1
|
Lin SJ, Gillespie NA, Notestine R, Gamst AC, Chen AM, McEvoy LK, Panizzon MS, Elman JA, Glatt SJ, Hagler DJ, Neale MC, Franz CE, Kremen WS, Fennema-Notestine C. The genetic and environmental etiology of novel frequency-driven regional parcellations of abnormal white matter. Am J Med Genet B Neuropsychiatr Genet 2025; 198:e33004. [PMID: 39148448 DOI: 10.1002/ajmg.b.33004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 06/28/2024] [Accepted: 08/05/2024] [Indexed: 08/17/2024]
Abstract
The prevalence of white matter disease increases with age and is associated with cerebrovascular disease, cognitive decline, and risk for dementia. MRI measures of abnormal signal in the white matter (AWM) provide estimates of damage, however, regional patterns of AWM may be differentially influenced by genetic or environmental factors. With our data-driven regional parcellation approach, we created a probability distribution atlas using Vietnam Era Twin Study of Aging (VETSA) data (n = 475, mean age 67.6 years) and applied a watershed algorithm to define separate regional parcellations. We report biometrical twin modeling for five anatomically distinct regions: (1) Posterior, (2) Superior frontal and parietal, (3) Anterior and inferior frontal with deep areas, (4) Occipital, and (5) Anterior periventricular. We tested competing multivariate hypotheses to identify unique influences and to explain sources of covariance among the parcellations. Family aggregation could be entirely explained by additive genetic influences, with additive genetic variance (heritability) ranging from 0.69 to 0.79. Most genetic correlations between parcellations ranged from moderate to high (rg = 0.57-0.85), although two were small (rg = 0.35-0.39), consistent with varying degrees of unique genetic influences. This proof-of-principle investigation demonstrated the value of our novel, data-driven parcellations, with identifiable genetic and environmental differences, for future exploration.
Collapse
Affiliation(s)
- Shu-Ju Lin
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
- Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, California, USA
| | - Nathan A Gillespie
- Virginia Institute for Psychiatric and Behavior Genetics, Richmond, Virginia, USA
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Randy Notestine
- Computational and Applied Statistics Laboratory (CASL) at the San Diego Supercomputer Center (SDSC), La Jolla, California, USA
| | - Anthony C Gamst
- Computational and Applied Statistics Laboratory (CASL) at the San Diego Supercomputer Center (SDSC), La Jolla, California, USA
- Department of Mathematics, University of California, San Diego, La Jolla, California, USA
| | - Anna M Chen
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
| | - Linda K McEvoy
- Department of Radiology, University of California, San Diego, La Jolla, California, USA
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, La Jolla, California, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, Washington, USA
| | - Matthew S Panizzon
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
- Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, California, USA
| | - Jeremy A Elman
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
- Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, California, USA
| | - Stephen J Glatt
- Psychiatric Genetic Epidemiology & Neurobiology Laboratory (PsychGENe Lab), Departments of Psychiatry and Behavioral Sciences, Neuroscience and Physiology, and Public Health and Preventive Medicine, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Donald J Hagler
- Department of Radiology, University of California, San Diego, La Jolla, California, USA
| | - Michael C Neale
- Virginia Institute for Psychiatric and Behavior Genetics, Richmond, Virginia, USA
| | - Carol E Franz
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
- Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, California, USA
| | - William S Kremen
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
- Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, California, USA
| | - Christine Fennema-Notestine
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
- Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, California, USA
- Department of Radiology, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
2
|
Díaz-Pérez S, DeLong JH, Rivier CA, Lee CY, Askenase MH, Zhu B, Zhang L, Brennand KJ, Martins AJ, Sansing LH. Single-nucleus RNA sequencing of human periventricular white matter in vascular dementia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.06.627202. [PMID: 39713290 PMCID: PMC11661092 DOI: 10.1101/2024.12.06.627202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Vascular dementia (VaD) refers to a variety of dementias driven by cerebrovascular disease and is the second leading cause of dementia globally. VaD may be caused by ischemic strokes, intracerebral hemorrhage, and/or cerebral small vessel disease, commonly identified as white matter hyperintensities on MRI. The mechanisms underlying these white matter lesions in the periventricular brain are poorly understood. In this study we perform an extensive transcriptomic analysis on human postmortem periventricular white matter lesions in patients with VaD with the goal of identifying molecular pathways in the disease. We find increased cellular stress responses in astrocytes, oligodendrocytes, and oligodendrocyte precursor cells as well as transcriptional and translational repression in microglia in our dataset. We show that several genes identified by GWAS as being associated with white matter disease are differentially expressed in cells in VaD. Finally, we compare our dataset to an independent snRNAseq dataset of PVWM in VaD and a scRNAseq dataset on human iPSC-derived microglia exposed to oxygen glucose deprivation (OGD). We identify the increase of the heat shock protein response as a conserved feature of VaD across celltypes and show that this increase is not linked to OGD exposure. Overall, our study is the first to show that increased heat shock protein responses are a common feature of lesioned PVWM in VaD and may represent a potential therapeutic target.
Collapse
Affiliation(s)
| | - Jonathan H. DeLong
- Department of Neurology, Yale University School of Medicine, New Haven, CT
| | - Cyprien A. Rivier
- Department of Neurology, Yale University School of Medicine, New Haven, CT
| | - Chia-Yi Lee
- Department of Genetics, Yale University School of Medicine, New Haven, CT
| | - Michael H. Askenase
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Biqing Zhu
- Program of Computational Biology and Bioinformatics, Yale University School of Medicine, New Haven, CT
| | - Le Zhang
- Department of Neurology, Yale University School of Medicine, New Haven, CT
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT
| | - Kristen J. Brennand
- Department of Genetics, Yale University School of Medicine, New Haven, CT
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT
| | - Andrew J. Martins
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
| | - Lauren H. Sansing
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT
- Department of Neurology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
3
|
Makkinejad N, Zanon Zotin MC, van den Brink H, Auger CA, Vom Eigen KA, Iglesias JE, Greenberg SM, Perosa V, van Veluw SJ. Neuropathological Correlates of White Matter Hyperintensities in Cerebral Amyloid Angiopathy. J Am Heart Assoc 2024; 13:e035744. [PMID: 39526350 DOI: 10.1161/jaha.124.035744] [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: 04/23/2024] [Accepted: 09/17/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND White matter hyperintensities (WMHs) are frequently observed on magnetic resonance imaging (MRI) in patients with cerebral amyloid angiopathy (CAA). The neuropathological substrates that underlie WMHs in CAA are unclear, and it remains largely unexplored whether the different WMH distribution patterns associated with CAA (posterior confluent and subcortical multispot) reflect alternative pathophysiological mechanisms. METHODS AND RESULTS We performed a combined in vivo MRI-ex vivo MRI-neuropathological study in patients with definite CAA. Formalin-fixed hemispheres from 19 patients with CAA, most of whom also had in vivo MRI available, underwent 3T MRI, followed by standard neuropathological examination of the hemispheres and targeted neuropathological assessment of WMH patterns. Ex vivo WMH volume was independently associated with CAA severity (P=0.046) but not with arteriolosclerosis (P=0.743). In targeted neuropathological examination, compared with normal-appearing white matter, posterior confluent WMHs were associated with activated microglia (P=0.043) and clasmatodendrosis (P=0.031), a form of astrocytic injury. Trends were found for an association with white matter rarefaction (P=0.074) and arteriolosclerosis (P=0.094). An exploratory descriptive analysis suggested that the histopathological correlates of WMH multispots were similar to those underlying posterior confluent WMHs. CONCLUSIONS This study confirmed that vascular amyloid β severity in the cortex is significantly associated with WMH volume in patients with definite CAA. The histopathological substrates of both posterior confluent and WMH multispots were comparable, suggesting overlapping pathophysiological mechanisms, although these exploratory observations require confirmation in larger studies.
Collapse
Affiliation(s)
- Nazanin Makkinejad
- J. Philip Kistler Stroke Research Center, Department of Neurology Massachusetts General Hospital, Harvard Medical School Boston MA USA
| | - Maria Clara Zanon Zotin
- J. Philip Kistler Stroke Research Center, Department of Neurology Massachusetts General Hospital, Harvard Medical School Boston MA USA
- Center for Imaging Sciences and Medical Physics, Department of Medical Imaging, Hematology and Clinical Oncology, Ribeirão Preto Medical School University of São Paulo Ribeirão Preto SP Brazil
| | - Hilde van den Brink
- J. Philip Kistler Stroke Research Center, Department of Neurology Massachusetts General Hospital, Harvard Medical School Boston MA USA
| | - Corinne A Auger
- Department of Neurology MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School Charlestown MA USA
| | - Kali A Vom Eigen
- Department of Neurology MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School Charlestown MA USA
| | - Juan Eugenio Iglesias
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology Massachusetts General Hospital, Harvard Medical School Charlestown MA USA
- Computer Science and Artificial Intelligence Laboratory, MIT Cambridge MA USA
- Centre for Medical Image Computing University College London London United Kingdom
| | - Steven M Greenberg
- J. Philip Kistler Stroke Research Center, Department of Neurology Massachusetts General Hospital, Harvard Medical School Boston MA USA
| | - Valentina Perosa
- J. Philip Kistler Stroke Research Center, Department of Neurology Massachusetts General Hospital, Harvard Medical School Boston MA USA
| | - Susanne J van Veluw
- J. Philip Kistler Stroke Research Center, Department of Neurology Massachusetts General Hospital, Harvard Medical School Boston MA USA
- Department of Neurology MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School Charlestown MA USA
| |
Collapse
|
4
|
Libecap TJ, Pappas CA, Bauer CE, Zachariou V, Raslau FD, Gold BT. Enlarged perivascular space burden predicts declines in cognitive and functional performance. J Neurol Sci 2024; 466:123232. [PMID: 39298972 PMCID: PMC11563846 DOI: 10.1016/j.jns.2024.123232] [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: 03/14/2024] [Revised: 09/07/2024] [Accepted: 09/08/2024] [Indexed: 09/22/2024]
Abstract
INTRODUCTION We evaluated the relationship between baseline enlarged perivascular space (ePVS) burden and later cognitive decline. METHODS 83 community-dwelling, older adults (aged 56-86) completed three annual cognitive assessments that included the Clinical Dementia Rating (CDR®) Dementia Staging Instrument Sum of Boxes (CDR-SB) and composite measures of executive function and episodic memory. An MRI scan at baseline was used to count ePVS in the basal ganglia and centrum semiovale. Mixed effects models were run with ePVS as the predictor variable and cognitive measures as the dependent variable. Covariates included age, sex, education, cerebral small vessel disease (cSVD) risk factors, and cSVD neuroimaging biomarkers. RESULTS At baseline, high basal ganglia ePVS counts were associated with lower executive function scores and episodic memory scores. Moreover, baseline basal ganglia ePVS predicted worse longitudinal CDR-SB scores over the study period. DISCUSSION Basal ganglia ePVS burden is a promising biomarker for cSVD-related cognitive and functional decline.
