atrophy. However, the potential effect of CAA on cerebellar tissue loss and its clinical implications have not been investigated. METHODS Our study included 70 non-demented patients with probable CAA, 70 age-matched healthy controls (HCs) and 70 age-matched patients with Alzheimer's disease (AD). The cerebellum was segmented into percent of cerebellar subcortical volume (pCbll-ScV) and percent of cerebellar cortical volume (pCbll-CV) represented as percent (p) of estimated total intracranial volume. We compared pCbll-ScV and pCbll-CV between patients with CAA, HCs and those with AD. Gait velocity (metres/second) was used to investigate gait function in patients with CAA. RESULTS Patients with CAA had significantly lower pCbll-ScV compared with both HC (1.49±0.1 vs 1.73±0.2, p<0.001) and AD (1.49±0.1 vs 1.66±0.24, p<0.001) and lower pCbll-CV compared with HCs (6.03±0.5 vs 6.23±0.6, p=0.028). Diagnosis of CAA was independently associated with lower pCbll-ScV compared with HCs (p<0.001) and patients with AD (p<0.001) in separate linear regression models adjusted for age, sex and presence of hypertension. Lower pCbll-ScV was independently associated with worse gait velocity (β=0.736, 95% CI 0.28 to 1.19, p=0.002) in a stepwise linear regression analysis including pCbll-CV along with other relevant variables. INTERPRETATION Patients with CAA show more subcortical cerebellar atrophy than HC or patients with AD and more cortical cerebellar atrophy than HCs. Reduced pCbll-ScV correlated with lower gait velocity in regression models including other relevant variables. Overall, this study suggests that CAA causes cerebellar injury, which might contribute to gait disturbance.
Collapse
Affiliation(s)
- Mitchell J Horn
- J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Elif Gokcal
- J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Alex J Becker
- Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Alvin S Das
- J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Andrew D Warren
- J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Kristin Schwab
- J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Joshua N Goldstein
- Emergency Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Alessandro Biffi
- J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Jonathan Rosand
- J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Jonathan R Polimeni
- Athinoula A Martinos Center for Biomedical Imaging, Charlestown, Massachusetts, USA
| | - Anand Viswanathan
- J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Steven M Greenberg
- J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - M Edip Gurol
- J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
21
|
Freeze WM, Zanon Zotin MC, Scherlek AA, Perosa V, Auger CA, Warren AD, van der Weerd L, Schoemaker D, Horn MJ, Gurol ME, Gokcal E, Bacskai BJ, Viswanathan A, Greenberg SM, Reijmer YD, van Veluw SJ. Corpus callosum lesions are associated with worse cognitive performance in cerebral amyloid angiopathy. Brain Commun 2022; 4:fcac105. [PMID: 35611313 PMCID: PMC9123849 DOI: 10.1093/braincomms/fcac105] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 02/20/2022] [Accepted: 04/21/2022] [Indexed: 11/19/2022] Open
Abstract
The impact of vascular lesions on cognition is location dependent. Here, we assessed the contribution of small vessel disease lesions in the corpus callosum to vascular cognitive impairment in cerebral amyloid angiopathy, as a model for cerebral small vessel disease. Sixty-five patients with probable cerebral amyloid angiopathy underwent 3T magnetic resonance imaging, including a diffusion tensor imaging scan, and neuropsychological testing. Microstructural white-matter integrity was quantified by fractional anisotropy and mean diffusivity. Z-scores on individual neuropsychological tests were averaged into five cognitive domains: information processing speed, executive functioning, memory, language and visuospatial ability. Corpus callosum lesions were defined as haemorrhagic (microbleeds or larger bleeds) or ischaemic (microinfarcts, larger infarcts and diffuse fluid-attenuated inversion recovery hyperintensities). Associations