1
|
Batallas D, Gallego JJ, Casanova-Ferrer F, López-Gramaje A, Rivas-Diaz P, Megías J, Escudero-García D, Durbán L, Benlloch S, Urios A, Hidalgo V, Salvador A, Montoliu C. Sex differences in the mediating role of brain-derived neurotrophic factor between inflammation and memory in cirrhotic patients with minimal hepatic encephalopathy. Brain Behav Immun Health 2025; 46:100998. [PMID: 40343108 PMCID: PMC12060516 DOI: 10.1016/j.bbih.2025.100998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/12/2025] [Accepted: 04/21/2025] [Indexed: 05/11/2025] Open
Abstract
Minimal hepatic encephalopathy (MHE) affects attention, visuo-motor coordination, and visual perception, with mixed evidence on its impact on memory. Brain-derived neurotrophic factor (BDNF) is associated with memory dysfunction, and plays a crucial role in modulating neuroplasticity. This study investigates the mediating role of BDNF in the relationship between pro-inflammatory cytokines (IL-6, IL-15, IL-18), and declarative memory performance, and the moderating effects of sex. Sixty-eight cirrhotic patients and 22 healthy volunteers performed the Psychometric Hepatic Encephalopathy Score for MHE diagnosis and logical memory subtest (Wechsler Memory Scale-III). Moderated mediation analysis using bias-corrected bootstrapping and multiple regression was performed. Results showed that increased levels of IL-18 and IL-15 were significantly associated with lower BDNF levels (p = 0.03 and p = 0.02 respectively). However, no direct effect was observed between IL-18 and IL-15 and memory. The conditional effects of BDNF on memory were significant only for women with and without MHE, and lower BDNF levels were associated with lower memory performance (without MHE: p = 0.002; MHE: p = 0.001). Moreover, BDNF mediated indirectly the relationship between pro-inflammatory cytokines and memory. IL-18 and IL-15 impacted memory through reduced BDNF levels only in women with and without MHE, whereas IL-6 showed no significant effect on BDNF or memory across groups. These findings underscore the important role of BDNF in memory in cirrhotic patients, especially women with MHE, by mediating the IL-18 and IL-15 effects. The study highlights the role of IL-18 and IL-15 cytokines in neuroplasticity-related memory decline, positioning BDNF as a key biomarker for inflammation-associated cognitive impairment in this population.
Collapse
Affiliation(s)
- Daniela Batallas
- Laboratory of Social Cognitive Neuroscience, Department of Psychobiology and IDOCAL, University of Valencia, 46010, Valencia, Spain
| | - Juan José Gallego
- Fundación Investigación Hospital Clínico Universitario de Valencia. INCLIVA, 46010, Valencia, Spain
- Department of Pathology. University of Valencia, 46010, Valencia, Spain
| | - Franc Casanova-Ferrer
- Fundación Investigación Hospital Clínico Universitario de Valencia. INCLIVA, 46010, Valencia, Spain
| | - Adriá López-Gramaje
- Fundación Investigación Hospital Clínico Universitario de Valencia. INCLIVA, 46010, Valencia, Spain
- Department of Pathology. University of Valencia, 46010, Valencia, Spain
| | - Pablo Rivas-Diaz
- Laboratory of Social Cognitive Neuroscience, Department of Psychobiology and IDOCAL, University of Valencia, 46010, Valencia, Spain
| | - Javier Megías
- Department of Pathology. University of Valencia, 46010, Valencia, Spain
| | - Desamparados Escudero-García
- Servicio de Medicina Digestiva, Hospital Clínico Universitario de Valencia, Spain
- Departamento de Medicina. University of Valencia, 46010 Valencia, Spain
| | - Lucía Durbán
- Servicio de Medicina Digestiva, Hospital Arnau de Vilanova, 46015, Valencia, Spain
| | - Salvador Benlloch
- Servicio de Medicina Digestiva, Hospital Arnau de Vilanova, 46015, Valencia, Spain
- CIBERehd, Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Amparo Urios
- Fundación Investigación Hospital Clínico Universitario de Valencia. INCLIVA, 46010, Valencia, Spain
| | - Vanesa Hidalgo
- Laboratory of Social Cognitive Neuroscience, Department of Psychobiology and IDOCAL, University of Valencia, 46010, Valencia, Spain
- Department of Psychology and Sociology, Area of Psychobiology, University of Zaragoza, Teruel, Spain
| | - Alicia Salvador
- Laboratory of Social Cognitive Neuroscience, Department of Psychobiology and IDOCAL, University of Valencia, 46010, Valencia, Spain
- Spanish National Network for Research in Mental Health CIBERSAM, 28029, Madrid, Spain
| | - Carmina Montoliu
- Fundación Investigación Hospital Clínico Universitario de Valencia. INCLIVA, 46010, Valencia, Spain
- Department of Pathology. University of Valencia, 46010, Valencia, Spain
| |
Collapse
|
2
|
Aghetti A, Bouteloup V, Lebenberg J, Chupin M, Gourieux E, Mangin JF, Chêne G, Dufouil C, Jouvent E. Superficial white matter hyperintensities are associated with mild tissue alterations in vascular aging. Rev Neurol (Paris) 2025:S0035-3787(25)00501-6. [PMID: 40287332 DOI: 10.1016/j.neurol.2025.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 02/26/2025] [Accepted: 03/31/2025] [Indexed: 04/29/2025]
Abstract
In the elderly, white matter hyperintensities (WMH) are usually rated as periventricular or deep. However, recent data suggest that superficial WMH may be associated with distinct mechanisms and may be associated with milder underlying tissue alterations. We developed and validated a new grading scale to differentiate superficial WMH from other WMH (either periventricular or deep). We evaluated individuals with high loads of WMH from MEMENTO, a multicenter memory-clinic study, to evaluate the links between superficial WMH and 1) MRI markers of cerebral small vessel disease (number of lacunes and microbleeds and normalized brain volume); 2) cognitive outcomes including global evaluation with Mini Mental State Examination (MMSE). Our analytical sample included 208 participants. Participants with higher grades of superficial WMH had larger normalized brain volumes (82.1±1.3% vs 81.0±1.1%, P<0.001) and were more frequently women (85.0% vs 51.4%, P=0.01). In total contrast but as expected, participants with higher grades of other WMH were older (79.8±8.1 vs 75.5±6.2 years, P<0.001), had more often lacunes (41.7% vs 7.1%, P<0.001) and performed worse at the MMSE (26.8±2.0 vs 28.1±1.7, P=0.01). Our results support the hypothesis that superficial WMH are distinct from other WMH and probably correspond to mild tissue alterations.
Collapse
Affiliation(s)
- A Aghetti
- AP-HP, Lariboisière Hospital, Department of Neurology and FHU NeuroVasc, Université de Paris Cité, 75475 Paris, France; Université Paris Cité, Inserm, NeuroDiderot, 75019 Paris, France
| | - V Bouteloup
- INSERM, Bordeaux Population Health Research Center, UMR 1219, University Bordeaux, ISPED, CIC 1401-EC, Univ Bordeaux, 33000 Bordeaux, France; CHU de Bordeaux, Pole de Santé Publique, 33000 Bordeaux, France
| | - J Lebenberg
- Université Paris Cité, Inserm, NeuroDiderot, 75019 Paris, France; AP-HP, Lariboisière Hospital, Translational Neurovascular Centre, FHU NeuroVasc, Université de Paris Cité, 75475 Paris, France
| | - M Chupin
- ICM, Sorbonne Université, CNRS, Paris, France; CATI Multicenter Neuroimaging Platform
| | - E Gourieux
- ICM, Sorbonne Université, CNRS, Paris, France; CATI Multicenter Neuroimaging Platform
| | - J-F Mangin
- CATI Multicenter Neuroimaging Platform; Université Paris-Saclay, CEA, CNRS, Neurospin, Baobab, Gif-sur-Yvette, France
| | - G Chêne
- INSERM, Bordeaux Population Health Research Center, UMR 1219, University Bordeaux, ISPED, CIC 1401-EC, Univ Bordeaux, 33000 Bordeaux, France; CHU de Bordeaux, Pole de Santé Publique, 33000 Bordeaux, France
| | - C Dufouil
- INSERM, Bordeaux Population Health Research Center, UMR 1219, University Bordeaux, ISPED, CIC 1401-EC, Univ Bordeaux, 33000 Bordeaux, France; CHU de Bordeaux, Pole de Santé Publique, 33000 Bordeaux, France
| | - E Jouvent
- AP-HP, Lariboisière Hospital, Department of Neurology and FHU NeuroVasc, Université de Paris Cité, 75475 Paris, France; Université Paris Cité, Inserm, NeuroDiderot, 75019 Paris, France; Université Paris-Cité, Paris, France.
| |
Collapse
|
3
|
Ma Y, Yang Y, Wang X, Huang Y, Nan J, Feng J, Yan F, Han L. Prevalence and Risk Factors of Poststroke Cognitive Impairment: A Systematic Review and Meta-Analysis. Public Health Nurs 2025; 42:1047-1059. [PMID: 39702976 DOI: 10.1111/phn.13503] [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: 07/31/2024] [Accepted: 11/18/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND Stroke is a common disease that poses a significant threat to human health. Approximately one-third of stroke patients experience poststroke cognitive impairment (PSCI), which severely impacts their quality of life and survival time. Although the prevalence and risk factors for PSCI have been widely reported, these results have not been synthesized. OBJECTIVES This systematic review was conducted to explore the prevalence and risk factors of PSCI. METHODS PubMed, EMBASE, Web of Science, and Cochrane Library were comprehensively searched for studies exploring the prevalence and risk factors of PSCI from inception to July 5, 2022. RESULTS A total of 49 articles were included for meta-analysis. It was found that the combined prevalence of PSCI was 39%-47%. Risk factors for PSCI include female gender, age, education level less than 7 years, atrial fibrillation, diabetes, smoking, drinking, hypertension, coronary artery disease, carotid artery plaque, admission NIHSS score ≥ 5, unemployment, and homocysteine. CONCLUSIONS This systematic review has revealed a combined prevalence of PSCI is ranging from 39% to 47% and identified several risk factors for PSCI. These findings indicate a high incidence of the condition and underscore the need for increased public awareness. Future investigations should prioritize the identification of PSCI risk factors, providing a theoretical basis for nursing professionals to effectively manage and treat PSCI patients.
Collapse
Affiliation(s)
- Yuxia Ma
- School of Nursing, Evidence-Based Nursing Center, Lanzhou University, Lanzhou, China
- First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Yifang Yang
- School of Nursing, Evidence-Based Nursing Center, Lanzhou University, Lanzhou, China
| | - Xinyu Wang
- Nursing Department, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanan Huang
- Nursing Department, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Jinhan Nan
- School of Nursing, Evidence-Based Nursing Center, Lanzhou University, Lanzhou, China
| | - Juanjuan Feng
- School of Nursing, Evidence-Based Nursing Center, Lanzhou University, Lanzhou, China
| | - Fanghong Yan
- School of Nursing, Evidence-Based Nursing Center, Lanzhou University, Lanzhou, China
| | - Lin Han
- School of Nursing, Evidence-Based Nursing Center, Lanzhou University, Lanzhou, China
- Department of Nursing, Gansu Provincial Hospital, Lanzhou, China
| |
Collapse
|
4
|
Garcia FJ, Heiman M. Molecular and cellular characteristics of cerebrovascular cell types and their contribution to neurodegenerative diseases. Mol Neurodegener 2025; 20:13. [PMID: 39881338 PMCID: PMC11780804 DOI: 10.1186/s13024-025-00799-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 01/07/2025] [Indexed: 01/31/2025] Open
Abstract
Many diseases and disorders of the nervous system suffer from a lack of adequate therapeutics to halt or slow disease progression, and to this day, no cure exists for any of the fatal neurodegenerative diseases. In part this is due to the incredible diversity of cell types that comprise the brain, knowledge gaps in understanding basic mechanisms of disease, as well as a lack of reliable strategies for delivering new therapeutic modalities to affected areas. With the advent of single cell genomics, it is now possible to interrogate the molecular characteristics of diverse cell populations and their alterations in diseased states. More recently, much attention has been devoted to cell populations that have historically been difficult to profile with bulk single cell technologies. In particular, cell types that comprise the cerebrovasculature have become increasingly better characterized in normal and neurodegenerative disease contexts. In this review, we describe the current understanding of cerebrovasculature structure, function, and cell type diversity and its role in the mechanisms underlying various neurodegenerative diseases. We focus on human and mouse cerebrovasculature studies and discuss both origins and consequences of cerebrovascular dysfunction, emphasizing known cell type-specific vulnerabilities in neuronal and cerebrovascular cell populations. Lastly, we highlight how novel insights into cerebrovascular biology have impacted the development of modern therapeutic approaches and discuss outstanding questions in the field.
Collapse
Affiliation(s)
- Francisco J Garcia
- The Picower Institute for Learning and Memory, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Myriam Heiman
- The Picower Institute for Learning and Memory, Cambridge, MA, USA.
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA.
| |
Collapse
|
5
|
Fislage M, Chen CH, Cheng YW, Chen YF, Tang SC. Subcortical volumes and cognition in CADASIL - A pilot study. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2024; 7:100371. [PMID: 39493517 PMCID: PMC11530611 DOI: 10.1016/j.cccb.2024.100371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 08/21/2024] [Accepted: 10/08/2024] [Indexed: 11/05/2024]
Abstract
Background Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CADASIL) represents the most common heritable cause of vascular dementia. Subcortical volumes might be proxies of brain reserve capacity and reflective of cognitive function. We explored the impact of subcortical volumes on cognition in CADASIL patients. Methods We included 90 patients with pathogenic NOTCH3 variants from the Taiwan Associated Genetic and Nongenetic Small Vessel Disease cohort. They underwent MRI sessions at baseline. The volumes of the putamen, caudate, pallidum, thalamus, cerebellum, cortical gray matter and brain parenchymal fraction (BPF) were calculated by using FreeSurfer. We tested the association of the subcortical volumes, cortical gray matter volume and BPF with scores of the Mini-mental state examination (MMSE), cognitive domains, and the diagnosis of vascular cognitive impairment (VCI) which was defined as MMSE score <24. Results The thalamus and putamen were consistently associated with the MMSE (thalamus adjusted beta per SD decrement -1.41 [95 % CI -2.68-(-0.14)], p = 0.03; R²=0.25; putamen -1.93 [95 % CI -2.99-(-0.86)], p < 0.001; R²=0.36) and VCI (thalamus OR per SD-decrement 3.66 [95 % CI 1.38-9.72], p = 0.009), putamen (OR 3.06 [95 % CI 1.21-7.73], p = 0.02). A larger thalamus volume was also associated with better executive function and visuospatial perception. The cortical gray matter volume and the BPF showed associations with various cognitive outcomes in all analyses. Conclusion Although cortical gray matter volume and the BPF still appear to be robust markers of cognitive performance in CADASIL, the volumes of the thalamus and the putamen might also be promising regions of interest for future research.
Collapse
Affiliation(s)
- Marinus Fislage
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Anesthesiology and Operative Intensive Care Medicine (CCM, CVK), Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Chih-Hao Chen
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yu-Wen Cheng
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ya-Fang Chen
- Department of Medical Imaging, National Taiwan University Hospital, Taipei, Taiwan
| | - Sung-Chun Tang
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
6
|
Manco C, Cortese R, Leoncini M, Plantone D, Gentile G, Luchetti L, Zhang J, Di Donato I, Salvadori E, Poggesi A, Cosottini M, Mascalchi M, Federico A, Dotti MT, Battaglini M, Inzitari D, Pantoni L, De Stefano N. Hippocampal atrophy and white matter lesions characteristics can predict evolution to dementia in patients with vascular mild cognitive impairment. J Neurol Sci 2024; 464:123163. [PMID: 39128160 DOI: 10.1016/j.jns.2024.123163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/01/2024] [Accepted: 08/04/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND Vascular mild cognitive impairment (VMCI) is a transitional condition that may evolve into Vascular Dementia(VaD). Hippocampal volume (HV) is suggested as an early marker for VaD, the role of white matter lesions (WMLs) in neurodegeneration remains debated. OBJECTIVES Evaluate HV and WMLs as predictive markers of VaD in VMCI patients by assessing: (i)baseline differences in HV and WMLs between converters to VaD and non-converters, (ii) predictive power of HV and WMLs for VaD, (iii) associations between HV, WMLs, and cognitive decline, (iv)the role of WMLs on HV. METHODS This longitudinal multicenter study included 110 VMCI subjects (mean age:74.33 ± 6.63 years, 60males/50females) from the VMCI-Tuscany Study database. Subjects underwent brain MRI and cognitive testing, with 2-year follow-up data on VaD progression. HV and WMLs were semi-automatically segmented and measured. ANCOVA assessed group differences, while linear and logistic regression models evaluated predictive power. RESULTS After 2 years, 32/110 VMCI patients progressed to VaD. Converting patients had lower HV(p = 0.015) and higher lesion volumes in the posterior thalamic radiation (p = 0.046), splenium of the corpus callosum (p = 0.016), cingulate gyrus (p = 0.041), and cingulum hippocampus(p = 0.038). HV alone did not fully explain progression (p = 0.059), but combined with WMLs volume, the model was significant (p = 0.035). The best prediction model (p = 0.001) included total HV (p = 0.004) and total WMLs volume of the posterior thalamic radiation (p = 0.005) and cingulate gyrus (p = 0.005), achieving 80% precision, 81% specificity, and 74% sensitivity. Lower HV were linked to poorer performance on the Rey Auditory-Verbal Learning Test delayed recall (RAVLT) and Mini Mental State Examination (MMSE). CONCLUSIONS HV and WMLs are significant predictors of progression from VMCI to VaD. Lower HV correlate with worse cognitive performance on RAVLT and MMSE tests.