Collapse
Affiliation(s)
- T J Libecap
- MD/PhD Program, University of Kentucky College of Medicine, Lexington, KY, USA; Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Colleen A Pappas
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Christopher E Bauer
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Valentinos Zachariou
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Flavius D Raslau
- Department of Radiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Brian T Gold
- Department of Neuroscience, University of Kentucky College of Medicine, Lexington, KY, USA; Department of Radiology, University of Kentucky College of Medicine, Lexington, KY, USA; Magnetic Resonance Imaging and Spectroscopy Center, University of Kentucky, Lexington, KY, USA; Sanders-Brown Center on Aging University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
5
|
Bernal J, Menze I, Yakupov R, Peters O, Hellmann-Regen J, Freiesleben SD, Priller J, Spruth EJ, Altenstein S, Schneider A, Fliessbach K, Wiltfang J, Schott BH, Jessen F, Rostamzadeh A, Glanz W, Incesoy EI, Buerger K, Janowitz D, Ewers M, Perneczky R, Rauchmann BS, Teipel S, Kilimann I, Laske C, Sodenkamp S, Spottke A, Esser A, Lüsebrink F, Dechent P, Hetzer S, Scheffler K, Schreiber S, Düzel E, Ziegler G. Longitudinal evidence for a mutually reinforcing relationship between white matter hyperintensities and cortical thickness in cognitively unimpaired older adults. Alzheimers Res Ther 2024; 16:240. [PMID: 39465440 PMCID: PMC11520063 DOI: 10.1186/s13195-024-01606-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/18/2024] [Indexed: 10/29/2024]
Abstract
BACKGROUND For over three decades, the concomitance of cortical neurodegeneration and white matter hyperintensities (WMH) has sparked discussions about their coupled temporal dynamics. Longitudinal studies supporting this hypothesis nonetheless remain scarce. METHODS We applied global and regional bivariate latent growth curve modelling to determine the extent to which WMH and cortical thickness were interrelated over a four-year period. For this purpose, we leveraged longitudinal MRI data from 451 cognitively unimpaired participants (DELCODE; median age 69.71 [IQR 65.51, 75.50] years; 52.32% female). Participants underwent MRI sessions annually over a four-year period (1815 sessions in total, with roughly four MRI sessions per participant). We adjusted all models for demographics and cardiovascular risk. RESULTS Our findings were three-fold. First, larger WMH volumes were linked to lower cortical thickness (σ = -0.165, SE = 0.047, Z = -3.515, P < 0.001). Second, individuals with higher WMH volumes experienced more rapid cortical thinning (σ = -0.226, SE = 0.093, Z = -2.443, P = 0.007), particularly in temporal, cingulate, and insular regions. Similarly, those with lower initial cortical thickness had faster WMH progression (σ = -0.141, SE = 0.060, Z = -2.336, P = 0.009), with this effect being most pronounced in temporal, cingulate, and insular cortices. Third, faster WMH progression was associated with accelerated cortical thinning (σ = -0.239, SE = 0.139, Z = -1.710, P = 0.044), particularly in frontal, occipital, and insular cortical regions. CONCLUSIONS Our study suggests that cortical thinning and WMH progression could be mutually reinforcing rather than parallel, unrelated processes, which become entangled before cognitive deficits are detectable. TRIAL REGISTRATION German Clinical Trials Register (DRKS00007966, 04/05/2015).
Collapse
Affiliation(s)
- Jose Bernal
- Institute of Cognitive Neurology and Dementia Research, Otto-Von-Guericke University Magdeburg, Magdeburg, Germany.
- German Centre for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.
- Centre for Clinical Brain Sciences, the University of Edinburgh, Edinburgh, UK.
- UK Dementia Research Institute Centre at the University of Edinburgh, Edinburgh, UK.
| | - Inga Menze
- Institute of Cognitive Neurology and Dementia Research, Otto-Von-Guericke University Magdeburg, Magdeburg, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Renat Yakupov
- Institute of Cognitive Neurology and Dementia Research, Otto-Von-Guericke University Magdeburg, Magdeburg, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Oliver Peters
- German Centre for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Charité - Universitätsmedizin Berlin, Institute of Psychiatry and Psychotherapy, Berlin, Germany
| | - Julian Hellmann-Regen
- German Centre for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Charité - Universitätsmedizin Berlin, Department of Psychiatry and Neurosciences, Campus Benjamin Franklin, Berlin, Germany
- German Centre for Mental Health (DZPG), Berlin, Germany
| | - Silka Dawn Freiesleben
- German Centre for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Charité - Universitätsmedizin Berlin, Institute of Psychiatry and Psychotherapy, Berlin, Germany
| | - Josef Priller
- UK Dementia Research Institute Centre at the University of Edinburgh, Edinburgh, UK
- German Centre for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany
- School of Medicine, Department of Psychiatry and Psychotherapy, Technical University of Munich, Munich, Germany
| | - Eike Jakob Spruth
- German Centre for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany
| | - Slawek Altenstein
- German Centre for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Berlin, Germany
| | - Anja Schneider
- German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn and University of Bonn, Bonn, Germany
| | - Klaus Fliessbach
- German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Old Age Psychiatry and Cognitive Disorders, University Hospital Bonn and University of Bonn, Bonn, Germany
| | - Jens Wiltfang
- German Centre for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Centre Göttingen, University of Göttingen, Göttingen, Germany
- Neurosciences and Signalling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Björn H Schott
- German Centre for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Department of Psychiatry and Psychotherapy, University Medical Centre Göttingen, University of Göttingen, Göttingen, Germany
- Leibniz Institute for Neurobiology, Brenneckestr. 6, 39118, Magdeburg, Germany
| | - Frank Jessen
- German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Psychiatry, Medical Faculty, University of Cologne, Cologne, Germany
- Excellence Cluster On Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Ayda Rostamzadeh
- Department of Psychiatry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Wenzel Glanz
- German Centre for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Enise I Incesoy
- Institute of Cognitive Neurology and Dementia Research, Otto-Von-Guericke University Magdeburg, Magdeburg, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Department for Psychiatry and Psychotherapy, University Clinic Magdeburg, Magdeburg, Germany
| | - Katharina Buerger
- German Centre for Neurodegenerative Diseases (DZNE), Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Michael Ewers
- German Centre for Neurodegenerative Diseases (DZNE), Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
| | - Robert Perneczky
- German Centre for Neurodegenerative Diseases (DZNE), Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy) Munich, Munich, Germany
- Ageing Epidemiology Research Unit (AGE), School of Public Health, Imperial College London, London, UK
| | - Boris-Stephan Rauchmann
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
- Department of Neuroradiology, University Hospital LMU, Munich, Germany
| | - Stefan Teipel
- German Centre for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Centre, Rostock, Germany
| | - Ingo Kilimann
- German Centre for Neurodegenerative Diseases (DZNE), Rostock, Germany
- Department of Psychosomatic Medicine, Rostock University Medical Centre, Rostock, Germany
| | - Christoph Laske
- German Centre for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Section for Dementia Research, Hertie Institute for Clinical Brain Research and Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Sebastian Sodenkamp
- German Centre for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Psychiatry and Psychotherapy, University of Tübingen, Tübingen, Germany
| | - Annika Spottke
- German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurology, University of Bonn, Bonn, Germany
| | - Anna Esser
- German Centre for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Falk Lüsebrink
- German Centre for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Peter Dechent
- Department of Cognitive Neurology, MR-Research in Neurosciences, Georg-August-University, Göttingen, Germany
| | - Stefan Hetzer
- Berlin Centre for Advanced Neuroimaging, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Klaus Scheffler
- Department for Biomedical Magnetic Resonance, University of Tübingen, Tübingen, Germany
| | - Stefanie Schreiber
- German Centre for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Department of Neurology, University Hospital Magdeburg, Magdeburg, Germany
| | - Emrah Düzel
- Institute of Cognitive Neurology and Dementia Research, Otto-Von-Guericke University Magdeburg, Magdeburg, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Gabriel Ziegler
- Institute of Cognitive Neurology and Dementia Research, Otto-Von-Guericke University Magdeburg, Magdeburg, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| |
Collapse
|
6
|
Hayes A, Kasner SE, Favilla CG, Rothstein A, Witsch J, Hamilton RH, Sloane KL. Not So Transient?: A Narrative Review on Cognitive Impairment After Transient Ischemic Attack. Stroke 2024; 55:2558-2566. [PMID: 39212043 PMCID: PMC11421974 DOI: 10.1161/strokeaha.124.046821] [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] [Indexed: 09/04/2024]
Abstract
Transient ischemic attack (TIA) is traditionally viewed as a self-resolving episode of neurological change without persistent impairments and without evidence of acute brain injury on neuroimaging. However, emerging evidence suggests that TIA may be associated with lingering cognitive dysfunction. Cognitive impairment is a prevalent and disabling sequela of ischemic stroke, but the clinical relevance of this phenomenon after TIA is less commonly recognized. We performed a literature search of observational studies of cognitive function after TIA. There is a consistent body of literature suggesting that rates of cognitive impairment following TIA are higher than healthy controls, but the studies included here are limited by heterogeneity in design and analysis methods. We go on to summarize recent literature on proposed pathophysiological mechanisms underlying the development of cognitive impairment following TIA and finally suggest future directions for further research in this field.
Collapse
Affiliation(s)
| | - Scott E. Kasner
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher G. Favilla
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Aaron Rothstein
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jens Witsch
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Roy H. Hamilton
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physical Medicine and Rehabilitation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kelly L. Sloane
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physical Medicine and Rehabilitation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
7
|
Bermudez C, Lesnick TG, More SS, Ramanan VK, Knopman DS, Rabinstein AA, Cogswell PM, Jack CR, Vemuri P, Petersen RC, Graff-Radford J, Chen JJ. Optical Coherence Tomography Angiography Retinal Imaging Associations With Burden of Small Vessel Disease and Amyloid Positivity in the Brain. J Neuroophthalmol 2024:00041327-990000000-00691. [PMID: 39085998 DOI: 10.1097/wno.0000000000002230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
BACKGROUND Alzheimer disease (AD) and other dementias are associated with vascular changes and amyloid deposition, which may be reflected as density changes in the retinal capillaries. These changes may can be directly visualized and quantified with optical coherence tomography angiography (OCTA), making OCTA a potential noninvasive preclinical biomarker of small vessel disease and amyloid positivity. Our objective was to investigate the feasibility of retinal imaging metrics as noninvasive biomarkers of small vessel disease and amyloid positivity in the brain. METHODS We investigated associations between OCTA and neuroimaging and cognitive metrics in 41 participants without dementia from the Mayo Clinic Study of Aging and Alzheimer's Disease Research Center. OCTA metrics included superficial, deep, and full retina capillary density of the fovea, parafovea, and macula as well as the area of the foveal avascular zone (FAZ). Neuroimaging metrics included a high burden of white matter hyperintensity (WMH), presence of cerebral microbleeds (CMB), lacunar infarcts, and amyloid positivity as evidenced on positron emission tomography (PET), whereas cognitive metrics included mini-mental status examination (MMSE) score. We performed generalized estimating equations to account for measurements in each eye while controlling for age and sex to estimate associations between OCTA metrics and neuroimaging and cognitive scores. RESULTS Associations between OCTA and neuroimaging metrics were restricted to the fovea. OCTA showed decreased capillary density with high burden of WMH in both the superficial (P = 0.003), deep (P = 0.004), and full retina (P = 0.01) in the fovea but not the parafovea or whole macula. Similarly, participants with amyloid PET positivity had significantly decreased capillary density in the superficial fovea (P = 0.027) and deep fovea (P = 0.03) but higher density in the superficial parafovea (P = 0.038). Participants with amyloid PET positivity also had a significantly larger FAZ (P = 0.031), whereas in those with high WMH burden the difference did not reach statistical significance (P = 0.075). There was also a positive association between MMSE and capillary density of the full retina within the fovea (P = 0.037) and in the superficial parafovea (P = 0.046). No associations were found between OCTA metrics and presence of CMB or presence of lacunar infarcts. CONCLUSION The associations of lower foveal capillary density with cerebral WMH and amyloid positivity suggest that further research is warranted to evaluate for shared mechanisms of disease between small vessel disease and AD pathologies.