between corpus callosum lesion presence, microstructural white-matter integrity and cognitive performance were examined with multiple regression models. The prevalence of corpus callosum lesions was confirmed in an independent cohort of memory clinic patients with and without cerebral amyloid angiopathy (n = 82). In parallel, we assessed corpus callosum lesions on ex vivo magnetic resonance imaging in cerebral amyloid angiopathy patients (n = 19) and controls (n = 5) and determined associated tissue abnormalities with histopathology. A total number of 21 corpus callosum lesions was found in 19/65 (29%) cerebral amyloid angiopathy patients. Corpus callosum lesion presence was associated with reduced microstructural white-matter integrity within the corpus callosum and in the whole-brain white matter. Patients with corpus callosum lesions performed significantly worse on all cognitive domains except language, compared with those without corpus callosum lesions after correcting for age, sex, education and time between magnetic resonance imaging and neuropsychological assessment. This association was independent of the presence of intracerebral haemorrhage, whole-brain fractional anisotropy and mean diffusivity, and white-matter hyperintensity volume and brain volume for the domains of information processing speed and executive functioning. In the memory clinic patient cohort, corpus callosum lesions were present in 14/54 (26%) patients with probable and 2/8 (25%) patients with possible cerebral amyloid angiopathy, and in 3/20 (15%) patients without cerebral amyloid angiopathy. In the ex vivo cohort, corpus callosum lesions were present in 10/19 (53%) patients and 2/5 (40%) controls. On histopathology, ischaemic corpus callosum lesions were associated with tissue loss and demyelination, which extended beyond the lesion core. Together, these data suggest that corpus callosum lesions are a frequent finding in cerebral amyloid angiopathy, and that they independently contribute to cognitive impairment through strategic microstructural disruption of white-matter tracts.
Collapse
Affiliation(s)
- Whitney M. Freeze
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Neuropsychology and Psychiatry, Maastricht University, Maastricht, The Netherlands
| | - Maria Clara Zanon Zotin
- Department of Neurology, J. Philip Kistler Stroke Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Medical Imaging, Hematology and Clinical Oncology, Ribeirão Preto Medical School, USP, SP, Brazil
| | - Ashley A. Scherlek
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Valentina Perosa
- Department of Neurology, J. Philip Kistler Stroke Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Corinne A. Auger
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Andrew D. Warren
- Department of Neurology, J. Philip Kistler Stroke Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Louise van der Weerd
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Mitchell J. Horn
- Department of Neurology, J. Philip Kistler Stroke Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - M. Edip Gurol
- Department of Neurology, J. Philip Kistler Stroke Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Elif Gokcal
- Department of Neurology, J. Philip Kistler Stroke Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Brian J. Bacskai
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Anand Viswanathan
- Department of Neurology, J. Philip Kistler Stroke Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Steven M. Greenberg
- Department of Neurology, J. Philip Kistler Stroke Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Yael D. Reijmer
- Department of Neurology, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Susanne J. van Veluw
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Neurology, J. Philip Kistler Stroke Research Center, Massachusetts General Hospital, Boston, MA, USA
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, MA 02129, USA
| |
Collapse
|
22
|