Collapse
Affiliation(s)
- Carlo Manco
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
| | - Rosa Cortese
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy.
| | | | - Domenico Plantone
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
| | - Giordano Gentile
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy; Siena Imaging SRL, 53100 Siena, Italy
| | - Ludovico Luchetti
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy; Siena Imaging SRL, 53100 Siena, Italy
| | | | | | - Emilia Salvadori
- Department of Biomedical and Clinical Sciences, University of Milano, Italy
| | - Anna Poggesi
- NEUROFARBA Department, Neuroscience Section, University of Florence, Florence, Italy
| | - Mirco Cosottini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Mario Mascalchi
- Department of Clinical and Experimental Biomedical Sciences -"Mario Serio", University of Florence, Florence, Italy
| | - Antonio Federico
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
| | - Maria Teresa Dotti
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
| | - Marco Battaglini
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy; Siena Imaging SRL, 53100 Siena, Italy
| | - Domenico Inzitari
- NEUROFARBA Department, Neuroscience Section, University of Florence, Florence, Italy
| | - Leonardo Pantoni
- Department of Biomedical and Clinical Sciences, University of Milano, Italy
| | - Nicola De Stefano
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
| |
Collapse
|
7
|
Yeshaw Y, Madakkatel I, Mulugeta A, Lumsden A, Hyppönen E. Uncovering Predictors of Low Hippocampal Volume: Evidence from a Large-Scale Machine-Learning-Based Study in the UK Biobank. Neuroepidemiology 2024; 58:369-382. [PMID: 38560977 PMCID: PMC11449190 DOI: 10.1159/000538565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 03/20/2024] [Indexed: 04/04/2024] Open
Abstract
INTRODUCTION Hippocampal atrophy is an established biomarker for conversion from the normal ageing process to developing cognitive impairment and dementia. This study used a novel hypothesis-free machine-learning approach, to uncover potential risk factors of lower hippocampal volume using information from the world's largest brain imaging study. METHODS A combination of machine learning and conventional statistical methods were used to identify predictors of low hippocampal volume. We run gradient boosting decision tree modelling including 2,891 input features measured before magnetic resonance imaging assessments (median 9.2 years, range 4.2-13.8 years) using data from 42,152 dementia-free UK Biobank participants. Logistic regression analyses were run on 87 factors identified as important for prediction based on Shapley values. False discovery rate-adjusted p value <0.05 was used to declare statistical significance. RESULTS Older age, male sex, greater height, and whole-body fat-free mass were the main predictors of low hippocampal volume with the model also identifying associations with lung function and lifestyle factors including smoking, physical activity, and coffee intake (corrected p < 0.05 for all). Red blood cell count and several red blood cell indices such as haemoglobin concentration, mean corpuscular haemoglobin, mean corpuscular volume, mean reticulocyte volume, mean sphered cell volume, and red blood cell distribution width were among many biomarkers associated with low hippocampal volume. CONCLUSION Lifestyles, physical measures, and biomarkers may affect hippocampal volume, with many of the characteristics potentially reflecting oxygen supply to the brain. Further studies are required to establish causality and clinical relevance of these findings.
Collapse
Affiliation(s)
- Yigizie Yeshaw
- Australian Centre for Precision Health, University of South Australia, Adelaide, South Australia, Australia,
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia,
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia,
- Department of Epidemiology and Biostatistics, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia,
| | - Iqbal Madakkatel
- Australian Centre for Precision Health, University of South Australia, Adelaide, South Australia, Australia
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Anwar Mulugeta
- Australian Centre for Precision Health, University of South Australia, Adelaide, South Australia, Australia
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Department of Pharmacology and Clinical Pharmacy, College of Health Sciences, Addis Ababa, Ethiopia
| | - Amanda Lumsden
- Australian Centre for Precision Health, University of South Australia, Adelaide, South Australia, Australia
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Elina Hyppönen
- Australian Centre for Precision Health, University of South Australia, Adelaide, South Australia, Australia
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| |
Collapse
|
8
|
Sakurai R, Pieruccini‐Faria F, Cornish B, Fraser J, Binns MA, Beaton D, Dilliott AA, Kwan D, Ramirez J, Tan B, Scott CJM, Sunderland KM, Tartaglia C, Finger E, Zinman L, Freedman M, McLaughlin PM, Swartz RH, Symons S, Lang AE, Bartha R, Black SE, Masellis M, Hegele RA, McIlroy W, Montero‐Odasso M. Link among apolipoprotein E E4, gait, and cognition in neurodegenerative diseases: ONDRI study. Alzheimers Dement 2024; 20:2968-2979. [PMID: 38470007 PMCID: PMC11032526 DOI: 10.1002/alz.13740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 12/08/2023] [Accepted: 12/14/2023] [Indexed: 03/13/2024]
Abstract
INTRODUCTION Apolipoprotein E E4 allele (APOE E4) and slow gait are independently associated with cognitive impairment and dementia. However, it is unknown whether their coexistence is associated with poorer cognitive performance and its underlying mechanism in neurodegenerative diseases. METHODS Gait speed, APOE E4, cognition, and neuroimaging were assessed in 480 older adults with neurodegeneration. Participants were grouped by APOE E4 presence and slow gait. Mediation analyses were conducted to determine if brain structures could explain the link between these factors and cognitive performance. RESULTS APOE E4 carriers with slow gait had the lowest global cognitive performance and smaller gray matter volumes compared to non-APOE E4 carriers with normal gait. Coexistence of APOE E4 and slow gait best predicted global and domain-specific poorer cognitive performances, mediated by smaller gray matter volume. DISCUSSION Gait slowness in APOE E4 carriers with neurodegenerative diseases may indicate extensive gray matter changes associated with poor cognition. HIGHLIGHTS APOE E4 and slow gait are risk factors for cognitive decline in neurodegenerative diseases. Slow gait and smaller gray matter volumes are associated, independently of APOE E4. Worse cognition in APOE E4 carriers with slow gait is explained by smaller GM volume. Gait slowness in APOE E4 carriers indicates poorer cognition-related brain changes.
Collapse
Affiliation(s)
- Ryota Sakurai
- Research Team for Social Participation and Healthy AgingTokyo Metropolitan Institute for Geriatrics and GerontologyItabashi‐kuTokyoJapan
- Gait & Brain Lab, St. Joseph' Health Care London, Lawson Health Research, Western University, Division of Geriatric MedicineLondonOntarioCanada
| | - Frederico Pieruccini‐Faria
- Gait & Brain Lab, St. Joseph' Health Care London, Lawson Health Research, Western University, Division of Geriatric MedicineLondonOntarioCanada
- Department of MedicineDivision of Geriatric MedicineParkwood HospitalWestern University, Parkwood InstituteLondonOntarioCanada
| | - Benjamin Cornish
- Neuroscience, Mobility and Balance Lab, Department of Kinesiology and Health SciencesUniversity of WaterlooWaterlooOntarioCanada
| | - Julia Fraser
- Neuroscience, Mobility and Balance Lab, Department of Kinesiology and Health SciencesUniversity of WaterlooWaterlooOntarioCanada
| | - Malcolm A. Binns
- Rotman Research Institute, Baycrest Health SciencesTorontoOntarioCanada
| | - Derek Beaton
- Data Science and Advanced Analytics, St. Michael's Hospital, Unity Health TorontoTorontoOntarioCanada
| | - Allison Ann Dilliott
- Department of Neurology and NeurosurgeryMontreal Neurological Institute, McGill UniversityMontréalQuebecCanada
| | - Donna Kwan
- Centre for Neuroscience Studies, Queen's UniversityKingstonOntarioCanada
| | - Joel Ramirez
- L.C. Campbell Cognitive Neurology Research Unit, Hurvitz Brain Sciences Program, Department of Medicine (Neurology)Sunnybrook Research Institute, Sunnybrook HSC, University of TorontoTorontoOntarioCanada
| | - Brian Tan
- Rotman Research Institute, Baycrest Health SciencesTorontoOntarioCanada
| | | | | | - Carmela Tartaglia
- Krembil Brain InstituteUniversity Health Network Memory Clinic, Toronto Western HospitalTorontoOntarioCanada
- Tanz Centre for Research in Neurodegenerative Diseases, University of TorontoTorontoOntarioCanada
| | - Elizabeth Finger
- Department of Clinical Neurological SciencesSchulich School of Medicine and Dentistry, Western UniversityLondonOntarioCanada
| | - Lorne Zinman
- Sunnybrook Research Institute, Sunnybrook Health Sciences CentreTorontoOntarioCanada
- Department of Medicine (Neurology)University of TorontoTorontoOntarioCanada
| | - Morris Freedman
- Rotman Research Institute, Baycrest Health SciencesTorontoOntarioCanada
- Department of Medicine (Neurology)University of TorontoTorontoOntarioCanada
- Division of NeurologyBaycrest Health SciencesTorontoOntarioCanada
| | - Paula M. McLaughlin
- Halifax Clinical Psychology Residency ProgramNova Scotia Health AuthorityHalifaxNova ScotiaCanada
| | - Richard H. Swartz
- Sunnybrook Research Institute, Sunnybrook Health Sciences CentreTorontoOntarioCanada
- Department of Medicine (Neurology)University of TorontoTorontoOntarioCanada
| | - Sean Symons
- L.C. Campbell Cognitive Neurology Research Unit, Hurvitz Brain Sciences Program, Department of Medicine (Neurology)Sunnybrook Research Institute, Sunnybrook HSC, University of TorontoTorontoOntarioCanada
| | - Anthony E. Lang
- Division of NeurologyDepartment of MedicineEdmond J Safra Program in Parkinson's Disease and Morton and Gloria Shulman Movement Disorders ClinicToronto Western HospitalUniversity of TorontoTorontoOntarioCanada
| | - Robert Bartha
- Department of Medical BiophysicsSchulich School of Medicine and Dentistry, Robarts Research Institute, Western UniversityLondonOntarioCanada
| | - Sandra E. Black
- L.C. Campbell Cognitive Neurology Research Unit, Hurvitz Brain Sciences Program, Department of Medicine (Neurology)Sunnybrook Research Institute, Sunnybrook HSC, University of TorontoTorontoOntarioCanada
| | - Mario Masellis
- L.C. Campbell Cognitive Neurology Research Unit, Hurvitz Brain Sciences Program, Department of Medicine (Neurology)Sunnybrook Research Institute, Sunnybrook HSC, University of TorontoTorontoOntarioCanada
| | - Robert A. Hegele
- Schulich School of Medicine and Dentistry, Western UniversityLondonOntarioCanada
- Robarts Research Institute, Western UniversityLondonOntarioCanada
| | - William McIlroy
- Neuroscience, Mobility and Balance Laboratory, Department of Kinesiology and Health SciencesUniversity of WaterlooWaterlooOntarioCanada
| | - ONDRI Investigators
- Research Team for Social Participation and Healthy AgingTokyo Metropolitan Institute for Geriatrics and GerontologyItabashi‐kuTokyoJapan
| | - Manuel Montero‐Odasso
- Gait & Brain Lab, St. Joseph' Health Care London, Lawson Health Research, Western University, Division of Geriatric MedicineLondonOntarioCanada
- Gait and Brain Lab, Division of Geriatric Medicineand Lawson Health Research InstituteParkwood Institute, Western UniversityLondonOntarioCanada
- Division of Geriatric MedicineDepartment of MedicineSchulich School of Medicine and Dentistry, Western University, Parkwood InstituteLondonOntarioCanada
| |
Collapse
|
9
|
Perosa V, Zanon Zotin MC, Schoemaker D, Sveikata L, Etherton MR, Charidimou A, Greenberg SM, Viswanathan A. Association Between Hippocampal Volumes and Cognition in Cerebral Amyloid Angiopathy. Neurology 2024; 102:e207854. [PMID: 38165326 PMCID: PMC10870737 DOI: 10.1212/wnl.0000000000207854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 10/03/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Accumulating evidence suggests that gray matter atrophy, often considered a marker of Alzheimer disease (AD), can also result from cerebral small vessel disease (CSVD). Cerebral amyloid angiopathy (CAA) is a form of sporadic CSVD, diagnosed through neuroimaging criteria, that often co-occurs with AD pathology and leads to cognitive impairment. We sought to identify the role of hippocampal integrity in the development of cognitive impairment in a cohort of patients with possible and probable CAA. METHODS Patients were recruited from an ongoing CAA study at Massachusetts General Hospital. Composite scores defined performance in the cognitive domains of memory, language, executive function, and processing speed. Hippocampal subfields' volumes were measured from 3T MRI, using an automated method, and multivariate linear regression models were used to estimate their association with each cognitive domain and relationship to CAA-related neuroimaging markers. RESULTS One hundred twenty patients, 36 with possible (age mean [range]: 75.6 [65.6-88.9]), 67 with probable CAA (75.9 [59.0-94.0]), and 17 controls without cognitive impairment and CSVD (72.4 [62.5-82.7]; 76.4% female patients), were included in this study. We found a positive association between all investigated hippocampal subfields and memory and language, whereas specific subfields accounted for executive function (CA4 [Estimate = 5.43; 95% CI 1.26-9.61; p = 0.020], subiculum [Estimate = 2.85; 95% CI 0.67-5.02; p = 0.022]), and processing speed (subiculum [Estimate = 1.99; 95% CI 0.13-3.85; p = 0.036]). These findings were independent of other CAA-related markers, which did not have an influence on cognition in this cohort. Peak width of skeletonized mean diffusivity (PSMD), a measure of white matter integrity, was negatively associated with hippocampal subfields' volumes (CA3 [Estimate = -0.012; 95% CI -0.020 to -0.004; p = 0.034], CA4 [Estimate = -0.010; 95% CI -0.020 to -0.0007; p = 0.037], subiculum [Estimate = -0.019; 95% CI -0.042 to -0.0001; p = 0.003]). DISCUSSION These results suggest that hippocampal integrity is an independent contributor to cognitive impairment in patients with CAA and that it might be related to loss of integrity in the white matter. Further studies exploring potential causes and directionality of the relationship between white matter and hippocampal integrity may be warranted.
Collapse
Affiliation(s)
- Valentina Perosa
- From the J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Maria Clara Zanon Zotin
- From the J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Dorothee Schoemaker
- From the J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Lukas Sveikata
- From the J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Mark R Etherton
- From the J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Andreas Charidimou
- From the J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Steven M Greenberg
- From the J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Anand Viswanathan
- From the J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston
| |
Collapse
|
10
|
Cheng H, Teng J, Jia L, Xu L, Yang F, Li H, Ling C, Liu W, Li J, Li Y, Guo Z, Geng X, Guo J, Zhang D. Association between morphologic features of intracranial distal arteries and brain atrophy indexes in cerebral small vessel disease: a voxel-based morphometry study. Front Neurol 2023; 14:1198402. [PMID: 37396753 PMCID: PMC10313400 DOI: 10.3389/fneur.2023.1198402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 05/24/2023] [Indexed: 07/04/2023] Open
Abstract
Background Brain atrophy represents a final common pathway for pathological processes in patients with cerebral small vessel disease (CSVD) and is now recognized as a strong independent predictor of clinical status and progression. The mechanism underlying brain atrophy in patients with CSVD is not yet fully comprehended. This study aims to investigate the association of morphologic features of intracranial distal arteries (A2, M2, P2 and more distal) with different brain structures [gray matter volume (GMV), white matter volume (WMV), and cerebrospinal fluid volume (CSFV)]. Furthermore, we also examined whether a correlation existed between these cerebrovascular characteristics and GMV in different brain regions. Method A total of 39 participants were eventually enrolled. The morphologic features of intracranial distal arteries based on TOF-MRA were extracted and quantified using the intracranial artery feature extraction technique (iCafe). The brain 3D-T1 images were segmented into gray matter (GM), white matter (WM), and cerebrospinal fluid (CSF) using the "Segment" tool in CAT12 for the voxel-based morphometry (VBM) analysis. Univariable and multivariable linear regression models were used to investigate the relationship between these cerebrovascular features and different brain structures. Partial correlation analysis with a one-tailed method was used to evaluate the relationship between these cerebrovascular features and GMV in different brain regions. Results Our findings indicate that both distal artery length and density were positively correlated with GM fraction in CSVD patients, regardless of whether univariable or multivariable linear regression analyses were performed. In addition, distal artery length (β = -0.428, p = 0.007) and density (β = -0.337, p = 0.036) were also found to be negative associated with CSF fraction, although this relationship disappeared after adjusting for potential confounders. Additional adjustment for the effect of WMHs volume did not change these results. In subgroup anasysis, we found that participants in the highest distal artery length tertile had significantly higher GM fraction and lower CSF fraction level than participants in the lowest distal artery length tertile. In partial correlation analysis, we also found that these cerebrovascular characteristics associated with regional GMV, especially subcortical nuclear. Conclusion The morphologic features of intracranial distal arteries, including artery length, density and average tortuosity, measured from 3D-TOF MRA, are associated with generalized or focal atrophy indexes of CSVD.