Collapse
Affiliation(s)
- Camilo Bermudez
- Department of Neurology (CB, VKR, DSK, AAR, RCP, JG-R, JJC), Mayo Clinic, Rochester, Minnesota; Center for Drug Design (SSM), College of Pharmacy, University of Minnesota, Minneapolis, Minnesota; Departments of Radiology (PMC, CRJ, PV) and Ophthalmology (JJC), Mayo Clinic, Rochester, Minnesota; and Department of Quantitative Health Sciences (TGL), Mayo Clinic, Rochester, Minnesota
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Javierre-Petit C, Kontzialis M, Leurgans SE, Bennett DA, Schneider JA, Arfanakis K. Quantitative assessment of enlarged perivascular spaces via deep-learning in community-based older adults reveals independent associations with vascular neuropathologies, vascular risk factors and cognition. Brain Commun 2024; 6:fcae252. [PMID: 39130513 PMCID: PMC11316207 DOI: 10.1093/braincomms/fcae252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/24/2024] [Accepted: 07/27/2024] [Indexed: 08/13/2024] Open
Abstract
Enlarged perivascular spaces (EPVS) are common in older adults, but their neuropathologic correlates are unclear mainly because most work to date has relied on visual rating scales and/or clinical cohorts. The present study first developed a deep-learning model for automatic segmentation, localization and quantification of EPVS in ex vivo brain MRI, and then used this model to investigate the neuropathologic, clinical and cognitive correlates of EPVS in 817 community-based older adults that underwent autopsy. The new method exhibited high sensitivity in detecting EPVS as small as 3 mm3, good segmentation accuracy and consistency. Most EPVS were located in the frontal lobe, but the highest density was observed in the basal ganglia. EPVS in the cerebrum and specifically in the frontal lobe were associated with infarcts independent of other neuropathologies, while temporal and occipital EPVS were associated with cerebral amyloid angiopathy. EPVS in most brain lobes were also associated with diabetes mellitus independently of neuropathologies, while basal ganglia EPVS were independently associated with hypertension, supporting the notion of independent pathways from diabetes and hypertension to EPVS. Finally, EPVS were associated with lower cognitive performance independently of neuropathologies and clinical variables, suggesting that EPVS represent additional abnormalities contributing to lower cognition.
Collapse
Affiliation(s)
- Carles Javierre-Petit
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL 60616, USA
| | - Marinos Kontzialis
- Department of Diagnostic Radiology and Nuclear Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Sue E Leurgans
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | - David A Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
| | - Julie A Schneider
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Pathology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Konstantinos Arfanakis
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL 60616, USA
- Department of Diagnostic Radiology and Nuclear Medicine, Rush University Medical Center, Chicago, IL 60612, USA
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
9
|
Bachmann D, von Rickenbach B, Buchmann A, Hüllner M, Zuber I, Studer S, Saake A, Rauen K, Gruber E, Nitsch RM, Hock C, Treyer V, Gietl A. White matter hyperintensity patterns: associations with comorbidities, amyloid, and cognition. Alzheimers Res Ther 2024; 16:67. [PMID: 38561806 PMCID: PMC10983708 DOI: 10.1186/s13195-024-01435-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 03/23/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND White matter hyperintensities (WMHs) are often measured globally, but spatial patterns of WMHs could underlie different risk factors and neuropathological and clinical correlates. We investigated the spatial heterogeneity of WMHs and their association with comorbidities, Alzheimer's disease (AD) risk factors, and cognition. METHODS In this cross-sectional study, we studied 171 cognitively unimpaired (CU; median age: 65 years, range: 50 to 89) and 51 mildly cognitively impaired (MCI; median age: 72, range: 53 to 89) individuals with available amyloid (18F-flutementamol) PET and FLAIR-weighted images. Comorbidities were assessed using the Cumulative Illness Rating Scale (CIRS). Each participant's white matter was segmented into 38 parcels, and WMH volume was calculated in each parcel. Correlated principal component analysis was applied to the parceled WMH data to determine patterns of WMH covariation. Adjusted and unadjusted linear regression models were used to investigate associations of component scores with comorbidities and AD-related factors. Using multiple linear regression, we tested whether WMH component scores predicted cognitive performance. RESULTS Principal component analysis identified four WMH components that broadly describe FLAIR signal hyperintensities in posterior, periventricular, and deep white matter regions, as well as basal ganglia and thalamic structures. In CU individuals, hypertension was associated with all patterns except the periventricular component. MCI individuals showed more diverse associations. The posterior and deep components were associated with renal disorders, the periventricular component was associated with increased amyloid, and the subcortical gray matter structures was associated with sleep disorders, endocrine/metabolic disorders, and increased amyloid. In the combined sample (CU + MCI), the main effects of WMH components were not associated with cognition but predicted poorer episodic memory performance in the presence of increased amyloid. No interaction between hypertension and the number of comorbidities on component scores was observed. CONCLUSION Our study underscores the significance of understanding the regional distribution patterns of WMHs and the valuable insights that risk factors can offer regarding their underlying causes. Moreover, patterns of hyperintensities in periventricular regions and deep gray matter structures may have more pronounced cognitive implications, especially when amyloid pathology is also present.
Collapse
Affiliation(s)
- Dario Bachmann
- Institute for Regenerative Medicine, University of Zurich, Campus Schlieren, Wagistrasse 12, 8952, Zurich, Schlieren, Switzerland.
- Department of Health Sciences and Technology, ETH Zürich, 8093, Zurich, Switzerland.
| | | | - Andreas Buchmann
- Institute for Regenerative Medicine, University of Zurich, Campus Schlieren, Wagistrasse 12, 8952, Zurich, Schlieren, Switzerland
| | - Martin Hüllner
- Department of Nuclear Medicine, University Hospital of Zurich, University of Zurich, 8091, Zurich, Switzerland
| | - Isabelle Zuber
- Institute for Regenerative Medicine, University of Zurich, Campus Schlieren, Wagistrasse 12, 8952, Zurich, Schlieren, Switzerland
| | - Sandro Studer
- Institute for Regenerative Medicine, University of Zurich, Campus Schlieren, Wagistrasse 12, 8952, Zurich, Schlieren, Switzerland
| | - Antje Saake
- Institute for Regenerative Medicine, University of Zurich, Campus Schlieren, Wagistrasse 12, 8952, Zurich, Schlieren, Switzerland
| | - Katrin Rauen
- Institute for Regenerative Medicine, University of Zurich, Campus Schlieren, Wagistrasse 12, 8952, Zurich, Schlieren, Switzerland
- Department of Geriatric Psychiatry, Psychiatric Hospital Zurich, 8032, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich, 8057, Zurich, Switzerland
| | - Esmeralda Gruber
- Institute for Regenerative Medicine, University of Zurich, Campus Schlieren, Wagistrasse 12, 8952, Zurich, Schlieren, Switzerland
| | - Roger M Nitsch
- Institute for Regenerative Medicine, University of Zurich, Campus Schlieren, Wagistrasse 12, 8952, Zurich, Schlieren, Switzerland
- Neurimmune AG, 8952, Zurich, Schlieren, Switzerland
| | - Christoph Hock
- Institute for Regenerative Medicine, University of Zurich, Campus Schlieren, Wagistrasse 12, 8952, Zurich, Schlieren, Switzerland
- Neurimmune AG, 8952, Zurich, Schlieren, Switzerland
| | - Valerie Treyer
- Institute for Regenerative Medicine, University of Zurich, Campus Schlieren, Wagistrasse 12, 8952, Zurich, Schlieren, Switzerland
- Department of Nuclear Medicine, University Hospital of Zurich, University of Zurich, 8091, Zurich, Switzerland
| | - Anton Gietl
- Institute for Regenerative Medicine, University of Zurich, Campus Schlieren, Wagistrasse 12, 8952, Zurich, Schlieren, Switzerland
- Department of Geriatric Psychiatry, Psychiatric Hospital Zurich, 8032, Zurich, Switzerland
| |
Collapse
|
10
|
Zhilan T, Zengyu Z, Pengpeng J, Hualan Y, Chao L, Yan X, Zimin G, Shuangxing H, Weiwei L. Salidroside promotes pro-angiogenesis and repair of blood brain barrier via Notch/ITGB1 signal path in CSVD Model. J Adv Res 2024:S2090-1232(24)00081-X. [PMID: 38417575 DOI: 10.1016/j.jare.2024.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/05/2023] [Accepted: 02/25/2024] [Indexed: 03/01/2024] Open
Abstract
INTRODUCTION Salidroside (SAL), extracted from Rhodiola rosea, has been widely used in coronary heart disease and myocardial ischemia for decades. Previous studies have demonstrated that SAL could reduce arteriosclerosis, and thus combat ischemic brain damage. However, the in-depth function of the salidroside in Cerebral Small Vascular Disease (CSVD) has not been discovered, and related molecular mechanism is still unclear. OBJECTIVES The present study aims to explore the effects of salidroside in angiogenesis as well as repair of blood brain barrier (BBB) and its possible mechanisms. METHODS We established a rat model of SHR via 2-vessel gradual occlusion (SHR-2VGO) to mimic the CSVD. Subsequently, the MRI, pathomorphism, as well as Morriss water maze test were conducted to determine CSVD-related indicators. 8 weeks post-surgery, animals were randomly administered SAL, DAPT, ATN161 or saline.The aim was to explore the protective effects of SAL in CSVD as well as its possible mechanism. RESULTS Here we found that SAL could attenuate cerebral hypoperfusion-induced BBB disruption, promote the pro-angiogenesis through enhancing the cell budding. Further investigations demonstrated that SAL could significantly increase the expression of Notch1, Hes1, Hes5, and ITGB1. In addition, we confirmed that SAL could activate Notch signal path, and then up-regulate ITGB1 to promote pro-angiogenesis and thus protect BBB from disruption. CONCLUSION The aforementioned findings demonstrated that SAL could protect BBB integrity through Notch-ITGB1 signaling path in CSVD, which indicated that SAL could be a potential medicine candidate for CSVD treatment.