Beaman C, Kozii K, Hilal S, Liu M, Spagnolo-Allende AJ, Polanco-Serra G, Chen C, Cheng CY, Zambrano D, Arikan B, Del Brutto VJ, Wright C, Flowers XE, Leskinen SP, Rundek T, Mitchell A, Vonsattel JP, Cortes E, Teich AF, Sacco RL, Elkind MSV, Roh D, Gutierrez J. Cerebral Microbleeds, Cerebral Amyloid Angiopathy, and Their Relationships to Quantitative Markers of Neurodegeneration. Neurology 2022; 98:e1605-e1616. [PMID: 35228332 PMCID: PMC9052569 DOI: 10.1212/wnl.0000000000200142] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 01/18/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Age-related cognitive impairment is driven by the complex interplay of neurovascular and neurodegenerative disease. There is a strong relationship between cerebral microbleeds (CMBs), cerebral amyloid angiopathy (CAA), and the cognitive decline observed in conditions such as Alzheimer disease. However, in the early, preclinical phase of cognitive impairment, the extent to which CMBs and underlying CAA affect volumetric changes in the brain related to neurodegenerative disease remains unclear. METHODS We performed cross-sectional analyses from 3 large cohorts: The Northern Manhattan Study (NOMAS), Alzheimer's Disease Neuroimaging Initiative (ADNI), and the Epidemiology of Dementia in Singapore study (EDIS). We conducted a confirmatory analysis of 82 autopsied cases from the Brain Arterial Remodeling Study (BARS). We implemented multivariate regression analyses to study the association between 2 related markers of cerebrovascular disease-MRI-based CMBs and autopsy-based CAA-as independent variables and volumetric markers of neurodegeneration as dependent variables. NOMAS included mostly dementia-free participants age 55 years or older from northern Manhattan. ADNI included participants living in the United States age 55-90 years with a range of cognitive status. EDIS included community-based participants living in Singapore age 60 years and older with a range of cognitive status. BARS included postmortem pathologic samples. RESULTS We included 2,657 participants with available MRI data and 82 autopsy cases from BARS. In a meta-analysis of NOMAS, ADNI, and EDIS, superficial CMBs were associated with larger gray matter (β = 4.49 ± 1.13, p = 0.04) and white matter (β = 4.72 ± 2.1, p = 0.03) volumes. The association between superficial CMBs and larger white matter volume was more evident in participants with 1 CMB (β = 5.17 ± 2.47, p = 0.04) than in those with ≥2 CMBs (β = 1.97 ± 3.41, p = 0.56). In BARS, CAA was associated with increased cortical thickness (β = 6.5 ± 2.3, p = 0.016) but not with increased brain weight (β = 1.54 ± 1.29, p = 0.26). DISCUSSION Superficial CMBs are associated with larger morphometric brain measures, specifically white matter volume. This association is strongest in brains with fewer CMBs, suggesting that the CMB/CAA contribution to neurodegeneration may not relate to tissue loss, at least in early stages of disease.
Collapse
Affiliation(s)
- Charles Beaman
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Krystyna Kozii
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Saima Hilal
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Minghua Liu
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Anthony J Spagnolo-Allende
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Guillermo Polanco-Serra
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Christopher Chen
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Ching-Yu Cheng
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Daniela Zambrano
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Burak Arikan
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Victor J Del Brutto
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Clinton Wright
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Xena E Flowers
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Sandra P Leskinen
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Tatjana Rundek
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Amanda Mitchell
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Jean Paul Vonsattel
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Etty Cortes
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Andrew F Teich
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Ralph L Sacco