Collapse
Affiliation(s)
- Hongjiang Cheng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Junfang Teng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Longbin Jia
- Department of Neurology, Jincheng People’s Hospital, Jincheng, Shanxi, China
| | - Lina Xu
- Department of Neurology, Jincheng People’s Hospital, Jincheng, Shanxi, China
| | - Fengbing Yang
- Department of Neurology, Jincheng People’s Hospital, Jincheng, Shanxi, China
| | - Huimin Li
- Department of Neurology, Jincheng People’s Hospital, Jincheng, Shanxi, China
| | - Chen Ling
- Graduate School, Changzhi Medical College, Changzhi, Shanxi, China
| | - Wei Liu
- Department of Neurology, Jincheng People’s Hospital, Jincheng, Shanxi, China
| | - Jinna Li
- Department of Neurology, Jincheng People’s Hospital, Jincheng, Shanxi, China
| | - Yujuan Li
- Department of Neurology, Jincheng People’s Hospital, Jincheng, Shanxi, China
| | - Zixuan Guo
- Department of Neurology, Jincheng People’s Hospital, Jincheng, Shanxi, China
| | - Xia Geng
- Department of Neurology, Jincheng People’s Hospital, Jincheng, Shanxi, China
| | - Jiaying Guo
- Department of Neurology, Jincheng People’s Hospital, Jincheng, Shanxi, China
| | - Dandan Zhang
- Department of Neurology, Jincheng People’s Hospital, Jincheng, Shanxi, China
| |
Collapse
|
11
|
Cheng YW, Chao CC, Chen CH, Yeh TY, Jeng JS, Tang SC, Hsieh ST. Small Fiber Pathology in CADASIL: Clinical Correlation With Cognitive Impairment. Neurology 2022; 99:e583-e593. [PMID: 35584924 PMCID: PMC9442619 DOI: 10.1212/wnl.0000000000200672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/16/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES This study investigated the cutaneous small fiber pathology of cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) and its clinical significance, that is, the NOTCH3 deposition in cutaneous vasculatures and CNS neurodegeneration focusing on cognitive impairment. METHODS Thirty-seven patients with CADASIL and 59 age-matched healthy controls were enrolled to evaluate cutaneous small fiber pathology by quantitative measures of intraepidermal nerve fiber density (IENFD), sweat gland innervation, and vascular innervation. Cognitive performance of patients with CADASIL was evaluated by a comprehensive neuropsychological assessment, and its association with small fiber pathology was tested using multivariable linear regression analysis adjusted for age and diabetes mellitus. We further assessed the relationships of IENFD with cutaneous vascular NOTCH3 ectodomain (NOTCH3ECD) deposition and biomarkers of neurodegeneration including structural brain MRI measures, serum neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP), tau, and ubiquitin carboxy-terminal hydrolase L1. RESULTS Patients with CADASIL showed reduced IENFD (5.22 ± 2.42 vs 7.88 ± 2.89 fibers/mm, p = 0.0001) and reduced sweat gland (p < 0.0001) and vascular (p < 0.0001) innervations compared with age-matched controls. Reduced IENFD was associated with impaired global cognition measured by Mini-Mental State Examination (B = 1.062, 95% CI = 0.370-1.753, p = 0.004), and this association remained after adjustment for age and diabetes mellitus (p = 0.043). In addition, IENFD in patients with CADASIL was associated with mean cortical thickness (Pearson r = 0.565, p = 0.0023) but not white matter hyperintensity volume, total lacune count, or total microbleed count. Reduced IENFD was associated with cutaneous vascular NOTCH3ECD deposition amount among patients harboring pathogenic variants in exon 11 (mainly p.R544C) (B = -0.092, 95% CI = -0.175 to -0.009, p = 0.031). Compared with those with normal cognition, patients with CADASIL with cognitive impairment had an elevated plasma NfL level regardless of concurrent small fiber denervation, whereas only patients with both cognitive impairment and small fiber denervation showed an elevated plasma GFAP level. DISCUSSION Cutaneous small fiber pathology correlates with cognitive impairment and CNS neurodegeneration in patients with CADASIL, indicating a peripheral neurodegenerative process related to NOTCH3ECD aggregation.
Collapse
Affiliation(s)
- Yu-Wen Cheng
- From the Department of Neurology (Y.-W.C.), National Taiwan University Hospital, Hsin-Chu Branch; Graduate Institute of Clinical Medicine (Y.-W.C.), College of Medicine, National Taiwan University; Department of Neurology (C.-C.C., C.-H.C., J.-S.J., S.-C.T., S.-T.H.), National Taiwan University Hospital; Department of Anatomy and Cell Biology (T.-Y.Y., S.-T.H.), College of Medicine, National Taiwan University; Graduate Institute of Brain and Mind Sciences (S.-T.H.), College of Medicine, National Taiwan University; and Center of Precision Medicine (S.-T.H.), National Taiwan University College of Medicine, Taipei
| | - Chi-Chao Chao
- From the Department of Neurology (Y.-W.C.), National Taiwan University Hospital, Hsin-Chu Branch; Graduate Institute of Clinical Medicine (Y.-W.C.), College of Medicine, National Taiwan University; Department of Neurology (C.-C.C., C.-H.C., J.-S.J., S.-C.T., S.-T.H.), National Taiwan University Hospital; Department of Anatomy and Cell Biology (T.-Y.Y., S.-T.H.), College of Medicine, National Taiwan University; Graduate Institute of Brain and Mind Sciences (S.-T.H.), College of Medicine, National Taiwan University; and Center of Precision Medicine (S.-T.H.), National Taiwan University College of Medicine, Taipei
| | - Chih-Hao Chen
- From the Department of Neurology (Y.-W.C.), National Taiwan University Hospital, Hsin-Chu Branch; Graduate Institute of Clinical Medicine (Y.-W.C.), College of Medicine, National Taiwan University; Department of Neurology (C.-C.C., C.-H.C., J.-S.J., S.-C.T., S.-T.H.), National Taiwan University Hospital; Department of Anatomy and Cell Biology (T.-Y.Y., S.-T.H.), College of Medicine, National Taiwan University; Graduate Institute of Brain and Mind Sciences (S.-T.H.), College of Medicine, National Taiwan University; and Center of Precision Medicine (S.-T.H.), National Taiwan University College of Medicine, Taipei
| | - Ti-Yen Yeh
- From the Department of Neurology (Y.-W.C.), National Taiwan University Hospital, Hsin-Chu Branch; Graduate Institute of Clinical Medicine (Y.-W.C.), College of Medicine, National Taiwan University; Department of Neurology (C.-C.C., C.-H.C., J.-S.J., S.-C.T., S.-T.H.), National Taiwan University Hospital; Department of Anatomy and Cell Biology (T.-Y.Y., S.-T.H.), College of Medicine, National Taiwan University; Graduate Institute of Brain and Mind Sciences (S.-T.H.), College of Medicine, National Taiwan University; and Center of Precision Medicine (S.-T.H.), National Taiwan University College of Medicine, Taipei
| | - Jiann-Shing Jeng
- From the Department of Neurology (Y.-W.C.), National Taiwan University Hospital, Hsin-Chu Branch; Graduate Institute of Clinical Medicine (Y.-W.C.), College of Medicine, National Taiwan University; Department of Neurology (C.-C.C., C.-H.C., J.-S.J., S.-C.T., S.-T.H.), National Taiwan University Hospital; Department of Anatomy and Cell Biology (T.-Y.Y., S.-T.H.), College of Medicine, National Taiwan University; Graduate Institute of Brain and Mind Sciences (S.-T.H.), College of Medicine, National Taiwan University; and Center of Precision Medicine (S.-T.H.), National Taiwan University College of Medicine, Taipei
| | - Sung-Chun Tang
- From the Department of Neurology (Y.-W.C.), National Taiwan University Hospital, Hsin-Chu Branch; Graduate Institute of Clinical Medicine (Y.-W.C.), College of Medicine, National Taiwan University; Department of Neurology (C.-C.C., C.-H.C., J.-S.J., S.-C.T., S.-T.H.), National Taiwan University Hospital; Department of Anatomy and Cell Biology (T.-Y.Y., S.-T.H.), College of Medicine, National Taiwan University; Graduate Institute of Brain and Mind Sciences (S.-T.H.), College of Medicine, National Taiwan University; and Center of Precision Medicine (S.-T.H.), National Taiwan University College of Medicine, Taipei
| | - Sung-Tsang Hsieh
- From the Department of Neurology (Y.-W.C.), National Taiwan University Hospital, Hsin-Chu Branch; Graduate Institute of Clinical Medicine (Y.-W.C.), College of Medicine, National Taiwan University; Department of Neurology (C.-C.C., C.-H.C., J.-S.J., S.-C.T., S.-T.H.), National Taiwan University Hospital; Department of Anatomy and Cell Biology (T.-Y.Y., S.-T.H.), College of Medicine, National Taiwan University; Graduate Institute of Brain and Mind Sciences (S.-T.H.), College of Medicine, National Taiwan University; and Center of Precision Medicine (S.-T.H.), National Taiwan University College of Medicine, Taipei.
| |
Collapse
|
12
|
Jolly AA, Nannoni S, Edwards H, Morris RG, Markus HS. Prevalence and Predictors of Vascular Cognitive Impairment in Patients With CADASIL. Neurology 2022; 99:e453-e461. [PMID: 35606149 PMCID: PMC9421594 DOI: 10.1212/wnl.0000000000200607] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 03/11/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is the most common monogenic form of stroke and early-onset dementia. We determined the prevalence of vascular cognitive impairment (VCI) in a group of patients with CADASIL and investigated which factors were associated with VCI risk, including clinical, genetic, and MRI parameters. METHODS Cognition was assessed in patients with genetically confirmed CADASIL (n = 176) and healthy controls (n = 265) (mean [SD] age 50.95 [11.35] vs 52.37 [7.93] years) using the Brief Memory and Executive Test (BMET) and the Montreal Cognitive Assessment (MoCA). VCI was defined according to previously validated cutoffs. We determined the prevalence of VCI and its associations with clinical risk factors, mutation location (epidermal growth factor-like repeats [EGFr] 1-6 vs EGFr 7-34), and MRI markers of small vessel disease. RESULTS VCI was more common in patients with CADASIL than in controls; 39.8 vs 10.2% on the BMET and 47.7% vs 19.6% on the MOCA. Patients with CADASIL had worse performance across all cognitive domains. A history of stroke was associated with VCI on the BMET (OR 2.12, 95% CI [1.05, 4.27] p = 0.04) and MoCA (OR 2.55 [1.21, 5.41] p = 0.01), after controlling for age and sex. There was no association of VCI with mutation site. Lacune count was the only MRI parameter independently associated with VCI on the BMET (OR: 1.63, 95% CI [1.10, 2.41], p = 0.014), after controlling for other MRI parameters. These associations persisted after controlling for education in the sensitivity analyses. DISCUSSION VCI is present in almost half of the patients with CADASIL with a mean age of 50 years. Stroke and lacune count on MRI were both independent predictors of VCI on the BMET.
Collapse
Affiliation(s)
- Amy A Jolly
- From the Stroke Research Group, Department of Clinical Neurosciences (A.A.J., S.N., H.E., H.S.M.), University of Cambridge, Cambridge Biomedical Campus, United Kingdom; and Department of Psychology (R.G.M.), King's College Institute of Psychiatry, Psychology and Neurosciences, Institute of Psychiatry, London, United Kingdom.
| | - Stefania Nannoni
- From the Stroke Research Group, Department of Clinical Neurosciences (A.A.J., S.N., H.E., H.S.M.), University of Cambridge, Cambridge Biomedical Campus, United Kingdom; and Department of Psychology (R.G.M.), King's College Institute of Psychiatry, Psychology and Neurosciences, Institute of Psychiatry, London, United Kingdom
| | - Hayley Edwards
- From the Stroke Research Group, Department of Clinical Neurosciences (A.A.J., S.N., H.E., H.S.M.), University of Cambridge, Cambridge Biomedical Campus, United Kingdom; and Department of Psychology (R.G.M.), King's College Institute of Psychiatry, Psychology and Neurosciences, Institute of Psychiatry, London, United Kingdom
| | - Robin G Morris
- From the Stroke Research Group, Department of Clinical Neurosciences (A.A.J., S.N., H.E., H.S.M.), University of Cambridge, Cambridge Biomedical Campus, United Kingdom; and Department of Psychology (R.G.M.), King's College Institute of Psychiatry, Psychology and Neurosciences, Institute of Psychiatry, London, United Kingdom
| | - Hugh S Markus
- From the Stroke Research Group, Department of Clinical Neurosciences (A.A.J., S.N., H.E., H.S.M.), University of Cambridge, Cambridge Biomedical Campus, United Kingdom; and Department of Psychology (R.G.M.), King's College Institute of Psychiatry, Psychology and Neurosciences, Institute of Psychiatry, London, United Kingdom
| |
Collapse
|
13
|
Chen CH, Chu YT, Chen YF, Ko TY, Cheng YW, Lee MJ, Chen PL, Tang SC, Jeng JS. Comparison of clinical and neuroimaging features between NOTCH3 mutations and nongenetic spontaneous intracerebral haemorrhage. Eur J Neurol 2022; 29:3243-3254. [PMID: 35781912 DOI: 10.1111/ene.15485] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/25/2022] [Accepted: 06/29/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND The NOTCH3 mutation is a common cause of hereditary cerebral small vessel disease (CSVD) and may be a cause of spontaneous intracerebral haemorrhage (ICH). We aimed to investigate the clinical/imaging features for identifying the NOTCH3 mutations related ICH. METHODS The study was based on a cohort of 749 CSVD patients in Taiwan who received next-generation sequencing of CSVD genes including NOTCH3. Patients with history of ICH (n=206) were included for analysis. The CSVD neuroimaging markers were compared between the patients with NOTCH3 and without known genetic mutations. RESULTS After excluding the patients with other causes of ICH (structural lesions, systemic/medication-related, or amyloid angiopathy) and those without neuroimaging, 45 NOTCH3 mutation patients and 109 nongenetic ICH patients were included. The NOTCH3 mutation patients were more likely to have thalamic haemorrhage, a family history of stroke, and more severe CSVD neuroimaging markers. A five-point NOTCH3-ICH score was constructed and consisted of: history of stroke in siblings, thalamic haemorrhage, any deep nuclei lacunae, any hippocampal cerebral microbleed (CMB), and a thalamic CMB >5 (one point for each). A score ≥2 had a sensitivity of 88.9% and a specificity of 64.2% in identifying the NOTCH3 mutation. The NOTCH3 mutation patients had a higher risk of recurrent stroke (9.1 vs. 4.5 per 100 person-years; log-rank p = 0.03) during follow-up. CONCLUSION The patients with NOTCH3 mutation-related ICH had a higher burden of CMB in the hippocampus/thalamus and a higher recurrent stroke risk. The NOTCH3-ICH score may assist identifying genetic causes of ICH.
Collapse
Affiliation(s)
- Chih-Hao Chen
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yung-Tsai Chu
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ya-Fang Chen
- Department of Medical Imaging, National Taiwan University Hospital, Taipei, Taiwan
| | - Tzu-Yu Ko
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Wen Cheng
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan.,Department of Neurology, National Taiwan University Hospital Hsinchu Branch, Hsinchu, Taiwan
| | - Ming-Jen Lee
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan.,Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | - Pei-Lung Chen
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, National Taiwan University, Taipei, Taiwan
| | - Sung-Chun Tang
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Jiann-Shing Jeng
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
14
|
Sun W, Huang L, Cheng Y, Qin R, Xu H, Shao P, Ma J, Yao Z, Shi L, Xu Y. Medial Temporal Atrophy Contributes to Cognitive Impairment in Cerebral Small Vessel Disease. Front Neurol 2022; 13:858171. [PMID: 35665031 PMCID: PMC9159509 DOI: 10.3389/fneur.2022.858171] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 04/14/2022] [Indexed: 11/18/2022] Open
Abstract
Background The role of brain atrophy in cognitive decline related to cerebral small vessel disease (CSVD) remains unclear. This study used AccuBrain™ to identify major CSVD-related brain changes and verified the relationship between brain atrophy and different cognition domains in CSVD patients. Methods All enrolled 242 CSVD patients and 76 healthy participants underwent magnetic resonance imaging examinations and detailed neuropsychological scale assessments were collected at the same time. The AccuBrain™ technology was applied to fully automated image segmentation, measurement, and calculation of the acquired imaging results to obtain the volumes of different brain partitions and the volume of WMH for quantitative analysis. Correlation analyses were used to estimate the relationship between MRI features and different cognitive domains. Multifactor linear regression models were performed to analyze independent predictors of MTA and cognitive decline. Results CSVD patients exhibited multiple gray matter nucleus volume decreases in the basal ganglia regions and brain lobes, including the temporal lobe (P = 0.019), especially in the medial temporal lobe (p < 0.001), parietal lobe (p = 0.013), and cingulate lobe (p = 0.036) compare to HC. The volume of PWMH was an independent predictor of MTA for CSVD patients. Both medial temporal atrophy (MTA) and PWMH were associated with cognition impairment in CSVD-CI patients. MTA mediated the effect of PWMH on executive function in CSVD-CI patients. Conclusions Our results showed that MTA was related to cognition impairment in CSVD patients, which might become a potential imaging marker for CSVD-CI.
Collapse
Affiliation(s)
- Wenshan Sun
- Department of Neurology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Jiangsu Key Laboratory for Molecular Medicine, Nanjing University, Nanjing, China
- Department of Neurology, Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Lili Huang
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Jiangsu Key Laboratory for Molecular Medicine, Nanjing University, Nanjing, China
| | - Yue Cheng
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Jiangsu Key Laboratory for Molecular Medicine, Nanjing University, Nanjing, China
| | - Ruomeng Qin
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Jiangsu Key Laboratory for Molecular Medicine, Nanjing University, Nanjing, China
| | - Hengheng Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Jiangsu Key Laboratory for Molecular Medicine, Nanjing University, Nanjing, China
| | - Pengfei Shao
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Jiangsu Key Laboratory for Molecular Medicine, Nanjing University, Nanjing, China
| | - Junyi Ma
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Jiangsu Key Laboratory for Molecular Medicine, Nanjing University, Nanjing, China
| | - Zhelv Yao
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Jiangsu Key Laboratory for Molecular Medicine, Nanjing University, Nanjing, China
| | - Lin Shi
- Department of Imaging and Interventional Radiology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- BrainNow Research Institute, Hong Kong Science and Technology Park, Hong Kong, Hong Kong SAR, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Jiangsu Key Laboratory for Molecular Medicine, Nanjing University, Nanjing, China
- *Correspondence: Yun Xu
| |
Collapse
|
15
|
Li P, Huang Q, Ban S, Qiao Y, Wu J, Zhai Y, Du X, Hua F, Su J. Altered Default Mode Network Is Associated With Cognitive Impairment in CADASIL as Revealed by Multimodal Neu roimaging. Front Neurol 2021; 12:735033. [PMID: 34938255 PMCID: PMC8685443 DOI: 10.3389/fneur.2021.735033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 10/08/2021] [Indexed: 11/13/2022] Open
Abstract
Background and Purpose: Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy caused by mutations in the NOTCH3 gene is a hereditary cerebral small vessel disease, manifesting with stroke, cognitive impairment, and mood disturbances. Functional or structural changes in the default mode network (DMN), which plays important role in cognitive and mental maintenance, have been found in several neurological and mental diseases. However, it remains unclear whether DMN is altered in patients with cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL). Methods: Multimodal imaging methods, including MRI and positron emission tomography (PET), were applied to evaluate the functional, structural, and metabolic characteristics of DMN in 25 patients with CADASIL and 42 healthy controls. Results: Compared with controls, patients with CADASIL had decreased nodal efficiency and degree centrality of the dorsal medial pre-frontal cortex and hippocampal formation within DMN. Structural MRI and diffusion tensor imaging (DTI) showed decreased gray matter volume and fiber tracks presented in the bilateral hippocampal formation. Meanwhile, PET imaging showed decreased metabolism within the whole DMN in CADASIL. Furthermore, correlation analyses showed that these nodal characteristics, gray matter volume, and metabolic signals of DMN were related to cognitive scores in CADASIL. Conclusions: Our results suggested that altered network characteristics of DMN might play important roles in cognitive deficits of CADASIL.