Collapse
Affiliation(s)
- Tu Zhilan
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Zhang Zengyu
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China; Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jin Pengpeng
- Department of Chronic Disease Management, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Yang Hualan
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Li Chao
- Vasculocardiology Department, Change County Hospital of Traditional Chinese Medicine, Shandong Province 261300, China
| | - Xi Yan
- Department of Radiology, Shanghai TCM-Integrated Hospital, 200082 Shanghai, China
| | - Guo Zimin
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Hou Shuangxing
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China.
| | - Li Weiwei
- Institute of Pediatrics, Children's Hospital of Fudan University, Fudan University, Shanghai 201102, China.
| |
Collapse
|
11
|
Hainsworth AH, Markus HS, Schneider JA. Cerebral Small Vessel Disease, Hypertension, and Vascular Contributions to Cognitive Impairment and Dementia. Hypertension 2024; 81:75-86. [PMID: 38044814 PMCID: PMC10734789 DOI: 10.1161/hypertensionaha.123.19943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Hypertension-associated cerebral small vessel disease is a common finding in older people. Strongly associated with age and hypertension, small vessel disease is found at autopsy in over 50% of people aged ≥65 years, with a spectrum of clinical manifestations. It is the main cause of lacunar stroke and a major source of vascular contributions to cognitive impairment and dementia. The brain areas affected are subcortical and periventricular white matter and deep gray nuclei. Neuropathological sequelae are diffuse white matter lesions (seen as white matter hyperintensities on T2-weighted magnetic resonance imaging), small ischemic foci (lacunes or microinfarcts), and less commonly, subcortical microhemorrhages. The most common form of cerebral small vessel disease is concentric, fibrotic thickening of small penetrating arteries (up to 300 microns outer diameter) termed arteriolosclerosis. Less common forms are small artery atheroma and lipohyalinosis (the lesions described by C. Miller Fisher adjacent to lacunes). Other microvascular lesions that are not reviewed here include cerebral amyloid angiopathy and venous collagenosis. Here, we review the epidemiology, neuropathology, clinical management, genetics, preclinical models, and pathogenesis of hypertensive small vessel disease. Knowledge gaps include initiating factors, molecular pathogenesis, relationships between arterial pathology and tissue damage, possible reversibility, pharmacological targets, and molecular biomarkers. Progress is anticipated from multicell transcriptomic and proteomic profiling, novel experimental models and further target-finding and interventional clinical studies.
Collapse
Affiliation(s)
- Atticus H. Hainsworth
- Molecular and Clinical Sciences Research Institute, St George’s University of London, United Kingdom (A.H.H.)
- Department of Neurology, St George’s University Hospitals NHS Foundation Trust, London, United Kingdom (A.H.H.)
| | - Hugh S. Markus
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, United Kingdom (H.S.M.)
| | - Julie A. Schneider
- Rush Alzheimer’s Disease Center, Departments of Pathology and Neurological Sciences, Rush University Medical Center, Chicago, IL (J.A.S.)
| |
Collapse
|
12
|
Keller JA, Sigurdsson S, Klaassen K, Hirschler L, van Buchem MA, Launer LJ, van Osch MJ, Gudnason V, de Bresser J. White matter hyperintensity shape is associated with long-term dementia risk. Alzheimers Dement 2023; 19:5632-5641. [PMID: 37303267 PMCID: PMC10713858 DOI: 10.1002/alz.13345] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 04/11/2023] [Accepted: 05/05/2023] [Indexed: 06/13/2023]
Abstract
INTRODUCTION We aimed to investigate the association between white matter hyperintensity (WMH) shape and volume and the long-term dementia risk in community-dwelling older adults. METHODS Three thousand seventy-seven participants (mean age: 75.6 ± 5.2 years) of the Age Gene/Environment Susceptibility (AGES)-Reykjavik study underwent baseline 1.5T brain magnetic resonance imaging and were followed up for dementia (mean follow-up: 9.9 ± 2.6 years). RESULTS More irregular shape of periventricular/confluent WMH (lower solidity (hazard ratio (95% confidence interval) 1.34 (1.17 to 1.52), p < .001) and convexity 1.38 (1.28 to 1.49), p < .001); higher concavity index 1.43 (1.32 to 1.54), p < .001) and fractal dimension 1.45 (1.32 to 1.58), p < .001)), higher total WMH volume (1.68 (1.54 to 1.87), p < .001), higher periventricular/confluent WMH volume (1.71 (1.55 to 1.89), p < .001), and higher deep WMH volume (1.17 (1.08 to 1.27), p < .001) were associated with an increased long-term dementia risk. DISCUSSION WMH shape markers may in the future be useful in determining patient prognosis and may aid in patient selection for future preventive treatments in community-dwelling older adults.
Collapse
Affiliation(s)
- Jasmin A. Keller
- Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | | | - Kelly Klaassen
- Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Lydiane Hirschler
- Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Mark A. van Buchem
- Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Lenore J. Launer
- Laboratory of Epidemiology and Population Science, National Institute on Aging, Bethesda, MD 20898, United States
| | - Matthias J.P. van Osch
- Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Vilmundur Gudnason
- Icelandic Heart Association, 201 Kopavogur, Iceland
- Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - Jeroen de Bresser
- Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| |
Collapse
|
13
|
Faigle W, Piccirelli M, Hortobágyi T, Frontzek K, Cannon AE, Zürrer WE, Granberg T, Kulcsar Z, Ludersdorfer T, Frauenknecht KBM, Reimann R, Ineichen BV. The Brainbox -a tool to facilitate correlation of brain magnetic resonance imaging features to histopathology. Brain Commun 2023; 5:fcad307. [PMID: 38025281 PMCID: PMC10664401 DOI: 10.1093/braincomms/fcad307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/20/2023] [Accepted: 11/07/2023] [Indexed: 12/01/2023] Open
Abstract
Magnetic resonance imaging (MRI) has limitations in identifying underlying tissue pathology, which is relevant for neurological diseases such as multiple sclerosis, stroke or brain tumours. However, there are no standardized methods for correlating MRI features with histopathology. Thus, here we aimed to develop and validate a tool that can facilitate the correlation of brain MRI features to corresponding histopathology. For this, we designed the Brainbox, a waterproof and MRI-compatible 3D printed container with an integrated 3D coordinate system. We used the Brainbox to acquire post-mortem ex vivo MRI of eight human brains, fresh and formalin-fixed, and correlated focal imaging features to histopathology using the built-in 3D coordinate system. With its built-in 3D coordinate system, the Brainbox allowed correlation of MRI features to corresponding tissue substrates. The Brainbox was used to correlate different MR image features of interest to the respective tissue substrate, including normal anatomical structures such as the hippocampus or perivascular spaces, as well as a lacunar stroke. Brain volume decreased upon fixation by 7% (P = 0.01). The Brainbox enabled degassing of specimens before scanning, reducing susceptibility artefacts and minimizing bulk motion during scanning. In conclusion, our proof-of-principle experiments demonstrate the usability of the Brainbox, which can contribute to improving the specificity of MRI and the standardization of the correlation between post-mortem ex vivo human brain MRI and histopathology. Brainboxes are available upon request from our institution.
Collapse
Affiliation(s)
- Wolfgang Faigle
- Neuroimmunology and MS Research Section, Neurology Clinic, University Zurich, University Hospital Zurich, CH-8091 Zurich, Switzerland
| | - Marco Piccirelli
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, CH-8091 Zurich, Switzerland
| | - Tibor Hortobágyi
- Institute of Neuropathology, University of Zurich, CH-8091 Zurich, Switzerland
| | - Karl Frontzek
- Institute of Neuropathology, University of Zurich, CH-8091 Zurich, Switzerland
- Queen Square Brain Bank for Neurological Disorders, UCL Queen Square Institute of Neurology, WC1N 1PJ London, United Kingdom
| | - Amelia Elaine Cannon
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, CH-8091 Zurich, Switzerland
| | - Wolfgang Emanuel Zürrer
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, CH-8091 Zurich, Switzerland
| | - Tobias Granberg
- Department of Neuroradiology, Karolinska University Hospital, S-141 86 Stockholm, Sweden
| | - Zsolt Kulcsar
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, CH-8091 Zurich, Switzerland
| | - Thomas Ludersdorfer
- Neuroimmunology and MS Research Section, Neurology Clinic, University Zurich, University Hospital Zurich, CH-8091 Zurich, Switzerland
| | - Katrin B M Frauenknecht
- Institute of Neuropathology, University of Zurich, CH-8091 Zurich, Switzerland
- Luxembourg Center of Neuropathology (LCNP), Laboratoire National de Santé, 3555 Dudelange, Luxembourg
- National Center of Pathology (NCP), Laboratoire National de Santé, 3555 Dudelange, Luxembourg
| | - Regina Reimann
- Institute of Neuropathology, University of Zurich, CH-8091 Zurich, Switzerland
| | - Benjamin Victor Ineichen
- Department of Neuroradiology, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, CH-8091 Zurich, Switzerland
- Center for Reproducible Science, University of Zurich, CH-8001 Zurich, Switzerland
| |
Collapse
|
14
|
Autio AH, Paavola J, Tervonen J, Lång M, Huuskonen TJ, Huttunen J, Kärkkäinen V, von Und Zu Fraunberg M, Lindgren AE, Koivisto T, Kurola J, Jääskeläinen JE, Kämäräinen OP. Should individual timeline and serial CT/MRI panels of all patients be presented in acute brain insult cohorts? A pilot study of 45 patients with decompressive craniectomy after aneurysmal subarachnoid hemorrhage. Acta Neurochir (Wien) 2023; 165:3299-3323. [PMID: 36715752 PMCID: PMC10624760 DOI: 10.1007/s00701-022-05473-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 12/20/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE Our review of acute brain insult articles indicated that the patients' individual (i) timeline panels with the defined time points since the emergency call and (ii) serial brain CT/MRI slice panels through the neurointensive care until death or final brain tissue outcome at 12 months or later are not presented. METHODS We retrospectively constructed such panels for the 45 aneurysmal subarachnoid hemorrhage (aSAH) patients with a secondary decompressive craniectomy (DC) after the acute admission to neurointensive care at Kuopio University Hospital (KUH) from a defined population from 2005 to 2018. The patients were indicated by numbers (1.-45.) in the pseudonymized panels, tables, results, and discussion. The timelines contained up to ten defined time points on a logarithmic time axis until death ([Formula: see text]; 56%) or 3 years ([Formula: see text]; 44%). The brain CT/MRI panels contained a representative slice from the following time points: SAH diagnosis, after aneurysm closure, after DC, at about 12 months (20 survivors). RESULTS The timelines indicated re-bleeds and allowed to compare the times elapsed between any two time points, in terms of workflow swiftness. The serial CT/MRI slices illustrated the presence and course of intracerebral hemorrhage (ICH), perihematomal edema, intraventricular hemorrhage (IVH), hydrocephalus, delayed brain injury, and, in the 20 (44%) survivors, the brain tissue outcome. CONCLUSIONS The pseudonymized timeline panels and serial brain imaging panels, indicating the patients by numbers, allowed the presentation and comparison of individual clinical courses. An obvious application would be the quality control in acute or elective medicine for timely and equal access to clinical care.
