1
|
Yang J, Liu Y, Ma Y, Zhang W, Han L, Feng H, Chen M, Zhong J. Association of glymphatic clearance function with imaging markers and risk factors of cerebral small vessel disease. J Stroke Cerebrovasc Dis 2025; 34:108187. [PMID: 39667440 DOI: 10.1016/j.jstrokecerebrovasdis.2024.108187] [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: 10/31/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 12/14/2024] Open
Abstract
BACKGROUND Cerebral small vessel disease (CSVD) is a common disease in the elderly, and its pathogenesis is still being explored. Glymphatic clearance function can be evaluated by diffusion tensor imaging analysis along the perivascular space (DTI-ALPS) index. This study aims to investigate the changes in glymphatic clearance function in CSVD patients and its relationship with imaging markers and risk factors of CSVD. METHODS The DTI-ALPS index of all participants was calculated. The DTI-ALPS index was compared between the patient group and healthy controls (HCs) group. Pearson correlation analysis was used to analyze the relation between the DTI-ALPS index and CSVD imaging markers, and to explore the effect of mean diffusivity (MD) as a covariate. Regression analysis was used to investigate the correlation between DTI-ALPS index and risk factors. RESULTS The DTI-ALPS index in the bilateral hemispheres of CSVD patients was significantly lower than that in the HCs group (p < 0.001). The DTI-ALPS index in the bilateral hemisphere of CSVD patients was negatively correlated with the grade of EPVS in basal ganglia. There was a significant negative correlation between the left DTI-ALPS index and lacunas, the right DTI-ALPS index and DWMHs. After removing the covariate MD, there was no significant correlation between the DTI-ALPS index and CSVD imaging markers. The DTI-ALPS index was associated with gender, diabetes, drinking and smoking. CONCLUSIONS The CSVD patients have glymphatic clearance dysfunction, which may be related to the imaging features and CSVD risk factors. Meanwhile, it's recommended to consider removing MD as mixed signal.
Collapse
Affiliation(s)
- Jie Yang
- Department of Radiology, Zigong First People's Hospital, Zigong, PR China
| | - Yujian Liu
- Sichuan Vocational College of Health and Rehabilitation, Zigong, PR China
| | - Yuanying Ma
- Department of Radiology, Zigong First People's Hospital, Zigong, PR China
| | - Wei Zhang
- Department of Radiology, Zigong First People's Hospital, Zigong, PR China
| | - Limei Han
- Department of Radiology, Zigong First People's Hospital, Zigong, PR China
| | - Hao Feng
- Department of Radiology, Zigong First People's Hospital, Zigong, PR China
| | - Meining Chen
- MR Research Collaboration, Siemens Healthineers, Shanghai, PR China
| | - Jianquan Zhong
- Department of Radiology, Zigong First People's Hospital, Zigong, PR China.
| |
Collapse
|
2
|
Ford AL, Fellah S, Wang Y, Unger-Levinson K, Hagan M, Reis MN, Mirro A, Lewis JB, Ying C, Guilliams KP, Fields ME, An H, King AA, Chen Y. Brain Age Modeling and Cognitive Outcomes in Young Adults With and Without Sickle Cell Anemia. JAMA Netw Open 2025; 8:e2453669. [PMID: 39821401 PMCID: PMC11742535 DOI: 10.1001/jamanetworkopen.2024.53669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 10/31/2024] [Indexed: 01/19/2025] Open
Abstract
Importance Both sickle cell anemia (SCA) and socioeconomic status have been associated with altered brain structure and cognitive disability, yet precise mechanisms underlying these associations are unclear. Objective To determine whether brains of individuals with and without SCA appear older than chronological age and if brain age modeling using brain age gap (BAG) can estimate cognitive outcomes and mediate the association of socioeconomic status and disease with these outcomes. Design, Setting, and Participants In this cross-sectional study of 230 adults with and without SCA, individuals underwent brain magnetic resonance imaging (MRI) and cognitive assessment. Brain age was estimated using DeepBrainNet, a model trained to estimate chronological age from 14 468 structural MRIs from healthy individuals across the lifespan. BAG was defined as estimated brain age minus chronological age. Linear regression examined clinical factors associated with BAG and the ability of BAG to estimate cognitive performance compared to neuroimaging metrics of brain health and ischemic brain injury, such as normalized whole brain volume, white matter mean diffusivity (MD), and infarct volume. BAG and white matter MD were tested further as mediators of the association of socioeconomic status and SCA with cognitive performance. Data were analyzed from October 15, 2023, to July 1, 2024. Exposures SCA disease status and economic deprivation as measured using the area deprivation index (ADI). Main Outcome and Measures Executive function, crystallized function, processing speed, and full-scale intelligence quotient (FSIQ) were derived from the National Institutes of Health (NIH) Toolbox and Wechsler Abbreviated Scale of Intelligence, Second Edition. Results Among 230 included adults, 123 individuals had SCA (median [IQR] age, 26.4 [21.8-34.3] years; 77 female [63%]) and 107 individuals did not (control cohort; median [IQR] age, 30.1 [26.3-34.8] years; 77 female [72%]). Participants with SCA had a larger median (IQR) BAG compared to individuals in the control cohort (14.2 [8.0-19.2] vs 7.3 [3.2-11.1] years; median difference, 6.13 years; 95% CI, 4.29-8.05 years; P < .001). Individuals in the control cohort demonstrated a larger BAG relative to the reference population (mean difference, 7.52 years; 95% CI, 6.32-8.72 years; P < .001). Higher economic deprivation was associated with BAG in the control cohort (β [SE] per 1% ADI increase, 0.079 [0.028]; 95% CI, 0.023 to 0.135; P = .006), while intracranial vasculopathy (β [SE], 6.562 [1.883]; 95% CI, 2.828 to 10.296; P < .001) and hemoglobin S percentage (β [SE] per 1% increase, 0.089 [0.032]; 95% CI, 0.026 to 0.151; P = .006) were associated with BAG in participants with SCA. Across neuroimaging metrics of brain health, BAG demonstrated the largest effect size for cognitive outcomes in the control cohort (eg, executive function: r = -0.430; P = .001), while white matter MD demonstrated the largest effect size for cognitive outcomes (eg, executive function: r = -0.365; P = .001) in the SCA cohort. Across the study population, BAG mediated the association of ADI with cognitive performance (eg, executive function: β [SE] per 1-unit decrease in ADI, -0.031 [0.014]; 95% CI, -0.061 to -0.006), while BAG (eg, FSIQ: β [SE], -3.79 [1.42]; 95% CI, -6.87 to -1.40) and white matter MD (eg, FSIQ: β [SE], -4.55 [1.82]; 95% CI, -8.14 to -0.94) mediated the association of SCA with cognitive performance. Conclusions and Relevance Adults with SCA and a healthy control cohort with greater economic deprivation demonstrated older brain age, suggestive of insufficient brain development, premature brain aging, or both. Brain estimates of chronological age may inform mechanisms of the association between chronic disease and socioeconomic status with cognitive outcomes in healthy and SCA populations, yet will require confirmation in larger and longitudinal studies.
Collapse
Affiliation(s)
- Andria L. Ford
- Department of Neurology, Washington University in St Louis School of Medicine, St Louis, Missouri
- Mallinckrodt Institute of Radiology, Washington University in St Louis School of Medicine, St Louis, Missouri
| | - Slim Fellah
- Department of Neurology, Washington University in St Louis School of Medicine, St Louis, Missouri
| | - Yan Wang
- Department of Neurology, Washington University in St Louis School of Medicine, St Louis, Missouri
| | - Kira Unger-Levinson
- Department of Neurology, Washington University in St Louis School of Medicine, St Louis, Missouri
| | - Maria Hagan
- Department of Neurology, Washington University in St Louis School of Medicine, St Louis, Missouri
| | - Martin N. Reis
- Mallinckrodt Institute of Radiology, Washington University in St Louis School of Medicine, St Louis, Missouri
| | - Amy Mirro
- Department of Pediatrics, Washington University in St Louis School of Medicine, St Louis, Missouri
| | - Josiah B. Lewis
- Department of Neurology, Washington University in St Louis School of Medicine, St Louis, Missouri
| | - Chunwei Ying
- Mallinckrodt Institute of Radiology, Washington University in St Louis School of Medicine, St Louis, Missouri
| | - Kristin P. Guilliams
- Department of Neurology, Washington University in St Louis School of Medicine, St Louis, Missouri
- Mallinckrodt Institute of Radiology, Washington University in St Louis School of Medicine, St Louis, Missouri
- Department of Pediatrics, Washington University in St Louis School of Medicine, St Louis, Missouri
| | - Melanie E. Fields
- Department of Pediatrics, Washington University in St Louis School of Medicine, St Louis, Missouri
| | - Hongyu An
- Mallinckrodt Institute of Radiology, Washington University in St Louis School of Medicine, St Louis, Missouri
| | - Allison A. King
- Department of Pediatrics, Washington University in St Louis School of Medicine, St Louis, Missouri
- Program in Occupational Therapy, Washington University in St Louis School of Medicine, St Louis, Missouri
| | - Yasheng Chen
- Department of Neurology, Washington University in St Louis School of Medicine, St Louis, Missouri
| |
Collapse
|
3
|
Feng M, Song Z, Zhou Z, Wu Z, Ma M, Liu Y, Wang Y, Dai H. Cognitive impairment mediates the white matter injury load and gait disorders in subcortical ischemic vascular disease. Brain Imaging Behav 2024; 18:1418-1427. [PMID: 39316311 DOI: 10.1007/s11682-024-00941-3] [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] [Accepted: 09/17/2024] [Indexed: 09/25/2024]
Abstract
Gait disorders are common in patients with subcortical ischemic vascular disease (SIVD). We aim to explore the impact of white matter (WM) damage on gait disorders in SIVD. 21 SIVD patients and 20 normal controls (NC) were included in the study. Montreal Cognitive Assessment (MoCA) was used to evaluate general cognition, while Speed-Accuracy Trade-Off (SAT) was used to assess executive function. Gait velocity, cadence, and stride length were measured. Diffusion Tensor Imaging (DTI) data were analyzed using Tract-Based Spatial Statistics (TBSS) and Peak Width of Skeletonized Mean Diffusivity (PSMD). The relationships among WM damage, gait disorders, and cognitive function were examined through mediation analysis. SIVD scored lower than NC in MoCA and SAT tests (P < 0.001). Gait velocity and stride length were decreased in SIVD. SIVD had lower PSMD (P < 0.001). PSMD correlated with gait parameters, which were totally mediated by MoCA and partially mediated by SAT. The fractional anisotropy (FA) and mean diffusivity (MD) of the genu of the corpus callosum (GCC) and body of CC (BCC) were correlated with gait parameters. The FA of the bilateral anterior corona radiata (ACR) was positively correlated with gait parameters, while the MD of the bilateral superior corona radiata (SCR), bilateral superior longitudinal fasciculus (SLF), and left external capsule (EC) were negatively correlated with them (P < 0.05). Gait impairments in SIVD were associated with cognitive deficits. Cognitive impairment mediated the WM damage and gait disorders. The microstructural alterations of CC, SLF, EC, and CR may be related to changes in gait.
Collapse
Affiliation(s)
- Mengmeng Feng
- Department of Radiology, the First Affiliated Hospital of Soochow University, Suzhou city, 215000, Jiangsu province, P.R. China
| | - Ziyang Song
- Department of Radiology, the First Affiliated Hospital of Soochow University, Suzhou city, 215000, Jiangsu province, P.R. China
| | - Zheping Zhou
- Department of Geratology, the First Affiliated Hospital of Soochow University, Suzhou city, 215000, Jiangsu province, P.R. China
| | - Zhiwei Wu
- Department of Radiology, the First Affiliated Hospital of Soochow University, Suzhou city, 215000, Jiangsu province, P.R. China
| | - Mengya Ma
- Department of Radiology, the First Affiliated Hospital of Soochow University, Suzhou city, 215000, Jiangsu province, P.R. China
| | - Yuanqing Liu
- Department of Radiology, the First Affiliated Hospital of Soochow University, Suzhou city, 215000, Jiangsu province, P.R. China
| | - Yueju Wang
- Department of Geratology, the First Affiliated Hospital of Soochow University, Suzhou city, 215000, Jiangsu province, P.R. China
| | - Hui Dai
- Department of Radiology, the First Affiliated Hospital of Soochow University, Suzhou city, 215000, Jiangsu province, P.R. China.
- Institute of Medical Imaging, Soochow University, Suzhou city, 215000, Jiangsu province, P.R. China.
- Suzhou Key Laboratory of Intelligent Medicine and Equipment, Suzhou city, 215123, Jiangsu province, P.R. China.
| |
Collapse
|
4
|
Song Y, Zhou X, Zhao H, Zhao W, Sun Z, Zhu J, Yu Y. Characterizing the role of the microbiota-gut-brain axis in cerebral small vessel disease: An integrative multi‑omics study. Neuroimage 2024; 303:120918. [PMID: 39505226 DOI: 10.1016/j.neuroimage.2024.120918] [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: 04/16/2024] [Revised: 10/22/2024] [Accepted: 11/04/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Prior efforts have revealed changes in gut microbiome, circulating metabolome, and multimodal neuroimaging features in cerebral small vessel disease (CSVD). However, there is a paucity of research integrating the multi-omic information to characterize the role of the microbiota-gut-brain axis in CSVD. METHODS We collected gut microbiome, fecal and blood metabolome, multimodal magnetic resonance imaging data from 37 CSVD patients with white matter hyperintensities and 46 healthy controls. Between-group comparison was performed to identify the differential gut microbial taxa, followed by performance of multi-stage microbiome-metabolome-neuroimaging-neuropsychology correlation analyses in CSVD patients. RESULTS Our data showed both depleted and enriched gut microbes in CSVD patients. Among the differential microbes, Haemophilus and Akkermansia were associated with a range of metabolites enriched for Aminoacyl-tRNA biosynthesis pathway. Furthermore, the affected metabolites were associated with neuroimaging measures involving gray matter morphology, spontaneous intrinsic brain activity, white matter integrity, and global structural network topology, which were in turn related to cognition and emotion in CSVD patients. CONCLUSION Our findings provide an integrative framework to understand the pathophysiological mechanisms underlying the interplay between gut microbiota dysbiosis and CSVD, highlighting the potential of targeting the microbiota-gut-brain axis as a therapeutic strategy in CSVD patients.
Collapse
Affiliation(s)
- Yu Song
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, PR China; Research Center of Clinical Medical Imaging, Anhui Province, Hefei 230032, PR China; Anhui Provincial Institute of Translational Medicine, Hefei 230032, PR China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, PR China
| | - Xia Zhou
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, PR China
| | - Han Zhao
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, PR China; Research Center of Clinical Medical Imaging, Anhui Province, Hefei 230032, PR China; Anhui Provincial Institute of Translational Medicine, Hefei 230032, PR China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, PR China
| | - Wenming Zhao
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, PR China; Research Center of Clinical Medical Imaging, Anhui Province, Hefei 230032, PR China; Anhui Provincial Institute of Translational Medicine, Hefei 230032, PR China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, PR China
| | - Zhongwu Sun
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, PR China.
| | - Jiajia Zhu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, PR China; Research Center of Clinical Medical Imaging, Anhui Province, Hefei 230032, PR China; Anhui Provincial Institute of Translational Medicine, Hefei 230032, PR China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, PR China.
| | - Yongqiang Yu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, PR China; Research Center of Clinical Medical Imaging, Anhui Province, Hefei 230032, PR China; Anhui Provincial Institute of Translational Medicine, Hefei 230032, PR China; Anhui Provincial Key Laboratory for Brain Bank Construction and Resource Utilization, Hefei 230032, PR China.
| |
Collapse
|
5
|
Chen Y, Tozer D, Li R, Li H, Tuladhar A, De Leeuw FE, Markus HS. Improved Dementia Prediction in Cerebral Small Vessel Disease Using Deep Learning-Derived Diffusion Scalar Maps From T1. Stroke 2024; 55:2254-2263. [PMID: 39145386 PMCID: PMC11346716 DOI: 10.1161/strokeaha.124.047449] [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: 04/10/2024] [Revised: 06/23/2024] [Accepted: 07/22/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Cerebral small vessel disease is the most common pathology underlying vascular dementia. In small vessel disease, diffusion tensor imaging is more sensitive to white matter damage and better predicts dementia risk than conventional magnetic resonance imaging sequences, such as T1 and fluid attenuation inversion recovery, but diffusion tensor imaging takes longer to acquire and is not routinely available in clinical practice. As diffusion tensor imaging-derived scalar maps-fractional anisotropy (FA) and mean diffusivity (MD)-are frequently used in clinical settings, one solution is to synthesize FA/MD from T1 images. METHODS We developed a deep learning model to synthesize FA/MD from T1. The training data set consisted of 4998 participants with the highest white matter hyperintensity volumes in the UK Biobank. Four external validations data sets with small vessel disease were included: SCANS (St George's Cognition and Neuroimaging in Stroke; n=120), RUN DMC (Radboud University Nijmegen Diffusion Tensor and Magnetic Resonance Imaging Cohort; n=502), PRESERVE (Blood Pressure in Established Cerebral Small Vessel Disease; n=105), and NETWORKS (n=26), along with 1000 normal controls from the UK Biobank. RESULTS The synthetic maps resembled ground-truth maps (structural similarity index >0.89 for MD maps and >0.80 for FA maps across all external validation data sets except for SCANS). The prediction accuracy of dementia using whole-brain median MD from the synthetic maps is comparable to the ground truth (SCANS ground-truth c-index, 0.822 and synthetic, 0.821; RUN DMC ground truth, 0.816 and synthetic, 0.812) and better than white matter hyperintensity volume (SCANS, 0.534; RUN DMC, 0.710). CONCLUSIONS We have developed a fast and generalizable method to synthesize FA/MD maps from T1 to improve the prediction accuracy of dementia in small vessel disease when diffusion tensor imaging data have not been acquired.