Collapse
Affiliation(s)
- Panlong Li
- School of Electrical and Information Engineering, Zhengzhou University of Light Industry, Zhengzhou, China
| | - Qi Huang
- Positron Emission Tomography (PET) Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Shiyu Ban
- Shanghai Key Laboratory of Magnetic Resonance and Department of Physics, School of Physics and Materials Science, East China Normal University, Shanghai, China
| | - Yuan Qiao
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Wu
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Zhai
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoxia Du
- Shanghai Key Laboratory of Magnetic Resonance and Department of Physics, School of Physics and Materials Science, East China Normal University, Shanghai, China
| | - Fengchun Hua
- Department of Nuclear Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jingjing Su
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
16
|
Schoemaker D, Velilla-Jimenez L, Zuluaga Y, Baena A, Ospina C, Bocanegra Y, Alvarez S, Ochoa-Escudero M, Guzmán-Vélez E, Martinez J, Lopera F, Arboleda-Velasquez JF, Quiroz YT. Global Cardiovascular Risk Profile and Cerebrovascular Abnormalities in Presymptomatic Individuals with CADASIL or Autosomal Dominant Alzheimer's Disease. J Alzheimers Dis 2021; 82:841-853. [PMID: 34092645 DOI: 10.3233/jad-210313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Cardiovascular risk factors increase the risk of developing dementia, including Alzheimer's disease and vascular dementia. OBJECTIVE Studying individuals with autosomal dominant mutations leading to the early onset of dementia, this study examines the effect of the global cardiovascular risk profile on early cognitive and neuroimaging features of Alzheimer's disease and vascular dementia. METHODS We studied 85 non-demented and stroke-free individuals, including 20 subjects with Presenilin1 (PSEN1) E280A mutation leading to the early onset of autosomal dominant Alzheimer's disease (ADAD), 20 subjects with NOTCH3 mutations leading to cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) and to the early onset of vascular dementia, and 45 non-affected family members (non-carriers). All subjects underwent clinical and neuropsychological evaluations and an MRI. The global cardiovascular risk profile was estimated using the office-based Framingham Cardiovascular Risk Profile (FCRP) score. RESULTS In individuals with CADASIL, a higher FCRP score was associated with a reduced hippocampal volume (B = -0.06, p < 0.05) and an increased severity of cerebral microbleeds (B = 0.13, p < 0.001), lacunes (B = 0.30, p < 0.001), and perivascular space enlargement in the basal ganglia (B = 0.50, p < 0.05). There was no significant association between the FCRP score and neuroimaging measures in ADAD or non-carrier subjects. While the FCRP score was related to performance in executive function in non-carrier subjects (B = 0.06, p < 0.05), it was not significantly associated with cognitive performance in individuals with CADASIL or ADAD. CONCLUSION Our results suggest that individuals with CADASIL and other forms of vascular cognitive impairment might particularly benefit from early interventions aimed at controlling cardiovascular risks.
Collapse
Affiliation(s)
- Dorothee Schoemaker
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | | | - Yesica Zuluaga
- Grupo de Neurociencias, Universidad de Antioquia, Medellín, Colombia
| | - Ana Baena
- Grupo de Neurociencias, Universidad de Antioquia, Medellín, Colombia
| | - Carolina Ospina
- Grupo de Neurociencias, Universidad de Antioquia, Medellín, Colombia
| | - Yamile Bocanegra
- Grupo de Neurociencias, Universidad de Antioquia, Medellín, Colombia
| | - Sergio Alvarez
- Department of Radiology, Hospital Pablo Tobon Uribe, Medellín, Colombia
| | | | - Edmarie Guzmán-Vélez
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jairo Martinez
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Francisco Lopera
- Grupo de Neurociencias, Universidad de Antioquia, Medellín, Colombia
| | - Joseph F Arboleda-Velasquez
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Yakeel T Quiroz
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Grupo de Neurociencias, Universidad de Antioquia, Medellín, Colombia.,Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
17
|
Ehret F, Moreno Traspas R, Neumuth MT, Hamann B, Lasse D, Kempermann G. Notch3-Dependent Effects on Adult Neurogenesis and Hippocampus-Dependent Learning in a Modified Transgenic Model of CADASIL. Front Aging Neurosci 2021; 13:617733. [PMID: 34093162 PMCID: PMC8177050 DOI: 10.3389/fnagi.2021.617733] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 04/08/2021] [Indexed: 11/13/2022] Open
Abstract
We and others have reported that Notch3 is a regulator of adult hippocampal neurogenesis. Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CADASIL), the most common genetic form of vascular dementia, is caused by mutations in Notch3. The present study intended to investigate whether there is a correlation between altered adult hippocampal neurogenesis and spatial memory performance in CADASIL transgenic mice. To overcome visual disabilities that hampered behavioral testing of the original mice (on an FVB background) we back-crossed the existing TgN3 R169C CADASIL mouse model onto the C57BL/6J background. These animals showed an age-dependent increase in the pathognomonic granular osmiophilic material (GOM) deposition in the hippocampus. Analysis in the Morris water maze task at an age of 6 and 12 months revealed deficits in re-learning and perseverance in the CADASIL transgenic mice. Overexpression of Notch3 alone resulted in deficits in the use of spatial strategies and diminished adult neurogenesis in both age groups. The additional CADASIL mutation compensated the effect on strategy usage but not on adult neurogenesis. In brain bank tissue samples from deceased CADASIL patients we found signs of new neurons, as assessed by calretinin immunohistochemistry, but no conclusive quantification was possible. In summary, while our study confirmed the role of Notch3 in adult neurogenesis, we found a specific effect of the CADASIL mutation only on the reversion of the Notch3 effect on behavior, particularly visible at 6 months of age, consistent with a loss of function. The mutation did not revert the Notch3-dependent changes in adult neurogenesis or otherwise affected adult neurogenesis in this model.
Collapse
Affiliation(s)
- Fanny Ehret
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
| | | | | | - Bianca Hamann
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
| | - Daniela Lasse
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
| | - Gerd Kempermann
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
- Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany
| |
Collapse
|
18
|
Ulivi L, Cosottini M, Migaleddu G, Orlandi G, Giannini N, Siciliano G, Mancuso M. Brain MRI in Monogenic Cerebral Small Vessel Diseases: A Practical Handbook. Curr Mol Med 2021; 22:300-311. [DOI: 10.2174/1566524021666210510164003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 01/28/2021] [Accepted: 02/09/2021] [Indexed: 11/22/2022]
Abstract
:
Monogenic cerebral small vessel diseases are a topic of growing interest, as several genes responsible have been recently described and new sequencing techniques such as Next generation sequencing are available. Brain imaging is a key exam in these diseases. First, since it is often the first exam performed, an MRI is key in selecting patients for genetic testing and for interpreting Next generation sequencing reports. In addition, neuroimaging can be helpful in describing the underlying pathological mechanisms involved in cerebral small vessel disease. With this review, we aim to provide Neurologists and Stroke physicians with an up-to date overview of the current neuroimaging knowledge on monogenic small vessel diseases.
Collapse
Affiliation(s)
- Leonardo Ulivi
- Department of Experimental and Clinical Medicine, Neurological Clinic, Pisa University, Via Roma 67, Pisa, Italy
| | - Mirco Cosottini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Gianmichele Migaleddu
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Giovanni Orlandi
- Department of Experimental and Clinical Medicine, Neurological Clinic, Pisa University, Via Roma 67, Pisa, Italy
| | - Nicola Giannini
- Department of Experimental and Clinical Medicine, Neurological Clinic, Pisa University, Via Roma 67, Pisa, Italy
| | - Gabriele Siciliano
- Department of Experimental and Clinical Medicine, Neurological Clinic, Pisa University, Via Roma 67, Pisa, Italy
| | - Michelangelo Mancuso
- Department of Experimental and Clinical Medicine, Neurological Clinic, Pisa University, Via Roma 67, Pisa, Italy
| |
Collapse
|
19
|
Schellhorn T, Aamodt EB, Lydersen S, Aam S, Wyller TB, Saltvedt I, Beyer MK. Clinically accessible neuroimaging predictors of post-stroke neurocognitive disorder: a prospective observational study. BMC Neurol 2021; 21:89. [PMID: 33632149 PMCID: PMC7905565 DOI: 10.1186/s12883-021-02117-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 02/19/2021] [Indexed: 01/21/2023] Open
Abstract
Background Neurocognitive disorder (NCD) is common in stroke survivors. We aimed to identify clinically accessible imaging markers of stroke and chronic pathology that are associated with early post-stroke NCD. Methods We included 231 stroke survivors from the “Norwegian Cognitive Impairment after Stroke (Nor-COAST)” study who underwent a standardized cognitive assessment 3 months after the stroke. Any NCD (mild cognitive impairment and dementia) and major NCD (dementia) were diagnosed according to “Diagnostic and Statistical Manual of Mental Disorders (DSM-5)” criteria. Clinically accessible imaging findings were analyzed on study-specific brain MRIs in the early phase after stroke. Stroke lesion volumes were semi automatically quantified and strategic stroke locations were determined by an atlas based coregistration. White matter hyperintensities (WMH) and medial temporal lobe atrophy (MTA) were visually scored. Logistic regression was used to identify neuroimaging findings associated with major NCD and any NCD. Results Mean age was 71.8 years (SD 11.1), 101 (43.7%) were females, mean time from stroke to imaging was 8 (SD 16) days. At 3 months 63 (27.3%) had mild NCD and 65 (28.1%) had major NCD. Any NCD was significantly associated with WMH pathology (odds ratio (OR) = 2.73 [1.56 to 4.77], p = 0.001), MTA pathology (OR = 1.95 [1.12 to 3.41], p = 0.019), and left hemispheric stroke (OR = 1.8 [1.05 to 3.09], p = 0.032). Major NCD was significantly associated with WMH pathology (OR = 2.54 [1.33 to 4.84], p = 0.005) and stroke lesion volume (OR (per ml) =1.04 [1.01 to 1.06], p = 0.001). Conclusion WMH pathology, MTA pathology and left hemispheric stroke were associated with the development of any NCD. Stroke lesion volume and WMH pathology were associated with the development of major NCD 3 months after stroke. These imaging findings may be used in the routine clinical setting to identify patients at risk for early post-stroke NCD. Trial registration ClinicalTrials.gov, NCT02650531, Registered 8 January 2016 – Retrospectively registered. Supplementary Information The online version contains supplementary material available at 10.1186/s12883-021-02117-8.
Collapse
Affiliation(s)
- Till Schellhorn
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway. .,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Eva Birgitte Aamodt
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Stian Lydersen
- Regional Centre for Child and Youth Mental Health and Child Welfare, Department of Mental Health, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Stina Aam
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Geriatric Medicine, Clinic of Medicine St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Torgeir Bruun Wyller
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Geriatric Medicine, Oslo University Hospital, Oslo, Norway
| | - Ingvild Saltvedt
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Science, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Geriatric Medicine, Clinic of Medicine St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Mona Kristiansen Beyer
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
20
|
Fontaine JT, Rosehart AC, Joutel A, Dabertrand F. HB-EGF depolarizes hippocampal arterioles to restore myogenic tone in a genetic model of small vessel disease. Mech Ageing Dev 2020; 192:111389. [PMID: 33127441 PMCID: PMC7683376 DOI: 10.1016/j.mad.2020.111389] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/16/2020] [Accepted: 10/22/2020] [Indexed: 12/26/2022]
Abstract
Vascular cognitive impairment, the second most common cause of dementia, profoundly affects hippocampal-dependent functions. However, while the growing literature covers complex neuronal interactions, little is known about the sustaining hippocampal microcirculation. Here we examined vasoconstriction to physiological pressures of hippocampal arterioles, a fundamental feature of small arteries, in a genetic mouse model of CADASIL, an archetypal cerebral small vessel disease. Using diameter and membrane potential recordings on isolated arterioles, we observed both blunted pressure-induced vasoconstriction and smooth muscle cell depolarization in CADASIL. This impairment was abolished in the presence of voltage-gated potassium (KV1) channel blocker 4-aminopyridine, or by application of heparin-binding EGF-like growth factor (HB-EGF), which promotes KV1 channel down-regulations. Interestingly, we observed that HB-EGF induced a depolarization of the myocyte plasma membrane within the arteriolar wall in CADASIL, but not wild-type, arterioles. Collectively, our results indicate that hippocampal arterioles in CADASIL mice display a blunted contractile response to luminal pressure, similar to the defect we previously reported in cortical arterioles and pial arteries, that is rescued by HB-EGF. Hippocampal vascular dysfunction in CADASIL could then contribute to the decreased vascular reserve associated with decreased cognitive performance, and its correction may provide a therapeutic option for treating vascular cognitive impairment.
Collapse
Affiliation(s)
- Jackson T Fontaine
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Amanda C Rosehart
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Anne Joutel
- Department of Pharmacology, Larner College of Medicine University of Vermont, Burlington, VT, USA; Institute of Psychiatry and Neurosciences of Paris, INSERM UMR1266, University of Paris, GHU Paris Psychiatrie et Neurosciences, France
| | - Fabrice Dabertrand
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
21
|
Han JW, Maillard P, Harvey D, Fletcher E, Martinez O, Johnson DK, Olichney JM, Farias ST, Villeneuve S, Jagust W, Mungas D, DeCarli C. Association of vascular brain injury, neurodegeneration, amyloid, and cognitive trajectory. Neurology 2020; 95:e2622-e2634. [PMID: 32732300 PMCID: PMC7713731 DOI: 10.1212/wnl.0000000000010531] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 05/08/2020] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To determine whether vascular and neurodegenerative factors influence cognition before clinically relevant Alzheimer disease pathology, we analyzed MRI measures and amyloid imaging in an ethnoracially diverse cohort of cognitively normal individuals older than 60 years. METHODS Participants (n = 154; mean age 74.15 ± 6.94; 50% female; 54% Caucasian, 22.1% Hispanic, 14.9% African American) were recruited from the University of California, Davis Alzheimer's Disease Research Center, who were cognitively normal at baseline, time of PET, and MRI, and received yearly cognitive assessment for 6.23 ± 4.16 years. Mixed model regression with random slope and intercept was calculated for episodic memory and executive function, adjusting for age, sex, education, and ethnicity. RESULTS Vascular burden score was associated with total white matter hyperintensity (WMH) volume (β, 0.171; 95% confidence interval [CI], 0.024-0.318). WMH volume was associated with low baseline executive function (-0.115; -0.226 to -0.003) and rate of change in memory (-0.029; -0.045 to -0.012). Hippocampal volume was associated with the rate of change in memory (0.040; 0.021-0.059) and executive function (0.024; 0.008-0.039). Continuous measures of amyloid status influenced change in memory (-0.026; -0.044 to -0.008) and executive function (-0.033; -0.046 to -0.021) independently of MRI measures. CONCLUSION Vascular brain injury and neurodegeneration are associated with baseline cognitive performance and the rate of longitudinal change independent of amyloid status among community-dwelling, ethnicity diverse cognitively normal individuals, supporting the role of vascular diseases as risk factors for later-life dementia.