Collapse
Affiliation(s)
- Anniina H Autio
- Neurosurgery, NeuroCenter, Kuopio University Hospital, PL 100, 70029, Kuopio, Finland.
- Institute of Clinical Medicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland.
| | - Juho Paavola
- Neurosurgery, NeuroCenter, Kuopio University Hospital, PL 100, 70029, Kuopio, Finland
- Institute of Clinical Medicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Joona Tervonen
- Neurosurgery, NeuroCenter, Kuopio University Hospital, PL 100, 70029, Kuopio, Finland
- Institute of Clinical Medicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Maarit Lång
- Institute of Clinical Medicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
- Neurointensive Care Unit, Kuopio University Hospital, Kuopio, Finland
| | - Terhi J Huuskonen
- Neurosurgery, NeuroCenter, Kuopio University Hospital, PL 100, 70029, Kuopio, Finland
- Institute of Clinical Medicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jukka Huttunen
- Neurosurgery, NeuroCenter, Kuopio University Hospital, PL 100, 70029, Kuopio, Finland
- Institute of Clinical Medicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Virve Kärkkäinen
- Neurosurgery, NeuroCenter, Kuopio University Hospital, PL 100, 70029, Kuopio, Finland
| | - Mikael von Und Zu Fraunberg
- Neurosurgery, NeuroCenter, Kuopio University Hospital, PL 100, 70029, Kuopio, Finland
- Institute of Clinical Medicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
- Department of Neurosurgery, Oulu University Hospital, Oulu, Finland
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
| | - Antti E Lindgren
- Neurosurgery, NeuroCenter, Kuopio University Hospital, PL 100, 70029, Kuopio, Finland
- Institute of Clinical Medicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
- Clinical Radiology, Kuopio University Hospital, Kuopio, Finland
| | - Timo Koivisto
- Neurosurgery, NeuroCenter, Kuopio University Hospital, PL 100, 70029, Kuopio, Finland
- Institute of Clinical Medicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jouni Kurola
- Institute of Clinical Medicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
- Center for Prehospital Emergency Care, Kuopio University Hospital, Kuopio, Finland
| | - Juha E Jääskeläinen
- Neurosurgery, NeuroCenter, Kuopio University Hospital, PL 100, 70029, Kuopio, Finland
- Institute of Clinical Medicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Olli-Pekka Kämäräinen
- Neurosurgery, NeuroCenter, Kuopio University Hospital, PL 100, 70029, Kuopio, Finland
- Institute of Clinical Medicine, School of Medicine, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
15
|
Ishikawa H, Shindo A, Mizutani A, Tomimoto H, Lo EH, Arai K. A brief overview of a mouse model of cerebral hypoperfusion by bilateral carotid artery stenosis. J Cereb Blood Flow Metab 2023; 43:18-36. [PMID: 36883344 PMCID: PMC10638994 DOI: 10.1177/0271678x231154597] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/23/2022] [Accepted: 01/04/2023] [Indexed: 03/09/2023]
Abstract
Vascular cognitive impairment (VCI) refers to all forms of cognitive disorder related to cerebrovascular diseases, including vascular mild cognitive impairment, post-stroke dementia, multi-infarct dementia, subcortical ischemic vascular dementia (SIVD), and mixed dementia. Among the causes of VCI, more attention has been paid to SIVD because the causative cerebral small vessel pathologies are frequently observed in elderly people and because the gradual progression of cognitive decline often mimics Alzheimer's disease. In most cases, small vessel diseases are accompanied by cerebral hypoperfusion. In mice, prolonged cerebral hypoperfusion is induced by bilateral carotid artery stenosis (BCAS) with surgically implanted metal micro-coils. This cerebral hypoperfusion BCAS model was proposed as a SIVD mouse model in 2004, and the spreading use of this mouse SIVD model has provided novel data regarding cognitive dysfunction and histological/genetic changes by cerebral hypoperfusion. Oxidative stress, microvascular injury, excitotoxicity, blood-brain barrier dysfunction, and secondary inflammation may be the main mechanisms of brain damage due to prolonged cerebral hypoperfusion, and some potential therapeutic targets for SIVD have been proposed by using transgenic mice or clinically used drugs in BCAS studies. This review article overviews findings from the studies that used this hypoperfused-SIVD mouse model, which were published between 2004 and 2021.
Collapse
Affiliation(s)
- Hidehiro Ishikawa
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Akihiro Shindo
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Akane Mizutani
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hidekazu Tomimoto
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Eng H Lo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
16
|
Li L, He G, Shi M, Zhu J, Cheng Y, Chen Y, Chen J, Xue Q. Edaravone dexborneol ameliorates cognitive impairment by regulating the NF-κB pathway through AHR and promoting microglial polarization towards the M2 phenotype in mice with bilateral carotid artery stenosis (BCAS). Eur J Pharmacol 2023; 957:176036. [PMID: 37673366 DOI: 10.1016/j.ejphar.2023.176036] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 08/29/2023] [Accepted: 08/29/2023] [Indexed: 09/08/2023]
Abstract
Cerebral small vessel disease (CSVD) is one of the most important causes of stroke and vascular dementia, so exploring effective treatment modalities for CSVD is warranted. This study aimed to explore the anti-inflammatory effects of Edaravone dexborneol (C.EDA) in a CSVD model. Mice with CSVD showed distinct cognitive decline, as assessed by the Morris water maze (MWM). Pathological staining verified leakage across the blood‒brain barrier (BBB), microglial proliferation, neuronal loss and demyelination. Western blot analysis demonstrated that M1 microglia dominated prophase and released proinflammatory molecules; the aryl hydrocarbon receptor (AHR) was found to participate in modulating nuclear factor-kappa B (NF-κB) signalling activation through tumour necrosis factor receptor-associated factor-6 (TRAF6). C.EDA treatment resulted in the polarization of microglia from the M1 to the M2 phenotype. Mice sequentially treated with C.EDA exhibited a significant improvement in cognitive function; expression of the anti-inflammatory cytokines and modulatory proteins AHR and TRAF6 was upregulated, while the levels of pNF-κBp65 and pIΚBα were downregulated. C.EDA promoted microglial activation towards the M2 phenotype by upregulating AHR expression, which prevented TRAF6 ubiquitination, promoted NF-κB RelA/p65 protein degradation and inhibited subsequent NF-κB phosphorylation. Mechanistically, the anti-inflammatory effect of C.EDA alleviated neuronal loss and myelin damage, while at the functional level, C.EDA improved cognitive function and thus showed good application prospects.
Collapse
Affiliation(s)
- Lei Li
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006, China; Department of Neurology, The First People's Hospital of Yancheng, Affiliated Hospital of Nanjing University Medical School, Yancheng, Jiangsu Province, 224005, China
| | - Guojun He
- Department of Neurology, The First People's Hospital of Yancheng, Affiliated Hospital of Nanjing University Medical School, Yancheng, Jiangsu Province, 224005, China
| | - Mingyu Shi
- Department of Neurology, The First People's Hospital of Yancheng, Affiliated Hospital of Nanjing University Medical School, Yancheng, Jiangsu Province, 224005, China
| | - Juehua Zhu
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006, China
| | - Yongqing Cheng
- Department of Neurology, The First People's Hospital of Yancheng, Affiliated Hospital of Nanjing University Medical School, Yancheng, Jiangsu Province, 224005, China
| | - Yang Chen
- Department of Neurology, The First People's Hospital of Yancheng, Affiliated Hospital of Nanjing University Medical School, Yancheng, Jiangsu Province, 224005, China
| | - Jin Chen
- Department of Neurology, The First People's Hospital of Yancheng, Affiliated Hospital of Nanjing University Medical School, Yancheng, Jiangsu Province, 224005, China
| | - Qun Xue
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215006, China.
| |
Collapse
|
17
|
Caprihan A, Hillmer L, Erhardt EB, Adair JC, Knoefel JE, Prestopnik J, Rosenberg GA. A trichotomy method for defining homogeneous subgroups in a dementia population. Ann Clin Transl Neurol 2023; 10:1802-1815. [PMID: 37602520 PMCID: PMC10578887 DOI: 10.1002/acn3.51869] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/11/2023] [Accepted: 07/22/2023] [Indexed: 08/22/2023] Open
Abstract
INTRODUCTION Diagnosis of dementia in the aging brain is confounded by the presence of multiple pathologies. Mixed dementia (MX), a combination of Alzheimer's disease (AD) proteins with vascular disease (VD), is frequently found at autopsy, and has been difficult to diagnose during life. This report develops a method for separating the MX group and defining preclinical AD (presence of AD factors with normal cognition) and preclinical VD subgroups (presence of white matter damage with normal cognition). METHODS Clustering was based on three diagnostic axes: (1) AD factor (ADF) derived from cerebrospinal fluid proteins (Aβ42 and pTau), (2) VD factor (VDF) calculated from mean free water and peak width of skeletonized mean diffusivity in the white matter, and (3) Cognition (Cog) based on memory and executive function. The trichotomy method was applied to an Alzheimer's Disease Neuroimaging Initiative cohort (N = 538). RESULTS Eight biologically defined subgroups were identified which included the MX group with both high ADF and VDF (9.3%) and a preclinical VD group (3.9%), and a preclinical AD group (13.6%). Cog is significantly associated with both ADF and VDF, and the partial-correlation remains significant even when the effect of the other variable is removed (r(Cog, ADF/VDF removed) = 0.46, p < 10-28 and r(Cog, VDF/ADF removed) = 0.24, p < 10-7 ). DISCUSSION The trichotomy method creates eight biologically characterized patient groups, which includes MX, preclinical AD, and preclinical VD subgroups. Further longitudinal studies are needed to determine the utility of the 3-way clustering method with multimodal biological biomarkers.