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Mitchell S V Elkind
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - David Roh
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| | - Jose Gutierrez
- From the Departments of Neurology (C.B., K.K., M.L., A.J.S.-A., D.Z., A.M., A.F.T., M.S.V.E., D.R., J.G.) and Pathology and Cell Biology (X.E.F., S.P.L., J.P.V., A.F.T.), Columbia University Irving Medical Center, New York, NY; Department of Neurology (C.B.), UCLA Medical Center, Los Angeles, CA; Memory Aging and Cognition Center (S.H., C.C.), National University Health System, Singapore; Department of Pharmacology (S.H., C.C.), Yong Loo Lin School of Medicine, National University of Singapore; Saw Swee Hock School of Public Health (S.H.), National University of Singapore and National University Health System, Singapore; College of Medicine (G.P.-S.), SUNY Upstate Medical University, Syracuse, NY; Singapore Eye Research Institute (C.-Y.C.), Singapore National Eye Centre; Ophthalmology and Visual Sciences Academic Clinical Program (C.-Y.C.), Duke-NUS Medical School, National University of Singapore; Istanbul University Cerrahpasa School of Medicine (B.A.), Turkey; Department of Neurology and Evelyn F. McKnight Brain Institute (V.J.D.B., T.R., R.L.S.), Miller School of Medicine, University of Miami Miller School of Medicine, FL; National Institutes of Health (C.W.), Bethesda, MD; Department of Pathology (E.C.), Icahn School of Medicine at Mount Sinai, New York, NY; and Department of Epidemiology (M.S.V.E.), Mailman School of Public Health, Columbia University, New York, NY
| |
Collapse
|
23
|
DiFrancesco JC, Stanzani L. Increased Brain Volume: A Novel Biomarker of Neurodegeneration? Neurology 2022; 98:649-650. [PMID: 35228330 DOI: 10.1212/wnl.0000000000200166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- Jacopo C DiFrancesco
- Department of Neurology, ASST San Gerardo Hosp., Univ. Milano-Bicocca, Monza, Italy
| | - Lorenzo Stanzani
- Department of Neurology, ASST San Gerardo Hosp., Univ. Milano-Bicocca, Monza, Italy
| |
Collapse
|
24
|
Jo S, Cheong EN, Kim N, Oh JS, Shim WH, Kim HJ, Lee SJ, Lee Y, Oh M, Kim JS, Kim BJ, Roh JH, Kim SJ, Lee JH. Role of White Matter Abnormalities in the Relationship Between Microbleed Burden and Cognitive Impairment in Cerebral Amyloid Angiopathy. J Alzheimers Dis 2022; 86:667-678. [DOI: 10.3233/jad-215094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Cerebral amyloid angiopathy (CAA) often presents as cognitive impairment, but the mechanism of cognitive decline is unclear. Recent studies showed that number of microbleeds were associated with cognitive decline. Objective: We aimed to investigate how microbleeds contribute to cognitive impairment in association with white matter tract abnormalities or cortical thickness in CAA. Methods: This retrospective comparative study involved patients with probable CAA according to the Boston criteria (Aβ + CAA) and patients with Alzheimer’s disease (Aβ + AD), all of whom showed severe amyloid deposition on amyloid PET. Using mediation analysis, we investigated how FA or cortical thickness mediates the correlation between the number of lobar microbleeds and cognition. Results: We analyzed 30 patients with Aβ + CAA (age 72.2±7.6, female 53.3%) and 30 patients with Aβ + AD (age 71.5±7.6, female 53.3%). The two groups showed similar degrees of cortical amyloid deposition in AD-related regions. The Aβ + CAA group had significantly lower FA values in the clusters of the posterior area than did the Aβ + AD group (family-wise error-corrected p < 0.05). The correlation between the number of lobar microbleeds and visuospatial function was indirectly mediated by white matter tract abnormality of right posterior thalamic radiation (PTR) and tapetum, while lobar microbleeds and language function was indirectly mediated by the abnormality of left PTR and sagittal stratum. Cortical thickness did not mediate the association between lobar microbleeds and cognition. Conclusion: This result supports the hypothesis that microbleeds burden leads to white matter tract damage and subsequent cognitive decline in CAA.