Collapse
Affiliation(s)
- Ji Won Han
- From the Department of Neurology (J.W.H., P.M., E.F., O.M., D.K.J., J.M.O., S.T.F., D.M., C.D.), Imaging of Dementia and Aging (IDeA) Laboratory (J.W.H., P.M., E.F., O.M., C.D.), and Division of Biostatistics, School of Medicine (D.H.), University of California at Davis; Department of Neuropsychiatry (J.W.H.), Seoul National University Bundang Hospital, Seoul National University, Seongnam, Republic of Korea; Douglas Mental Health University Institute (S.V.), McGill University, Montreal, Canada; and Helen Wills Neuroscience Institute (W.J.), University of California, Berkeley
| | - Pauline Maillard
- From the Department of Neurology (J.W.H., P.M., E.F., O.M., D.K.J., J.M.O., S.T.F., D.M., C.D.), Imaging of Dementia and Aging (IDeA) Laboratory (J.W.H., P.M., E.F., O.M., C.D.), and Division of Biostatistics, School of Medicine (D.H.), University of California at Davis; Department of Neuropsychiatry (J.W.H.), Seoul National University Bundang Hospital, Seoul National University, Seongnam, Republic of Korea; Douglas Mental Health University Institute (S.V.), McGill University, Montreal, Canada; and Helen Wills Neuroscience Institute (W.J.), University of California, Berkeley
| | - Danielle Harvey
- From the Department of Neurology (J.W.H., P.M., E.F., O.M., D.K.J., J.M.O., S.T.F., D.M., C.D.), Imaging of Dementia and Aging (IDeA) Laboratory (J.W.H., P.M., E.F., O.M., C.D.), and Division of Biostatistics, School of Medicine (D.H.), University of California at Davis; Department of Neuropsychiatry (J.W.H.), Seoul National University Bundang Hospital, Seoul National University, Seongnam, Republic of Korea; Douglas Mental Health University Institute (S.V.), McGill University, Montreal, Canada; and Helen Wills Neuroscience Institute (W.J.), University of California, Berkeley
| | - Evan Fletcher
- From the Department of Neurology (J.W.H., P.M., E.F., O.M., D.K.J., J.M.O., S.T.F., D.M., C.D.), Imaging of Dementia and Aging (IDeA) Laboratory (J.W.H., P.M., E.F., O.M., C.D.), and Division of Biostatistics, School of Medicine (D.H.), University of California at Davis; Department of Neuropsychiatry (J.W.H.), Seoul National University Bundang Hospital, Seoul National University, Seongnam, Republic of Korea; Douglas Mental Health University Institute (S.V.), McGill University, Montreal, Canada; and Helen Wills Neuroscience Institute (W.J.), University of California, Berkeley
| | - Oliver Martinez
- From the Department of Neurology (J.W.H., P.M., E.F., O.M., D.K.J., J.M.O., S.T.F., D.M., C.D.), Imaging of Dementia and Aging (IDeA) Laboratory (J.W.H., P.M., E.F., O.M., C.D.), and Division of Biostatistics, School of Medicine (D.H.), University of California at Davis; Department of Neuropsychiatry (J.W.H.), Seoul National University Bundang Hospital, Seoul National University, Seongnam, Republic of Korea; Douglas Mental Health University Institute (S.V.), McGill University, Montreal, Canada; and Helen Wills Neuroscience Institute (W.J.), University of California, Berkeley
| | - David K Johnson
- From the Department of Neurology (J.W.H., P.M., E.F., O.M., D.K.J., J.M.O., S.T.F., D.M., C.D.), Imaging of Dementia and Aging (IDeA) Laboratory (J.W.H., P.M., E.F., O.M., C.D.), and Division of Biostatistics, School of Medicine (D.H.), University of California at Davis; Department of Neuropsychiatry (J.W.H.), Seoul National University Bundang Hospital, Seoul National University, Seongnam, Republic of Korea; Douglas Mental Health University Institute (S.V.), McGill University, Montreal, Canada; and Helen Wills Neuroscience Institute (W.J.), University of California, Berkeley
| | - John M Olichney
- From the Department of Neurology (J.W.H., P.M., E.F., O.M., D.K.J., J.M.O., S.T.F., D.M., C.D.), Imaging of Dementia and Aging (IDeA) Laboratory (J.W.H., P.M., E.F., O.M., C.D.), and Division of Biostatistics, School of Medicine (D.H.), University of California at Davis; Department of Neuropsychiatry (J.W.H.), Seoul National University Bundang Hospital, Seoul National University, Seongnam, Republic of Korea; Douglas Mental Health University Institute (S.V.), McGill University, Montreal, Canada; and Helen Wills Neuroscience Institute (W.J.), University of California, Berkeley
| | - Sarah T Farias
- From the Department of Neurology (J.W.H., P.M., E.F., O.M., D.K.J., J.M.O., S.T.F., D.M., C.D.), Imaging of Dementia and Aging (IDeA) Laboratory (J.W.H., P.M., E.F., O.M., C.D.), and Division of Biostatistics, School of Medicine (D.H.), University of California at Davis; Department of Neuropsychiatry (J.W.H.), Seoul National University Bundang Hospital, Seoul National University, Seongnam, Republic of Korea; Douglas Mental Health University Institute (S.V.), McGill University, Montreal, Canada; and Helen Wills Neuroscience Institute (W.J.), University of California, Berkeley
| | - Sylvia Villeneuve
- From the Department of Neurology (J.W.H., P.M., E.F., O.M., D.K.J., J.M.O., S.T.F., D.M., C.D.), Imaging of Dementia and Aging (IDeA) Laboratory (J.W.H., P.M., E.F., O.M., C.D.), and Division of Biostatistics, School of Medicine (D.H.), University of California at Davis; Department of Neuropsychiatry (J.W.H.), Seoul National University Bundang Hospital, Seoul National University, Seongnam, Republic of Korea; Douglas Mental Health University Institute (S.V.), McGill University, Montreal, Canada; and Helen Wills Neuroscience Institute (W.J.), University of California, Berkeley
| | - William Jagust
- From the Department of Neurology (J.W.H., P.M., E.F., O.M., D.K.J., J.M.O., S.T.F., D.M., C.D.), Imaging of Dementia and Aging (IDeA) Laboratory (J.W.H., P.M., E.F., O.M., C.D.), and Division of Biostatistics, School of Medicine (D.H.), University of California at Davis; Department of Neuropsychiatry (J.W.H.), Seoul National University Bundang Hospital, Seoul National University, Seongnam, Republic of Korea; Douglas Mental Health University Institute (S.V.), McGill University, Montreal, Canada; and Helen Wills Neuroscience Institute (W.J.), University of California, Berkeley
| | - Dan Mungas
- From the Department of Neurology (J.W.H., P.M., E.F., O.M., D.K.J., J.M.O., S.T.F., D.M., C.D.), Imaging of Dementia and Aging (IDeA) Laboratory (J.W.H., P.M., E.F., O.M., C.D.), and Division of Biostatistics, School of Medicine (D.H.), University of California at Davis; Department of Neuropsychiatry (J.W.H.), Seoul National University Bundang Hospital, Seoul National University, Seongnam, Republic of Korea; Douglas Mental Health University Institute (S.V.), McGill University, Montreal, Canada; and Helen Wills Neuroscience Institute (W.J.), University of California, Berkeley
| | - Charles DeCarli
- From the Department of Neurology (J.W.H., P.M., E.F., O.M., D.K.J., J.M.O., S.T.F., D.M., C.D.), Imaging of Dementia and Aging (IDeA) Laboratory (J.W.H., P.M., E.F., O.M., C.D.), and Division of Biostatistics, School of Medicine (D.H.), University of California at Davis; Department of Neuropsychiatry (J.W.H.), Seoul National University Bundang Hospital, Seoul National University, Seongnam, Republic of Korea; Douglas Mental Health University Institute (S.V.), McGill University, Montreal, Canada; and Helen Wills Neuroscience Institute (W.J.), University of California, Berkeley.
| |
Collapse
|
22
|
Yamamoto Y, Hase Y, Ihara M, Khundakar A, Roeber S, Duering M, Kalaria RN. Neuronal densities and vascular pathology in the hippocampal formation in CADASIL. Neurobiol Aging 2020; 97:33-40. [PMID: 33130454 PMCID: PMC7758782 DOI: 10.1016/j.neurobiolaging.2020.09.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 09/21/2020] [Accepted: 09/21/2020] [Indexed: 11/17/2022]
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is the most common form of hereditary cerebral small vessel disease. Previous neuroimaging studies have suggested loss of hippocampal volume is a pathway for cognitive impairment in CADASIL. We used unbiased stereological methods to estimate SMI32-positive and total numbers and volumes of neurons in the hippocampal formation of 12 patients with CADASIL and similar age controls (young controls) and older controls. We found densities of SMI32-positive neurons in the entorhinal cortex, layer V, and cornu ammonis CA2 regions were reduced by 26%–50% in patients with CADASIL compared with young controls (p < 0.01), with a decreasing trend observed in older controls in the order of young controls> older controls ≥ CADASIL. These changes were not explained by any hippocampal infarct or vascular pathology or glial changes. Our results suggest notable loss of subsets of projection neurons within the hippocampal formation that may contribute to certain memory deficits in CADASIL, which is purely a vascular disease. It is likely that the severe arteriopathy leads to white matter damage which disconnects cortico-cortical and subcortical-cortical networks including the hippocampal formation. Hippocampal volume loss was associated with cognitive dysfunction in CADASIL. SMI32+ neurons in entorhinal cortex (V) and CA2 regions were reduced in CADASIL. Hippocampal cellular changes were not explained by any infarct pathology. Neuronal volumes or glial cell numbers per neuron were not changed. Selective loss of projection neurons within the hippocampal formation in CADASIL.
Collapse
Affiliation(s)
- Yumi Yamamoto
- Translational and Clinical Research Institute, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, UK; Department of Molecular Innovation in Lipidemiology, Suita, Osaka, Japan; Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Yoshiki Hase
- Translational and Clinical Research Institute, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, UK
| | - Masafumi Ihara
- Department of Molecular Innovation in Lipidemiology, Suita, Osaka, Japan; Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Ahmad Khundakar
- School of Health and Life Sciences, Teesside University, Middlesbrough, Tees Valley, UK
| | | | - Marco Duering
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich & Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Raj N Kalaria
- Translational and Clinical Research Institute, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, UK.
| |
Collapse
|
23
|
Pan H, Huang Q, Ban S, Du X, Su J, Liu J. Brain structural changes in CADASIL patients: A morphometric magnetic resonance imaging study. Neurosci Lett 2020; 738:135388. [PMID: 32949660 DOI: 10.1016/j.neulet.2020.135388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 09/07/2020] [Accepted: 09/11/2020] [Indexed: 01/23/2023]
Abstract
BACKGROUND Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is a subcortical, inherited, cerebral small vessel disease. Several studies have revealed the involvement of specific cortical regions. However, the structural brain alterations and their clinical correlations remain largely undetermined. METHODS We evaluated 22 CADASIL patients and 22 age- and sex-matched healthy controls. We used surface- and voxel-based morphometric data derived from 3.0-T magnetic resonance imaging (MRI) to explore structural changes in gray and white matter. We calculated Pearson correlations between such data and clinical and MRI metrics. RESULTS Compared with controls, CADASIL patients exhibited significantly decreased cortical thickness in the left supramarginal gyrus, superior temporal gyrus, transverse temporal gyrus, insula, lateral orbitofrontal gyrus, isthmus cingulate gyrus and precentral gyrus. An extensive decrease in the white (but not gray) matter volume was also evident, predominantly in the frontal, parietal, temporal, and occipital lobes. The number of previous strokes or transient ischemic attacks was negatively associated with the cortical thickness of the left pars opercularis and right posterior cingulate gyrus. CONCLUSION Reductions in cortical thickness and white matter volume were evident in CADASIL patients compared with controls, and higher numbers of strokes and transient ischemic attacks were associated with regional cortical thinning.
Collapse
Affiliation(s)
- Hui Pan
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China
| | - Qi Huang
- PET Center, Huashan Hospital, Fudan University, 518 East Wuzhong Road, Shanghai, 200235, People's Republic of China
| | - Shiyu Ban
- Shanghai Key Laboratory of Magnetic Resonance and Department of Physics, School of Physics and Materials Science, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, People's Republic of China
| | - Xiaoxia Du
- Shanghai Key Laboratory of Magnetic Resonance and Department of Physics, School of Physics and Materials Science, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, People's Republic of China.
| | - Jingjing Su
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China.
| | - Jianren Liu
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, People's Republic of China.
| |
Collapse
|
24
|
Savic I. MRS Shows Regionally Increased Glutamate Levels among Patients with Exhaustion Syndrome Due to Occupational Stress. Cereb Cortex 2020; 30:3759-3770. [PMID: 32195540 DOI: 10.1093/cercor/bhz340] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 12/17/2019] [Indexed: 12/21/2022] Open
Abstract
Despite the rapid increase of reports of exhaustion syndrome (ES) due to daily occupational stress, the mechanisms underlying ES are unknown. We used voxel-based 1H-MR spectroscopy to examine the potential role of glutamate in this condition. The levels of glutamate were found to be elevated among ES patients (n = 30, 16 females) compared with controls (n = 31, 15 females). Notably, this increase was detected only in the anterior cingulate and mesial prefrontal cortex (ACC/mPFC), and the glutamate levels were linearly correlated with the degree of perceived stress. Furthermore, there was a sex by group interaction, as the glutamate elevation was present only in female patients. Female but not male ES patients also showed an increase in N-acetyl aspartate (NAA) levels in the amygdala. No group differences were detected in glutamine concentration (also measured). These data show the key role of glutamate in stress-related neuronal signaling and the specific roles of the amygdala and ACC/mPFC. The data extend previous reports about the neurochemical basis of stress and identify a potential neural marker and mediator of ES due to occupational stress. The observation of specific sex differences provides a tentative explanation to the well-known female predominance in stress-related psychopathology.
Collapse
Affiliation(s)
- Ivanka Savic
- Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, SE-171 76 Stockholm, Sweden.,Department of Neurology, Karolinska University Hospital, SE-171 76 Stockholm, Sweden.,Department of Neurology, UCLA, Los Angeles, CA 90095-1769, USA
| |
Collapse
|
25
|
Chabriat H, Jouvent E. Imaging of the aging brain and development of MRI signal abnormalities. Rev Neurol (Paris) 2020; 176:661-669. [PMID: 32229042 DOI: 10.1016/j.neurol.2019.12.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 12/14/2019] [Accepted: 12/16/2019] [Indexed: 02/04/2023]
Abstract
Major changes occur at the cerebral level with aging. Cerebral atrophy develops progressively. Multiple lesions related to small-vessel diseases are detected in association with cerebral atrophy including white-matter hyperintensities, lacunes, microbleeds, dilated perivascular spaces and cerebral, including cortex, atrophy. The clinical impact and predictive value of these Imaging makers were examined.
Collapse
Affiliation(s)
- H Chabriat
- Inserm U1161 and DHU NeuroVasc, department of neurology, Paris University, Lariboisiere Hospital,Assistance Publique-Hopitaux de Paris, Paris, France.
| | - E Jouvent
- Inserm U1161 and DHU NeuroVasc, department of neurology, Paris University, Lariboisiere Hospital,Assistance Publique-Hopitaux de Paris, Paris, France
| |
Collapse
|
26
|
Suda S, Nishimura T, Ishiwata A, Muraga K, Aoki J, Kanamaru T, Suzuki K, Sakamoto Y, Katano T, Nishiyama Y, Mishina M, Kimura K. Early Cognitive Impairment after Minor Stroke: Associated Factors and Functional Outcome. J Stroke Cerebrovasc Dis 2020; 29:104749. [PMID: 32178931 DOI: 10.1016/j.jstrokecerebrovasdis.2020.104749] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/18/2020] [Accepted: 02/09/2020] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVES Evaluation of cognitive status is not performed routinely in the acute stroke setting. This study aimed to evaluate the frequency of early cognitive impairment in patients with minor ischemic stroke, analyze the factors associated with early cognitive impairment, and assess functional outcomes. METHODS In this prospective study, 112 consecutive patients with acute minor ischemic stroke were enrolled. Neuroimages were assessed for semiquantitative evaluation of brain atrophy and small vessel disease (SVD) markers. Cognitive performance was measured within 5 days of onset using Montreal Cognitive Assessment (MoCA) scores. Functional outcome analyses were adjusted for demographic variables, premorbid cognitive status, education level, vascular risk factors, neuroimaging characteristics, stroke severity, and MoCA scores. RESULTS The median MoCA score was 22, and 63% of patients had cognitive impairment. Factors independently associated with cognitive impairment were education (odds ratios [OR], .79; confidence intervals [CI], .63-.99), smoking (OR, .26; 95%CI, .073-.89), and temporal horn atrophy (OR, 4.73; 95% CI, 1.66-13.49). Factors independently associated with poor functional outcome were total MoCA score (OR, .78; 95%CI, .62-.95) and the sum of 4 MoCA subscores (visuospatial/executive, attention, language, and orientation; OR, .72; 95%CI, .53-.92). The cutoff value of the sum of 4 MoCA subscores for predicting poor outcome was 13 points with 76.5% sensitivity and 81.1% specificity. CONCLUSIONS Early cognitive impairment was common after minor ischemic stroke and was associated with preexisting temporal horn atrophy but not SVD markers. The sum of 4 MoCA subscores was useful in predicting the functional outcome.
Collapse
Affiliation(s)
- Satoshi Suda
- Department of Neurology, Nippon Medical School, Bunkyo-ku, Tokyo, Japan.
| | - Takuya Nishimura
- Department of Neurology, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| | - Akiko Ishiwata
- Department of Neurology, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| | - Kanako Muraga
- Department of Neurology, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| | - Junya Aoki
- Department of Neurology, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| | - Takuya Kanamaru
- Department of Neurology, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| | - Kentaro Suzuki
- Department of Neurology, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| | - Yuki Sakamoto
- Department of Neurology, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| | - Takehiro Katano
- Department of Neurology, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| | | | - Masahiro Mishina
- Department of Neurology, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| | - Kazumi Kimura
- Department of Neurology, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
27
|
Takahashi Y, Saito S, Yamamoto Y, Uehara T, Yokota C, Sakai G, Nishida N, Takahashi R, Kalaria RN, Toyoda K, Nagatsuka K, Ihara M. Visually-Rated Medial Temporal Lobe Atrophy with Lower Educational History as a Quick Indicator of Amnestic Cognitive Impairment after Stroke. J Alzheimers Dis 2020; 67:621-629. [PMID: 30584149 DOI: 10.3233/jad-180976] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Time and resource limitations prevent cognitive assessment in acute-to-subacute settings, even in comprehensive stroke centers. OBJECTIVE To assess cognitive function in acute stroke patients undergoing routine clinical, laboratory, and radiological investigations, with a view to improving post-stroke care and treatment. METHODS Sixty-nine patients (72.6±11.1 years; 65% male) were prospectively enrolled within 14 days of acute ischemic stroke. Patients with altered consciousness, aphasia, or dysarthria were excluded. Clinical features including modified Rankin and NIH stroke scales, and vascular risk factors were assessed, as well as neuroimaging parameters by semi-quantitative evaluation of medial temporal lobe atrophy (MTLA) using MRA source images, FLAIR images for white matter changes (Fazekas scores), and T2∗ images for cerebral microbleeds. Neuropsychological screening was conducted using the Montreal Cognitive Assessment (MoCA) test. Univariate and multivariate analyses were used to evaluate the influence of variables on MoCA total and subscale scores. RESULTS Lower MoCA scores of 22 or less were associated with MTLA [OR (95% CI), 5.3 (1.0-27.5); p = 0.045], education years [OR (95% CI), 0.71 (0.55-0.91); p = 0.007], and modified Rankin scale at discharge [OR (95% CI), 2.4 (1.3-4.5); p = 0.007]. The delayed recall MoCA score was correlated with MTLA (r = - 0.452, p < 0.001), periventricular (r = - 0.273, p = 0.024), and deep (r = - 0.242, p = 0.046), white matter changes. CONCLUSIONS MTLA, together with lower educational history, are quick indicators of amnestic cognitive impairment after stroke. The association between cognitive impairment and physical disability at discharge may signify the importance of earlier cognitive assessment.