Collapse
Affiliation(s)
| | - Laura Hillmer
- Center for Memory and AgingUniversity of New Mexico School of MedicineAlbuquerqueNew Mexico87106USA
| | - Erik Barry Erhardt
- Departments of Mathematics and StatisticsUniversity of New Mexico College of Arts and SciencesAlbuquerqueNew Mexico87106USA
| | - John C. Adair
- Center for Memory and AgingUniversity of New Mexico School of MedicineAlbuquerqueNew Mexico87106USA
- Department of NeurologyUniversity of New MexicoAlbuquerqueNew Mexico87106USA
| | - Janice E. Knoefel
- Center for Memory and AgingUniversity of New Mexico School of MedicineAlbuquerqueNew Mexico87106USA
- Department of NeurologyUniversity of New MexicoAlbuquerqueNew Mexico87106USA
| | - Jillian Prestopnik
- Center for Memory and AgingUniversity of New Mexico School of MedicineAlbuquerqueNew Mexico87106USA
| | - Gary A. Rosenberg
- Center for Memory and AgingUniversity of New Mexico School of MedicineAlbuquerqueNew Mexico87106USA
- Department of NeurologyUniversity of New MexicoAlbuquerqueNew Mexico87106USA
| | | |
Collapse
|
18
|
Sperber C, Hakim A, Gallucci L, Seiffge D, Rezny-Kasprzak B, Jäger E, Meinel T, Wiest R, Fischer U, Arnold M, Umarova R. A typology of cerebral small vessel disease based on imaging markers. J Neurol 2023; 270:4985-4994. [PMID: 37368130 PMCID: PMC10511610 DOI: 10.1007/s00415-023-11831-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 06/16/2023] [Accepted: 06/17/2023] [Indexed: 06/28/2023]
Abstract
BACKGROUND Lacunes, microbleeds, enlarged perivascular spaces (EPVS), and white matter hyperintensities (WMH) are brain imaging features of cerebral small vessel disease (SVD). Based on these imaging markers, we aimed to identify subtypes of SVD and to evaluate the validity of these markers as part of clinical ratings and as biomarkers for stroke outcome. METHODS In a cross-sectional study, we examined 1207 first-ever anterior circulation ischemic stroke patients (mean age 69.1 ± 15.4 years; mean NIHSS 5.3 ± 6.8). On acute stroke MRI, we assessed the numbers of lacunes and microbleeds and rated EPVS and deep and periventricular WMH. We used unsupervised learning to cluster patients based on these variables. RESULTS We identified five clusters, of which the last three appeared to represent distinct late stages of SVD. The two largest clusters had no to only mild or moderate WMH and EPVS, respectively, and favorable stroke outcome. The third cluster was characterized by the largest number of lacunes and a likewise favorable outcome. The fourth cluster had the highest age, most pronounced WMH, and poor outcome. Showing the worst outcome, the fifth cluster presented pronounced microbleeds and the most severe SVD burden. CONCLUSION The study confirmed the existence of different SVD types with different relationships to stroke outcome. EPVS and WMH were identified as imaging features of presumably early progression. The number of microbleeds and WMH severity appear to be promising biomarkers for distinguishing clinical subgroups. Further understanding of SVD progression might require consideration of refined SVD features, e.g., for EPVS and type of lacunes.
Collapse
Affiliation(s)
- Christoph Sperber
- Department of Neurology, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Arsany Hakim
- University Institute of Diagnostic and Interventional Neuroradiology, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Laura Gallucci
- Department of Neurology, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - David Seiffge
- Department of Neurology, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Beata Rezny-Kasprzak
- University Institute of Diagnostic and Interventional Neuroradiology, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Eugen Jäger
- University Institute of Diagnostic and Interventional Neuroradiology, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Thomas Meinel
- Department of Neurology, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Roland Wiest
- University Institute of Diagnostic and Interventional Neuroradiology, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Urs Fischer
- Department of Neurology, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
- Department of Neurology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Marcel Arnold
- Department of Neurology, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Roza Umarova
- Department of Neurology, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland.
| |
Collapse
|
19
|
Pauls MMH, Fish J, Binnie LR, Benjamin P, Betteridge S, Clarke B, Dhillon MPK, Ghatala R, Hainsworth FAH, Howe FA, Khan U, Kruuse C, Madigan JB, Moynihan B, Patel B, Pereira AC, Rostrup E, Shtaya ABY, Spilling CA, Trippier S, Williams R, Young R, Barrick TR, Isaacs JD, Hainsworth AH. Testing the cognitive effects of tadalafil. Neuropsychological secondary outcomes from the PASTIS trial. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2023; 5:100187. [PMID: 37811523 PMCID: PMC10550803 DOI: 10.1016/j.cccb.2023.100187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/25/2023] [Accepted: 09/25/2023] [Indexed: 10/10/2023]
Abstract
Cerebral small vessel disease (SVD) is a major cause of cognitive impairment in older people. As secondary endpoints in a phase-2 randomised clinical trial, we tested the effects of single administration of a widely-used PDE5 inhibitor, tadalafil, on cognitive performance in older people with SVD. In a double-blinded, placebo-controlled, cross-over trial, participants received tadalafil (20 mg) and placebo on two visits ≥ 7 days apart (randomised to order of treatment). The Montreal Cognitive Assessment (MOCA) was administered at baseline, alongside a measure to estimate optimal intellectual ability (Test of Premorbid Function). Then, before and after treatment, a battery of neuropsychological tests was administered, assessing aspects of attention, information processing speed, working memory and executive function. Sixty-five participants were recruited and 55 completed the protocol (N = 55, age: 66.8 (8.6) years, range 52-87; 15/40 female/male). Median MOCA score was 26 (IQR: 23, 27], range 15-30). No significant treatment effects were seen in any of the neuropsychological tests. There was a trend towards improved performance on Digit Span Forward (treatment effect 0.37, C.I. 0.01, 0.72; P = 0.0521). We did not identify significant treatment effects of single-administration tadalafil on neuropsychological performance in older people with SVD. The trend observed on Digit Span Forward may help to inform future studies. Clinical trial registration http://www.clinicaltrials.gov. Unique identifier: NCT00123456, https://eudract.ema.europa.eu. Unique identifier: 2015-001,235-20NCT00123456.
Collapse
Affiliation(s)
- Mathilde MH Pauls
- Molecular & Clinical Sciences Research Institute, St George's University of London, UK
- Department of Neurology, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Jessica Fish
- Neuropsychology, St George's University Hospitals NHS Foundation Trust, London, UK
- School of Health & Wellbeing, University of Glasgow, UK
| | - Lauren R Binnie
- Molecular & Clinical Sciences Research Institute, St George's University of London, UK
| | - Philip Benjamin
- Molecular & Clinical Sciences Research Institute, St George's University of London, UK
- Neuroradiology, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Shai Betteridge
- Neuropsychology, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Brian Clarke
- Department of Neurology, St George's University Hospitals NHS Foundation Trust, London, UK
| | | | - Rita Ghatala
- South London Stroke Research Network, London, UK
| | | | - Franklyn A Howe
- Molecular & Clinical Sciences Research Institute, St George's University of London, UK
| | - Usman Khan
- Department of Neurology, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Christina Kruuse
- Department of Neurology and Neurovascular Research Unit, Herlev Gentofte Hospital, Denmark
| | - Jeremy B Madigan
- Neuroradiology, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Barry Moynihan
- Department of Neurology, St George's University Hospitals NHS Foundation Trust, London, UK
- Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Bhavini Patel
- Department of Neurology, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Anthony C Pereira
- Molecular & Clinical Sciences Research Institute, St George's University of London, UK
- Department of Neurology, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Egill Rostrup
- Mental Health Centre, University of Copenhagen, Glostrup, Denmark
| | - Anan BY Shtaya
- Molecular & Clinical Sciences Research Institute, St George's University of London, UK
| | - Catherine A Spilling
- Molecular & Clinical Sciences Research Institute, St George's University of London, UK
| | | | | | - Robin Young
- Robertson Centre for Biostatistics, University of Glasgow, UK
| | - Thomas R Barrick
- Molecular & Clinical Sciences Research Institute, St George's University of London, UK
| | - Jeremy D Isaacs
- Molecular & Clinical Sciences Research Institute, St George's University of London, UK
- Department of Neurology, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Atticus H Hainsworth
- Molecular & Clinical Sciences Research Institute, St George's University of London, UK
- Department of Neurology, St George's University Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
20
|
Nikseresht G, Evia AM, Nag S, Leurgans SE, Capuano AW, Agam G, Barnes LL, Bennett DA, Schneider JA, Arfanakis K. Neuropathologic correlates of cerebral microbleeds in community-based older adults. Neurobiol Aging 2023; 129:89-98. [PMID: 37279617 PMCID: PMC10524842 DOI: 10.1016/j.neurobiolaging.2023.05.005] [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: 01/05/2023] [Revised: 04/14/2023] [Accepted: 05/04/2023] [Indexed: 06/08/2023]
Abstract
Cerebral microbleeds (CMBs) appearing as hypointense foci on T2*-weighted magnetic resonance images are small hemorrhages that have been linked to cognitive decline and increased mortality. However, the neuropathologic correlates of CMBs in community-based older adults are poorly understood. The present study investigated the association of age-related neuropathologies with CMBs in community-based older adults. Cerebral hemispheres from 289 participants of the Rush Memory and Aging Project, Religious Orders Study, Minority Aging Research Study, and Rush Alzheimer's Disease Clinical Core underwent ex vivo MRI and detailed neuropathologic examination. Following Bonferroni correction, CMBs in the cerebrum overall and in the frontal lobe were associated with cerebral amyloid angiopathy, CMBs in the frontal lobe were also associated with arteriolosclerosis, and CMBs in the basal ganglia showed a borderline significant association with microinfarcts. These findings suggest that CMBs can aid in the prediction of small vessel disease in community-based older adults. Finally, CMBs were not associated with dementia, suggesting that CMBs in community-based older adults may not be linked to substantial cognitive impairment.
Collapse
Affiliation(s)
- Grant Nikseresht
- Department of Computer Science, Illinois Institute of Technology, Chicago, IL, USA; Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, USA
| | - Arnold M Evia
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Sukriti Nag
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Sue E Leurgans
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Ana W Capuano
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Gady Agam
- Department of Computer Science, Illinois Institute of Technology, Chicago, IL, USA
| | - Lisa L Barnes
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Konstantinos Arfanakis
- Department of Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, USA; Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA; Department of Diagnostic Radiology, Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
21
|
Affleck AJ, Sachdev PS, Halliday GM. Past antihypertensive medication use is associated with lower levels of small vessel disease and lower Aβ plaque stage in the brains of older individuals. Neuropathol Appl Neurobiol 2023; 49:e12922. [PMID: 37431095 PMCID: PMC10947144 DOI: 10.1111/nan.12922] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 05/22/2023] [Accepted: 06/24/2023] [Indexed: 07/12/2023]
Abstract
AIMS This study assesses the association of antihypertensive medication use on the severities of neuropathological cerebrovascular disease (CVD excluding lobar infarction) in older individuals. METHODS Clinical and neuropathological data were retrieved for 149 autopsy cases >75 years old with or without CVD or Alzheimer's disease and no other neuropathological diagnoses. Clinical data included hypertension status, hypertension diagnosis, antihypertensive medication use, antihypertensive medication dose (where available) and clinical dementia rating (CDR). Neuropathological CVD severity was evaluated for differences with anti-hypertensive medication usage. RESULTS Antihypertensive medication use was associated with less severe white matter small vessel disease (SVD, mainly perivascular dilatation and rarefaction), with a 5.6-14.4 times greater likelihood of less severe SVD if medicated. No significant relationship was detected between infarction (presence, type, number and size), lacunes or cerebral amyloid angiopathy and antihypertensive medication use. Only increased white matter rarefaction/oedema and not perivascular dilation was associated with Alzheimer's pathology, with a 4.3 times greater likelihood of reduced Aβ progression through the brain if white matter rarefaction severity was none or mild. Antihypertensive medication use was associated with reduced Aβ progression but only in those with moderate to severe white matter SVD. CONCLUSIONS This histopathological study provides further evidence that antihypertensive medication use in older individuals is associated with white matter SVD and not with other CVD pathologies. This is mainly due to a reduction in white matter perivascular dilation and rarefaction/oedema. Even in those with moderate to severe white matter SVD, antihypertensive medication use reduced rarefaction and Aβ propagation through the brain.