Collapse
Affiliation(s)
- Yutong Chen
- Department of Clinical Neuroscience, Stroke Research Group, University of Cambridge, United Kingdom (Y.C., D.T., R.L., H.S.M.)
| | - Daniel Tozer
- Department of Clinical Neuroscience, Stroke Research Group, University of Cambridge, United Kingdom (Y.C., D.T., R.L., H.S.M.)
| | - Rui Li
- Department of Clinical Neuroscience, Stroke Research Group, University of Cambridge, United Kingdom (Y.C., D.T., R.L., H.S.M.)
| | - Hao Li
- Department of Clinical Neuroscience, Stroke Research Group, University of Cambridge, United Kingdom (Y.C., D.T., R.L., H.S.M.)
- Department of Neurology, Radboud University Medical Center, Donders Center for Medical Neurosciences, Nijmegen, the Netherlands (H.L., A.T., F.E.D.L.)
| | - Anil Tuladhar
- Department of Neurology, Radboud University Medical Center, Donders Center for Medical Neurosciences, Nijmegen, the Netherlands (H.L., A.T., F.E.D.L.)
| | - Frank Erik De Leeuw
- Department of Neurology, Radboud University Medical Center, Donders Center for Medical Neurosciences, Nijmegen, the Netherlands (H.L., A.T., F.E.D.L.)
| | - Hugh S. Markus
- Department of Clinical Neuroscience, Stroke Research Group, University of Cambridge, United Kingdom (Y.C., D.T., R.L., H.S.M.)
| |
Collapse
|
6
|
Wang Y, Wang T, Yu Z, Wang J, Liu F, Ye M, Fang X, Liu Y, Liu J. Alterations in structural integrity of superior longitudinal fasciculus III associated with cognitive performance in cerebral small vessel disease. BMC Med Imaging 2024; 24:138. [PMID: 38858645 PMCID: PMC11165890 DOI: 10.1186/s12880-024-01324-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 06/05/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND This study aimed to investigate the alterations in structural integrity of superior longitudinal fasciculus subcomponents with increasing white matter hyperintensity severity as well as the relationship to cognitive performance in cerebral small vessel disease. METHODS 110 cerebral small vessel disease study participants with white matter hyperintensities were recruited. According to Fazekas grade scale, white matter hyperintensities of each subject were graded. All subjects were divided into two groups. The probabilistic fiber tracking method was used for analyzing microstructure characteristics of superior longitudinal fasciculus subcomponents. RESULTS Probabilistic fiber tracking results showed that mean diffusion, radial diffusion, and axial diffusion values of the left arcuate fasciculus as well as the mean diffusion value of the right arcuate fasciculus and left superior longitudinal fasciculus III in high white matter hyperintensities rating group were significantly higher than those in low white matter hyperintensities rating group (p < 0.05). The mean diffusion value of the left superior longitudinal fasciculus III was negatively related to the Montreal Cognitive Assessment score of study participants (p < 0.05). CONCLUSIONS The structural integrity injury of bilateral arcuate fasciculus and left superior longitudinal fasciculus III is more severe with the aggravation of white matter hyperintensities. The structural integrity injury of the left superior longitudinal fasciculus III correlates to cognitive impairment in cerebral small vessel disease.
Collapse
Affiliation(s)
- Yifan Wang
- Department of Radiology, Eye& ENT Hospital of Shanghai Medical School, Fudan University, Shanghai, China
| | - Tianyao Wang
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zekuan Yu
- Academy for Engineering and Technology, Fudan University, Shanghai, 200433, China
| | - Junjie Wang
- Department of Neurosurgery, Beijing Hospital, National Center of Gerontology, Beijing, China
- Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Fang Liu
- Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
- Department of Neurology, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Mengwen Ye
- Academy for Engineering and Technology, Fudan University, Shanghai, 200433, China
| | - Xianjin Fang
- Anhui University of Science and Technology, Anhui, China
| | - Yinhong Liu
- Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China.
- Department of Neurology, Beijing Hospital, National Center of Gerontology, Beijing, China.
| | - Jun Liu
- Department of Radiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 XianXia Road, Shanghai, 200050, China.
| |
Collapse
|
7
|
Xu K, Wang Y, Jiang Y, Wang Y, Li P, Lu H, Suo C, Yuan Z, Yang Q, Dong Q, Jin L, Cui M, Chen X. Analysis of gait pattern related to high cerebral small vessel disease burden using quantitative gait data from wearable sensors. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 250:108162. [PMID: 38631129 DOI: 10.1016/j.cmpb.2024.108162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 03/28/2024] [Accepted: 04/03/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND AND OBJECTIVES Sensor-based wearable devices help to obtain a wide range of quantitative gait parameters, which provides sufficient data to investigate disease-specific gait patterns. Although cerebral small vessel disease (CSVD) plays a significant role in gait impairment, the specific gait pattern associated with a high burden of CSVD remains to be explored. METHODS We analyzed the gait pattern related to high CSVD burden from 720 participants (aged 55-65 years, 42.5 % male) free of neurological disease in the Taizhou Imaging Study. All participants underwent detailed quantitative gait assessments (obtained from an insole-like wearable gait tracking device) and brain magnetic resonance imaging examinations. Thirty-three gait parameters were summarized into five gait domains. Sparse sliced inverse regression was developed to extract the gait pattern related to high CSVD burden. RESULTS The specific gait pattern derived from several gait domains (i.e., angles, phases, variability, and spatio-temporal) was significantly associated with the CSVD burden (OR=1.250, 95 % CI: 1.011-1.546). The gait pattern indicates that people with a high CSVD burden were prone to have smaller gait angles, more stance time, more double support time, larger gait variability, and slower gait velocity. Furthermore, people with this gait pattern had a 25 % higher risk of a high CSVD burden. CONCLUSIONS We established a more stable and disease-specific quantitative gait pattern related to high CSVD burden, which is prone to facilitate the identification of individuals with high CSVD burden among the community residents or the general population.
Collapse
Affiliation(s)
- Kelin Xu
- Department of Biostatistics, Ministry of Education Key Laboratory of Public Health Safety, School of Public Health, Fudan University, Shanghai, China
| | - Yingzhe Wang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Shanghai, China; Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yanfeng Jiang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Shanghai, China; Fudan University Taizhou Institute of Health Sciences, Taizhou, China
| | - Yawen Wang
- Department of Biostatistics, Ministry of Education Key Laboratory of Public Health Safety, School of Public Health, Fudan University, Shanghai, China
| | - Peixi Li
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Heyang Lu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Chen Suo
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China; Department of Epidemiology, Ministry of Education Key Laboratory of Public Health Safety, School of Public Health, Fudan University, Shanghai, China
| | - Ziyu Yuan
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China
| | - Qi Yang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiang Dong
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Shanghai, China; Fudan University Taizhou Institute of Health Sciences, Taizhou, China
| | - Mei Cui
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China.
| | - Xingdong Chen
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Shanghai, China; Fudan University Taizhou Institute of Health Sciences, Taizhou, China.
| |
Collapse
|
8
|
Wang Z, Xia K, Li J, Liu Y, Zhou Y, Zhang L, Tang L, Zeng X, Fan D, Yang Q. Essential Nutrients and White Matter Hyperintensities: A Two-Sample Mendelian Randomization Study. Biomedicines 2024; 12:810. [PMID: 38672165 PMCID: PMC11047968 DOI: 10.3390/biomedicines12040810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/24/2024] [Accepted: 04/03/2024] [Indexed: 04/28/2024] Open
Abstract
Stroke and dementia have been linked to the appearance of white matter hyperintensities (WMHs). Meanwhile, diffusion tensor imaging (DTI) might capture the microstructural change in white matter early. Specific dietary interventions may help to reduce the risk of WMHs. However, research on the relationship between specific nutrients and white matter changes is still lacking. We aimed to investigate the causal effects of essential nutrients (amino acids, fatty acids, mineral elements, and vitamins) on WMHs and DTI measures, including fraction anisotropy (FA) and mean diffusivity (MD), by a Mendelian randomization analysis. We selected single nucleotide polymorphisms (SNPs) associated with each nutrient as instrumental variables to assess the causal effects of nutrient-related exposures on WMHs, FA, and MD. The outcome was from a recently published large-scale European Genome Wide Association Studies pooled dataset, including WMHs (N = 18,381), FA (N = 17,663), and MD (N = 17,467) data. We used the inverse variance weighting (IVW) method as the primary method, and sensitivity analyses were conducted using the simple median, weighted median, and MR-Egger methods. Genetically predicted serum calcium level was positively associated with WMHs risk, with an 8.1% increase in WMHs risk per standard deviation unit increase in calcium concentration (OR = 1.081, 95% CI = 1.006-1.161, p = 0.035). The plasma linoleic acid level was negatively associated with FA (OR = 0.776, 95% CI = 0.616-0.978, p = 0.032). Our study demonstrated that genetically predicted calcium was a potential risk factor for WMHs, and linoleic acid may be negatively associated with FA, providing evidence for interventions from the perspective of gene-environment interactions.
Collapse
Affiliation(s)
- Zhengrui Wang
- Department of Neurology, Peking University Third Hospital, Beijing 100191, China
- Peking University Health Science Center, Beijing 100191, China
| | - Kailin Xia
- Department of Neurology, Peking University Third Hospital, Beijing 100191, China
| | - Jiayi Li
- Department of Neurology, Peking University Third Hospital, Beijing 100191, China
- Peking University Health Science Center, Beijing 100191, China
| | - Yanru Liu
- Department of Neurology, Peking University Third Hospital, Beijing 100191, China
- Peking University Health Science Center, Beijing 100191, China
| | - Yumou Zhou
- Department of Neurology, Peking University Third Hospital, Beijing 100191, China
- Peking University Health Science Center, Beijing 100191, China
| | - Linjing Zhang
- Department of Neurology, Peking University Third Hospital, Beijing 100191, China
| | - Lu Tang
- Department of Neurology, Peking University Third Hospital, Beijing 100191, China
| | - Xiangzhu Zeng
- Department of Radiology, Peking University Third Hospital, Beijing 100191, China
| | - Dongsheng Fan
- Department of Neurology, Peking University Third Hospital, Beijing 100191, China
- Beijing Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing 100191, China
- Key Laboratory for Neuroscience, National Health Commission, Ministry of Education, Peking University, Beijing 100191, China
| | - Qiong Yang
- Department of Neurology, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
9
|
Hong H, Tozer DJ, Markus HS. Relationship of Perivascular Space Markers With Incident Dementia in Cerebral Small Vessel Disease. Stroke 2024; 55:1032-1040. [PMID: 38465597 PMCID: PMC10962441 DOI: 10.1161/strokeaha.123.045857] [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: 11/14/2023] [Revised: 02/02/2024] [Accepted: 02/13/2024] [Indexed: 03/12/2024]
Abstract
BACKGROUND Recent studies, using diffusion tensor image analysis along the perivascular space (DTI-ALPS), suggest impaired perivascular space (PVS) function in cerebral small vessel disease, but they were cross-sectional, making inferences on causality difficult. We determined associations between impaired PVS, measured using DTI-ALPS and PVS volume, and cognition and incident dementia. METHODS In patients with lacunar stroke and confluent white matter hyperintensities, without dementia at baseline, recruited prospectively in a single center, magnetic resonance imaging was performed annually for 3 years, and cognitive assessments, including global, memory, executive function, and processing speed, were performed annually for 5 years. We determined associations between DTI-ALPS and PVS volume with cerebral small vessel disease imaging markers (white matter hyperintensity volume, lacunes, and microbleeds) at baseline and with changes in imaging markers. We determined whether DTI-ALPS and PVS volume at baseline and change over 3 years predicted incident dementia. Analyses were controlled for conventional diffusion tensor image metrics using 2 markers (median mean diffusivity [MD] and peak width of skeletonized MD) and adjusted for age, sex, and vascular risk factors. RESULTS A total of 120 patients, mean age 70.0 years and 65.0% male, were included. DTI-ALPS declined over 3 years, while no change in PVS volume was found. Neither DTI-ALPS nor PVS volume was associated with cerebral small vessel disease imaging marker progression. Baseline DTI-ALPS was associated with changes in global cognition (β=0.142, P=0.032), executive function (β=0.287, P=0.027), and long-term memory (β=0.228, P=0.027). Higher DTI-ALPS at baseline predicted a lower risk of dementia (hazard ratio, 0.328 [0.183-0.588]; P<0.001), and this remained significant after including median MD as a covariate (hazard ratio, 0.290 [0.139-0.602]; P<0.001). Change in DTI-ALPS predicted dementia conversion (hazard ratio, 0.630 [0.428-0.964]; P=0.048), but when peak width of skeletonized MD and median MD were entered as covariates, the association was not significant. There was no association between baseline PVS volume, or PVS change over 3 years, and conversion to dementia. CONCLUSIONS DTI-ALPS predicts future dementia risk in patients with lacunar strokes and confluent white matter hyperintensities. However, the weakening of the association between change in DTI-ALPS and incident dementia after controlling for peak width of skeletonized MD and median MD suggests part of the signal may represent conventional diffusion tensor image metrics. PVS volume is not a predictor of future dementia risk.
Collapse
Affiliation(s)
- Hui Hong
- Department of Clinical Neurosciences, University of Cambridge, United Kingdom (H.H., D.J.T., H.S.M.)
- Department of Radiology, Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China (H.H.)
| | - Daniel J. Tozer
- Department of Clinical Neurosciences, University of Cambridge, United Kingdom (H.H., D.J.T., H.S.M.)
| | - Hugh S. Markus
- Department of Clinical Neurosciences, University of Cambridge, United Kingdom (H.H., D.J.T., H.S.M.)
| |
Collapse
|
10
|
Zhang Y, Hamidi RE, Hadi M. Cerebral Small Vessel Ischemic Disease: A Source of Patient Panic or a Case of Pragmatic Reporting? Semin Roentgenol 2024; 59:157-164. [PMID: 38880514 DOI: 10.1053/j.ro.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 06/18/2024]
Affiliation(s)
- Yi Zhang
- Department of Radiology, University of Louisville, 530 South Jackson Street, CCB-C07, Louisville, KY
| | - Ramin E Hamidi
- Department of Radiology, University of Louisville, 530 South Jackson Street, CCB-C07, Louisville, KY.
| | - Mohiuddin Hadi
- Department of Radiology, University of Louisville, 530 South Jackson Street, CCB-C07, Louisville, KY
| |
Collapse
|
11
|
Guo C, Harshfield EL, Markus HS. Sleep Characteristics and Risk of Stroke and Dementia: An Observational and Mendelian Randomization Study. Neurology 2024; 102:e209141. [PMID: 38350061 PMCID: PMC11067695 DOI: 10.1212/wnl.0000000000209141] [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: 08/18/2023] [Accepted: 11/16/2023] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Sleep disturbances are implicated as risk factors of both stroke and dementia. However, whether these associations are causal and whether treatment of sleep disorders could reduce stroke and dementia risk remain uncertain. We aimed to evaluate associations and ascertain causal relationships between sleep characteristics and stroke/dementia risk and MRI markers of small vessel disease (SVD). METHODS We used data sets from a multicenter population-based study and summary statistics from genome-wide association studies (GWASs) of sleep characteristics and outcomes. We analyzed 502,383 UK Biobank participants with self-reported sleep measurements, including sleep duration, insomnia, chronotype, napping, daytime dozing, and snoring. In observational analyses, the primary outcomes were incident stroke, dementia, and their subtypes, alongside SVD markers. Hazard ratios (HRs) and odds ratios (ORs) were adjusted for age, sex, and ethnicity, and additional vascular risk factors. In Mendelian randomization (MR) analyses, ORs or risk ratios are reported for the association of each genetic score with clinical or MRI end points. RESULTS Among 502,383 participants (mean [SD] age, 56.5 [8.1] years; 54.4% female), there were 7,668 cases of all-cause dementia and 10,334 strokes. In longitudinal analyses, after controlling for cardiovascular risk factors, participants with insomnia, daytime napping, and dozing were associated with increased risk of any stroke (HR 1.05, 95% CI 1.01-1.11, p = 8.53 × 10-3; HR 1.09, 95% CI 1.05-1.14, p = 3.20 × 10-5; HR 1.19, 95% CI 1.08-1.32, p = 4.89 × 10-4, respectively). Almost all sleep measures were associated with dementia risk (all p < 0.001, except insomnia). Cross-sectional analyses identified associations between napping, snoring, and MRI markers of SVD (all p < 0.001). MR analyses supported a causal link between genetically predicted insomnia and increased stroke risk (OR 1.31, 95% CI 1.13-1.51, p = 0.00072), but not with dementia or SVD markers. DISCUSSION We found that multiple sleep measures predicted future risk of stroke and dementia, but these associations were attenuated after controlling for cardiovascular risk factors and were absent in MR analyses for Alzheimer disease. This suggests possible confounding or reverse causation, implying caution before proposing sleep disorder modifications for dementia treatment.
Collapse
Affiliation(s)
- Chutian Guo
- From the Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, United Kingdom
| | - Eric L Harshfield
- From the Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, United Kingdom
| | - Hugh S Markus
- From the Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, United Kingdom
| |
Collapse
|
12
|
Wang D, Wang L, Wang J, Du Y, Wang K, Wang M, Yang L, Zhao X. Retinal structure and vessel density changes in cerebral small vessel disease. Front Neurosci 2024; 18:1288380. [PMID: 38469574 PMCID: PMC10925719 DOI: 10.3389/fnins.2024.1288380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 02/14/2024] [Indexed: 03/13/2024] Open
Abstract
Background Cerebral small vessel disease (CSVD) attaches people's attention in recent years. In this study, we aim to explore retinal structure and vessel density changes in CSVD patients. Methods We collected information on retinal metrics assessed by optical coherence tomography (OCT) and OCT angiography and CSVD characters. Logistic and liner regression was used to analyze the relationship between retinal metrics and CSVD. Results Vessel density of superficial retinal capillary plexus (SRCP), foveal density- 300 length (FD-300), radial peripapillary capillary (RPC) and thickness of retina were significantly lower in CSVD patients, the difference only existed in the thickness of retina after adjusted relevant risk factors (OR (95% CI): 0.954 (0.912, 0.997), p = 0.037). SRCP vessel density showed a significant downward trend with the increase of CSVD scores (β: -0.087, 95%CI: -0.166, -0.008, p = 0.031). SRCP and FD-300 were significantly lower in patients with lacunar infarctions and white matter hypertensions separately [OR (95% CI): 0.857 (0.736, 0.998), p = 0.047 and OR (95% CI): 0.636 (0.434, 0.932), p = 0.020, separately]. Conclusion SRCP, FD-300 and thickness of retina were associated with the occurrence and severity of total CSVD scores and its different radiological manifestations. Exploring CSVD by observing alterations in retinal metrics has become an optional research direction in future.