Collapse
Affiliation(s)
- Sungyang Jo
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - E-Nae Cheong
- Department of Medical Science and Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Nayoung Kim
- Department of Clinical Epidemiology and Biostatistics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jungsu S. Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Woo Hyun Shim
- Department of Medical Science and Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hyung-Ji Kim
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sun Ju Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yoojin Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Minyoung Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jae Seung Kim
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Bum Joon Kim
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jee Hoon Roh
- Department of Physiology, Neuroscience Research Institute, Korea University College of Medicine, Seoul, Republic of Korea
| | - Sang Joon Kim
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jae-Hong Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
25
|
Smirnov M, Destrieux C, Maldonado IL. Cerebral white matter vasculature: still uncharted? Brain 2021; 144:3561-3575. [PMID: 34718425 DOI: 10.1093/brain/awab273] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/17/2021] [Accepted: 07/11/2021] [Indexed: 11/14/2022] Open
Abstract
White matter vasculature plays a major role in the pathophysiology of permanent neurological deficits following a stroke or progressive cognitive alteration related to small vessel disease. Thus, knowledge of the complex vascularization and functional aspects of the deep white matter territories is paramount to comprehend clinical manifestations of brain ischemia. This review provides a structured presentation of the existing knowledge of the vascularization of the human cerebral white matter from seminal historical studies to the current literature. First, we revisit the highlights of prenatal development of the endoparenchymal telencephalic vascular system that are crucial for the understanding of vessel organization in the adult. Second, we reveal the tangled history of debates on the existence, clinical significance, and physiological role of leptomeningeal anastomoses. Then, we present how conceptions on white matter vascularization transitioned from the mixed ventriculopetal/ventriculofugal theory, in which a low-flow area was interposed in between concurrent arterial flows, to the purely ventriculopetal theory. The latter model explains variable white matter sensitivity to ischemia by various organizations of ventriculopetal vessel terminals having different origin/length properties and interconnection patterns. Next, arteries supplying primarily the white matter are described according to their length and overall structure. Furthermore, the known distribution territories, to date, are studied in relation to primary anatomical structures of the human cerebral white matter, emphasizing the sparsity of the "ground-truth" data available in the literature. Finally, the implications for both large vessel occlusion and chronic small vessel disease are discussed, as well as the insights from neuroimaging. All things considered, we identify the need for further research on deep white matter vascularization, especially regarding the arterial supply of white matter fiber tracts.
Collapse
Affiliation(s)
- Mykyta Smirnov
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Christophe Destrieux
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.,CHRU de Tours, Tours, France
| | | |
Collapse
|
26
|
Kozberg MG, Perosa V, Gurol ME, van Veluw SJ. A practical approach to the management of cerebral amyloid angiopathy. Int J Stroke 2021; 16:356-369. [PMID: 33252026 PMCID: PMC9097498 DOI: 10.1177/1747493020974464] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cerebral amyloid angiopathy is a common small vessel disease in the elderly involving vascular amyloid-β deposition. Cerebral amyloid angiopathy is one of the leading causes of intracerebral hemorrhage and a significant contributor to age-related cognitive decline. The awareness of a diagnosis of cerebral amyloid angiopathy is important in clinical practice as it impacts decisions to use lifelong anticoagulation or nonpharmacological alternatives to anticoagulation such as left atrial appendage closure in patients who have concurrent atrial fibrillation, another common condition in older adults. This review summarizes the latest literature regarding the management of patients with sporadic cerebral amyloid angiopathy, including diagnostic criteria, imaging biomarkers for cerebral amyloid angiopathy severity, and management strategies to decrease intracerebral hemorrhage risk. In a minority of patients, the presence of cerebral amyloid angiopathy triggers an autoimmune inflammatory reaction, referred to as cerebral amyloid angiopathy-related inflammation, which is often responsive to immunosuppressive treatment in the acute phase. Diagnosis and management of cerebral amyloid angiopathy-related inflammation will be presented separately. While there are currently no effective therapeutics available to cure or halt the progression of cerebral amyloid angiopathy, we discuss emerging avenues for potential future interventions.