Collapse
Affiliation(s)
- Andrew J. Affleck
- Neuroscience Research Australia (NeuRA)SydneyAustralia
- Centre for Health Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, Faculty of MedicineUniversity of New South WalesSydneyAustralia
| | - Perminder S. Sachdev
- Centre for Health Brain Ageing (CHeBA), Discipline of Psychiatry and Mental Health, Faculty of MedicineUniversity of New South WalesSydneyAustralia
- Neuropsychiatric InstituteThe Prince of Wales HospitalSydneyAustralia
| | - Glenda M. Halliday
- Neuroscience Research Australia (NeuRA)SydneyAustralia
- School of Medical Sciences, Faculty of MedicineUniversity of New South WalesSydneyAustralia
- Brain and Mind Centre & Faculty of Medicine and Health School of Medical SciencesUniversity of SydneySydneyAustralia
| |
Collapse
|
22
|
Tang B, Wang Y, Ren J. Basic information about memantine and its treatment of Alzheimer's disease and other clinical applications. IBRAIN 2023; 9:340-348. [PMID: 37786758 PMCID: PMC10527776 DOI: 10.1002/ibra.12098] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 10/04/2023]
Abstract
Memantine is a noncompetitive moderate-affinity strong voltage-dependent N-methyl-D-aspartate receptor antagonist. It has been used to treat Alzheimer's disease (AD) since 1989. In 2018, it became the second most commonly used drug for the treatment of dementia in the world. AD is nonreversible, and memantine can only relieve the symptoms of AD but not cure it. Over the past half-century, memantine's research and clinical application have been extensively developed. In this review, the basic composition of memantine, the mechanism and limitations of memantine in the treatment of AD, memantine combination therapy, comparison of memantine with other drugs for AD, and clinical studies of memantine in other diseases are reviewed to provide a valuable reference for further research and application of memantine for the treatment of AD.
Collapse
Affiliation(s)
- Bin‐Can Tang
- Department of AnesthesiologySouthwest Medical UniversityLuzhouChina
| | - Ya‐Ting Wang
- Department of AnesthesiologySouthwest Medical UniversityLuzhouChina
| | - Jie Ren
- Department of NeuroscienceThe University of SheffieldSheffieldUK
| |
Collapse
|
23
|
Ying C, Kang P, Binkley MM, Ford AL, Chen Y, Hassenstab J, Wang Q, Strain J, Morris JC, Lee JM, Benzinger TLS, An H. Neuroinflammation and amyloid deposition in the progression of mixed Alzheimer and vascular dementia. Neuroimage Clin 2023; 38:103373. [PMID: 36933348 PMCID: PMC10036862 DOI: 10.1016/j.nicl.2023.103373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 01/18/2023] [Accepted: 03/08/2023] [Indexed: 03/17/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) and vascular contributions to cognitive impairment and dementia (VCID) pathologies coexist in patients with cognitive impairment. Abnormal amyloid beta (Aβ) deposition is the hallmark pathologic biomarker for AD. Neuroinflammation may be a pathophysiological mechanism in both AD and VCID. In this study, we aimed to understand the role of neuroinflammation and Aβ deposition in white matter hyperintensities (WMH) progression and cognitive decline over a decade in patients with mixed AD and VCID pathologies. METHODS Twenty-four elderly participants (median [interquartile range] age 78 [64.8, 83] years old, 14 female) were recruited from the Knight Alzheimer Disease Research Center. 11C-PK11195 standard uptake value ratio (SUVR) and 11C-PiB mean cortical binding potential (MCBP) were used to evaluate neuroinflammation and Aβ deposition in-vivo, respectively. Fluid-attenuated inversion recovery MR images were acquired to obtain baseline WMH volume and its progression over 11.5 years. Composite cognitive scores (global, processing speed and memory) were computed at baseline and follow-up over 7.5 years. Multiple linear regression models evaluated the association between PET biomarkers (11C-PK11195 SUVR and 11C-PiB MCBP) and baseline WMH volume and cognitive function. Moreover, linear mixed-effects models evaluated whether PET biomarkers predicted greater WMH progression or cognitive decline over a decade. RESULTS Fifteen participants (62.5%) had mixed AD (positive PiB) and VCID (at least one vascular risk factor) pathologies. Elevated 11C-PK11195 SUVR, but not 11C-PiB MCBP, was associated with greater baseline WMH volume and predicted greater WMH progression. Elevated 11C-PiB MCBP was associated with baseline memory and global cognition. Elevated 11C-PK11195 SUVR and elevated 11C-PiB MCBP independently predicted greater global cognition and processing speed declines. No association was found between 11C-PK11195 SUVR and 11C-PiB MCBP. CONCLUSIONS Neuroinflammation and Aβ deposition may represent two distinct pathophysiological pathways, and both independently contributed to the progression of cognitive impairment in mixed AD and VCID pathologies. Neuroinflammation, but not Aβ deposition, contributed to WMH volume and progression.
Collapse
Affiliation(s)
- Chunwei Ying
- Department of Biomedical Engineering, Washington University in St. Louis, USA; Mallinckrodt Institute of Radiology, Washington University School of Medicine, USA
| | - Peter Kang
- Department of Neurology, Washington University School of Medicine, USA
| | - Michael M Binkley
- Department of Neurology, Washington University School of Medicine, USA
| | - Andria L Ford
- Department of Neurology, Washington University School of Medicine, USA; Mallinckrodt Institute of Radiology, Washington University School of Medicine, USA
| | - Yasheng Chen
- Department of Neurology, Washington University School of Medicine, USA
| | - Jason Hassenstab
- Department of Neurology, Washington University School of Medicine, USA; Knight Alzheimer Disease Research Center, Washington University School of Medicine, USA
| | - Qing Wang
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, USA; Knight Alzheimer Disease Research Center, Washington University School of Medicine, USA
| | - Jeremy Strain
- Department of Neurology, Washington University School of Medicine, USA
| | - John C Morris
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, USA
| | - Jin-Moo Lee
- Department of Biomedical Engineering, Washington University in St. Louis, USA; Department of Neurology, Washington University School of Medicine, USA; Mallinckrodt Institute of Radiology, Washington University School of Medicine, USA
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, USA; Knight Alzheimer Disease Research Center, Washington University School of Medicine, USA; Department of Neurosurgery, Washington University School of Medicine, USA
| | - Hongyu An
- Department of Biomedical Engineering, Washington University in St. Louis, USA; Department of Neurology, Washington University School of Medicine, USA; Mallinckrodt Institute of Radiology, Washington University School of Medicine, USA.
| |
Collapse
|
24
|
Toledo JB, Abdelnour C, Weil RS, Ferreira D, Rodriguez-Porcel F, Pilotto A, Wyman-Chick KA, Grothe MJ, Kane JPM, Taylor A, Rongve A, Scholz S, Leverenz JB, Boeve BF, Aarsland D, McKeith IG, Lewis S, Leroi I, Taylor JP. Dementia with Lewy bodies: Impact of co-pathologies and implications for clinical trial design. Alzheimers Dement 2023; 19:318-332. [PMID: 36239924 PMCID: PMC9881193 DOI: 10.1002/alz.12814] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 08/29/2022] [Accepted: 09/09/2022] [Indexed: 02/01/2023]
Abstract
Dementia with Lewy bodies (DLB) is clinically defined by the presence of visual hallucinations, fluctuations, rapid eye movement (REM) sleep behavioral disorder, and parkinsonism. Neuropathologically, it is characterized by the presence of Lewy pathology. However, neuropathological studies have demonstrated the high prevalence of coexistent Alzheimer's disease, TAR DNA-binding protein 43 (TDP-43), and cerebrovascular pathologic cases. Due to their high prevalence and clinical impact on DLB individuals, clinical trials should account for these co-pathologies in their design and selection and the interpretation of biomarkers values and outcomes. Here we discuss the frequency of the different co-pathologies in DLB and their cross-sectional and longitudinal clinical impact. We then evaluate the utility and possible applications of disease-specific and disease-nonspecific biomarkers and how co-pathologies can impact these biomarkers. We propose a framework for integrating multi-modal biomarker fingerprints and step-wise selection and assessment of DLB individuals for clinical trials, monitoring target engagement, and interpreting outcomes in the setting of co-pathologies.
Collapse
Affiliation(s)
- Jon B Toledo
- Nantz National Alzheimer Center, Stanley H. Appel Department of Neurology, Houston Methodist Hospital, Houston, Texas, USA
| | - Carla Abdelnour
- Fundació ACE. Barcelona Alzheimer Treatment and Research Center, Universitat Autónoma de Barcelona, Barcelona, Spain
| | - Rimona S Weil
- Dementia Research Centre, Wellcome Centre for Human Neuroimaging, Movement Disorders Consortium, National Hospital for Neurology and Neurosurgery, University College London, London, UK
| | - Daniel Ferreira
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer's Research, Karolinska Institutet, Stockholm, Sweden
| | | | - Andrea Pilotto
- Department of Clinical and Experimental Sciences, University of Brescia, Parkinson's Disease Rehabilitation Centre, FERB ONLUS-S, Isidoro Hospital, Trescore Balneario (BG), Italy
| | - Kathryn A Wyman-Chick
- HealthPartners Center for Memory and Aging and Struthers Parkinson's Center, Saint Paul, Minnesota, USA
| | - Michel J Grothe
- Instituto de Biomedicina de Sevilla (IBiS), Unidad de Trastornos del Movimiento, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Joseph P M Kane
- Centre for Public Health, Queen's University Belfast, Belfast, UK
| | - Angela Taylor
- Lewy Body Dementia Association, Lilburn, Georgia, USA
| | - Arvid Rongve
- Department of Research and Innovation, Institute of Clinical Medicine (K1), Haugesund Hospital, Norway and The University of Bergen, Bergen, Norway
| | - Sonja Scholz
- Department of Neurology, National Institute of Neurological Disorders and Stroke, Neurodegenerative Diseases Research Unit, Johns Hopkins University Medical Center, Baltimore, Maryland, USA
| | - James B Leverenz
- Lou Ruvo Center for Brain Health, Cleveland Clinic, Cleveland, Ohio, USA
| | - Bradley F Boeve
- Department of Neurology and Center for Sleep Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Dag Aarsland
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, UK
| | - Ian G McKeith
- Newcastle University Translational and Clinical Research Institute (NUTCRI, Newcastle upon Tyne, UK
| | - Simon Lewis
- ForeFront Parkinson's Disease Research Clinic, School of Medical Sciences, Brain and Mind Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Iracema Leroi
- Global Brain Health Institute, Trinity College Dublin, Dublin, Ireland
| | - John P Taylor
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
25
|
van Veluw SJ, Barkhof F, Schirmer MD. White Matter Hyperintensity Spatial Patterns Provide Clues About Underlying Disease: Location Matters! Neurology 2022; 99:1017-1018. [PMID: 36123129 DOI: 10.1212/wnl.0000000000201398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 08/30/2022] [Indexed: 11/15/2022] Open
Affiliation(s)
- Susanne J van Veluw
- From the Department of Neurology (S.J.v.V., M.D.S.), Massachusetts General Hospital/Harvard Medical School, Boston, MA; Department of Radiology and Nuclear Medicine (F.B.), Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands; and Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, UK.