Collapse
Affiliation(s)
- Dandan Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Lina Wang
- Department of Ophthalmology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jinjin Wang
- Department of Neurology, Peking University Sixth Hospital, Peking University Institute of Mental Health, Beijing, China
| | - Yang Du
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Kaiyue Wang
- Department of Ophthalmology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Meizi Wang
- Department of Ophthalmology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Liu Yang
- Department of Ophthalmology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xingquan Zhao
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
13
|
Liu L, Shen Q, Zhang D, Bao Y, Xu F, Huang H, Xu Y. Mendelian Randomization Analysis to Assess Whether Magnetic Resonance Imaging Signs of Cerebral Small Vessel Disease Can Cause Cognitive Decline and Dementia. J Prev Alzheimers Dis 2024; 11:1390-1396. [PMID: 39350385 DOI: 10.14283/jpad.2024.95] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2025]
Abstract
OBJECTIVE Cognitive decline and dementia have been linked to cerebral small vessel disease, so we explored using Mendelian randomization whether cerebral small vessel disease visible as 10 neuroimaging signs may cause cognitive decline and dementia. METHODS We analyzed publicly available data from genome-wide association studies using two-sample Mendelian randomization involving inverse variance weighting, weighted median, MR-Egger, and MR-PRESSO approaches. RESULTS Mendelian randomization suggested that cognitive decline can be caused by lacunar stroke (inverse variance weighting, β = -0.012, 95% CI -0.024 to -0.001, P = 0.033). Furthermore, an elevated burden of white matter hyperintensities was associated with an increased risk of Dementia due to Parkinson's disease (inverse variance weighting, OR 2.035, 95% CI 1.105 to 3.745, P = 0.023). Notably, no significant associations were observed between neuroimaging markers of Cerebral Small Vessel Disease and other types of dementia. CONCLUSION This Mendelian randomization study provides evidence that lacunar stroke and white matter lesions can cause cognitive decline, and that white matter hyperintensity may increase risk of dementia due to Parkinson's disease. These results underscore the need for further investigations into the neurocognitive effects of cerebral small vessel disease.
Collapse
Affiliation(s)
- L Liu
- Yanming Xu, Sichuan University West China Hospital, West China Hospital of Sichuan University, China,
| | | | | | | | | | | | | |
Collapse
|
14
|
Marzi C, Scheda R, Salvadori E, Giorgio A, De Stefano N, Poggesi A, Inzitari D, Pantoni L, Mascalchi M, Diciotti S. Fractal dimension of the cortical gray matter outweighs other brain MRI features as a predictor of transition to dementia in patients with mild cognitive impairment and leukoaraiosis. Front Hum Neurosci 2023; 17:1231513. [PMID: 37822707 PMCID: PMC10562576 DOI: 10.3389/fnhum.2023.1231513] [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: 05/30/2023] [Accepted: 08/31/2023] [Indexed: 10/13/2023] Open
Abstract
Background The relative contribution of changes in the cerebral white matter (WM) and cortical gray matter (GM) to the transition to dementia in patients with mild cognitive impairment (MCI) is not yet established. In this longitudinal study, we aimed to analyze MRI features that may predict the transition to dementia in patients with MCI and T2 hyperintensities in the cerebral WM, also known as leukoaraiosis. Methods Sixty-four participants with MCI and moderate to severe leukoaraiosis underwent baseline MRI examinations and annual neuropsychological testing over a 2 year period. The diagnosis of dementia was based on established criteria. We evaluated demographic, neuropsychological, and several MRI features at baseline as predictors of the clinical transition. The MRI features included visually assessed MRI features, such as the number of lacunes, microbleeds, and dilated perivascular spaces, and quantitative MRI features, such as volumes of the cortical GM, hippocampus, T2 hyperintensities, and diffusion indices of the cerebral WM. Additionally, we examined advanced quantitative features such as the fractal dimension (FD) of cortical GM and WM, which represents an index of tissue structural complexity derived from 3D-T1 weighted images. To assess the prediction of transition to dementia, we employed an XGBoost-based machine learning system using SHapley Additive exPlanations (SHAP) values to provide explainability to the machine learning model. Results After 2 years, 18 (28.1%) participants had transitioned from MCI to dementia. The area under the receiving operator characteristic curve was 0.69 (0.53, 0.85) [mean (90% confidence interval)]. The cortical GM-FD emerged as the top-ranking predictive feature of transition. Furthermore, aggregated quantitative neuroimaging features outperformed visually assessed MRI features in predicting conversion to dementia. Discussion Our findings confirm the complementary roles of cortical GM and WM changes as underlying factors in the development of dementia in subjects with MCI and leukoaraiosis. FD appears to be a biomarker potentially more sensitive than other brain features.
Collapse
Affiliation(s)
- Chiara Marzi
- Department of Statistics, Computer Science, Applications “Giuseppe Parenti, ” University of Florence, Florence, Italy
| | - Riccardo Scheda
- Department of Electrical, Electronic, and Information Engineering “Guglielmo Marconi, ” University of Bologna, Cesena, Italy
| | - Emilia Salvadori
- NEUROFARBA Department, Neuroscience Section, University of Florence, Florence, Italy
| | - Antonio Giorgio
- Department of Medicine, Surgery, and Neuroscience, University of Siena, Siena, Italy
| | - Nicola De Stefano
- Department of Medicine, Surgery, and Neuroscience, University of Siena, Siena, Italy
| | - Anna Poggesi
- NEUROFARBA Department, Neuroscience Section, University of Florence, Florence, Italy
| | - Domenico Inzitari
- NEUROFARBA Department, Neuroscience Section, University of Florence, Florence, Italy
| | - Leonardo Pantoni
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Mario Mascalchi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio, ” University of Florence, Florence, Italy
- Division of Epidemiology and Clinical Governance, Institute for Study, Prevention and Network in Oncology (ISPRO), Florence, Italy
| | - Stefano Diciotti
- Department of Electrical, Electronic, and Information Engineering “Guglielmo Marconi, ” University of Bologna, Cesena, Italy
- Alma Mater Research Institute for Human-Centered Artificial Intelligence, University of Bologna, Bologna, Italy
| |
Collapse
|
15
|
Zhang Y, Chang P, Liu N, Jiang Y, Chu Y, Du W, Lin L, Gao B, Li Y, Qu M, Yang C, Miao Y. Correlation between lenticulostriate arteries and white matter microstructure changes in patients with cerebral small vessel disease. Front Neurosci 2023; 17:1202538. [PMID: 37817799 PMCID: PMC10560852 DOI: 10.3389/fnins.2023.1202538] [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: 07/23/2023] [Accepted: 09/05/2023] [Indexed: 10/12/2023] Open
Abstract
To explore the correlation between the number of lenticulostriate arteries (LSAs) and the white matter features in cerebral small vessel diseases (CSVD) by 3T magnetic resonance imaging (MRI). Seventy-one patients with diagnoses of CSVD were prospectively enrolled to undergo 3T MRI examination, including high-resolution vascular wall imaging (VWI) and diffusion tensor imaging (DTI). The LSAs were observed and counted on VWI, and the patients were divided into three groups according to the LSA counts. The presence of white matter hyperintensities (WMHs), lacunes, cerebral microbleeds (CMBs), and enlarged perivascular spaces (EPVS) was assessed in each patient, and a composite CSVD score was calculated. Periventricular and deep white matter hyperintensity (PVWMH, DWMH) volume ratios were obtained based on automatic segmentation. Fractional anisotropy (FA) and mean diffusivity (MD) were processed by using tract-based spatial statistics (TBSS) analysis. These parameters were compared among the three groups. Correlations between the LSA counts and white matter features were also analyzed. There were differences in WMHs (P = 0.001), CMBs (P < 0.001), EPVS (P = 0.017), composite CSVD scores (P < 0.001), PVWMH volume ratios (P = 0.001), DWMH volume ratios (P < 0.001), global FA (P = 0.001), and global MD (P = 0.002) among the three groups. There were correlations between the LSA counts and WMHs (r = -0.45, P < 0.001), CMBs (r = -0.44, P < 0.001), EPVS (r = -0.28, P = 0.020), the composite CSVD score (r = -0.52, P < 0.001), DWMH volume ratio (r = -0.47, P < 0.001), PWMH volume ratio (r = -0.34, P = 0.004), global FA (r = 0.36, P = 0.002), and global MD (r = -0.33, P = 0.005). Diabetes mellitus (OR 3.36, 95% CI 1.06-10.63; P = 0.039) and increased DWMH volume ratios (OR 1.04, 95% CI 1.00-1.08; P = 0.048) were independent risk factors for a decrease in LSA counts. TBSS analysis showed differences among the three groups in global FA and MD after adjusting for age and sex (P < 0.05). The LSA counts was associated with white matter microstructure changes in CSVD and has the potential to represent the extent of subcortical microvascular damage in CSVD patients.
Collapse
Affiliation(s)
- Yukun Zhang
- Department of Radiology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Peipei Chang
- Department of Radiology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Na Liu
- Department of Radiology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yuhan Jiang
- Department of Radiology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Ying Chu
- Department of Neurology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Wei Du
- Department of Radiology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | | | - Bingbing Gao
- Department of Radiology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yuan Li
- Department of Radiology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Mingrui Qu
- Department of Radiology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Chao Yang
- Department of Radiology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - YanWei Miao
- Department of Radiology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
16
|
Badji A, Youwakim J, Cooper A, Westman E, Marseglia A. Vascular cognitive impairment - Past, present, and future challenges. Ageing Res Rev 2023; 90:102042. [PMID: 37634888 DOI: 10.1016/j.arr.2023.102042] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 08/29/2023]
Abstract
Vascular cognitive impairment (VCI) is a lifelong process encompassing a broad spectrum of cognitive disorders, ranging from subtle or mild deficits to prodromal and fully developed dementia, originating from cerebrovascular lesions such as large and small vessel disease. Genetic predisposition and environmental exposure to risk factors such as unhealthy lifestyles, hypertension, cardiovascular disease, and metabolic disorders will synergistically interact, yielding biochemical and structural brain changes, ultimately culminating in VCI. However, little is known about the pathological processes underlying VCI and the temporal dynamics between risk factors and disease mechanisms (biochemical and structural brain changes). This narrative review aims to provide an evidence-based summary of the link between individual vascular risk/disorders and cognitive dysfunction and the potential structural and biochemical pathophysiological processes. We also discuss some key challenges for future research on VCI. There is a need to shift from individual risk factors/disorders to comorbid vascular burden, identifying and integrating imaging and fluid biomarkers, implementing a life-course approach, considering possible neuroprotective influences of positive life exposures, and addressing biological sex at birth and gender differences. Finally, this review highlights the need for future researchers to leverage and integrate multidimensional data to advance our understanding of the mechanisms and pathophysiology of VCI.
Collapse
Affiliation(s)
- Atef Badji
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging, Karolinska University Hospital, Stockholm, Sweden
| | - Jessica Youwakim
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada; Centre interdisciplinaire de recherche sur le cerveau et l'apprentissage (CIRCA), Montreal, QC, Canada; Groupe de Recherche sur la Signalisation Neuronal et la Circuiterie (SNC), Montreal, QC, Canada
| | - Alexandra Cooper
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Unit of Integrative Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Eric Westman
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, UK
| | - Anna Marseglia
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
17
|
Li R, Harshfield EL, Bell S, Burkhart M, Tuladhar AM, Hilal S, Tozer DJ, Chappell FM, Makin SD, Lo JW, Wardlaw JM, de Leeuw FE, Chen C, Kourtzi Z, Markus HS. Predicting incident dementia in cerebral small vessel disease: comparison of machine learning and traditional statistical models. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2023; 5:100179. [PMID: 37593075 PMCID: PMC10428032 DOI: 10.1016/j.cccb.2023.100179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/07/2023] [Accepted: 08/08/2023] [Indexed: 08/19/2023]
Abstract
Background Cerebral small vessel disease (SVD) contributes to 45% of dementia cases worldwide, yet we lack a reliable model for predicting dementia in SVD. Past attempts largely relied on traditional statistical approaches. Here, we investigated whether machine learning (ML) methods improved prediction of incident dementia in SVD from baseline SVD-related features over traditional statistical methods. Methods We included three cohorts with varying SVD severity (RUN DMC, n = 503; SCANS, n = 121; HARMONISATION, n = 265). Baseline demographics, vascular risk factors, cognitive scores, and magnetic resonance imaging (MRI) features of SVD were used for prediction. We conducted both survival analysis and classification analysis predicting 3-year dementia risk. For each analysis, several ML methods were evaluated against standard Cox or logistic regression. Finally, we compared the feature importance ranked by different models. Results We included 789 participants without missing data in the survival analysis, amongst whom 108 (13.7%) developed dementia during a median follow-up of 5.4 years. Excluding those censored before three years, we included 750 participants in the classification analysis, amongst whom 48 (6.4%) developed dementia by year 3. Comparing statistical and ML models, only regularised Cox/logistic regression outperformed their statistical counterparts overall, but not significantly so in survival analysis. Baseline cognition was highly predictive, and global cognition was the most important feature. Conclusions When using baseline SVD-related features to predict dementia in SVD, the ML survival or classification models we evaluated brought little improvement over traditional statistical approaches. The benefits of ML should be evaluated with caution, especially given limited sample size and features.
Collapse
Affiliation(s)
- Rui Li
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, United Kingdom of Great Britain and Northern Ireland
| | - Eric L. Harshfield
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, United Kingdom of Great Britain and Northern Ireland
- Heart and Lung Research Institute, University of Cambridge, United Kingdom of Great Britain and Northern Ireland
| | - Steven Bell
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, United Kingdom of Great Britain and Northern Ireland
- Heart and Lung Research Institute, University of Cambridge, United Kingdom of Great Britain and Northern Ireland
- Precision Breast Cancer Institute, Department of Oncology, University of Cambridge, United Kingdom of Great Britain and Northern Ireland
| | - Michael Burkhart
- Adaptive Brain Lab, Department of Psychology, University of Cambridge, United Kingdom of Great Britain and Northern Ireland
| | - Anil M. Tuladhar
- Department of Neurology, Donders Centre for Medical Neuroscience, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Saima Hilal
- Memory Aging and Cognition Centre, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Daniel J. Tozer
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, United Kingdom of Great Britain and Northern Ireland
| | - Francesca M. Chappell
- Centre for Clinical Brain Sciences, University of Edinburgh, United Kingdom of Great Britain and Northern Ireland
| | - Stephen D.J. Makin
- Centre for Rural Health, Institute of Applied Health Sciences, University of Aberdeen, United Kingdom of Great Britain and Northern Ireland
| | - Jessica W. Lo
- Centre for Healthy Brain Ageing, University of New South Wales, Sydney, Australia
| | - Joanna M. Wardlaw
- Centre for Clinical Brain Sciences, University of Edinburgh, United Kingdom of Great Britain and Northern Ireland
| | - Frank-Erik de Leeuw
- Department of Neurology, Donders Centre for Medical Neuroscience, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Christopher Chen
- Memory Aging and Cognition Centre, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Zoe Kourtzi
- Adaptive Brain Lab, Department of Psychology, University of Cambridge, United Kingdom of Great Britain and Northern Ireland
| | - Hugh S. Markus
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, United Kingdom of Great Britain and Northern Ireland
- Heart and Lung Research Institute, University of Cambridge, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
18
|
Ng YL, Tan CS, Egle M, Gyanwali B, Tozer DJ, Markus HS, Chen C, Hilal S. The association of diffusion tensor MRI measures of normal appearing white matter and cognition. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2023; 5:100174. [PMID: 37457665 PMCID: PMC10344700 DOI: 10.1016/j.cccb.2023.100174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 05/29/2023] [Accepted: 07/01/2023] [Indexed: 07/18/2023]
Abstract
Objective Median and peak height of fractional anisotropy (FA) and mean diffusivity (MD) are diffusion tensor imaging (DTI) markers used to quantify white matter microstructure changes. We examine the association of DTI histogram-derived measures in global normal appearing white matter (NAWM) and cognitive decline in patients with normal cognition and cognitive impairment no dementia from a memory clinic in Singapore. Methods A total of 252 patients (mean age: 71.1 ± 7.6 years, 53.2% women) were included. All patients underwent clinical assessments, a brain MRI scan at baseline, and neuropsychological assessments annually for 2 years. DTI scans were processed to obtain MD and FA histogram-derived measures. The National Institute of Neurological Disorders and Stroke and the Canadian Stroke Network harmonization neuropsychological battery were used to assess cognitive function. Linear regression models with generalised estimating equation (GEE) and logistic regression models were used to examine the association between DTI histogram measures and cognitive decline. Results When compared to baseline, MD and FA measures at Year 2 were associated with an accelerated worsening in global cognition (all p for interaction <0.001; Year 0 vs 2, MD median: -0.29 (95%CI: -0.49, -0.09) vs -0.45 (95%CI: -0.65,-0.25); MD peak height: 0.22 (95%CI: 0.07, 0.37) vs 0.37 (95%CI: 0.21, 0.53); FA median: 0.11 (95%CI: -0.05, 0.26) vs 0.22 (95%CI: 0.07, 0.37); FA peak height: -0.14 (95%CI: -0.28, 0.00) vs -0.24 (95%CI: -0.38, -0.10);). Similar findings were observed for executive function and visuomotor speed while only MD measures predicted worsening in memory domain. Interpretation This study shows that DTI histogram measures are associated with accelerated cognitive decline suggesting the utility of DTI as a pre-clinical marker in predicting the worsening of cognition in clinical trials.