Collapse
Affiliation(s)
- Mariel G Kozberg
- MassGeneral Institute for Neurodegenerative Disease, 2348Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Neurology, 2348Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Hemorrhagic Stroke Research Program, J. Philip Kistler Stroke Research Center, Department of Neurology, 2348Massachusetts General Hospital, Harvard Medical School, Boston, MA,USA
| | - Valentina Perosa
- MassGeneral Institute for Neurodegenerative Disease, 2348Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Neurology, 2348Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Hemorrhagic Stroke Research Program, J. Philip Kistler Stroke Research Center, Department of Neurology, 2348Massachusetts General Hospital, Harvard Medical School, Boston, MA,USA
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany
| | - M Edip Gurol
- Department of Neurology, 2348Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Hemorrhagic Stroke Research Program, J. Philip Kistler Stroke Research Center, Department of Neurology, 2348Massachusetts General Hospital, Harvard Medical School, Boston, MA,USA
| | - Susanne J van Veluw
- MassGeneral Institute for Neurodegenerative Disease, 2348Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
- Department of Neurology, 2348Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Hemorrhagic Stroke Research Program, J. Philip Kistler Stroke Research Center, Department of Neurology, 2348Massachusetts General Hospital, Harvard Medical School, Boston, MA,USA
| |
Collapse
|
27
|
Fotiadis P, Pasi M, Charidimou A, Warren AD, Schwab KM, Rosand J, van der Grond J, van Buchem MA, Viswanathan A, Gurol ME, Greenberg SM. Decreased Basal Ganglia Volume in Cerebral Amyloid Angiopathy. J Stroke 2021; 23:223-233. [PMID: 34102757 PMCID: PMC8189850 DOI: 10.5853/jos.2020.04280] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 01/06/2021] [Indexed: 11/25/2022] Open
Abstract
Background and Purpose Cerebral amyloid angiopathy (CAA) is a common pathology of the leptomeningeal and cortical small vessels associated with hemorrhagic and non-hemorrhagic brain injury. Given previous evidence for CAA-related loss of cortical thickness and white matter volume, we hypothesized that CAA might also cause tissue loss in the basal ganglia.
Methods We compared basal ganglia volumes expressed as a percentage of total intracranial volume (pBGV) of non-demented patients with sporadic and hereditary CAA to age-matched healthy control (HC) and Alzheimer’s disease (AD) cohorts.
Results Patients with sporadic CAA had lower pBGV (n=80, 1.16%±0.14%) compared to HC (n=80, 1.30%±0.13%, P<0.0001) and AD patients (n=80, 1.23%±0.11%, P=0.001). Similarly, patients with hereditary CAA demonstrated lower pBGV (n=25, 1.26%±0.17%) compared to their matched HC (n=25, 1.36%±0.15%, P=0.036). Using a measurement of normalized basal ganglia width developed for analysis of clinical-grade magnetic resonance images, we found smaller basal ganglia width in patients with CAA-related lobar intracerebral hemorrhage (ICH; n=93, 12.35±1.47) compared to age-matched patients with hypertension-related deep ICH (n=93, 13.46±1.51, P<0.0001) or HC (n=93, 15.45±1.22, P<0.0001). Within the sporadic CAA research cohort, decreased basal ganglia volume was independently correlated with greater cortical gray matter atrophy (r=0.45, P<0.0001), increased basal ganglia fractional anisotropy (r=–0.36, P=0.001), and worse performance on language processing (r=0.35, P=0.003), but not with cognitive tests of executive function or processing speed.
Conclusions These findings suggest an independent effect of CAA on basal ganglia tissue loss, indicating a novel mechanism for CAA-related brain injury and neurologic dysfunction.
Collapse
Affiliation(s)
- Panagiotis Fotiadis
- Department of Neurology, J.P. Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marco Pasi
- Department of Neurology, J.P. Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Stroke Unit, Department of Neurology, University of Lille, INSERM U1171, CHU Lille, Lille, France
| | - Andreas Charidimou
- Department of Neurology, J.P. Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrew D Warren
- Department of Neurology, J.P. Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kristin M Schwab
- Department of Neurology, J.P. Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Jonathan Rosand
- Department of Neurology, J.P. Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jeroen van der Grond
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Mark A van Buchem
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Anand Viswanathan
- Department of Neurology, J.P. Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - M Edip Gurol
- Department of Neurology, J.P. Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Steven M Greenberg
- Department of Neurology, J.P. Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
28
|
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: 84] [Impact Index Per Article: 21.0] [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
|