| | - Frederik Barkhof
- From the Department of Neurology (S.J.v.V., M.D.S.), Massachusetts General Hospital/Harvard Medical School, Boston, MA; Department of Radiology and Nuclear Medicine (F.B.), Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands; and Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, UK
| | - Markus D Schirmer
- From the Department of Neurology (S.J.v.V., M.D.S.), Massachusetts General Hospital/Harvard Medical School, Boston, MA; Department of Radiology and Nuclear Medicine (F.B.), Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, The Netherlands; and Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, UK
| |
Collapse
|
26
|
Li J, Shen D, Zhou Y, Jin Y, Jin L, Ye X, Tong L, Gao F. Underlying microangiopathy and functional outcome of simultaneous multiple intracerebral hemorrhage. Front Aging Neurosci 2022; 14:1000573. [PMID: 36425320 PMCID: PMC9679501 DOI: 10.3389/fnagi.2022.1000573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/17/2022] [Indexed: 11/10/2022] Open
Abstract
Objective To identify the predominant type of cerebral small vessel disease (SVD) and outcomes in patients with simultaneous multiple intracerebral hemorrhages (SMICH). Methods Consecutive patients with intracerebral hemorrhage (ICH) from a single-center prospective cohort were retrospectively reviewed. Presumed etiology was classified according to the SMASH-U criteria. Demographics, clinical and laboratory variables, and neuroimaging data were compared between patients with primary SMICH and those with single ICH. Functional outcomes were assessed using the modified Rankin scale 90 days after ICH. Results Of the 598 enrolled patients, 37 (6.2%) met the criteria for SMICH. Risk factors for SMICH included a high burden of deep cerebral microbleeds (CMBs) (odds ratio [OR] 1.06, 95% confidence interval [CI], 1.00–1.12; p = 0.040), white matter hyperintensity scores (OR 1.27, 95% CI 1.04–1.57; p = 0.021), history of ICH (OR 3.38, 95% CI 1.31–8.05; p = 0.008), and low serum magnesium levels (OR 0.01, 95% CI 0.00–0.25; p = 0.007). Based on the SMASH-U classification, 15(40.5%) SMICH were classified as hypertension, whereas 17 (45.9%) as undetermined-etiology. To further explore the potential microangiopathy underlying undetermined-SMICH, these patients with undetermined-etiology were compared to those with cerebral amyloid angiopathy-ICH, and were associated with a higher burden of deep CMBs but less severe centrum semiovale enlarged perivascular spaces. Likewise, compared with hypertension-ICH patients, those with undetermined SMICH were consistently associated with a higher deep CMB counts. Moreover, multivariate analysis revealed that SMICH was independently associated with poor outcomes (OR 2.23, 95%CI 1.03–4.76; p = 0.038). Conclusion Our results suggest that most patients with primary SMICH harbor hypertensive-SVD as principal angiopathy. Patients with SMICH are at a high risk of poor outcomes. (ClinicalTrials.gov Identifier: NCT 04803292).
Collapse
|
27
|
Libecap TJ, Zachariou V, Bauer CE, Wilcock DM, Jicha GA, Raslau FD, Gold BT. Enlarged Perivascular Spaces Are Negatively Associated With Montreal Cognitive Assessment Scores in Older Adults. Front Neurol 2022; 13:888511. [PMID: 35847209 PMCID: PMC9283758 DOI: 10.3389/fneur.2022.888511] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 06/06/2022] [Indexed: 11/30/2022] Open
Abstract
Emerging evidence suggests that enlarged perivascular spaces (ePVS) may be a clinically significant neuroimaging marker of global cognitive function related to cerebral small vessel disease (cSVD). We tested this possibility by assessing the relationship between ePVS and both a standardized measure of global cognitive function, the Montreal Cognitive Assessment (MoCA), and an established marker of cSVD, white matter hyperintensity volume (WMH) volume. One hundred and eleven community-dwelling older adults (56-86) underwent neuroimaging and MoCA testing. Quantification of region-specific ePVS burden was performed using a previously validated visual rating method and WMH volumes were computed using the standard ADNI pipeline. Separate linear regression models were run with ePVS as a predictor of MoCA scores and whole brain WMH volume. Results indicated a negative association between MoCA scores and both total ePVS counts (P ≤ 0.001) and centrum semiovale ePVS counts (P ≤ 0.001), after controlling for other relevant cSVD variables. Further, WMH volumes were positively associated with total ePVS (P = 0.010), basal ganglia ePVS (P ≤ 0.001), and centrum semiovale ePVS (P = 0.027). Our results suggest that ePVS burden, particularly in the centrum semiovale, may be a clinically significant neuroimaging marker of global cognitive dysfunction related to cSVD.
Collapse
Affiliation(s)
- Timothy J. Libecap
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Valentinos Zachariou
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Christopher E. Bauer
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Donna M. Wilcock
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Gregory A. Jicha
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, United States
- Department of Neurology, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Flavius D. Raslau
- Department of Radiology, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Brian T. Gold
- Department of Neuroscience, College of Medicine, University of Kentucky, Lexington, KY, United States
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, United States
- Magnetic Resonance Imaging and Spectroscopy Center, College of Medicine, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
28
|
Rost NS, Brodtmann A, Pase MP, van Veluw SJ, Biffi A, Duering M, Hinman JD, Dichgans M. Post-Stroke Cognitive Impairment and Dementia. Circ Res 2022; 130:1252-1271. [PMID: 35420911 DOI: 10.1161/circresaha.122.319951] [Citation(s) in RCA: 290] [Impact Index Per Article: 96.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Poststroke cognitive impairment and dementia (PSCID) is a major source of morbidity and mortality after stroke worldwide. PSCID occurs as a consequence of ischemic stroke, intracerebral hemorrhage, or subarachnoid hemorrhage. Cognitive impairment and dementia manifesting after a clinical stroke is categorized as vascular even in people with comorbid neurodegenerative pathology, which is common in elderly individuals and can contribute to the clinical expression of PSCID. Manifestations of cerebral small vessel disease, such as covert brain infarcts, white matter lesions, microbleeds, and cortical microinfarcts, are also common in patients with stroke and likewise contribute to cognitive outcomes. Although studies of PSCID historically varied in the approach to timing and methods of diagnosis, most of them demonstrate that older age, lower educational status, socioeconomic disparities, premorbid cognitive or functional decline, life-course exposure to vascular risk factors, and a history of prior stroke increase risk of PSCID. Stroke characteristics, in particular stroke severity, lesion volume, lesion location, multiplicity and recurrence, also influence PSCID risk. Understanding the complex interaction between an acute stroke event and preexisting brain pathology remains a priority and will be critical for developing strategies for personalized prediction, prevention, targeted interventions, and rehabilitation. Current challenges in the field relate to a lack of harmonization of definition and classification of PSCID, timing of diagnosis, approaches to neurocognitive assessment, and duration of follow-up after stroke. However, evolving knowledge on pathophysiology, neuroimaging, and biomarkers offers potential for clinical applications and may inform clinical trials. Preventing stroke and PSCID remains a cornerstone of any strategy to achieve optimal brain health. We summarize recent developments in the field and discuss future directions closing with a call for action to systematically include cognitive outcome assessment into any clinical studies of poststroke outcome.
Collapse
Affiliation(s)
- Natalia S Rost
- J. Philip Kistler Stroke Research Center (N.S.R., S.J.v.V., A. Biffi), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Amy Brodtmann
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Australia (A. Brodtmann).,Turner Institute for Brain and Mental Health, Monash University, Melbourne, Australia (A. Brodtmann. M.P.P.)
| | - Matthew P Pase
- Turner Institute for Brain and Mental Health, Monash University, Melbourne, Australia (A. Brodtmann. M.P.P.).,Harvard T.H. Chan School of Public Health, Boston (M.P.P.)
| | - Susanne J van Veluw
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown (S.J.v.V.)
| | - Alessandro Biffi
- J. Philip Kistler Stroke Research Center (N.S.R., S.J.v.V., A. Biffi), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston.,Divisions of Memory Disorders and Behavioral Neurology (A. Biffi), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Marco Duering
- J. Philip Kistler Stroke Research Center (N.S.R., S.J.v.V., A. Biffi), Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston.,Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Germany (M. Duering, M. Dichgans).,Medical Image Analysis Center and Department of Biomedical Engineering, University of Basel, Switzerland (M. Duering)
| | - Jason D Hinman
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles (J.D.H.).,Department of Neurology, West Los Angeles VA Medical Center, CA (J.D.H.)
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Germany (M. Duering, M. Dichgans).,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany (M. Dichgans).,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (M. Dichgans)
| |
Collapse
|
29
|
Smith EE. What Turns the White Matter White? Metabolomic Clues to the Origin of Age-Related Cerebral White Matter Hyperintensities. Circulation 2022; 145:1053-1055. [PMID: 35377744 DOI: 10.1161/circulationaha.122.059281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Eric E Smith
- Department of Clinical Neurosciences and Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
30
|
Abstract
As life expectancy grows, brain health is increasingly seen as central to what we mean by successful aging-and vascular brain health as central to overall brain health. Cerebrovascular pathologies are highly prevalent independent contributors to age-related cognitive impairment and at least partly modifiable with available treatments. The current Focused Update addresses vascular brain health from multiple angles, ranging from pathophysiologic mechanisms and neuroimaging features to epidemiologic risk factors, social determinants, and candidate treatments. Here we highlight some of the shared themes that cut across these distinct perspectives: 1) the lifetime course of vascular brain injury pathogenesis and progression; 2) the scientific and ethical imperative to extend vascular brain health research in non-White and non-affluent populations; 3) the need for improved tools to study the cerebral small vessels themselves; 4) the potential role for brain recovery mechanisms in determining vascular brain health and resilience; and 5) the cross-pathway mechanisms by which vascular and neurodegenerative processes may interact. The diverse perspectives featured in this Focused Update offer a sense of the multidisciplinary approaches and collaborations that will be required to launch our populations towards improved brain health and successful aging.
Collapse
Affiliation(s)
- Steven M Greenberg
- J. Philip Kistler Stroke Research Center, Massachusetts General Hospital, Harvard Medical School, Boston
| |
Collapse
|