Collapse
Affiliation(s)
- Yi Lin Ng
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Chuen Seng Tan
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Marco Egle
- Department of Clinical Neurosciences, Stroke Research Group, University of Cambridge, Cambridge, United Kingdom
| | - Bibek Gyanwali
- Memory Aging and Cognition Centre, National University Health System, Singapore
| | - Daniel J. Tozer
- Department of Clinical Neurosciences, Stroke Research Group, University of Cambridge, Cambridge, United Kingdom
| | - Hugh S. Markus
- Department of Clinical Neurosciences, Stroke Research Group, University of Cambridge, Cambridge, United Kingdom
| | - Christopher Chen
- Memory Aging and Cognition Centre, National University Health System, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Saima Hilal
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
19
|
Reekes TH, Ledbetter CR, Alexander JS, Stokes KY, Pardue S, Bhuiyan MAN, Patterson JC, Lofton KT, Kevil CG, Disbrow EA. Elevated plasma sulfides are associated with cognitive dysfunction and brain atrophy in human Alzheimer's disease and related dementias. Redox Biol 2023; 62:102633. [PMID: 36924684 PMCID: PMC10026043 DOI: 10.1016/j.redox.2023.102633] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 02/10/2023] [Indexed: 02/19/2023] Open
Abstract
Emerging evidence indicates that vascular stress is an important contributor to the pathophysiology of Alzheimer's disease and related dementias (ADRD). Hydrogen sulfide (H2S) and its metabolites (acid-labile (e.g., iron-sulfur clusters) and bound (e.g., per-, poly-) sulfides) have been shown to modulate both vascular and neuronal homeostasis. We recently reported that elevated plasma sulfides were associated with cognitive dysfunction and measures of microvascular disease in ADRD. Here we extend our previous work to show associations between elevated sulfides and magnetic resonance-based metrics of brain atrophy and white matter integrity. Elevated bound sulfides were associated with decreased grey matter volume, while increased acid labile sulfides were associated with decreased white matter integrity and greater ventricular volume. These findings are consistent with alterations in sulfide metabolism in ADRD which may represent maladaptive responses to oxidative stress.
Collapse
Affiliation(s)
- Tyler H Reekes
- Department of Pharmacology, Toxicology & Neuroscience, LSU Health Shreveport, United States; Center for Brain Health, LSU Health Shreveport, United States
| | - Christina R Ledbetter
- Center for Brain Health, LSU Health Shreveport, United States; Department of Neurosurgery, LSU Health Shreveport, United States
| | - J Steven Alexander
- Center for Brain Health, LSU Health Shreveport, United States; Center for Cardiovascular Diseases and Sciences, LSU Health Shreveport, United States; Department of Neurology, LSU Health Shreveport, United States; Department of Molecular and Cellular Physiology, LSU Health Shreveport, United States
| | - Karen Y Stokes
- Center for Brain Health, LSU Health Shreveport, United States; Center for Cardiovascular Diseases and Sciences, LSU Health Shreveport, United States; Department of Molecular and Cellular Physiology, LSU Health Shreveport, United States
| | - Sibile Pardue
- Center for Cardiovascular Diseases and Sciences, LSU Health Shreveport, United States; Department of Pathology and Translational Pathobiology, LSU Health Shreveport, United States
| | | | - James C Patterson
- Center for Brain Health, LSU Health Shreveport, United States; Department of Psychiatry and Behavioral Medicine, LSU Health Shreveport, United States
| | - Katelyn T Lofton
- Center for Brain Health, LSU Health Shreveport, United States; Department of Neurology, LSU Health Shreveport, United States; Department of Psychiatry and Behavioral Medicine, LSU Health Shreveport, United States
| | - Christopher G Kevil
- Center for Brain Health, LSU Health Shreveport, United States; Center for Cardiovascular Diseases and Sciences, LSU Health Shreveport, United States; Department of Pathology and Translational Pathobiology, LSU Health Shreveport, United States.
| | - Elizabeth A Disbrow
- Department of Pharmacology, Toxicology & Neuroscience, LSU Health Shreveport, United States; Center for Brain Health, LSU Health Shreveport, United States; Center for Cardiovascular Diseases and Sciences, LSU Health Shreveport, United States; Department of Neurology, LSU Health Shreveport, United States.
| |
Collapse
|
20
|
Tozer DJ, Pflanz CP, Markus HS. Reproducibility of regional structural and functional MRI networks in cerebral small vessel disease compared to age matched and stroke-free controls. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2023; 4:100167. [PMID: 37397269 PMCID: PMC10313873 DOI: 10.1016/j.cccb.2023.100167] [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: 11/10/2022] [Revised: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 07/04/2023]
Abstract
Abnormalities in structural and functional MRI connectivity measures have been reported in cerebral small vessel disease (SVD). Previous research has shown that whole-brain structural connectivity was highly reproducible in SVD patients, while whole-brain functional connectivity showed low reproducibility. It remains unclear whether the lower reproducibility of functional networks reported in SVD is due to selective disruption of reproducibility in specific networks or is generalised in patients with SVD. In this case-control study 15 SVD and 10 age-matched control participants were imaged twice with diffusion tensor imaging and resting state fMRI. Structural and functional connectivity matrices were constructed from this data and the default mode, fronto-parietal, limbic, salience, somatomotor and visual networks were extracted and the average connectivity between connections calculated and used to determine their reproducibility. Regional structural networks were more reproducible than functional networks, all structural networks showed ICC values ≥0.64 (except the salience network in SVD). The functional networks showed greater reproducibility in the controls compared to SVD with ICC values >0.7 for control participants and <=0.5 for the SVD group. The default mode network showed the greatest reproducibility for both control and SVD groups. Reproducibility of functional networks was affected by disease status with lower reproducibility in SVD compared with controls.
Collapse
Affiliation(s)
- Daniel J. Tozer
- Corresponding author at: University of Cambridge, Department of Clinical Neurosciences, Neurology Unit, R3, Box 83, Cambridge Biomedical Campus, Cambridge CB2 0QQ, United Kingdom.
| | | | | |
Collapse
|
21
|
Schwarz G, Kanber B, Prados F, Browning S, Simister R, Jäger HR, Ambler G, Gandini Wheeler-Kingshott CAM, Werring DJ. Whole-brain diffusion tensor imaging predicts 6-month functional outcome in acute intracerebral haemorrhage. J Neurol 2023; 270:2640-2648. [PMID: 36806785 PMCID: PMC10129992 DOI: 10.1007/s00415-023-11592-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 01/25/2023] [Accepted: 01/27/2023] [Indexed: 02/23/2023]
Abstract
INTRODUCTION Small vessel disease (SVD) causes most spontaneous intracerebral haemorrhage (ICH) and is associated with widespread microstructural brain tissue disruption, which can be quantified via diffusion tensor imaging (DTI) metrics: mean diffusivity (MD) and fractional anisotropy (FA). Little is known about the impact of whole-brain microstructural alterations after SVD-related ICH. We aimed to investigate: (1) association between whole-brain DTI metrics and functional outcome after ICH; and (2) predictive ability of these metrics compared to the pre-existing ICH score. METHODS Sixty-eight patients (38.2% lobar) were retrospectively included. We assessed whole-brain DTI metrics (obtained within 5 days after ICH) in cortical and deep grey matter and white matter. We used univariable logistic regression to assess the associations between DTI and clinical-radiological variables and poor outcome (modified Rankin Scale > 2). We determined the optimal predictive variables (via LASSO estimation) in: model 1 (DTI variables only), model 2 (DTI plus non-DTI variables), model 3 (DTI plus ICH score). Optimism-adjusted C-statistics were calculated for each model and compared (likelihood ratio test) against the ICH score. RESULTS Deep grey matter MD (OR 1.04 [95% CI 1.01-1.07], p = 0.010) and white matter MD (OR 1.11 [95% CI 1.01-1.23], p = 0.044) were associated (univariate analysis) with poor outcome. Discrimination values for model 1 (0.67 [95% CI 0.52-0.83]), model 2 (0.71 [95% CI 0.57-0.85) and model 3 (0.66 [95% CI 0.52-0.82]) were all significantly higher than the ICH score (0.62 [95% CI 0.49-0.75]). CONCLUSION Our exploratory study suggests that whole-brain microstructural disruption measured by DTI is associated with poor 6-month functional outcome after SVD-related ICH. Whole-brain DTI metrics performed better at predicting recovery than the existing ICH score.
Collapse
Affiliation(s)
- G Schwarz
- Neurologia-Stroke Unit ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
- Stroke Research Centre, Department of Brain Repair and Rehabilitation, Queen Square Institute of Neurology, University College London, and National Hospital for Neurology and Neurosurgery, London, UK
| | - B Kanber
- NMR Research Unit, Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, University College London (UCL) Queen Square Institute of Neurology, Faculty of Brain Sciences, UCL, London, UK
- Department of Medical Physics and Biomedical Engineering, Centre for Medical Image Computing, UCL, London, UK
- National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, UK
| | - F Prados
- NMR Research Unit, Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, University College London (UCL) Queen Square Institute of Neurology, Faculty of Brain Sciences, UCL, London, UK
- Department of Medical Physics and Biomedical Engineering, Centre for Medical Image Computing, UCL, London, UK
- National Institute for Health Research, University College London Hospitals, Biomedical Research Centre, London, UK
- E-Health Center, Universitat Oberta de Catalunya, Barcelona, Spain
| | - S Browning
- Stroke Research Centre, Department of Brain Repair and Rehabilitation, Queen Square Institute of Neurology, University College London, and National Hospital for Neurology and Neurosurgery, London, UK
| | - R Simister
- Stroke Research Centre, Department of Brain Repair and Rehabilitation, Queen Square Institute of Neurology, University College London, and National Hospital for Neurology and Neurosurgery, London, UK
| | - H R Jäger
- Lysholm Department of Neuroradiology and the Neuroradiological Academic Unit, Department of Brain Repair and Rehabilitation, UCL Institute of Neurology, Queen Square, London, UK
| | - G Ambler
- Department of Statistical Science, University College London, Gower Street, London, UK
| | - C A M Gandini Wheeler-Kingshott
- NMR Research Unit, Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, University College London (UCL) Queen Square Institute of Neurology, Faculty of Brain Sciences, UCL, London, UK
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
- Brain Connectivity Center, IRCCS Mondino Foundation, Pavia, Italy
| | - D J Werring
- Stroke Research Centre, Department of Brain Repair and Rehabilitation, Queen Square Institute of Neurology, University College London, and National Hospital for Neurology and Neurosurgery, London, UK.
| |
Collapse
|
22
|
Trofimova O, Latypova A, DiDomenicantonio G, Lutti A, de Lange AMG, Kliegel M, Stringhini S, Marques-Vidal P, Vaucher J, Vollenweider P, Strippoli MPF, Preisig M, Kherif F, Draganski B. Topography of associations between cardiovascular risk factors and myelin loss in the ageing human brain. Commun Biol 2023; 6:392. [PMID: 37037939 PMCID: PMC10086032 DOI: 10.1038/s42003-023-04741-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 03/21/2023] [Indexed: 04/12/2023] Open
Abstract
Our knowledge of the mechanisms underlying the vulnerability of the brain's white matter microstructure to cardiovascular risk factors (CVRFs) is still limited. We used a quantitative magnetic resonance imaging (MRI) protocol in a single centre setting to investigate the cross-sectional association between CVRFs and brain tissue properties of white matter tracts in a large community-dwelling cohort (n = 1104, age range 46-87 years). Arterial hypertension was associated with lower myelin and axonal density MRI indices, paralleled by higher extracellular water content. Obesity showed similar associations, though with myelin difference only in male participants. Associations between CVRFs and white matter microstructure were observed predominantly in limbic and prefrontal tracts. Additional genetic, lifestyle and psychiatric factors did not modulate these results, but moderate-to-vigorous physical activity was linked to higher myelin content independently of CVRFs. Our findings complement previously described CVRF-related changes in brain water diffusion properties pointing towards myelin loss and neuroinflammation rather than neurodegeneration.
Collapse
Affiliation(s)
- Olga Trofimova
- Laboratory for Research in Neuroimaging LREN, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Adeliya Latypova
- Laboratory for Research in Neuroimaging LREN, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Giulia DiDomenicantonio
- Laboratory for Research in Neuroimaging LREN, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Antoine Lutti
- Laboratory for Research in Neuroimaging LREN, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Ann-Marie G de Lange
- Laboratory for Research in Neuroimaging LREN, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Department of Psychology, University of Oslo, Oslo, Norway
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Matthias Kliegel
- Department of Psychology, University of Geneva, Geneva, Switzerland
| | - Silvia Stringhini
- Center for Primary Care and Public Health (Unisanté), University of Lausanne, Lausanne, Switzerland
- Institute of Social and Preventive Medicine, Lausanne University Hospital, Lausanne, Switzerland
- Unit of Population Epidemiology, Division of Primary Care Medicine, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Pedro Marques-Vidal
- Department of Medicine, Internal Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Julien Vaucher
- Department of Medicine, Internal Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Peter Vollenweider
- Department of Medicine, Internal Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Marie-Pierre F Strippoli
- Center for Research in Psychiatric Epidemiology and Psychopathology, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Martin Preisig
- Center for Research in Psychiatric Epidemiology and Psychopathology, Department of Psychiatry, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Ferath Kherif
- Laboratory for Research in Neuroimaging LREN, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Bogdan Draganski
- Laboratory for Research in Neuroimaging LREN, Centre for Research in Neurosciences, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
- Neurology Department, Max-Planck-Institute for Human Cognitive and Brain Sciences, Leipzig, Germany.
| |
Collapse
|
23
|
Xiao Y, Teng Z, Xu J, Qi Q, Guan T, Jiang X, Chen H, Xie X, Dong Y, Lv P. Systemic Immune-Inflammation Index is Associated with Cerebral Small Vessel Disease Burden and Cognitive Impairment. Neuropsychiatr Dis Treat 2023; 19:403-413. [PMID: 36852257 PMCID: PMC9960781 DOI: 10.2147/ndt.s401098] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 02/15/2023] [Indexed: 02/23/2023] Open
Abstract
OBJECTIVE This study sought to explore the associations of the systemic immune-inflammation index (SII) with total cerebral small vessel disease (CSVD) burden and cognitive impairment. METHODS We enrolled 201 patients in the retrospective study with complete clinical and laboratory data. The SII was calculated as platelet count × neutrophil count/lymphocyte count. Cognitive function was evaluated by the Mini-Mental State Examination (MMSE). Total CSVD burden was assessed based on magnetic resonance imaging. We performed logistic regression models, Spearman correlation, and mediation analysis to evaluate the associations of SII with CSVD burden and cognitive impairment. RESULTS After adjustment for confounding factors in the multivariate binary logistic regression model, elevated SII (odds ratio [OR], 3.263; 95% confidence interval [CI], 1.577-6.752; P = 0.001) or severe CSVD burden (OR, 2.794; 95% CI, 1.342-5.817; P = 0.006) was significantly associated with the risk of cognitive impairment. Correlation analyses revealed that SII levels were negatively associated with MMSE scores (rs = -0.391, P < 0.001), and positively associated with the total CSVD burden score (rs = 0.361, P < 0.001). Moreover, SII was significantly related to the severity of the CSVD burden (OR, 2.674; 95% CI, 1.359-5.263; P = 0.004). The multivariable-adjusted odds ratios (95% CI) in highest tertile versus lowest tertile of SII were 8.947 (3.315-24.145) for cognitive impairment and 4.945 (2.063-11.854) for severe CSVD burden, respectively. The effect of higher SII on cognitive impairment development was partly mediated by severe CSVD burden. CONCLUSION Elevated SII is associated with severe CSVD burden and cognitive impairment. The mediating role of severe CSVD burden suggests that higher SII may contribute to cognitive impairment through aggravating CSVD burden.
Collapse
Affiliation(s)
- Yining Xiao
- Department of Neurology, Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China.,Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, People's Republic of China.,Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Shijiazhuang, Hebei, People's Republic of China
| | - Zhenjie Teng
- Department of Neurology, Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China.,Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, People's Republic of China.,Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Shijiazhuang, Hebei, People's Republic of China
| | - Jing Xu
- Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, People's Republic of China.,Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Shijiazhuang, Hebei, People's Republic of China
| | - Qianqian Qi
- Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, People's Republic of China
| | - Tianyuan Guan
- Department of Neurology, Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China.,Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, People's Republic of China
| | - Xin Jiang
- Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, People's Republic of China.,Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Shijiazhuang, Hebei, People's Republic of China
| | - Huifang Chen
- Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, People's Republic of China
| | - Xiaohua Xie
- Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, People's Republic of China
| | - Yanhong Dong
- Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, People's Republic of China.,Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Shijiazhuang, Hebei, People's Republic of China
| | - Peiyuan Lv
- Department of Neurology, Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China.,Department of Neurology, Hebei General Hospital, Shijiazhuang, Hebei, People's Republic of China.,Hebei Provincial Key Laboratory of Cerebral Networks and Cognitive Disorders, Shijiazhuang, Hebei, People's Republic of China
| |
Collapse
|
24
|
Ip BYM, Lam BYK, Hui VMH, Au LWC, Liu MWT, Shi L, Lee VWY, Chu WCW, Leung TW, Ko H, Mok VCT. Efficacy and safety of cilostazol in decreasing progression of cerebral white matter hyperintensities-A randomized controlled trial. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12369. [PMID: 36583111 PMCID: PMC9793825 DOI: 10.1002/trc2.12369] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/20/2022] [Accepted: 11/21/2022] [Indexed: 12/28/2022]
Abstract
Introduction Cerebral small vessel disease (SVD) is an important cause of dementia that lacks effective treatment. We evaluated the efficacy and safety of cilostazol, an antiplatelet agent with potential neurovascular protective effects, in slowing the progression of white matter hyperintensities (WMHs) in stroke- and dementia-free subjects harboring confluent WMH on magnetic resonance imaging (MRI). Methods In this single-center, randomized, double-blind, placebo-controlled study, we randomized stroke- and dementia-free subjects with confluent WMHs to receive cilostazol or placebo for 2 years in a 1:1 ratio. The primary outcome was change in WMH volume over 2 years. Secondary outcomes were changes in brain volumes, lacunes, cerebral microbleeds, perivascular space, and alterations in white matter microstructural integrity, cognition, motor function, and mood. Results We recruited 120 subjects from October 27, 2014, to January 21, 2019. A total of 55 subjects in the cilostazol group and 54 subjects in the control group were included for intention-to-treat analysis. At 2-year follow-up, the changes in WMH volume were not statistically different between cilostazol treatment and placebo (0.3±1.0 mL vs -0.1±0.8 mL, p = 0.167). Secondary outcomes, bleeding and vascular events, were also not statistically different between the two groups. Discussion In this trial with stroke- and dementia-free subjects with confluent WMHs, cilostazol did not impact WMH progression but demonstrated an acceptable safety profile. Future studies should address the treatment effects of cilostazol on subjects at different clinical stages of SVD.