Collapse
Affiliation(s)
- Yukako Takahashi
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan.,Department of Neurology, Kyoto University Graduate School of Medicine, Osaka, Japan
| | - Satoshi Saito
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Yumi Yamamoto
- Department of Regenerative Medicine and Tissue Engineering, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Toshiyuki Uehara
- Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan.,Department of Stroke Rehabilitation, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Chiaki Yokota
- Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan.,Department of Stroke Rehabilitation, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Go Sakai
- Department of Diagnostic Radiology, Osaka Saiseikai Nakatsu Hospital, Osaka, Japan
| | - Norifumi Nishida
- Department of Diagnostic Radiology, Osaka Saiseikai Nakatsu Hospital, Osaka, Japan
| | - Ryosuke Takahashi
- Department of Neurology, Kyoto University Graduate School of Medicine, Osaka, Japan
| | - Raj N Kalaria
- Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle, UK
| | - Kazunori Toyoda
- Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Kazuyuki Nagatsuka
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| |
Collapse
|
28
|
Perosa V, Priester A, Ziegler G, Cardenas-Blanco A, Dobisch L, Spallazzi M, Assmann A, Maass A, Speck O, Oltmer J, Heinze HJ, Schreiber S, Düzel E. Hippocampal vascular reserve associated with cognitive performance and hippocampal volume. Brain 2020; 143:622-634. [PMID: 31994699 PMCID: PMC7009470 DOI: 10.1093/brain/awz383] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/12/2019] [Accepted: 10/07/2019] [Indexed: 01/14/2023] Open
Abstract
Medial temporal lobe dependent cognitive functions are highly vulnerable to hypoxia in the hippocampal region, yet little is known about the relationship between the richness of hippocampal vascular supply and cognition. Hippocampal vascularization patterns have been categorized into a mixed supply from both the posterior cerebral artery and the anterior choroidal artery or a single supply by the posterior cerebral artery only. Hippocampal arteries are small and affected by pathological changes when cerebral small vessel disease is present. We hypothesized, that hippocampal vascularization patterns may be important trait markers for vascular reserve and modulate (i) cognitive performance; (ii) structural hippocampal integrity; and (iii) the effect of cerebral small vessel disease on cognition. Using high-resolution 7 T time-of-flight angiography we manually classified hippocampal vascularization patterns in older adults with and without cerebral small vessel disease in vivo. The presence of a mixed supplied hippocampus was an advantage in several cognitive domains, including verbal list learning and global cognition. A mixed supplied hippocampus also was an advantage for verbal memory performance in cerebral small vessel disease. Voxel-based morphometry showed higher anterior hippocampal grey matter volume in mixed, compared to single supply. We discuss that a mixed hippocampal supply, as opposed to a single one, may increase the reliability of hippocampal blood supply and thereby provide a hippocampal vascular reserve that protects against cognitive impairment.
Collapse
Affiliation(s)
- Valentina Perosa
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke-University, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Anastasia Priester
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany
| | - Gabriel Ziegler
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke-University, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Arturo Cardenas-Blanco
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke-University, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Laura Dobisch
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke-University, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Marco Spallazzi
- Department of Neurology, Azienda Ospedaliero-Universitaria, Parma, Italy
| | - Anne Assmann
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany
| | - Anne Maass
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Oliver Speck
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
- Leibniz-Institute for Neurobiology (LIN), Magdeburg, Germany
- Institute of Physics, Otto-von-Guericke University, Magdeburg, Germany
| | - Jan Oltmer
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Hans-Jochen Heinze
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
- Leibniz-Institute for Neurobiology (LIN), Magdeburg, Germany
| | - Stefanie Schreiber
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Emrah Düzel
- Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke-University, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
- Institute of Cognitive Neuroscience, University College London, London, UK
| |
Collapse
|
29
|
Jokinen H, Koikkalainen J, Laakso HM, Melkas S, Nieminen T, Brander A, Korvenoja A, Rueckert D, Barkhof F, Scheltens P, Schmidt R, Fazekas F, Madureira S, Verdelho A, Wallin A, Wahlund LO, Waldemar G, Chabriat H, Hennerici M, O'Brien J, Inzitari D, Lötjönen J, Pantoni L, Erkinjuntti T. Global Burden of Small Vessel Disease-Related Brain Changes on MRI Predicts Cognitive and Functional Decline. Stroke 2019; 51:170-178. [PMID: 31699021 PMCID: PMC6924941 DOI: 10.1161/strokeaha.119.026170] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Supplemental Digital Content is available in the text. Cerebral small vessel disease is characterized by a wide range of focal and global brain changes. We used a magnetic resonance imaging segmentation tool to quantify multiple types of small vessel disease–related brain changes and examined their individual and combined predictive value on cognitive and functional abilities.
Collapse
Affiliation(s)
- Hanna Jokinen
- From the Clinical Neurosciences, Neurology, University of Helsinki and Helsinki University Hospital (H.J., H.M.L., S. Melkas, T.E.), Finland.,Department of Psychology and Logopedics, Faculty of Medicine (H.J., H.M.L.), Finland
| | - Juha Koikkalainen
- Combinostics, Ltd, Finland (J.K., T.N., J.L.).,VTT Technical Research Centre of Finland (J.K., J.L.).,Faculty of Health Sciences, University of Eastern Finland (J.K.)
| | - Hanna M Laakso
- From the Clinical Neurosciences, Neurology, University of Helsinki and Helsinki University Hospital (H.J., H.M.L., S. Melkas, T.E.), Finland.,Department of Psychology and Logopedics, Faculty of Medicine (H.J., H.M.L.), Finland
| | - Susanna Melkas
- From the Clinical Neurosciences, Neurology, University of Helsinki and Helsinki University Hospital (H.J., H.M.L., S. Melkas, T.E.), Finland
| | | | - Antti Brander
- Department of Radiology, Medical Imaging Center, Tampere University Hospital, Finland (A.B.)
| | - Antti Korvenoja
- Medical Imaging Center, Radiology, University of Helsinki and Helsinki University Hospital (A.K.), Finland
| | - Daniel Rueckert
- Biomedical Image Analysis Group, Department of Computing, Imperial College London, United Kingdom (D.R.)
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine (F.B.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands.,Institutes of Neurology and Healthcare Engineering, University College London, United Kingdom (F.B.)
| | - Philip Scheltens
- Alzheimer Center and Department of Neurology (P.S.), Neuroscience Campus Amsterdam, VU University Medical Center, the Netherlands.,NIHR Biomedical Research Centre at University College London Hospitals NHS Foundation Trust, University College London, United Kingdom (F.B.)
| | - Reinhold Schmidt
- Department of Neurology, Medical University of Graz, Austria (R.S., F.F.)
| | - Franz Fazekas
- Department of Neurology, Medical University of Graz, Austria (R.S., F.F.)
| | - Sofia Madureira
- Department of Neurosciences, Santa Maria Hospital, University of Lisbon, Portugal (S. Madureira, A.V.)
| | - Ana Verdelho
- Department of Neurosciences, Santa Maria Hospital, University of Lisbon, Portugal (S. Madureira, A.V.)
| | - Anders Wallin
- Sahlgrenska Academy, Institute of Neuroscience and Physiology, Section for Psychiatry and Neurochemistry, University of Gothenburg, Sweden (A.W.)
| | - Lars-Olof Wahlund
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Karolinska Institutet, Sweden (L.-O.W.)
| | - Gunhild Waldemar
- Department of Neurology, Danish Dementia Research Centre, Rigshospitalet, University of Copenhagen, Denmark (G.W.)
| | - Hugues Chabriat
- Department of Neurology, Hopital Lariboisiere, APHP and INSERM U1161-University Denis Diderot (DHU NeuroVasc), France (H.C.)
| | | | - John O'Brien
- Department of Psychiatry, University of Cambridge, United Kingdom (J.O.)
| | - Domenico Inzitari
- Institute of Neuroscience, Italian National Research Council (D.I.).,Department NEUROFARBA, University of Florence, Italy (D.I.)
| | - Jyrki Lötjönen
- Combinostics, Ltd, Finland (J.K., T.N., J.L.).,VTT Technical Research Centre of Finland (J.K., J.L.).,Department of Neuroscience and Biomedical Engineering, School of Science, Aalto University, Finland (J.L.)
| | - Leonardo Pantoni
- L. Sacco Department of Biomedical and Clinical Sciences, University of Milan, Italy (L.P.)
| | - Timo Erkinjuntti
- From the Clinical Neurosciences, Neurology, University of Helsinki and Helsinki University Hospital (H.J., H.M.L., S. Melkas, T.E.), Finland
| |
Collapse
|
30
|
Schoemaker D, Quiroz YT, Torrico-Teave H, Arboleda-Velasquez JF. Clinical and research applications of magnetic resonance imaging in the study of CADASIL. Neurosci Lett 2019; 698:173-179. [PMID: 30634011 PMCID: PMC6661177 DOI: 10.1016/j.neulet.2019.01.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 01/04/2019] [Accepted: 01/07/2019] [Indexed: 12/19/2022]
Abstract
Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CADASIL) is an inherited small vessel disease that leads to early cerebrovascular events and functional disability. It is the most common single-gene disorder leading to stroke. Magnetic resonance imaging (MRI) is a central component of the diagnosis and monitoring of CADASIL. Here we provide a descriptive review of the literature on three important aspects pertaining to the use of MRI in CADASIL. First, we review past research exploring MRI markers for this disease. Secondly, we describe results from studies investigating associations between neuroimaging abnormalities and neuropathology in CADASIL. Finally, we discuss previous findings relating MRI markers to clinical symptoms. This review thus provides a summary of the current state of knowledge regarding the use of MRI in CADASIL as well as suggestions for future research.
Collapse
Affiliation(s)
- Dorothee Schoemaker
- Schepens Eye Research Institute of Massachusetts Eye and Ear and Department of Ophthalmology, Harvard Medical School, Boston, MA, United States; Massachusetts General Hospital and Department of Psychiatry, Harvard Medical School, Boston, MA, United States.
| | - Yakeel T Quiroz
- Massachusetts General Hospital and Department of Psychiatry, Harvard Medical School, Boston, MA, United States
| | - Heirangi Torrico-Teave
- Massachusetts General Hospital and Department of Psychiatry, Harvard Medical School, Boston, MA, United States
| | - Joseph F Arboleda-Velasquez
- Schepens Eye Research Institute of Massachusetts Eye and Ear and Department of Ophthalmology, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
31
|
El Hajj H, Savage JC, Bisht K, Parent M, Vallières L, Rivest S, Tremblay MÈ. Ultrastructural evidence of microglial heterogeneity in Alzheimer's disease amyloid pathology. J Neuroinflammation 2019; 16:87. [PMID: 30992040 PMCID: PMC6469225 DOI: 10.1186/s12974-019-1473-9] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 04/01/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common neurodegenerative disease, characterized by the deposition of extracellular fibrillar amyloid β (fΑβ) and the intracellular accumulation of neurofibrillary tangles. As AD progresses, Aβ drives a robust and prolonged inflammatory response via its recognition by microglia, the brain's immune cells. Microglial reactivity to fAβ plaques may impair their normal surveillance duties, facilitating synaptic loss and neuronal death, as well as cognitive decline in AD. METHODS In the current study, we performed correlative light, transmission, and scanning electron microscopy to provide insights into microglial structural and functional heterogeneity. We analyzed microglial cell bodies and processes in areas containing fAβ plaques and neuronal dystrophy, dystrophy only, or appearing healthy, among the hippocampus CA1 of 14-month-old APPSwe-PS1Δe9 mice versus wild-type littermates. RESULTS Our quantitative analysis revealed that microglial cell bodies in the AD model mice were larger and displayed ultrastructural signs of cellular stress, especially nearby plaques. Microglial cell bodies and processes were overall less phagocytic in AD model mice. However, they contained increased fibrillar materials and non-empty inclusions proximal to plaques. Microglial cell bodies and processes in AD model mice also displayed reduced association with extracellular space pockets that contained debris. In addition, microglial processes in healthy subregions of AD model mice encircled synaptic elements more often compared with plaque-associated processes. These observations in mice were qualitatively replicated in post-mortem hippocampal samples from two patients with AD (Braak stage 5). CONCLUSION Together, our findings identify at the ultrastructural level distinct microglial transformations common to mouse and human in association with amyloid pathology.
Collapse
Affiliation(s)
- Hassan El Hajj
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, 2705, boulevard Laurier, T2-50, Quebec, QC G1V 4G2 Canada
| | - Julie C. Savage
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, 2705, boulevard Laurier, T2-50, Quebec, QC G1V 4G2 Canada
| | - Kanchan Bisht
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, 2705, boulevard Laurier, T2-50, Quebec, QC G1V 4G2 Canada
| | - Martin Parent
- Département de psychiatrie et de neurosciences, Faculté de médecine, Université Laval, Quebec, QC Canada
- CERVO Brain Research Center, Quebec, QC Canada
| | - Luc Vallières
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, 2705, boulevard Laurier, T2-50, Quebec, QC G1V 4G2 Canada
- Département de médecine moléculaire, Faculté de médecine, Université Laval, Quebec, QC Canada
| | - Serge Rivest
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, 2705, boulevard Laurier, T2-50, Quebec, QC G1V 4G2 Canada
- Département de médecine moléculaire, Faculté de médecine, Université Laval, Quebec, QC Canada
| | - Marie-Ève Tremblay
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, 2705, boulevard Laurier, T2-50, Quebec, QC G1V 4G2 Canada
- Département de médecine moléculaire, Faculté de médecine, Université Laval, Quebec, QC Canada
| |
Collapse
|
32
|
Tang Y, Xing Y, Zhu Z, He Y, Li F, Yang J, Liu Q, Li F, Teipel SJ, Zhao G, Jia J. The effects of 7‐week cognitive training in patients with vascular cognitive impairment, no dementia (the Cog‐VACCINE study): A randomized controlled trial. Alzheimers Dement 2019; 15:605-614. [DOI: 10.1016/j.jalz.2019.01.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 12/26/2018] [Accepted: 01/09/2019] [Indexed: 10/27/2022]
Affiliation(s)
- Yi Tang
- Department of NeurologyXuanwu HospitalCapital Medical UniversityBeijingChina
- Innovation Center for Neurological DisordersXuanwu HospitalCapital Medical UniversityBeijingChina
- Key Laboratory of Neurodegenerative DiseasesMinistry of Education of the People's Republic of ChinaBeijingChina
| | - Yi Xing
- Department of NeurologyXuanwu HospitalCapital Medical UniversityBeijingChina
- Innovation Center for Neurological DisordersXuanwu HospitalCapital Medical UniversityBeijingChina
- Key Laboratory of Neurodegenerative DiseasesMinistry of Education of the People's Republic of ChinaBeijingChina
| | - Zude Zhu
- Collaborative Innovation Center for Language AbilityJiangsu Normal UniversityXuzhouChina
| | - Yong He
- State Key Laboratory of Cognitive Neuroscience and LearningBeijingChina
- Beijing Key Laboratory of Brain Imaging and ConnectomicsBeijingChina
- IDG/McGovern Institute for Brain ResearchBeijing Normal UniversityBeijingChina
| | - Fang Li
- Department of GeriatricFu Xing HospitalCapital Medical UniversityBeijingChina
| | - Jianwei Yang
- Department of NeurologyXuanwu HospitalCapital Medical UniversityBeijingChina
- Innovation Center for Neurological DisordersXuanwu HospitalCapital Medical UniversityBeijingChina
- Key Laboratory of Neurodegenerative DiseasesMinistry of Education of the People's Republic of ChinaBeijingChina
| | - Qing Liu
- Department of NeurologyXuanwu HospitalCapital Medical UniversityBeijingChina
- Innovation Center for Neurological DisordersXuanwu HospitalCapital Medical UniversityBeijingChina
| | - Fangyu Li
- Department of NeurologyXuanwu HospitalCapital Medical UniversityBeijingChina
- Innovation Center for Neurological DisordersXuanwu HospitalCapital Medical UniversityBeijingChina
- Key Laboratory of Neurodegenerative DiseasesMinistry of Education of the People's Republic of ChinaBeijingChina
| | - Stefan J. Teipel
- Department of Psychosomatic MedicineUniversity Medicine RostockRostockGermany
- DZNEGerman Center for Neurodegenerative DiseasesRostockGermany
| | - Guoguang Zhao
- Department of NeurosurgeryXuanwu HospitalCapital Medical UniversityBeijingChina
| | - Jianping Jia
- Department of NeurologyXuanwu HospitalCapital Medical UniversityBeijingChina
- Innovation Center for Neurological DisordersXuanwu HospitalCapital Medical UniversityBeijingChina
- Key Laboratory of Neurodegenerative DiseasesMinistry of Education of the People's Republic of ChinaBeijingChina
- Center of Alzheimer's DiseaseBeijing Institute for Brain DisordersBeijingChina
- Beijing Key Laboratory of Geriatric Cognitive DisordersBeijingChina
| |
Collapse
|
33
|
Schaduangrat N, Prachayasittikul V, Choomwattana S, Wongchitrat P, Phopin K, Suwanjang W, Malik AA, Vincent B, Nantasenamat C. Multidisciplinary approaches for targeting the secretase protein family as a therapeutic route for Alzheimer's disease. Med Res Rev 2019; 39:1730-1778. [PMID: 30628099 DOI: 10.1002/med.21563] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 11/21/2018] [Accepted: 12/24/2018] [Indexed: 12/27/2022]
Abstract
The continual increase of the aging population worldwide renders Alzheimer's disease (AD) a global prime concern. Several attempts have been focused on understanding the intricate complexity of the disease's development along with the on- andgoing search for novel therapeutic strategies. Incapability of existing AD drugs to effectively modulate the pathogenesis or to delay the progression of the disease leads to a shift in the paradigm of AD drug discovery. Efforts aimed at identifying AD drugs have mostly focused on the development of disease-modifying agents in which effects are believed to be long lasting. Of particular note, the secretase enzymes, a group of proteases responsible for the metabolism of the β-amyloid precursor protein (βAPP) and β-amyloid (Aβ) peptides production, have been underlined for their promising therapeutic potential. This review article attempts to comprehensively cover aspects related to the identification and use of drugs targeting the secretase enzymes. Particularly, the roles of secretases in the pathogenesis of AD and their therapeutic modulation are provided herein. Moreover, an overview of the drug development process and the contribution of computational (in silico) approaches for facilitating successful drug discovery are also highlighted along with examples of relevant computational works. Promising chemical scaffolds, inhibitors, and modulators against each class of secretases are also summarized herein. Additionally, multitarget secretase modulators are also taken into consideration in light of the current growing interest in the polypharmacology of complex diseases. Finally, challenging issues and future outlook relevant to the discovery of drugs targeting secretases are also discussed.