Collapse
Affiliation(s)
- Bonaventure Y. M. Ip
- Division of NeurologyDepartment of Medicine and TherapeuticsThe Chinese University of Hong KongShatinHong Kong SARChina
- Gerald Choa Neuroscience InstituteMargaret K.L. Cheung Research Centre for Management of ParkinsonismTherese Pei Fong Chow Research Centre for Prevention of DementiaLui Che Woo Institute of Innovative MedicineLi Ka Shing Institute of Health ScienceLau Tat‐chuen Research Centre of Brain Degenerative Diseases in ChineseFaculty of MedicineThe Chinese University of Hong KongShatinHong Kong SARChina
| | - Bonnie Y. K. Lam
- Division of NeurologyDepartment of Medicine and TherapeuticsThe Chinese University of Hong KongShatinHong Kong SARChina
- Gerald Choa Neuroscience InstituteMargaret K.L. Cheung Research Centre for Management of ParkinsonismTherese Pei Fong Chow Research Centre for Prevention of DementiaLui Che Woo Institute of Innovative MedicineLi Ka Shing Institute of Health ScienceLau Tat‐chuen Research Centre of Brain Degenerative Diseases in ChineseFaculty of MedicineThe Chinese University of Hong KongShatinHong Kong SARChina
- Nuffield Department of Clinical NeurosciencesWellcome Centre for Integrative NeuroimagingUniversity of OxfordOxfordUK
| | - Vincent M. H. Hui
- Division of NeurologyDepartment of Medicine and TherapeuticsThe Chinese University of Hong KongShatinHong Kong SARChina
- Gerald Choa Neuroscience InstituteMargaret K.L. Cheung Research Centre for Management of ParkinsonismTherese Pei Fong Chow Research Centre for Prevention of DementiaLui Che Woo Institute of Innovative MedicineLi Ka Shing Institute of Health ScienceLau Tat‐chuen Research Centre of Brain Degenerative Diseases in ChineseFaculty of MedicineThe Chinese University of Hong KongShatinHong Kong SARChina
| | - Lisa W. C. Au
- Division of NeurologyDepartment of Medicine and TherapeuticsThe Chinese University of Hong KongShatinHong Kong SARChina
- Gerald Choa Neuroscience InstituteMargaret K.L. Cheung Research Centre for Management of ParkinsonismTherese Pei Fong Chow Research Centre for Prevention of DementiaLui Che Woo Institute of Innovative MedicineLi Ka Shing Institute of Health ScienceLau Tat‐chuen Research Centre of Brain Degenerative Diseases in ChineseFaculty of MedicineThe Chinese University of Hong KongShatinHong Kong SARChina
| | - Mandy W. T. Liu
- Division of NeurologyDepartment of Medicine and TherapeuticsThe Chinese University of Hong KongShatinHong Kong SARChina
- Gerald Choa Neuroscience InstituteMargaret K.L. Cheung Research Centre for Management of ParkinsonismTherese Pei Fong Chow Research Centre for Prevention of DementiaLui Che Woo Institute of Innovative MedicineLi Ka Shing Institute of Health ScienceLau Tat‐chuen Research Centre of Brain Degenerative Diseases in ChineseFaculty of MedicineThe Chinese University of Hong KongShatinHong Kong SARChina
| | - Lin Shi
- Department of Imaging and Interventional RadiologyThe Prince of Wale HospitalThe Chinese University of Hong KongShatinHong Kong SARChina
- BrainNow Research InstituteShenzhenGuangdong ProvinceChina
| | - Vivian W. Y. Lee
- Centre for Learning Enhancement and ResearchThe Chinese University of Hong KongHong Kong SARChina
| | - Winnie C. W. Chu
- Department of Imaging and Interventional RadiologyThe Prince of Wale HospitalThe Chinese University of Hong KongShatinHong Kong SARChina
| | - Thomas W. Leung
- Division of NeurologyDepartment of Medicine and TherapeuticsThe Chinese University of Hong KongShatinHong Kong SARChina
| | - Ho Ko
- Division of NeurologyDepartment of Medicine and TherapeuticsThe Chinese University of Hong KongShatinHong Kong SARChina
- Gerald Choa Neuroscience InstituteMargaret K.L. Cheung Research Centre for Management of ParkinsonismTherese Pei Fong Chow Research Centre for Prevention of DementiaLui Che Woo Institute of Innovative MedicineLi Ka Shing Institute of Health ScienceLau Tat‐chuen Research Centre of Brain Degenerative Diseases in ChineseFaculty of MedicineThe Chinese University of Hong KongShatinHong Kong SARChina
| | - Vincent C. T. Mok
- Division of NeurologyDepartment of Medicine and TherapeuticsThe Chinese University of Hong KongShatinHong Kong SARChina
- Gerald Choa Neuroscience InstituteMargaret K.L. Cheung Research Centre for Management of ParkinsonismTherese Pei Fong Chow Research Centre for Prevention of DementiaLui Che Woo Institute of Innovative MedicineLi Ka Shing Institute of Health ScienceLau Tat‐chuen Research Centre of Brain Degenerative Diseases in ChineseFaculty of MedicineThe Chinese University of Hong KongShatinHong Kong SARChina
| |
Collapse
|
25
|
Wang S, Zhang F, Huang P, Hong H, Jiaerken Y, Yu X, Zhang R, Zeng Q, Zhang Y, Kikinis R, Rathi Y, Makris N, Lou M, Pasternak O, Zhang M, O'Donnell LJ. Superficial white matter microstructure affects processing speed in cerebral small vessel disease. Hum Brain Mapp 2022; 43:5310-5325. [PMID: 35822593 PMCID: PMC9812245 DOI: 10.1002/hbm.26004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 06/10/2022] [Accepted: 06/15/2022] [Indexed: 01/15/2023] Open
Abstract
White matter hyperintensities (WMH) are a typical feature of cerebral small vessel disease (CSVD), which contributes to about 50% of dementias worldwide. Microstructural alterations in deep white matter (DWM) have been widely examined in CSVD. However, little is known about abnormalities in superficial white matter (SWM) and their relevance for processing speed, the main cognitive deficit in CSVD. In 141 CSVD patients, processing speed was assessed using Trail Making Test Part A. White matter abnormalities were assessed by WMH burden (volume on T2-FLAIR) and diffusion MRI measures. SWM imaging measures had a large contribution to processing speed, despite a relatively low SWM WMH burden. Across all imaging measures, SWM free water (FW) had the strongest association with processing speed, followed by SWM mean diffusivity (MD). SWM FW was the only marker to significantly increase between two subgroups with the lowest WMH burdens. When comparing two subgroups with the highest WMH burdens, the involvement of WMH in the SWM was accompanied by significant differences in processing speed and white matter microstructure. Mediation analysis revealed that SWM FW fully mediated the association between WMH volume and processing speed, while no mediation effect of MD or DWM FW was observed. Overall, results suggest that the SWM has an important contribution to processing speed, while SWM FW is a sensitive imaging marker associated with cognition in CSVD. This study extends the current understanding of CSVD-related dysfunction and suggests that the SWM, as an understudied region, can be a potential target for monitoring pathophysiological processes.
Collapse
Affiliation(s)
- Shuyue Wang
- Department of Radiologythe Second Affiliated Hospital of Zhejiang University School of MedicineChina
- Brigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Fan Zhang
- Brigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Peiyu Huang
- Department of Radiologythe Second Affiliated Hospital of Zhejiang University School of MedicineChina
| | - Hui Hong
- Department of Radiologythe Second Affiliated Hospital of Zhejiang University School of MedicineChina
| | - Yeerfan Jiaerken
- Department of Radiologythe Second Affiliated Hospital of Zhejiang University School of MedicineChina
| | - Xinfeng Yu
- Department of Radiologythe Second Affiliated Hospital of Zhejiang University School of MedicineChina
| | - Ruiting Zhang
- Department of Radiologythe Second Affiliated Hospital of Zhejiang University School of MedicineChina
| | - Qingze Zeng
- Department of Radiologythe Second Affiliated Hospital of Zhejiang University School of MedicineChina
| | - Yao Zhang
- Department of Radiologythe Second Affiliated Hospital of Zhejiang University School of MedicineChina
| | - Ron Kikinis
- Brigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Yogesh Rathi
- Brigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Nikos Makris
- Brigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Center for Morphometric AnalysisMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Min Lou
- Department of Neurologythe Second Affiliated Hospital of Zhejiang University School of MedicineChina
| | - Ofer Pasternak
- Brigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Minming Zhang
- Department of Radiologythe Second Affiliated Hospital of Zhejiang University School of MedicineChina
| | | |
Collapse
|
26
|
Liu Y, Cao S, Du B, Zhang J, Chen C, Hu P, Tian Y, Wang K, Ji GJ, Wei Q. Different Dynamic Nodal Properties Contribute to Cognitive Impairment in Patients with White Matter Hyperintensities. Brain Sci 2022; 12:1527. [PMID: 36421852 PMCID: PMC9688268 DOI: 10.3390/brainsci12111527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/09/2022] [Accepted: 10/25/2022] [Indexed: 12/31/2024] Open
Abstract
White matter hyperintensities (WMHs) are commonly observed in older adults and are associated with cognitive impairment. Although previous studies have found abnormal functional connectivities in patients with WMHs based on static functional magnetic resonance imaging (fMRI), the topological properties in the context of brain dynamics remain relatively unexplored. Herein, we explored disrupted dynamic topological properties of functional network connectivity in patients with WMHs and its relationship with cognitive impairment. We included 36 healthy controls (HC) and 104 patients with mild WMHs (n = 39), moderate WMHs (n = 37), and severe (n = 28) WMHs. The fMRI data of all participants were analyzed using Anatomical Automatic Labeling (AAL) and a sliding-window approach to generate dynamic functional connectivity matrics. Then, graph theory methods were applied to calculate the topological properties. Comprehensive neuropsychological scales were used to assess cognitive functions. Relationships between cognitive functions and abnormal dynamic topological properties were evaluated by Pearson's correlation. We found that the patients with WMHs had higher temporal variability in regional properties, including betweenness centrality, nodal efficiencies, and nodal clustering coefficient. Furthermore, we found that the degree of centrality was related to executive function and memory, and the local coefficient correlated to executive function. Our results indicate that patients with WMHs have higher temporal variabilities in regional properties and are associated with executive and memory function.
Collapse
Affiliation(s)
- Yuanyuan Liu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei 230032, China
| | - Shanshan Cao
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei 230032, China
| | - Baogen Du
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei 230032, China
| | - Jun Zhang
- Department of Neurology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Chen Chen
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei 230032, China
| | - Panpan Hu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei 230032, China
- The College of Mental Health and Psychological Sciences, Anhui Medical University, Hefei 230022, China
| | - Yanghua Tian
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei 230032, China
- Department of Neurology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
- The College of Mental Health and Psychological Sciences, Anhui Medical University, Hefei 230022, China
| | - Kai Wang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei 230032, China
- The College of Mental Health and Psychological Sciences, Anhui Medical University, Hefei 230022, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei 230032, China
| | - Gong-Jun Ji
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei 230032, China
| | - Qiang Wei
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
- Collaborative Innovation Center of Neuropsychiatric Disorders and Mental Health, Hefei 230032, China
- Anhui Province Key Laboratory of Cognition and Neuropsychiatric Disorders, Hefei 230032, China
| |
Collapse
|
27
|
Markus HS, van Der Flier WM, Smith EE, Bath P, Biessels GJ, Briceno E, Brodtman A, Chabriat H, Chen C, de Leeuw FE, Egle M, Ganesh A, Georgakis MK, Gottesman RF, Kwon S, Launer L, Mok V, O'Brien J, Ottenhoff L, Pendlebury S, Richard E, Sachdev P, Schmidt R, Springer M, Tiedt S, Wardlaw JM, Verdelho A, Webb A, Werring D, Duering M, Levine D, Dichgans M. Framework for Clinical Trials in Cerebral Small Vessel Disease (FINESSE): A Review. JAMA Neurol 2022; 79:1187-1198. [PMID: 35969390 PMCID: PMC11036410 DOI: 10.1001/jamaneurol.2022.2262] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Importance Cerebral small vessel disease (SVD) causes a quarter of strokes and is the most common pathology underlying vascular cognitive impairment and dementia. An important step to developing new treatments is better trial methodology. Disease mechanisms in SVD differ from other stroke etiologies; therefore, treatments need to be evaluated in cohorts in which SVD has been well characterized. Furthermore, SVD itself can be caused by a number of different pathologies, the most common of which are arteriosclerosis and cerebral amyloid angiopathy. To date, there have been few sufficiently powered high-quality randomized clinical trials in SVD, and inconsistent trial methodology has made interpretation of some findings difficult. Observations To address these issues and develop guidelines for optimizing design of clinical trials in SVD, the Framework for Clinical Trials in Cerebral Small Vessel Disease (FINESSE) was created under the auspices of the International Society of Vascular Behavioral and Cognitive Disorders. Experts in relevant aspects of SVD trial methodology were convened, and a structured Delphi consensus process was used to develop recommendations. Areas in which recommendations were developed included optimal choice of study populations, choice of clinical end points, use of brain imaging as a surrogate outcome measure, use of circulating biomarkers for participant selection and as surrogate markers, novel trial designs, and prioritization of therapeutic agents using genetic data via Mendelian randomization. Conclusions and Relevance The FINESSE provides recommendations for trial design in SVD for which there are currently few effective treatments. However, new insights into understanding disease pathogenesis, particularly from recent genetic studies, provide novel pathways that could be therapeutically targeted. In addition, whether other currently available cardiovascular interventions are specifically effective in SVD, as opposed to other subtypes of stroke, remains uncertain. FINESSE provides a framework for design of trials examining such therapeutic approaches.
Collapse
Affiliation(s)
- Hugh S Markus
- Alzheimer Center Amsterdam, Department of Neurology, Epidemiology and Data Science, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Wiesje M van Der Flier
- Alzheimer Center Amsterdam, Department of Neurology, Epidemiology and Data Science, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Eric E Smith
- Department of Clinical Neurosciences and Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Philip Bath
- Stroke Trials Unit, Mental Health & Clinical Neuroscience, University of Nottingham, Nottingham, United Kingdom
| | - Geert Jan Biessels
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Emily Briceno
- Department of Physical Medicine & Rehabilitation, University of Michigan Medical School, Ann Arbor
| | - Amy Brodtman
- Florey Institute of Neuroscience and Mental Health, Melbourne, Victoria, Australia
- University of Melbourne, Melbourne, Victoria, Australia
- Monash University, Melbourne, Victoria, Australia
| | - Hugues Chabriat
- Department of Neurology, FHU NeuroVasc, APHP, University of Paris, Paris, France
| | - Christopher Chen
- Memory Aging and Cognition Centre, Departments of Pharmacology and Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Frank-Erik de Leeuw
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijimegen, the Netherlands
| | - Marco Egle
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Aravind Ganesh
- Department of Clinical Neurosciences and Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Marios K Georgakis
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Munich, Germany
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Rebecca F Gottesman
- Now with National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, Maryland
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sun Kwon
- University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Lenore Launer
- Intramural Research Program, National Institute on Aging, Baltimore, Maryland
| | - Vincent Mok
- Gerald Choa Neuroscience Centre, Lui Che Woo Institute of Innovative Medicine, Margaret K.L. Cheung Research Centre for Management of Parkinsonism, Division of Neurology, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - John O'Brien
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Lois Ottenhoff
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam and the Netherlands and Brain Research Center Amsterdam, the Netherlands
| | - Sarah Pendlebury
- Wolfson Centre for Prevention of Stroke and Dementia, Nuffield Department of Clinical Neurosciences, University of Oxford, NIHR Oxford Biomedical Research Centre, Departments of General (internal) Medicine and Geratology, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Edo Richard
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, Nijimegen, the Netherlands
| | - Perminder Sachdev
- Centre for Healthy Brain Ageing (CHeBA), University of New South Wales, Sydney, New South Wales, Australia
| | - Reinhold Schmidt
- Department of Neurology, Clinical Division of Neurogeriatrics, Medical University Graz, Graz, Austria
| | | | - Stefan Tiedt
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Joanna M Wardlaw
- Centre for Clinical Brain Sciences, UK Dementia Research Institute Centre at the University of Edinburgh, Edinburgh, United Kingdom
| | - Ana Verdelho
- Faculdade de Medicina, Department of Neurosciences and Mental Health, CHULN-Hospital de Santa Maria Instituto de Medicina Molecular (IMM) e Instituto de Saúde Ambiental (ISAMB), University of Lisbon, Lisbon, Portugal
| | - Alastair Webb
- Wolfson Centre for Prevention of Stroke and Dementia, Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - David Werring
- Stroke Research Centre, Department of Brain Repair and Rehabilitation, UCL Queen Square Institute of Neurology and the National Hospital for Neurology and Neurosurgery, London, United Kingdom
| | - Marco Duering
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Medical Image Analysis Center (MIAC AG) and Quantitative Biomedical Imaging Group, Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Deborah Levine
- Departments of Internal Medicine and Neurology, University of Michigan, Ann Arbor
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
28
|
Pflanz CP, Egle MS, O'Brien JT, Morris RG, Barrick TR, Blamire AM, Ford GA, Tozer D, Markus HS. Association of Blood Pressure Lowering Intensity With White Matter Network Integrity in Patients With Cerebral Small Vessel Disease. Neurology 2022; 99:e1945-e1953. [PMID: 35977831 PMCID: PMC9620809 DOI: 10.1212/wnl.0000000000201018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 06/13/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Diffusion tensor imaging (DTI) networks integrate damage from a variety of pathologic processes in cerebral small vessel disease (SVD) and may be a sensitive marker to detect treatment effects. We determined whether brain network analysis could detect treatment effects in the PRESERVE trial data set, in which intensive vs standard blood pressure (BP) lowering was compared. The primary end point of DTI had not shown treatment differences. METHODS Participants with lacunar stroke were randomized to standard (systolic 130-140 mm Hg) or intensive (systolic ≤ 125 mm Hg) BP lowering and followed for 2 years with MRI at baseline and at 2 years. Graph theory-based metrics were derived from DTI data to produce a measure of network integrity weighted global efficiency and compared with individual MRI markers of DTI, brain volume, and white matter hyperintensities. RESULTS Data were available in 82 subjects: standard n = 40 (mean age 66.3 ± 1.5 years) and intensive n = 42 (mean age 69.6 ± 1.0 years). The mean (SD) systolic BP was reduced by 13(14) and 23(23) mm Hg in the standard and intensive groups, respectively (p < 0.001 between groups). Significant differences in diffusion network metrics were found, with improved network integrity (weighted global efficiency, p = 0.002) seen with intensive BP lowering. In contrast, there were no significant differences in individual MRI markers including DTI histogram metrics, brain volume, or white matter hyperintensities. DISCUSSION Brain network analysis may be a sensitive surrogate marker in trials in SVD. This work suggests that measures of brain network efficiency may be more sensitive to the effects of BP control treatment than conventional DTI metrics. TRIAL REGISTRATION INFORMATION The trial is registered with the ISRCTN Registry (ISRCTN37694103; doi.org/10.1186/ISRCTN37694103) and the NIHR Clinical Research Network (CRN 10962; public-odp.nihr.ac.uk/QvAJAXZfc/opendoc.htm?document=crncc_users%5Cfind%20a%20clinical%20research%20study.qvw&lang=en-US&host=QVS%40crn-prod-odp-pu&anonymous=true). CLASSIFICATION OF EVIDENCE This study provides Class II evidence that intensive BP lowering in patients with SVD results in improved brain network function when assessed by DTI-based brain network metrics.
Collapse
Affiliation(s)
- Chris Patrick Pflanz
- From the Stroke Research Group (C.P.P., M.S.E., D.T., H.S.M.), Department of Clinical Neuroscience, University of Cambridge; Department of Psychiatry (J.T.O.B.), University of Cambridge; Kings College Institute of Psychiatry (R.G.M.), Psychology and Neurosciences, London, UK; Molecular and Clinical Science Research Institute (T.R.B.), St George's, University of London, UK; Magnetic Resonance Centre (A.M.B.), Institute of Cellular Medicine, Newcastle University, UK; and Oxford University Hospitals NHS Foundation Trust & University of Oxford (G.A.F.)
| | - Marco S Egle
- From the Stroke Research Group (C.P.P., M.S.E., D.T., H.S.M.), Department of Clinical Neuroscience, University of Cambridge; Department of Psychiatry (J.T.O.B.), University of Cambridge; Kings College Institute of Psychiatry (R.G.M.), Psychology and Neurosciences, London, UK; Molecular and Clinical Science Research Institute (T.R.B.), St George's, University of London, UK; Magnetic Resonance Centre (A.M.B.), Institute of Cellular Medicine, Newcastle University, UK; and Oxford University Hospitals NHS Foundation Trust & University of Oxford (G.A.F.)
| | - John T O'Brien
- From the Stroke Research Group (C.P.P., M.S.E., D.T., H.S.M.), Department of Clinical Neuroscience, University of Cambridge; Department of Psychiatry (J.T.O.B.), University of Cambridge; Kings College Institute of Psychiatry (R.G.M.), Psychology and Neurosciences, London, UK; Molecular and Clinical Science Research Institute (T.R.B.), St George's, University of London, UK; Magnetic Resonance Centre (A.M.B.), Institute of Cellular Medicine, Newcastle University, UK; and Oxford University Hospitals NHS Foundation Trust & University of Oxford (G.A.F.)
| | - Robin G Morris
- From the Stroke Research Group (C.P.P., M.S.E., D.T., H.S.M.), Department of Clinical Neuroscience, University of Cambridge; Department of Psychiatry (J.T.O.B.), University of Cambridge; Kings College Institute of Psychiatry (R.G.M.), Psychology and Neurosciences, London, UK; Molecular and Clinical Science Research Institute (T.R.B.), St George's, University of London, UK; Magnetic Resonance Centre (A.M.B.), Institute of Cellular Medicine, Newcastle University, UK; and Oxford University Hospitals NHS Foundation Trust & University of Oxford (G.A.F.)
| | - Thomas R Barrick
- From the Stroke Research Group (C.P.P., M.S.E., D.T., H.S.M.), Department of Clinical Neuroscience, University of Cambridge; Department of Psychiatry (J.T.O.B.), University of Cambridge; Kings College Institute of Psychiatry (R.G.M.), Psychology and Neurosciences, London, UK; Molecular and Clinical Science Research Institute (T.R.B.), St George's, University of London, UK; Magnetic Resonance Centre (A.M.B.), Institute of Cellular Medicine, Newcastle University, UK; and Oxford University Hospitals NHS Foundation Trust & University of Oxford (G.A.F.)
| | - Andrew M Blamire
- From the Stroke Research Group (C.P.P., M.S.E., D.T., H.S.M.), Department of Clinical Neuroscience, University of Cambridge; Department of Psychiatry (J.T.O.B.), University of Cambridge; Kings College Institute of Psychiatry (R.G.M.), Psychology and Neurosciences, London, UK; Molecular and Clinical Science Research Institute (T.R.B.), St George's, University of London, UK; Magnetic Resonance Centre (A.M.B.), Institute of Cellular Medicine, Newcastle University, UK; and Oxford University Hospitals NHS Foundation Trust & University of Oxford (G.A.F.)
| | - Gary A Ford
- From the Stroke Research Group (C.P.P., M.S.E., D.T., H.S.M.), Department of Clinical Neuroscience, University of Cambridge; Department of Psychiatry (J.T.O.B.), University of Cambridge; Kings College Institute of Psychiatry (R.G.M.), Psychology and Neurosciences, London, UK; Molecular and Clinical Science Research Institute (T.R.B.), St George's, University of London, UK; Magnetic Resonance Centre (A.M.B.), Institute of Cellular Medicine, Newcastle University, UK; and Oxford University Hospitals NHS Foundation Trust & University of Oxford (G.A.F.)
| | - Daniel Tozer
- From the Stroke Research Group (C.P.P., M.S.E., D.T., H.S.M.), Department of Clinical Neuroscience, University of Cambridge; Department of Psychiatry (J.T.O.B.), University of Cambridge; Kings College Institute of Psychiatry (R.G.M.), Psychology and Neurosciences, London, UK; Molecular and Clinical Science Research Institute (T.R.B.), St George's, University of London, UK; Magnetic Resonance Centre (A.M.B.), Institute of Cellular Medicine, Newcastle University, UK; and Oxford University Hospitals NHS Foundation Trust & University of Oxford (G.A.F.)
| | - Hugh S Markus
- From the Stroke Research Group (C.P.P., M.S.E., D.T., H.S.M.), Department of Clinical Neuroscience, University of Cambridge; Department of Psychiatry (J.T.O.B.), University of Cambridge; Kings College Institute of Psychiatry (R.G.M.), Psychology and Neurosciences, London, UK; Molecular and Clinical Science Research Institute (T.R.B.), St George's, University of London, UK; Magnetic Resonance Centre (A.M.B.), Institute of Cellular Medicine, Newcastle University, UK; and Oxford University Hospitals NHS Foundation Trust & University of Oxford (G.A.F.).
| |
Collapse
|
29
|
Fouto AR, Nunes RG, Pinto J, Alves L, Calado S, Gonçalves C, Rebolo M, Viana-Baptista M, Vilela P, Figueiredo P. Impact of white-matter mask selection on DTI histogram-based metrics as potential biomarkers in cerebral small vessel disease. MAGMA (NEW YORK, N.Y.) 2022; 35:779-790. [PMID: 34997895 DOI: 10.1007/s10334-021-00991-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 12/09/2021] [Accepted: 12/11/2021] [Indexed: 06/14/2023]
Abstract
OBJECTIVE Histogram-based metrics extracted from diffusion-tensor imaging (DTI) have been suggested as potential biomarkers for cerebral small vessel disease (SVD), but methods and results have varied across studies. This work aims to assess the impact of mask selection for extracting histogram-based metrics of fractional anisotropy (FA) and mean diffusivity (MD) on their sensitivity as SVD biomarkers. METHODS DTI data were collected from 17 SVD patients and 12 healthy controls. FA and MD maps were estimated; from these, histograms were computed on two whole-brain white-matter masks: normal-appearing white-matter (NAWM) and mean FA tract skeleton (TBSS). Histogram-based metrics (median, peak height, peak width, peak value) were extracted from the FA and MD maps. These were compared between groups and correlated with the patients' cognitive scores (executive function and processing speed). RESULTS White-matter mask selection significantly impacted FA and MD histogram metrics. In particular, significant interactions were found between Mask and Group for FA peak height (p = 0.027), MD Median (p = 0.035) and MD peak width (p = 0.047); indicating that the mask used affected their ability to discriminate between groups. In fact, MD peak width showed a significant 8.8% increase in patients when using TBSS (p = 0.037), but not when using NAWM (p = 0.69). Moreover, the mask may have an effect on the correlations with cognitive measures. Nevertheless, MD peak width (TBSS: r = - 0.75, NAWM: r = - 0.71) and MD peak height (TBSS: r = 0.65, NAWM: r = 0.62) remained significantly correlated with executive function, regardless of the mask. CONCLUSION The impact of the processing methodology, in particular the choice of white-matter mask, highlights the need for standardized MRI data-processing pipelines.
Collapse
Affiliation(s)
- Ana R Fouto
- Institute for Systems and Robotics - Lisboa and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1, 1049-001, Lisbon, Portugal.
| | - Rita G Nunes
- Institute for Systems and Robotics - Lisboa and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1, 1049-001, Lisbon, Portugal
| | - Joana Pinto
- Institute for Systems and Robotics - Lisboa and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1, 1049-001, Lisbon, Portugal
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Luísa Alves
- Neurology Department, Hospital Egas Moniz, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
- CEDOC - NOVA Medical School, NOVA University of Lisbon, Lisbon, Portugal
| | - Sofia Calado
- Neurology Department, Hospital Egas Moniz, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
- CEDOC - NOVA Medical School, NOVA University of Lisbon, Lisbon, Portugal
| | - Carina Gonçalves
- Neurology Department, Hospital Egas Moniz, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
- CEDOC - NOVA Medical School, NOVA University of Lisbon, Lisbon, Portugal
| | | | - Miguel Viana-Baptista
- Neurology Department, Hospital Egas Moniz, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
- CEDOC - NOVA Medical School, NOVA University of Lisbon, Lisbon, Portugal
| | - Pedro Vilela
- Imaging Department, Hospital da Luz, Lisbon, Portugal
| | - Patrícia Figueiredo
- Institute for Systems and Robotics - Lisboa and Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1, 1049-001, Lisbon, Portugal
| |
Collapse
|
30
|
De Luca A, Kuijf H, Exalto L, Thiebaut de Schotten M, Biessels GJ. Multimodal tract-based MRI metrics outperform whole brain markers in determining cognitive impact of small vessel disease-related brain injury. Brain Struct Funct 2022; 227:2553-2567. [PMID: 35994115 PMCID: PMC9418106 DOI: 10.1007/s00429-022-02546-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/27/2022] [Indexed: 01/04/2023]
Abstract
In cerebral small vessel disease (cSVD), whole brain MRI markers of cSVD-related brain injury explain limited variance to support individualized prediction. Here, we investigate whether considering abnormalities in brain tracts by integrating multimodal metrics from diffusion MRI (dMRI) and structural MRI (sMRI), can better capture cognitive performance in cSVD patients than established approaches based on whole brain markers. We selected 102 patients (73.7 ± 10.2 years old, 59 males) with MRI-visible SVD lesions and both sMRI and dMRI. Conventional linear models using demographics and established whole brain markers were used as benchmark of predicting individual cognitive scores. Multi-modal metrics of 73 major brain tracts were derived from dMRI and sMRI, and used together with established markers as input of a feed-forward artificial neural network (ANN) to predict individual cognitive scores. A feature selection strategy was implemented to reduce the risk of overfitting. Prediction was performed with leave-one-out cross-validation and evaluated with the R2 of the correlation between measured and predicted cognitive scores. Linear models predicted memory and processing speed with R2 = 0.26 and R2 = 0.38, respectively. With ANN, feature selection resulted in 13 tract-specific metrics and 5 whole brain markers for predicting processing speed, and 28 tract-specific metrics and 4 whole brain markers for predicting memory. Leave-one-out ANN prediction with the selected features achieved R2 = 0.49 and R2 = 0.40 for processing speed and memory, respectively. Our results show proof-of-concept that combining tract-specific multimodal MRI metrics can improve the prediction of cognitive performance in cSVD by leveraging tract-specific multi-modal metrics.
Collapse
Affiliation(s)
- Alberto De Luca
- VCI Group, Neurology Department, UMC Utrecht Brain Center, UMC Utrecht, Utrecht, The Netherlands.
- Image Sciences Institute, Division Imaging and Oncology, UMC Utrecht, Utrecht, The Netherlands.
| | - Hugo Kuijf
- Image Sciences Institute, Division Imaging and Oncology, UMC Utrecht, Utrecht, The Netherlands
| | - Lieza Exalto
- VCI Group, Neurology Department, UMC Utrecht Brain Center, UMC Utrecht, Utrecht, The Netherlands
| | - Michel Thiebaut de Schotten
- Brain Connectivity and Behaviour Lab, Sorbonne University, Paris, France
- Institut des Maladies Neurodégénératives, Neurofunctional Imaging Group, University of Bordeaux, Bordeaux, France
| | - Geert-Jan Biessels
- VCI Group, Neurology Department, UMC Utrecht Brain Center, UMC Utrecht, Utrecht, The Netherlands
| |
Collapse
|
31
|
Biondo F, Jewell A, Pritchard M, Aarsland D, Steves CJ, Mueller C, Cole JH. Brain-age is associated with progression to dementia in memory clinic patients. Neuroimage Clin 2022; 36:103175. [PMID: 36087560 PMCID: PMC9467894 DOI: 10.1016/j.nicl.2022.103175] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 07/30/2022] [Accepted: 08/27/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Biomarkers for the early detection of dementia risk hold promise for better disease monitoring and targeted interventions. However, most biomarker studies, particularly in neuroimaging, have analysed artificially 'clean' research groups, free from comorbidities, erroneous referrals, contraindications and from a narrow sociodemographic pool. Such biases mean that neuroimaging samples are often unrepresentative of the target population for dementia risk (e.g., people referred to a memory clinic), limiting the generalisation of these studies to real-world clinical settings. To facilitate better translation from research to the clinic, datasets that are more representative of dementia patient groups are warranted. METHODS We analysed T1-weighted MRI scans from a real-world setting of patients referred to UK memory clinic services (n = 1140; 60.2 % female and mean [SD] age of 70.0[10.8] years) to derive 'brain-age'. Brain-age is an index of age-related brain health based on quantitative analysis of structural neuroimaging, largely reflecting brain atrophy. Brain-predicted age difference (brain-PAD) was calculated as brain-age minus chronological age. We determined which patients went on to develop dementia between three months and 7.8 years after neuroimaging assessment (n = 476) using linkage to electronic health records. RESULTS Survival analysis, using Cox regression, indicated a 3 % increased risk of dementia per brain-PAD year (hazard ratio [95 % CI] = 1.03 [1.02,1.04], p < 0.0001), adjusted for baseline age, age2, sex, Mini Mental State Examination (MMSE) score and normalised brain volume. In sensitivity analyses, brain-PAD remained significant when time-to-dementia was at least 3 years (hazard ratio [95 % CI] = 1.06 [1.02, 1.09], p = 0.0006), or when baseline MMSE score ≥ 27 (hazard ratio [95 % CI] = 1.03 [1.01, 1.05], p = 0.0006). CONCLUSIONS Memory clinic patients with older-appearing brains are more likely to receive a subsequent dementia diagnosis. Potentially, brain-age could aid decision-making during initial memory clinic assessment to improve early detection of dementia. Even when neuroimaging assessment was more than 3 years prior to diagnosis and when cognitive functioning was not clearly impaired, brain-age still proved informative. These real-world results support the use of quantitative neuroimaging biomarkers like brain-age in memory clinics.