Collapse
Affiliation(s)
- Nalini Schaduangrat
- Faculty of Medical Technology, Center of Data Mining and Biomedical Informatics, Mahidol University, Bangkok, Thailand
| | - Veda Prachayasittikul
- Faculty of Medical Technology, Center of Data Mining and Biomedical Informatics, Mahidol University, Bangkok, Thailand
| | - Saowapak Choomwattana
- Faculty of Medical Technology, Center of Data Mining and Biomedical Informatics, Mahidol University, Bangkok, Thailand
| | - Prapimpun Wongchitrat
- Faculty of Medical Technology, Center for Research and Innovation, Mahidol University, Bangkok, Thailand
| | - Kamonrat Phopin
- Faculty of Medical Technology, Center for Research and Innovation, Mahidol University, Bangkok, Thailand
| | - Wilasinee Suwanjang
- Faculty of Medical Technology, Center for Research and Innovation, Mahidol University, Bangkok, Thailand
| | - Aijaz Ahmad Malik
- Faculty of Medical Technology, Center of Data Mining and Biomedical Informatics, Mahidol University, Bangkok, Thailand
| | - Bruno Vincent
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand.,Centre National de la Recherche Scientifique, Paris, France
| | - Chanin Nantasenamat
- Faculty of Medical Technology, Center of Data Mining and Biomedical Informatics, Mahidol University, Bangkok, Thailand
| |
Collapse
|
34
|
Xiong L, Boulouis G, Charidimou A, Roongpiboonsopit D, Jessel MJ, Pasi M, Reijmer YD, Fotiadis P, Ayres A, Merrill E, Schwab K, Blacker D, Gurol ME, Greenberg SM, Viswanathan A. Dementia incidence and predictors in cerebral amyloid angiopathy patients without intracerebral hemorrhage. J Cereb Blood Flow Metab 2018; 38:241-249. [PMID: 28318355 PMCID: PMC5951014 DOI: 10.1177/0271678x17700435] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cerebral amyloid angiopathy (CAA) is a common cause of cognitive impairment in older individuals. This study aimed to investigate predictors of dementia in CAA patients without intracerebral hemorrhage (ICH). A total of 158 non-demented patients from the Stroke Service or the Memory Clinic who met the modified Boston Criteria for probable CAA were included. At baseline, neuroimaging markers, including lobar microbleeds (cerebral microbleeds (CMBs)), white matter hyperintensities (WMH), cortical superficial siderosis (cSS), magnetic resonance imaging (MRI)-visible centrum semiovale perivascular spaces (CSO-PVS), lacunes, and medial temporal atrophy (MTA) were assessed. The overall burden of small vessel disease (SVD) for CAA was calculated by a cumulative score based on CMB number, WMH severity, cSS presence and extent and CSO-PVS severity. The estimated cumulative dementia incidence at 1 year was 14% (95% confidence interval (CI): 5%-23%), and 5 years 73% (95% CI: 55%, 84%). Age (hazard ratio (HR) 1.05 per year, 95% CI: 1.01-1.08, p = 0.007), presence of MCI status (HR 3.40, 95% CI: 1.97-6.92, p < 0.001), MTA (HR 1.71 per point, 95% CI: 1.26-2.32, p = 0.001), and SVD score (HR 1.23 per point, 95% CI: 1.20-1.48, p = 0.030) at baseline were independent predictors for dementia conversion in these patients. Cognitive deterioration of CAA patients appears attributable to cumulative changes, from both vasculopathic and neurodegenerative lesions.
Collapse
Affiliation(s)
- Li Xiong
- 1 Department of Neurology, Harvard Medical School, Boston, USA
| | | | | | - Duangnapa Roongpiboonsopit
- 1 Department of Neurology, Harvard Medical School, Boston, USA.,2 Department of Medicine, Naresuan University, Phitsanulok, Thailand
| | | | - Marco Pasi
- 1 Department of Neurology, Harvard Medical School, Boston, USA
| | - Yael D Reijmer
- 1 Department of Neurology, Harvard Medical School, Boston, USA
| | | | - Alison Ayres
- 1 Department of Neurology, Harvard Medical School, Boston, USA
| | - Emily Merrill
- 3 MIND Informatics, Harvard Medical School, Boston, USA
| | - Kristin Schwab
- 1 Department of Neurology, Harvard Medical School, Boston, USA
| | - Deborah Blacker
- 4 Department of Psychiatry, Harvard Medical School, Boston, USA
| | - M Edip Gurol
- 1 Department of Neurology, Harvard Medical School, Boston, USA
| | | | | |
Collapse
|
35
|
Cadasil. Neurologia 2017. [DOI: 10.1016/s1634-7072(17)85562-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
|
36
|
Kalaria RN, Ihara M. Medial temporal lobe atrophy is the norm in cerebrovascular dementias. Eur J Neurol 2017; 24:539-540. [PMID: 28191732 DOI: 10.1111/ene.13243] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 01/04/2017] [Indexed: 11/27/2022]
Affiliation(s)
- R N Kalaria
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - M Ihara
- Department of Stroke and Cerebrovascular Diseases, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| |
Collapse
|
37
|
Xiong L, Davidsdottir S, Reijmer YD, Shoamanesh A, Roongpiboonsopit D, Thanprasertsuk S, Martinez-Ramirez S, Charidimou A, Ayres AM, Fotiadis P, Gurol E, Blacker DL, Greenberg SM, Viswanathan A. Cognitive Profile and its Association with Neuroimaging Markers of Non-Demented Cerebral Amyloid Angiopathy Patients in a Stroke Unit. J Alzheimers Dis 2017; 52:171-8. [PMID: 27060947 DOI: 10.3233/jad-150890] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Cerebral amyloid angiopathy (CAA) is increasingly recognized as a cause of cognitive impairment in the elderly, but the cognitive profile in patients with the disease has not been well characterized. OBJECTIVE To characterize the neuropsychological profile of CAA patients without dementia and to determine the association between cognitive performance in different domains and neuroimaging lesions characteristic of CAA. METHODS Fifty-eight non-demented CAA patients were compared to 138 cognitively normal subjects using a standard neuropsychological test battery. Total brain volume (TBV), white matter hyperintensities, number of lobar cerebral microbleeds, hippocampal volume, and cortical superficial siderosis in all CAA patients were assessed. The association between these neuroimaging markers and neuropsychological performance in different cognitive domains in the CAA group were analyzed. RESULTS Patients with CAA had significantly worse performance on all individual neuropsychological domains tested, when compared to the cognitive normal group. The cognitive decline of CAA patients was most noticeable in tests for processing speed with a Z score of -1.92±1.56 (mean±SD), then followed by executive function (-0.93±1.01), episodic memory (-0.87±1.29), semantic fluency (-0.73±1.06), and attention (-0.42±0.98). TBV of the CAA patients was correlated with processing speed (β= 0.335, p = 0.03) and executive function (β= 0.394, p = 0.01). CONCLUSIONS Non-demented patients with CAA had cognitive deficits in multiple areas. Lower TBV was related to slower processing speed and worse executive function.
Collapse
Affiliation(s)
- Li Xiong
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Department of Neurology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Sigurros Davidsdottir
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yael D Reijmer
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ashkan Shoamanesh
- Department of Neurology, McMaster University / Population Health Research Institute, Canada
| | - Duangnapa Roongpiboonsopit
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Department of Medicine, Naresuan University, Phitsanulok, Thailand
| | | | - Sergi Martinez-Ramirez
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Andreas Charidimou
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Alison M Ayres
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Panagiotis Fotiadis
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Edip Gurol
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Deborah L Blacker
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Steven M Greenberg
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Anand Viswanathan
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
38
|
Abstract
Vascular dementia (VaD) is a major contributor to the dementia syndrome and is described as having problems with reasoning, planning, judgment, and memory caused by impaired blood flow to the brain and damage to the blood vessels resulting from events such as stroke. There are a variety of etiologies that contribute to the development of vascular cognitive impairment and VaD, and these are often associated with other dementia-related pathologies such as Alzheimer disease. The diagnosis of VaD is difficult due to the number and types of lesions and their locations in the brain. Factors that increase the risk of vascular diseases such as stroke, high blood pressure, high cholesterol, and smoking also raise the risk of VaD. Therefore, controlling these risk factors can help lower the chances of developing VaD. This update describes the subtypes of VaD, with details of their complex presentation, associated pathological lesions, and issues with diagnosis, prevention, and treatment.
Collapse
Affiliation(s)
- Ayesha Khan
- Wolfson Centre for Age Related Diseases, Guys Campus, London, United Kingdom of Great Britain and Northern Ireland Institute of NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Raj N Kalaria
- Institute for Ageing and Health, Wolfson Research Centre, Campus for Ageing & Vitality, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Anne Corbett
- Wolfson Centre for Age Related Diseases, Guys Campus, London, United Kingdom of Great Britain and Northern Ireland
| | - Clive Ballard
- Wolfson Centre for Age Related Diseases, Guys Campus, London, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
39
|
Neuropathological diagnosis of vascular cognitive impairment and vascular dementia with implications for Alzheimer's disease. Acta Neuropathol 2016; 131:659-85. [PMID: 27062261 PMCID: PMC4835512 DOI: 10.1007/s00401-016-1571-z] [Citation(s) in RCA: 291] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 03/23/2016] [Accepted: 03/24/2016] [Indexed: 12/16/2022]
Abstract
Vascular dementia (VaD) is recognised as a neurocognitive disorder, which is explained by numerous vascular causes in the general absence of other pathologies. The heterogeneity of cerebrovascular disease makes it challenging to elucidate the neuropathological substrates and mechanisms of VaD as well as vascular cognitive impairment (VCI). Consensus and accurate diagnosis of VaD relies on wide-ranging clinical, neuropsychometric and neuroimaging measures with subsequent pathological confirmation. Pathological diagnosis of suspected clinical VaD requires adequate postmortem brain sampling and rigorous assessment methods to identify important substrates. Factors that define the subtypes of VaD include the nature and extent of vascular pathologies, degree of involvement of extra and intracranial vessels and the anatomical location of tissue changes. Atherosclerotic and cardioembolic diseases appear the most common substrates of vascular brain injury or infarction. Small vessel disease characterised by arteriolosclerosis and lacunar infarcts also causes cortical and subcortical microinfarcts, which appear to be the most robust substrates of cognitive impairment. Diffuse WM changes with loss of myelin and axonal abnormalities are common to almost all subtypes of VaD. Medial temporal lobe and hippocampal atrophy accompanied by variable hippocampal sclerosis are also features of VaD as they are of Alzheimer’s disease. Recent observations suggest that there is a vascular basis for neuronal atrophy in both the temporal and frontal lobes in VaD that is entirely independent of any Alzheimer pathology. Further knowledge on specific neuronal and dendro-synaptic changes in key regions resulting in executive dysfunction and other cognitive deficits, which define VCI and VaD, needs to be gathered. Hereditary arteriopathies such as cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy or CADASIL have provided insights into the mechanisms of dementia associated with cerebral small vessel disease. Greater understanding of the neurochemical and molecular investigations is needed to better define microvascular disease and vascular substrates of dementia. The investigation of relevant animal models would be valuable in exploring the pathogenesis as well as prevention of the vascular causes of cognitive impairment.
Collapse
|
40
|
Kebets V, Gregoire SM, Charidimou A, Barnes J, Rantell K, Brown MM, Jäger HR, Cipolotti L, Werring DJ. Prevalence and cognitive impact of medial temporal atrophy in a hospital stroke service: retrospective cohort study. Int J Stroke 2015; 10:861-7. [PMID: 26043795 DOI: 10.1111/ijs.12544] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 03/24/2015] [Indexed: 11/30/2022]
Abstract
BACKGROUND Cerebrovascular disease and neurodegeneration cause cognitive impairment and frequently coexist. AIMS Our objectives were to investigate the prevalence and cognitive impact of medial temporal lobe atrophy - a radiological marker often associated with Alzheimer's disease - in a hospital stroke service. METHODS Retrospective cohort study of patients from a hospital stroke service. Patients assessed for suspected ischemic stroke or transient ischemic attack, irrespective of final diagnosis, underwent neuropsychological testing and magnetic resonance imaging. medial temporal lobe atrophy, white matter hyperintensities, lacunes, and cerebral microbleeds were rated using established criteria and validated scales. The associations between medial temporal lobe atrophy and cognition were tested using multivariable logistic regression analyses, adjusted for age and imaging markers of cerebrovascular disease. RESULTS Three hundred and ninety-three patients were included, of whom 169 (43%; 95% confidence interval: 38·1-48·1%) had medial temporal lobe atrophy; in 38 patients (9·7%), medial temporal lobe atrophy was severe (mean score ≥2). In unadjusted logistic regression analyses in the whole cohort, mean medial temporal lobe atrophy score was associated with verbal memory, nominal and perceptual skills, executive function, and speed and attention. After adjustment for age, white matter hyperintensities, number of lacunes, presence of cerebral microbleeds, previous ischemic stroke or transient ischemic attack, and premorbid intelligence quotient, mean medial temporal lobe atrophy score remained associated with impairment in verbal memory (odds ratio: 1·64; 95% confidence interval 1·04-2·58) and nominal skills (odds ratio: 1·61; 95% confidence interval 1·04-2·48). CONCLUSIONS Medial temporal lobe atrophy is common and has an independent impact on cognitive function in a stroke service population, independent of confounding factors including age and magnetic resonance imaging markers of cerebrovascular disease. Medial temporal lobe atrophy is independently related to verbal memory and nominal skills, while small vessel pathology also contributes to speed and attention, and executive and perceptual functions.
Collapse
Affiliation(s)
- Valeria Kebets
- Stroke Research Group, Department of Brain Repair and Rehabilitation, UCL Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - Simone M Gregoire
- Stroke Research Group, Department of Brain Repair and Rehabilitation, UCL Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - Andreas Charidimou
- Stroke Research Group, Department of Brain Repair and Rehabilitation, UCL Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - Josephine Barnes
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
| | - Khadija Rantell
- Department of Biostatistics, Joint UCL/UCH/RFH Research Support Centre, National Hospital for Neurology and Neurosurgery, London, UK
| | - Martin M Brown
- Stroke Research Group, Department of Brain Repair and Rehabilitation, UCL Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - Hans R Jäger
- Neuroradiological Academic Unit, National Hospital for Neurology and Neurosurgery, London, UK
| | - Lisa Cipolotti
- Department of Neuropsychology, Università di Palermo, Palermo, Italy.,Dipartimento di Psicologia, Università di Palermo, Palermo, Italy
| | - David J Werring
- Stroke Research Group, Department of Brain Repair and Rehabilitation, UCL Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| |
Collapse
|
41
|
Xia W, Zhang B, Yang Y, Wang P, Yang Y, Wang S. Poorly controlled cholesterol is associated with cognitive impairment in T2DM: a resting-state fMRI study. Lipids Health Dis 2015; 14:47. [PMID: 25989796 PMCID: PMC4490615 DOI: 10.1186/s12944-015-0046-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Accepted: 05/13/2015] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Debate remains on whether hypercholesterolemia is associated with cognitive impairment. Hence, we investigated whether poorly controlled cholesterol impairs functional connectivity among patients with type 2 diabetes mellitus (T2DM). METHODS Resting-state functional connectivity infers to an interregional cooperation characterized by synchronous and low-frequency (<0.08 Hz) fluctuations on blood oxygen level-dependent functional magnetic resonance imaging (fMRI). We used resting-state fMRI to investigate the functional connectivity of 25 T2DM patients with poorly controlled cholesterol, 22 patients with target cholesterol and 26 healthy controls. Further correlation analysis was conducted between the functional connectivity and clinical data as well as neuropsychological tests. RESULTS The three groups did not statistically differ in age, sex, education level, body mass index, blood pressure, fasting C-peptides, and triglyceride. Compared with target cholesterol patients, patients with poorly controlled cholesterol showed significantly increased levels of serum cholesterol, low-density lipoprotein (LDL), and LDL/high-density lipoproteins (HDL) ratio, as well as poor performance in Trail Making Test B (TMT-B) (p<0.05). Disordered functional connectivity of bilateral hippocampus-middle frontal gyrus (MFG) in the poorly controlled group consistently existed when compared with the two other groups. Moreover, the aberrant functional connectivity was associated with the TMT-B scores and the LDL/HDL index in T2DM patients with poorly controlled cholesterol. CONCLUSIONS T2DM patients with poorly controlled cholesterol showed impaired attention and executive function. The resting-state connectivity disturbance of the hippocampus-MFG may be involved in this process. Decreasing the LDL/HDL ratio can be taken as precaution against cognitive decrements.
Collapse
Affiliation(s)
- Wenqing Xia
- Department of Endocrinology, ZhongDa Hospital of Southeast University, No.87 Dingjiaqiao Road, Nanjing, 210009, PR China.
- Medical school of Southeast University, No.87 Dingjiaqiao Road, Nanjing, 210009, PR China.
- Center for functional Neuroimaging, University of Pennsylvania, 3710 Hamilton Walk, Philadelphia, PA, 19104, USA.
| | - Bin Zhang
- Outpatient Depart, Panda Group Community Health Service Centre, No.4 Qingxi Road, Nanjing, 210009, PR China.
| | - Yang Yang
- Outpatient Depart, Panda Group Community Health Service Centre, No.4 Qingxi Road, Nanjing, 210009, PR China.
| | - Pin Wang
- Department of Endocrinology, ZhongDa Hospital of Southeast University, No.87 Dingjiaqiao Road, Nanjing, 210009, PR China.
| | - Yue Yang
- Department of Endocrinology, ZhongDa Hospital of Southeast University, No.87 Dingjiaqiao Road, Nanjing, 210009, PR China.
| | - Shaohua Wang
- Department of Endocrinology, ZhongDa Hospital of Southeast University, No.87 Dingjiaqiao Road, Nanjing, 210009, PR China.
- Medical school of Southeast University, No.87 Dingjiaqiao Road, Nanjing, 210009, PR China.
| |
Collapse
|
42
|
Craggs LJL, Yamamoto Y, Ihara M, Fenwick R, Burke M, Oakley AE, Roeber S, Duering M, Kretzschmar H, Kalaria RN. White matter pathology and disconnection in the frontal lobe in cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL). Neuropathol Appl Neurobiol 2015; 40:591-602. [PMID: 23844775 PMCID: PMC4282433 DOI: 10.1111/nan.12073] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 07/04/2013] [Indexed: 11/29/2022]
Abstract
BACKGROUND Magnetic resonance imaging indicates diffuse white matter (WM) changes are associated with cognitive impairment in cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL). We examined whether the distribution of axonal abnormalities is related to microvascular pathology in the underlying WM. METHODS We used post-mortem brains from CADASIL subjects and similar age cognitively normal controls to examine WM axonal changes, microvascular pathology, and glial reaction in up to 16 different regions extending rostro-caudally through the cerebrum. Using unbiased stereological methods, we estimated length densities of affected axons immunostained with neurofilament antibody SMI32. Standard immunohistochemistry was used to assess amyloid precursor protein immunoreactivity per WM area. To relate WM changes to microvascular pathology, we also determined the sclerotic index (SI) in WM arterioles. RESULTS The degree of WM pathology consistently scored higher across all brain regions in CADASIL subjects (P<0.01) with the WM underlying the primary motor cortex exhibiting the most severe change. SMI32 immunoreactive axons in CADASIL were invariably increased compared with controls (P<0.01), with most prominent axonal abnormalities observed in the frontal WM (P<0.05). The SIs of arterioles in CADASIL were increased by 25-45% throughout the regions assessed, with the highest change in the mid-frontal region (P=0.000). CONCLUSIONS Our results suggest disruption of either cortico-cortical or subcortical-cortical networks in the WM of the frontal lobe that may explain motor deficits and executive dysfunction in CADASIL. Widespread WM axonal changes arise from differential stenosis and sclerosis of arterioles in the WM of CADASIL subjects, possibly affecting some axons of projection neurones connecting to targets in the subcortical structures.
Collapse
Affiliation(s)
- Lucinda J L Craggs
- Institute for Ageing and Health, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Ehret F, Vogler S, Pojar S, Elliott DA, Bradke F, Steiner B, Kempermann G. Mouse model of CADASIL reveals novel insights into Notch3 function in adult hippocampal neurogenesis. Neurobiol Dis 2014; 75:131-41. [PMID: 25555543 DOI: 10.1016/j.nbd.2014.12.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 12/10/2014] [Accepted: 12/21/2014] [Indexed: 10/24/2022] Open
Abstract
Could impaired adult hippocampal neurogenesis be a relevant mechanism underlying CADASIL (cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy)? Memory symptoms in CADASIL, the most common hereditary form of vascular dementia, are usually thought to be primarily due to vascular degeneration and white matter lacunes. Since adult hippocampal neurogenesis, a process essential for the integration of new spatial memory occurs in a highly vascularized niche, we considered dysregulation of adult neurogenesis as a potential mechanism for the manifestation of dementia in CADASIL. Analysis in aged mice overexpressing Notch3 with a CADASIL mutation, revealed vascular deficits in arteries of the hippocampal fissure but not in the niche of the dentate gyrus. At 12 months of age, cell proliferation and survival of newborn neurons were reduced not only in CADASIL mice but also in transgenic controls overexpressing wild type Notch3. At 6 months, hippocampal neurogenesis was altered in CADASIL mice independent of overt vascular abnormalities in the fissure. Further, we identified Notch3 expression in hippocampal precursor cells and maturing neurons in vivo as well as in cultured hippocampal precursor cells. Overexpression and knockdown experiments showed that Notch3 signaling negatively regulated precursor cell proliferation. Notch3 overexpression also led to deficits in KCl-induced precursor cell activation. This suggests a cell-autonomous effect of Notch3 signaling in the regulation of precursor proliferation and activation and a loss-of-function effect in CADASIL. Consequently, besides vascular damage, aberrant precursor cell proliferation and differentiation due to Notch3 dysfunction might be an additional independent mechanism for the development of hippocampal dysfunction in CADASIL.
Collapse
Affiliation(s)
- Fanny Ehret
- German Center for Neurodegenerative Diseases (DZNE) Dresden, 01307 Dresden, Germany
| | - Steffen Vogler
- CRTD - Center for Regenerative Therapies Dresden, Genomics of Regeneration, Technische Universität Dresden, 01307 Dresden, Germany
| | - Sherin Pojar
- German Center for Neurodegenerative Diseases (DZNE) Dresden, 01307 Dresden, Germany
| | - David A Elliott
- German Center for Neurodegenerative Diseases (DZNE) Bonn, 53175 Bonn, Germany
| | - Frank Bradke
- German Center for Neurodegenerative Diseases (DZNE) Bonn, 53175 Bonn, Germany
| | - Barbara Steiner
- Department of Neurology, Charité University Medicine Berlin, 10117 Berlin, Germany
| | - Gerd Kempermann
- German Center for Neurodegenerative Diseases (DZNE) Dresden, 01307 Dresden, Germany; CRTD - Center for Regenerative Therapies Dresden, Genomics of Regeneration, Technische Universität Dresden, 01307 Dresden, Germany.
| |
Collapse
|
44
|
Staniloiu A, Woermann FG, Markowitsch HJ. Impairments in Episodic-Autobiographical Memory and Emotional and Social Information Processing in CADASIL during Mid-Adulthood. Front Behav Neurosci 2014; 8:227. [PMID: 25009481 PMCID: PMC4069576 DOI: 10.3389/fnbeh.2014.00227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 06/05/2014] [Indexed: 11/24/2022] Open
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) - is the most common genetic source of vascular dementia in adults, being caused by a mutation in NOTCH3 gene. Spontaneous de novo mutations may occur, but their frequency is largely unknown. Ischemic strokes and cognitive impairments are the most frequent manifestations, but seizures affect up to 10% of the patients. Herein, we describe a 47-year-old male scholar with a genetically confirmed diagnosis of CADASIL (Arg133Cys mutation in the NOTCH3 gene) and a seemingly negative family history of CADASIL illness, who was investigated with a comprehensive neuropsychological testing battery and neuroimaging methods. The patient demonstrated on one hand severe and accelerated deteriorations in multiple cognitive domains such as concentration, long-term memory (including the episodic-autobiographical memory domain), problem solving, cognitive flexibility and planning, affect recognition, discrimination and matching, and social cognition (theory of mind). Some of these impairments were even captured by abbreviated instruments for investigating suspicion of dementia. On the other hand the patient still possessed high crystallized (verbal) intelligence and a capacity to put forth a façade of well-preserved intellectual functioning. Although no definite conclusions can be drawn from a single case study, our findings point to the presence of additional cognitive changes in CADASIL in middle adulthood, in particular to impairments in the episodic-autobiographical memory domain and social information processing (e.g., social cognition). Whether these identified impairments are related to the patient's specific phenotype or to an ascertainment bias (e.g., a paucity of studies investigating these cognitive functions) requires elucidation by larger scale research.
Collapse
Affiliation(s)
- Angelica Staniloiu
- Physiological Psychology, University of Bielefeld, Bielefeld, Germany
- Hanse Institute of Advanced Science, Delmenhorst, Germany
| | | | - Hans J. Markowitsch
- Physiological Psychology, University of Bielefeld, Bielefeld, Germany
- Center of Excellence “Cognitive Interaction Technology” (CITEC), University of Bielefeld, Bielefeld, Germany
| |
Collapse
|
45
|
Liu C, Li C, Gui L, Zhao L, Evans AC, Xie B, Zhang J, Wei L, Zhou D, Wang J, Yin X. The pattern of brain gray matter impairments in patients with subcortical vascular dementia. J Neurol Sci 2014; 341:110-118. [PMID: 24798224 DOI: 10.1016/j.jns.2014.04.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 04/01/2014] [Accepted: 04/14/2014] [Indexed: 11/23/2022]
Abstract
Though subcortical ischemic vascular dementia (SIVD) is known to initially affect subcortical regions, numerous brain imaging studies have also documented the widespread cortical alternations. Here we collected brain structural magnetic resonance imaging data from 34 SIVD patients and 35 healthy controls. Voxel-based morphometry (VBM), cortical thickness (and surface area) analysis and deep gray matter volume measurements were performed. VBM analysis showed gray matter volume reduction in lateral and medial temporal lobes, as well as orbitofrontal cortex in SIVD patients. The surface-based analyses revealed more subtle structural differences in the perisylvian area, medial temporal lobe, anterior and posterior cingulate, as well as prefrontal areas. Furthermore, analyses of deep gray matter demonstrated significant atrophy of the hippocampus, amygdala, nucleus accumbens and other nuclei. Finally, we found that thinning in the hippocampus and anterior cingulate cortex, as well as the volume decline in thalamus, caudate nucleus and amygdala was correlated with the cognitive impairment in patients. In conclusion, our study showed the structural abnormalities of the hippocampus and its associated outflow areas, as well as cortices implicated in cholinergic circuits in SIVD. These findings may bring new insights into the dysfunction of brain gray matter in SIVD.
Collapse
Affiliation(s)
- Chen Liu
- Department of Radiology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Chuanming Li
- Department of Radiology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Li Gui
- Department of Neurology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Lu Zhao
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec H3A2B4, Canada
| | - Alan C Evans
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec H3A2B4, Canada
| | - Bing Xie
- Department of Radiology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Jiuquan Zhang
- Department of Radiology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Luqing Wei
- Department of Radiology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Daiquan Zhou
- Department of Radiology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Jian Wang
- Department of Radiology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China.
| | - Xuntao Yin
- Department of Radiology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China; McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec H3A2B4, Canada.
| |
Collapse
|
46
|
Yin X, Liu C, Gui L, Zhao L, Zhang J, Wei L, Xie B, Zhou D, Li C, Wang J. Comparison of medial temporal measures between Binswanger's disease and Alzheimer's disease. PLoS One 2014; 9:e86423. [PMID: 24466084 PMCID: PMC3900523 DOI: 10.1371/journal.pone.0086423] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 12/10/2013] [Indexed: 01/23/2023] Open
Abstract
Binswanger's disease (BD) is a common cause of vascular dementia in elderly patients; however, few studies have investigated the medial temporal lobe (MTL) atrophy in BD, and the differences in the atrophic patterns between BD and Alzheimer's disease (AD) remain largely unknown. Such knowledge is essential for understanding the pathologic basis of dementia. In this study, we collected structural magnetic resonance imaging (MRI) data from 16 normal controls, 14 patients with AD and 14 patients with BD. The volumes of the hippocampus and amygdala, and morphologic parameters (volume, surface area, cortical thickness and mean curvature) of the entorhinal cortex (ERC) and perirhinal cortex (PRC) were calculated using an automated approach. Volume reduction of the hippocampus, amygdala and ERC, and disturbance of the PRC curvature was found in both AD and BD patients compared with the controls (p<0.05, uncorrected). There were no significant differences among all the structural measures between the AD and BD patients. Finally, partial correlation analyses revealed that cognitive decline could be attributed to ERC thinning in AD and volume reduction of PRC in BD. We conclude that AD and BD exhibit similar atrophy patterns in the medial temporal cortices and deep gray matter but have distinct pathologic bases for cognitive impairments. Although atrophy of the MTL structures is a sensitive biomarker for AD, it is not superior for discrimination between AD and BD.
Collapse
Affiliation(s)
- Xuntao Yin
- Department of Radiology, Southwest Hospital, Third Military Medical University, Chongqing, China
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Chen Liu
- Department of Radiology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Li Gui
- Department of Neurology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Lu Zhao
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Jiuquan Zhang
- Department of Radiology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Luqing Wei
- Department of Radiology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Bing Xie
- Department of Radiology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Daiquan Zhou
- Department of Radiology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Chuanming Li
- Department of Radiology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jian Wang
- Department of Radiology, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
47
|
Ragno M, Trojano L. Management of CADASIL syndrome. Expert Opin Orphan Drugs 2013. [DOI: 10.1517/21678707.2013.828998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Michele Ragno
- Division of Neurology, Ospedale Madonna del Soccorso, San Benedetto del Tronto (AP), Italy
| | - Luigi Trojano
- Second University of Naples, Department of Psychology, Caserta, Italy ;
- Rehabilitation Institute of Telese Terme, Salvatore Maugeri Foundation, IRCCS, Telese Terme (BN), Italy
| |
Collapse
|
48
|
Ciolli L, Pescini F, Salvadori E, Del Bene A, Pracucci G, Poggesi A, Nannucci S, Valenti R, Basile AM, Squarzanti F, Bianchi S, Dotti MT, Adriano E, Balestrino M, Federico A, Gandolfo C, Inzitari D, Pantoni L. Influence of vascular risk factors and neuropsychological profile on functional performances in CADASIL: results from the MIcrovascular LEukoencephalopathy Study (MILES). Eur J Neurol 2013; 21:65-71. [PMID: 23869710 DOI: 10.1111/ene.12241] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2013] [Accepted: 06/17/2013] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND PURPOSE Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is an inherited cerebral small vessel disease that may lead to disability and whose phenotype modulators are still unknown. METHODS In the MIcrovascular LEukoencephalopathy Study (MILES), we assessed the influence of vascular risk factors and the effect of different cognitive domains (memory, psychomotor speed and executive functions) performances on functional abilities in CADASIL in comparison with age-related leukoencephalopathy (ARL). RESULTS We evaluated 51 CADASIL patients (mean age 50.3 ± 13.8 years, 47.1% males) and 68 ARL patients (70.6 ± 7.4 years, 58.8% males). Considering vascular risk factors, after adjustment for age, CADASIL patients had higher mean BMI values than ARL patients. Stroke history frequency was similar in the two groups. After adjustment for age, more CADASIL patients were disabled (impaired on ≥ 2 items of the Instrumental Activities of Daily Living scale) in comparison with ARL patients, and CADASIL patients had worse functional performances evaluated with the Disability Assessment for Dementia (DAD) scale. In CADASIL patients, hypertension was related to both DAD score and disability. The cognitive profile of CADASIL and ARL patients was similar, but on a stepwise linear regression analysis functional performances were mainly associated with the memory index (β = -0.418, P < 0.003) in CADASIL patients and the executive function index (β = -0.321, P = 0.028) in ARL. CONCLUSIONS This study suggests that hypertension may contribute to functional impairment in CADASIL and that memory impairment has a large influence on functional decline in contrast with that observed in a sample of subjects with ARL.
Collapse
Affiliation(s)
- L Ciolli
- NEUROFARBA Department, Neuroscience section, University of Florence, Florence, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Giorgio A, De Stefano N. Clinical use of brain volumetry. J Magn Reson Imaging 2013; 37:1-14. [PMID: 23255412 DOI: 10.1002/jmri.23671] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 03/12/2012] [Indexed: 12/13/2022] Open
Abstract
Magnetic resonance imaging (MRI)-based brain volumetry is increasingly being used in the clinical setting to assess brain volume changes from structural MR images in a range of neurologic conditions. Measures of brain volumes have been shown to be valid biomarkers of the clinical state and progression by offering high reliability and robust inferences on the underlying disease-related mechanisms. This review critically examines the different scenarios of the application of MRI-based brain volumetry in neurology: 1) supporting disease diagnosis, 2) understanding mechanisms and tracking clinical progression of disease, and 3) monitoring treatment effect. These aspects will be discussed in a wide range of neurologic conditions, with particular emphasis on Alzheimer's disease and multiple sclerosis.
Collapse
Affiliation(s)
- Antonio Giorgio
- Department of Neurological and Behavioral Sciences, University of Siena, Italy
| | | |
Collapse
|
50
|
Neuropathological substrates and structural changes in late-life depression: the impact of vascular burden. Acta Neuropathol 2012; 124:453-64. [PMID: 22836715 DOI: 10.1007/s00401-012-1021-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Revised: 07/13/2012] [Accepted: 07/17/2012] [Indexed: 01/01/2023]
Abstract
A first episode of depression after 65 years of age has long been associated with both severe macrovascular and small microvascular pathology. Among the three more frequent forms of depression in old age, post-stroke depression has been associated with an abrupt damage of cortical circuits involved in monoamine production and mood regulation. Late-onset depression (LOD) in the absence of stroke has been related to lacunes and white matter lesions that invade both the neocortex and subcortical nuclei. Recurrent late-life depression is thought to induce neuronal loss in the hippocampal formation and white matter lesions that affect limbic pathways. Despite an impressive number of magnetic resonance imaging (MRI) studies in this field, the presence of a causal relationship between structural changes in the human brain and LOD is still controversial. The present article provides a critical overview of the contribution of neuropathology in post-stroke, late-onset, and late-life recurrent depression. Recent autopsy findings challenge the role of stroke location in the occurrence of post-stroke depression by pointing to the deleterious effect of subcortical lacunes. Despite the lines of evidences supporting the association between MRI-assessed white matter changes and mood dysregulation, lacunes, periventricular and deep white matter demyelination are all unrelated to the occurrence of LOD. In the same line, neuropathological data show that early-onset depression is not associated with an acceleration of aging-related neurodegenerative changes in the human brain. However, they also provide data in favor of the neurotoxic theory of depression by showing that neuronal loss occurs in the hippocampus of chronically depressed patients. These three paradigms are discussed in the light of the complex relationships between psychosocial determinants and biological vulnerability in affective disorders.
Collapse
|