Collapse
Affiliation(s)
- Francesca Biondo
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, SE5 8AF, UK; South London and Maudsley NHS Foundation Trust, UK; Centre for Medical Image Computing, Department of Computer Science, University College London, WC1V 6LJ, UK.
| | | | | | - Dag Aarsland
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, SE5 8AF, UK; Centre for Age-Related Research, Stavanger University Hospital, Stavanger, Norway
| | - Claire J Steves
- Department of Ageing and Health, Guy's and St Thomas' NHS Foundation Trust, SE1 7EH, UK; Department of Twin Research and Genetic Epidemiology, King's College London, SE1 7EH, UK
| | - Christoph Mueller
- South London and Maudsley NHS Foundation Trust, UK; Department of Old Age Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, SE5 8AF, UK
| | - James H Cole
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, SE5 8AF, UK; South London and Maudsley NHS Foundation Trust, UK; Centre for Medical Image Computing, Department of Computer Science, University College London, WC1V 6LJ, UK; Dementia Research Centre, Institute of Neurology, University College London, WC1N 3AR, UK.
| |
Collapse
|
32
|
da Silva PHR, Paschoal AM, Secchinatto KF, Zotin MCZ, Dos Santos AC, Viswanathan A, Pontes-Neto OM, Leoni RF. Contrast agent-free state-of-the-art magnetic resonance imaging on cerebral small vessel disease - Part 2: Diffusion tensor imaging and functional magnetic resonance imaging. NMR IN BIOMEDICINE 2022; 35:e4743. [PMID: 35429070 DOI: 10.1002/nbm.4743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 04/04/2022] [Indexed: 06/14/2023]
Abstract
Cerebral small vessel disease (cSVD) has been widely studied using conventional magnetic resonance imaging (MRI) methods, although the association between MRI findings and clinical features of cSVD is not always concordant. We assessed the additional contribution of contrast agent-free, state-of-the-art MRI techniques, particularly diffusion tensor imaging (DTI) and functional magnetic resonance imaging (fMRI), to understand brain damage and structural and functional connectivity impairment related to cSVD. We performed a review following the PICOS worksheet and Search Strategy, including 152 original papers in English, published from 2000 to 2022. For each MRI method, we extracted information about their contributions regarding the origins, pathology, markers, and clinical outcomes in cSVD. In general, DTI studies have shown that changes in mean, radial, and axial diffusivity measures are related to the presence of cSVD. In addition to the classical deficit in executive functions and processing speed, fMRI studies indicate connectivity dysfunctions in other domains, such as sensorimotor, memory, and attention. Neuroimaging metrics have been correlated with the diagnosis, prognosis, and rehabilitation of patients with cSVD. In short, the application of contrast agent-free, state-of-the-art MRI techniques has provided a complete picture of cSVD markers and tools to explore questions that have not yet been clarified about this clinical condition. Longitudinal studies are desirable to look for causal relationships between image biomarkers and clinical outcomes.
Collapse
Affiliation(s)
| | - André Monteiro Paschoal
- Department of Physics, FFCLRP, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Medical Imaging, Hematology and Clinical Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Maria Clara Zanon Zotin
- Department of Medical Imaging, Hematology and Clinical Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Antônio Carlos Dos Santos
- Department of Medical Imaging, Hematology and Clinical Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Anand Viswanathan
- J Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Octavio M Pontes-Neto
- Department of Neurosciences and Behavioral Science, Ribeirão Preto Medical School, University of Sao Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Renata Ferranti Leoni
- Department of Physics, FFCLRP, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
33
|
Harshfield EL, Sands CJ, Tuladhar AM, de Leeuw FE, Lewis MR, Markus HS. Metabolomic profiling in small vessel disease identifies multiple associations with disease severity. Brain 2022; 145:2461-2471. [PMID: 35254405 PMCID: PMC9337813 DOI: 10.1093/brain/awac041] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 12/20/2021] [Accepted: 01/11/2022] [Indexed: 11/17/2022] Open
Abstract
Cerebral small vessel disease is a major cause of vascular cognitive impairment and dementia. There are few treatments, largely reflecting limited understanding of the underlying pathophysiology. Metabolomics can be used to identify novel risk factors to better understand pathogenesis and to predict disease progression and severity. We analysed data from 624 patients with symptomatic cerebral small vessel disease from two prospective cohort studies. Serum samples were collected at baseline and patients underwent MRI scans and cognitive testing at regular intervals with up to 14 years of follow-up. Using ultra-performance liquid chromatography-mass spectrometry and nuclear magnetic resonance spectroscopy, we obtained metabolic and lipidomic profiles from 369 annotated metabolites and 54 764 unannotated features and examined their association with respect to disease severity, assessed using MRI small vessel disease markers, cognition and future risk of all-cause dementia. Our analysis identified 28 metabolites that were significantly associated with small vessel disease imaging markers and cognition. Decreased levels of multiple glycerophospholipids and sphingolipids were associated with increased small vessel disease load as evidenced by higher white matter hyperintensity volume, lower mean diffusivity normalized peak height, greater brain atrophy and impaired cognition. Higher levels of creatine, FA(18:2(OH)) and SM(d18:2/24:1) were associated with increased lacune count, higher white matter hyperintensity volume and impaired cognition. Lower baseline levels of carnitines and creatinine were associated with higher annualized change in peak width of skeletonized mean diffusivity, and 25 metabolites, including lipoprotein subclasses, amino acids and xenobiotics, were associated with future dementia incidence. Our results show multiple distinct metabolic signatures that are associated with imaging markers of small vessel disease, cognition and conversion to dementia. Further research should assess causality and the use of metabolomic screening to improve the ability to predict future disease severity and dementia risk in small vessel disease. The metabolomic profiles may also provide novel insights into disease pathogenesis and help identify novel treatment approaches.
Collapse
Affiliation(s)
- Eric L Harshfield
- Correspondence to: Dr Eric L. Harshfield Stroke Research Group Department of Clinical Neurosciences University of Cambridge R3, Box 83, Cambridge Biomedical Campus Cambridge CB2 0QQ, UK E-mail:
| | - Caroline J Sands
- National Phenome Centre, Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW7 2AZ, UK
| | - Anil M Tuladhar
- Department of Neurology, Donders Center for Medical Neuroscience, Radboud University Nijmegen Medical Center, 6500 HB Nijmegen, The Netherlands
| | | | | | | |
Collapse
|
34
|
Ferris JK, Greeley B, Vavasour IM, Kraeutner SN, Rinat S, Ramirez J, Black SE, Boyd LA. In vivo myelin imaging and tissue microstructure in white matter hyperintensities and perilesional white matter. Brain Commun 2022; 4:fcac142. [PMID: 35694147 PMCID: PMC9178967 DOI: 10.1093/braincomms/fcac142] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 03/28/2022] [Accepted: 05/26/2022] [Indexed: 11/12/2022] Open
Abstract
White matter hyperintensities negatively impact white matter structure and relate to cognitive decline in aging. Diffusion tensor imaging detects changes to white matter microstructure, both within the white matter hyperintensity and extending into surrounding (perilesional) normal-appearing white matter. However, diffusion tensor imaging markers are not specific to tissue components, complicating the interpretation of previous microstructural findings. Myelin water imaging is a novel imaging technique that provides specific markers of myelin content (myelin water fraction) and interstitial fluid (geometric mean T2). Here we combined diffusion tensor imaging and myelin water imaging to examine tissue characteristics in white matter hyperintensities and perilesional white matter in 80 individuals (47 older adults and 33 individuals with chronic stroke). To measure perilesional normal-appearing white matter, white matter hyperintensity masks were dilated in 2 mm segments up to 10 mm in distance from the white matter hyperintensity. Fractional anisotropy, mean diffusivity, myelin water fraction, and geometric mean T2 were extracted from white matter hyperintensities and perilesional white matter. We observed a spatial gradient of higher mean diffusivity and geometric mean T2, and lower fractional anisotropy, in the white matter hyperintensity and perilesional white matter. In the chronic stroke group, myelin water fraction was reduced in the white matter hyperintensity but did not show a spatial gradient in perilesional white matter. Across the entire sample, white matter metrics within the white matter hyperintensity related to whole-brain white matter hyperintensity volume; with increasing white matter hyperintensity volume there was increased mean diffusivity and geometric mean T2, and decreased myelin water fraction in the white matter hyperintensity. Normal-appearing white matter adjacent to white matter hyperintensities exhibits characteristics of a transitional stage between healthy white matter and white matter hyperintensities. This effect was observed in markers sensitive to interstitial fluid, but not in myelin water fraction, the specific marker of myelin concentration. Within the white matter hyperintensity, interstitial fluid was higher and myelin concentration was lower in individuals with more severe cerebrovascular disease. Our data suggests white matter hyperintensities have penumbra-like effects in perilesional white matter that specifically reflect increased interstitial fluid, with no changes to myelin concentration. In contrast, within the white matter hyperintensity there are varying levels of demyelination, which vary based on the severity of cerebrovascular disease. Diffusion tensor imaging and myelin imaging may be useful clinical markers to predict white matter hyperintensity formation, and to stage neuronal damage within white matter hyperintensities.
Collapse
Affiliation(s)
- Jennifer K. Ferris
- University of British Columbia Graduate Programs in Rehabilitation Sciences, , Vancouver, Canada
| | - Brian Greeley
- University of British Columbia Department of Physical Therapy, , Vancouver, Canada
| | - Irene M. Vavasour
- The University of British Columbia Department of Radiology, , Vancouver, Canada
- University of British Columbia UBC MRI Research Centre, Faculty of Medicine, , Vancouver, Canada
| | - Sarah N. Kraeutner
- University of British Columbia Department of Psychology, , Okanagan, Kelowna, Canada
| | - Shie Rinat
- University of British Columbia Graduate Programs in Rehabilitation Sciences, , Vancouver, Canada
| | - Joel Ramirez
- LC Campbell Cognitive Neurology Research Unit, Dr Sandra Black Centre for Brain Resilience and Recovery , Toronto, Canada
- Sunnybrook Research Institute, University of Toronto Hurvitz Brain Sciences Research Program, , Toronto, Canada
| | - Sandra E. Black
- LC Campbell Cognitive Neurology Research Unit, Dr Sandra Black Centre for Brain Resilience and Recovery , Toronto, Canada
- Sunnybrook Research Institute, University of Toronto Hurvitz Brain Sciences Research Program, , Toronto, Canada
| | - Lara A. Boyd
- University of British Columbia Graduate Programs in Rehabilitation Sciences, , Vancouver, Canada
- University of British Columbia Department of Physical Therapy, , Vancouver, Canada
| |
Collapse
|
35
|
Brown RB, Tozer DJ, Loubière L, Hong YT, Fryer TD, Williams GB, Graves MJ, Aigbirhio FI, O’Brien JT, Markus HS. MINocyclinE to Reduce inflammation and blood brain barrier leakage in small Vessel diseAse (MINERVA) trial study protocol. Eur Stroke J 2022; 7:323-330. [PMID: 36082255 PMCID: PMC9445404 DOI: 10.1177/23969873221100338] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 04/24/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Cerebral small vessel disease (SVD) is a common cause of stroke and cognitive impairment. Recent data has implicated neuroinflammation and increased blood-brain barrier (BBB) permeability in its pathogenesis, but whether such processes are causal and can be therapeutically modified is uncertain. In a rodent model of SVD, minocycline was associated with reduced white matter lesions, inflammation and BBB permeability. Aims: To determine whether blood-brain barrier permeability (measured using dynamic contrast-enhanced MRI) and microglial activation (measured by positron emission tomography using the radioligand 11C-PK11195) can be modified in SVD. Design: Phase II randomised double blind, placebo-controlled trial of minocycline 100 mg twice daily for 3 months in 44 participants with moderate to severe SVD defined as a clinical lacunar stroke and confluent white matter hyperintensities. Outcomes: Primary outcome measures are volume and intensity of focal increases of blood-brain barrier permeability and microglial activation determined using PET-MRI imaging. Secondary outcome measures include inflammatory biomarkers in serum, and change in conventional MRI markers and cognitive performance over 1 year follow up. Discussion: The MINERVA trial aims to test whether minocycline can influence novel pathological processes thought to be involved in SVD progression, and will provide insights into whether central nervous system inflammation in SVD can be therapeutically modulated.
Collapse
Affiliation(s)
- Robin B Brown
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Daniel J Tozer
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Laurence Loubière
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Young T Hong
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Tim D Fryer
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Guy B Williams
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Martin J Graves
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - Franklin I Aigbirhio
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - John T O’Brien
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Hugh S Markus
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| |
Collapse
|
36
|
Wang Y, Liu X, Hu Y, Yu Z, Wu T, Wang J, Liu J, Liu J. Impaired functional network properties contribute to white matter hyperintensity related cognitive decline in patients with cerebral small vessel disease. BMC Med Imaging 2022; 22:40. [PMID: 35264145 PMCID: PMC8908649 DOI: 10.1186/s12880-022-00769-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 02/28/2022] [Indexed: 12/14/2022] Open
Abstract
Background White matter hyperintensity (WMH) is one of the typical neuroimaging manifestations of cerebral small vessel disease (CSVD), and the WMH correlates closely to cognitive impairment (CI). CSVD patients with WMH own altered topological properties of brain functional network, which is a possible mechanism that leads to CI. This study aims to identify differences in the characteristics of some brain functional network among patients with different grades of WMH and estimates the correlations between these different brain functional network characteristics and cognitive assessment scores. Methods 110 CSVD patients underwent 3.0 T Magnetic resonance imaging scans and neuropsychological cognitive assessments. WMH of each participant was graded on the basis of Fazekas grade scale and was divided into two groups: (A) WMH score of 1–2 points (n = 64), (B) WMH score of 3–6 points (n = 46). Topological indexes of brain functional network were analyzed using graph-theoretical method. T-test and Mann–Whitney U test was used to compare the differences in topological properties of brain functional network between groups. Partial correlation analysis was applied to explore the relationship between different topological properties of brain functional networks and overall cognitive function. Results Patients with high WMH scores exhibited decreased clustering coefficient values, global and local network efficiency along with increased shortest path length on whole brain level as well as decreased nodal efficiency in some brain regions on nodal level (p < 0.05). Nodal efficiency in the left lingual gyrus was significantly positively correlated with patients' total Montreal Cognitive Assessment (MoCA) scores (p < 0.05). No significant difference was found between two groups on the aspect of total MoCA and Mini-mental State Examination (MMSE) scores (p > 0.05). Conclusion Therefore, we come to conclusions that patients with high WMH scores showed less optimized small-world networks compared to patients with low WMH scores. Global and local network efficiency on the whole-brain level, as well as nodal efficiency in certain brain regions on the nodal level, can be viewed as markers to reflect the course of WMH. Supplementary Information The online version contains supplementary material available at 10.1186/s12880-022-00769-7.
Collapse
Affiliation(s)
- Yifan Wang
- Department of Radiology, Eye & ENT Hospital of Shanghai Medical School, Fudan University, Shanghai, China
| | - Xiao Liu
- School of Computer and Information Technology, Beijing Jiaotong University, Beijing, China
| | - Ying Hu
- Institute of Medical Imaging Engineering, School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Zekuan Yu
- Academy for Engineering and Technology, Fudan University, Yangpu District, No. 539 Handan Road, Shanghai, 200433, China. .,Key Laboratory of Industrial Dust Prevention and Control & Occupational Health and Safety, Ministry of Education, Beijing, China. .,Anhui Province Engineering Laboratory of Occupational Health and Safety, Huainan, China. .,Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Hefei, China.
| | - Tianhao Wu
- Department of Radiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 XianXia Road, Shanghai, 200050, China
| | - Junjie Wang
- Department of Neurosurgery, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Jie Liu
- School of Computer and Information Technology, Beijing Jiaotong University, No. 3, Shangyuan Village, Haidian District, Beijing, 100089, China.
| | - Jun Liu
- Department of Radiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 XianXia Road, Shanghai, 200050, China.
| |
Collapse
|
37
|
Clancy U, Makin SD, McHutchison CA, Cvoro V, Chappell FM, Hernández MDCV, Sakka E, Doubal F, Wardlaw JM. Impact of Small Vessel Disease Progression on Long-term Cognitive and Functional Changes After Stroke. Neurology 2022; 98:e1459-e1469. [PMID: 35131905 PMCID: PMC8992602 DOI: 10.1212/wnl.0000000000200005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 01/03/2022] [Indexed: 11/30/2022] Open
Abstract
Background and Objectives The severity of white matter hyperintensities (WMH) at presentation with stroke is associated with poststroke dementia and dependency. However, WMH can decrease or increase after stroke; prediction of cognitive decline is imprecise; and there are few data assessing longitudinal interrelationships among changing WMH, cognition, and function after stroke, despite the clinical importance. Methods We recruited patients within 3 months of a minor ischemic stroke, defined as NIH Stroke Scale (NIHSS) score <8 and not expected to result in a modified Rankin Scale (mRS) score >2. Participants repeated MRI at 1 year and cognitive and mRS assessments at 1 and 3 years. We ran longitudinal mixed-effects models assessing change in Addenbrooke’s Cognitive Examination–Revised (ACE-R) and mRS scores. For mRS score, we assessed longitudinal WMH volumes (cube root; percentage intracranial volume [ICV]), adjusting for age, NIHSS score, ACE-R, stroke subtype, and time to assessment. For ACE-R score, we additionally adjusted for ICV, mRS, premorbid IQ, and vascular risk factors. We then used a multivariate model to jointly assess changing cognition/mRS score, adjusted for prognostic variables, using all available data. Results We recruited 264 patients; mean age was 66.9 (SD 11.8) years; 41.7% were female; and median mRS score was 1 (interquartile range 1–2). One year after stroke, normalized WMH volumes were associated more strongly with 1-year ACE-R score (β = −0.259, 95% CI −0.407 to −0.111 more WMH per 1-point ACE-R decrease, p = 0.001) compared to subacute WMH volumes and ACE-R score (β = 0.105, 95% CI −0.265 to 0.054, p = 0.195). Three-year mRS score was associated with 3-year ACE-R score (β = −0.272, 95% CI −0.429 to −0.115, p = 0.001). Combined change in baseline-1-year jointly assessed ACE-R/mRS scores was associated with fluctuating WMH volumes (F = 9.3, p = 0.03). Discussion After stroke, fluctuating WMH mean that 1-year, but not baseline, WMH volumes are associated strongly with contemporaneous cognitive scores. Covarying longitudinal decline in cognition and independence after stroke, central to dementia diagnosis, is associated with increasing WMH volumes.
Collapse
Affiliation(s)
- Una Clancy
- Centre for Clinical Brain Sciences, Edinburgh Imaging and the UK Dementia Research Institute, University of Edinburgh, United Kingdom
| | - Stephen Dj Makin
- Centre for Clinical Brain Sciences, Edinburgh Imaging and the UK Dementia Research Institute, University of Edinburgh, United Kingdom.,Centre For Rural Health, Institute of Applied Health Sciences, University of Aberdeen, United Kingdom
| | - Caroline A McHutchison
- Centre for Clinical Brain Sciences, Edinburgh Imaging and the UK Dementia Research Institute, University of Edinburgh, United Kingdom
| | - Vera Cvoro
- Centre for Clinical Brain Sciences, Edinburgh Imaging and the UK Dementia Research Institute, University of Edinburgh, United Kingdom
| | - Francesca M Chappell
- Centre for Clinical Brain Sciences, Edinburgh Imaging and the UK Dementia Research Institute, University of Edinburgh, United Kingdom
| | - Maria Del C Valdés Hernández
- Centre for Clinical Brain Sciences, Edinburgh Imaging and the UK Dementia Research Institute, University of Edinburgh, United Kingdom
| | - Eleni Sakka
- Centre for Clinical Brain Sciences, Edinburgh Imaging and the UK Dementia Research Institute, University of Edinburgh, United Kingdom
| | - Fergus Doubal
- Centre for Clinical Brain Sciences, Edinburgh Imaging and the UK Dementia Research Institute, University of Edinburgh, United Kingdom
| | - Joanna M Wardlaw
- Centre for Clinical Brain Sciences, Edinburgh Imaging and the UK Dementia Research Institute, University of Edinburgh, United Kingdom
| |
Collapse
|
38
|
Vemuri P, Decarli CS, Duering M. Imaging Markers of Vascular Brain Health: Quantification, Clinical Implications, and Future Directions. Stroke 2022; 53:416-426. [PMID: 35000423 PMCID: PMC8830603 DOI: 10.1161/strokeaha.120.032611] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Cerebrovascular disease (CVD) manifests through a broad spectrum of mechanisms that negatively impact brain and cognitive health. Oftentimes, CVD changes (excluding acute stroke) are insufficiently considered in aging and dementia studies which can lead to an incomplete picture of the etiologies contributing to the burden of cognitive impairment. Our goal with this focused review is 3-fold. First, we provide a research update on the current magnetic resonance imaging methods that can measure CVD lesions as well as early CVD-related brain injury specifically related to small vessel disease. Second, we discuss the clinical implications and relevance of these CVD imaging markers for cognitive decline, incident dementia, and disease progression in Alzheimer disease, and Alzheimer-related dementias. Finally, we present our perspective on the outlook and challenges that remain in the field. With the increased research interest in this area, we believe that reliable CVD imaging biomarkers for aging and dementia studies are on the horizon.
Collapse
Affiliation(s)
| | - Charles S. Decarli
- Departments of Neurology and Center for Neuroscience, University of California at Davis, Sacramento, California, USA
| | - Marco Duering
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Germany
- Medical Image Analysis Center (MIAC AG) and qbig, Department of Biomedical Engineering, University of Basel, Switzerland
| |
Collapse
|
39
|
Egle M, Hilal S, Tuladhar AM, Pirpamer L, Bell S, Hofer E, Duering M, Wason J, Morris RG, Dichgans M, Schmidt R, Tozer DJ, Barrick TR, Chen C, de Leeuw FE, Markus HS. Determining the OPTIMAL DTI analysis method for application in cerebral small vessel disease. NEUROIMAGE: CLINICAL 2022; 35:103114. [PMID: 35908307 PMCID: PMC9421487 DOI: 10.1016/j.nicl.2022.103114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/24/2022] [Accepted: 07/10/2022] [Indexed: 11/23/2022] Open
Abstract
We were not able to identify a single optimal diffusion-weighted imaging analysis strategy across all 6 cohorts. Diffusion tensor imaging measures at baseline predicted dementia conversion in cerebral small vessel disease and mild cognitive impairment. Diffusion tensor imaging measures at baseline may be sensitive to differentiate between later vascular dementia vs Alzheimer’s disease dementia. Diffusion tensor imaging measures significantly changed over time in cohorts with cerebral small vessel disease and cohorts with mild cognitive impairment. Change in diffusion tensor imaging measures were only consistently associated with dementia conversion in severe SVD. The diffusion tensor imaging measures PSMD and DSEG required the lowest minimum sample sizes for a hypothetical clinical trial in patients with sporadic cerebral small vessel disease and mild cognitive impairment.
Background DTI is sensitive to white matter (WM) microstructural damage and has been suggested as a surrogate marker for phase 2 clinical trials in cerebral small vessel disease (SVD). The study’s objective is to establish the best way to analyse the diffusion-weighted imaging data in SVD for this purpose. The ideal method would be sensitive to change and predict dementia conversion, but also straightforward to implement and ideally automated. As part of the OPTIMAL collaboration, we evaluated five different DTI analysis strategies across six different cohorts with differing SVD severity. Methods Those 5 strategies were: (1) conventional mean diffusivity WM histogram measure (MD median), (2) a principal component-derived measure based on conventional WM histogram measures (PC1), (3) peak width skeletonized mean diffusivity (PSMD), (4) diffusion tensor image segmentation θ (DSEG θ) and (5) a WM measure of global network efficiency (Geff). The association between each measure and cognitive function was tested using a linear regression model adjusted by clinical markers. Changes in the imaging measures over time were determined. In three cohort studies, repeated imaging data together with data on incident dementia were available. The association between the baseline measure, change measure and incident dementia conversion was examined using Cox proportional-hazard regression or logistic regression models. Sample size estimates for a hypothetical clinical trial were furthermore computed for each DTI analysis strategy. Results There was a consistent cross-sectional association between the imaging measures and impaired cognitive function across all cohorts. All baseline measures predicted dementia conversion in severe SVD. In mild SVD, PC1, PSMD and Geff predicted dementia conversion. In MCI, all markers except Geff predicted dementia conversion. Baseline DTI was significantly different in patients converting to vascular dementia than to Alzheimer’ s disease. Significant change in all measures was associated with dementia conversion in severe but not in mild SVD. The automatic and semi-automatic measures PSMD and DSEG θ required the lowest minimum sample sizes for a hypothetical clinical trial in single-centre sporadic SVD cohorts. Conclusion DTI parameters obtained from all analysis methods predicted dementia, and there was no clear winner amongst the different analysis strategies. The fully automated analysis provided by PSMD offers advantages particularly for large datasets.
Collapse
Affiliation(s)
- Marco Egle
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom.
| | - Saima Hilal
- Department of Pharmacology, National University of Singapore, Singapore; Memory Ageing and Cognition Center, National University Health System, Singapore
| | - Anil M Tuladhar
- Department of Neurology, Donders Center for Medical Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Lukas Pirpamer
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Steven Bell
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Edith Hofer
- Department of Neurology, Medical University of Graz, Graz, Austria; Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, Graz, Austria
| | - Marco Duering
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany; Medical Image Analysis Center (MIAC) and Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - James Wason
- Population Health Sciences Institute, Newcastle University, Baddiley-Clark Building, Newcastle Upon Tyne, United Kingdom
| | - Robin G Morris
- Department of Psychology (R.G.M.), King's College, Institute of Psychiatry, Psychology and Neuroscience, London, United Kingdom
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Reinhold Schmidt
- Department of Neurology, Medical University of Graz, Graz, Austria
| | - Daniel J Tozer
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Thomas R Barrick
- Neurosciences Research Centre, Institute for Molecular and Clinical Sciences, St George's, University of London, United Kingdom
| | - Christopher Chen
- Department of Pharmacology, National University of Singapore, Singapore; Memory Ageing and Cognition Center, National University Health System, Singapore
| | - Frank-Erik de Leeuw
- Department of Neurology, Donders Center for Medical Neuroscience, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Hugh S Markus
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
40
|
Egle M, Hilal S, Tuladhar AM, Pirpamer L, Hofer E, Duering M, Wason J, Morris RG, Dichgans M, Schmidt R, Tozer D, Chen C, de Leeuw FE, Markus HS. Prediction of dementia using diffusion tensor MRI measures: the OPTIMAL collaboration. J Neurol Neurosurg Psychiatry 2022; 93:14-23. [PMID: 34509999 DOI: 10.1136/jnnp-2021-326571] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 08/21/2021] [Indexed: 11/03/2022]
Abstract
OBJECTIVES It has been suggested that diffusion tensor imaging (DTI) measures sensitive to white matter (WM) damage may predict future dementia risk not only in cerebral small vessel disease (SVD), but also in mild cognitive impairment. To determine whether DTI measures were associated with cognition cross-sectionally and predicted future dementia risk across the full range of SVD severity, we established the International OPtimising mulTImodal MRI markers for use as surrogate markers in trials of Vascular Cognitive Impairment due to cerebrAl small vesseL disease collaboration which included six cohorts. METHODS Among the six cohorts, prospective data with dementia incidences were available for three cohorts. The associations between six different DTI measures and cognition or dementia conversion were tested. The additional contribution to prediction of other MRI markers of SVD was also determined. RESULTS The DTI measure mean diffusivity (MD) median correlated with cognition in all cohorts, demonstrating the contribution of WM damage to cognition. Adding MD median significantly improved the model fit compared to the clinical risk model alone and further increased in all single-centre SVD cohorts when adding conventional MRI measures. Baseline MD median predicted dementia conversion. In a study with severe SVD (SCANS) change in MD median also predicted dementia conversion. The area under the curve was best when employing a multimodal MRI model using both DTI measures and other MRI measures. CONCLUSIONS Our results support a central role for WM alterations in dementia pathogenesis in all cohorts. DTI measures such as MD median may be a useful clinical risk predictor. The contribution of other MRI markers varied according to disease severity.
Collapse
Affiliation(s)
- Marco Egle
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Saima Hilal
- Memory Aging and Cognition Centre, Department of Pharmacology, Yong Loo Lim School of Medicine, National University of Singapore, Singapore.,Saw Swee Hock School of Public Health, National University of Singapore and National University Health System of Singapore, Singapore
| | - A M Tuladhar
- Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Department of Neurology, Nijmegen, The Netherlands
| | - Lukas Pirpamer
- Department of Neurology, Medical University Graz, Graz, Austria
| | - Edith Hofer
- Department of Neurology, Medical University Graz, Graz, Austria.,Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, Graz, Austria
| | - Marco Duering
- Institute for Stroke and Dementia Research, University Hospital, Ludwig Maximilian University Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - James Wason
- MRC Biostatistics Unit, School of Clinical Medicine, University of Cambridge, Cambridge Institute of Public Health, Cambridge, UK.,Population Health Sciences Institute, Newcastle University, Baddiley-Clark Building, Newcastle upon Tyne, UK
| | - Robin G Morris
- Department of Psychology (R.G.M), King's College London, Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, University Hospital, Ludwig Maximilian University Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | | | - Daniel Tozer
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Christopher Chen
- Memory Aging and Cognition Centre, Department of Pharmacology, Yong Loo Lim School of Medicine, National University of Singapore, Singapore
| | - Frank-Erik de Leeuw
- Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Department of Neurology, Nijmegen, The Netherlands
| | - Hugh S Markus
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| |
Collapse
|
41
|
Alme KN, Ulvik A, Askim T, Assmus J, Mollnes TE, Naik M, Næss H, Saltvedt I, Ueland PM, Knapskog AB. Neopterin and kynurenic acid as predictors of stroke recurrence and mortality: a multicentre prospective cohort study on biomarkers of inflammation measured three months after ischemic stroke. BMC Neurol 2021; 21:476. [PMID: 34879833 PMCID: PMC8653541 DOI: 10.1186/s12883-021-02498-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/15/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Chronic low-grade inflammation is associated with both ischemic stroke and sedentary behaviour. The aim of this study was to investigate the predictive abilities of biomarkers of inflammation and immune modulation associated with sedentary behaviour for ischemic stroke recurrence and mortality in a stroke population. METHODS Patients admitted to hospital for acute stroke were recruited to the prospective multicentre cohort study, the Norwegian Cognitive Impairment After Stroke (Nor-COAST) study, from May 2015 until March 2017. Patients with ischemic stroke, blood samples available from the three-month follow-up, and no stroke recurrence before the three-month follow-up were included. Serum was analysed for C-reactive protein (CRP) with high-sensitive technique, and plasma for interleukin-6 (IL-6), neopterin, pyridoxic acid ratio index (PAr-index: 4-pyridoxic acid: [pyrioxal+pyridoxal-5'-phosphate]) and kynurenic acid (KA). Ischemic stroke recurrence and death were identified by the Norwegian Stroke Registry and the Cause of Death Registry until 31 December 2018. RESULTS The study included 354 patients, 57% male, mean age 73 (SD 11) years, mean observation time 2.5 (SD 0.6) years, and median National Institute of Health Stroke Scale of 0 (IQR 1) at three months. CRP was associated with mortality (HR 1.40, CI 1.01, 1.96, p = 0.046), and neopterin was associated with the combined endpoint (recurrent ischemic stroke or death) (HR 1.52, CI 1.06, 2.20, p = 0.023), adjusted for age, sex, prior cerebrovascular disease, modified Rankin Scale, and creatinine. When adding neopterin and KA to the same model, KA was negatively associated (HR 0.57, CI 0.33, 0.97, p = 0.038), and neopterin was positively associated (HR 1.61, CI 1.02, 2.54, p = 0.040) with mortality. Patients with cardioembolic stroke at baseline had higher levels of inflammation at three months. CONCLUSION Neopterin might be a valuable prognostic biomarker in stroke patients. The use of KA as a measure of anti-inflammatory capacity should be investigated further. TRIAL REGISTRATION The study was registered at Clinicaltrials.gov ( NCT02650531 ). First posted on 08/01/2016.
Collapse
Affiliation(s)
- Katinka Nordheim Alme
- Institute of Clinical Medicine (K1), University of Bergen, Bergen, Norway. .,Department of Internal Medicine, Haraldsplass Deaconess Hospital, Bergen, Norway.
| | | | - Torunn Askim
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Science, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Jörg Assmus
- Centre for Clinical Research, Haukeland University Hospital, Bergen, Norway
| | - Tom Eirik Mollnes
- Department of Immunology, Oslo University Hospital and University of Oslo, Oslo, Norway.,Research Laboratory, Nordland Hospital, Bodø, and K.G. Jebsen TREC, University of Tromsø, Tromsø, Norway.,Centre of Molecular Inflammation Research, NTNU-Norwegian University of Science and Technology, Trondheim, Norway
| | - Mala Naik
- Department of Internal Medicine, Haraldsplass Deaconess Hospital, Bergen, Norway.,Department of Clinical Science (K2), University of Bergen, Bergen, Norway
| | - Halvor Næss
- Institute of Clinical Medicine (K1), University of Bergen, Bergen, Norway.,Department of Neurology, Haukeland University Hospital, Bergen, Norway.,Centre for age-related medicine, Stavanger University Hospital, Stavanger, Norway
| | - Ingvild Saltvedt
- Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Science, NTNU-Norwegian University of Science and Technology, Trondheim, Norway.,Department of Geriatrics, Clinic of internal medicine, St Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | | | - Anne-Brita Knapskog
- Department of Geriatric Medicine, Oslo University Hospital. Ullevaal, Oslo, Norway
| |
Collapse
|
42
|
Walsh J, Tozer DJ, Sari H, Hong YT, Drazyk A, Williams G, Shah NJ, O’Brien JT, Aigbirhio FI, Rosenberg G, Fryer TD, Markus HS. Microglial activation and blood-brain barrier permeability in cerebral small vessel disease. Brain 2021; 144:1361-1371. [PMID: 34000009 PMCID: PMC8874873 DOI: 10.1093/brain/awab003] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/12/2020] [Accepted: 10/27/2020] [Indexed: 11/28/2022] Open
Abstract
Cerebral small vessel disease (SVD) is a major cause of stroke and dementia. The underlying pathogenesis is poorly understood, but both neuroinflammation and increased blood-brain barrier permeability have been hypothesized to play a role, and preclinical studies suggest the two processes may be linked. We used PET magnetic resonance to simultaneously measure microglial activation using the translocator protein radioligand 11C-PK11195, and blood-brain barrier permeability using dynamic contrast enhanced MRI. A case control design was used with two disease groups with sporadic SVD (n = 20), monogenic SVD (cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy, CADASIL), and normal controls (n = 20) were studied. Hotspots of increased glial activation and blood-brain barrier permeability were identified as values greater than the 95th percentile of the distribution in controls. In sporadic SVD there was an increase in the volume of hotspots of both 11C-PK11195 binding (P = 0.003) and blood-brain barrier permeability (P = 0.007) in the normal appearing white matter, in addition to increased mean blood-brain barrier permeability (P < 0.001). In CADASIL no increase in blood-brain barrier permeability was detected; there was a non-significant trend to increased 11C-PK11195 binding (P = 0.073). Hotspots of 11C-PK11195 binding and blood-brain barrier permeability were not spatially related. A panel of 93 blood biomarkers relating to cardiovascular disease, inflammation and endothelial activation were measured in each participant; principal component analysis was performed and the first component related to blood-brain barrier permeability and microglial activation. Within the sporadic SVD group both hotspot and mean volume blood-brain barrier permeability values in the normal appearing white matter were associated with dimension 1 (β = 0.829, P = 0.017, and β = 0.976, P = 0.003, respectively). There was no association with 11C-PK11195 binding. No associations with blood markers were found in the CADASIL group. In conclusion, in sporadic SVD both microglial activation and increased blood-brain barrier permeability occur, but these are spatially distinct processes. No evidence of increased blood-brain barrier permeability was found in CADASIL.
Collapse
Affiliation(s)
- Jessica Walsh
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Dan J Tozer
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Hasan Sari
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Young T Hong
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Anna Drazyk
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Guy Williams
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - N Jon Shah
- Institute of Neuroscience and Medicine, Forschungszentrum Jülich, Jülich, Germany
- JARA–BRAIN–Translational Medicine, Aachen, and Department of Neurology, RWTH Aachen University, Aachen, Germany
| | - John T O’Brien
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | | | - Gary Rosenberg
- UNM Health Sciences Center, University of New Mexico, Albuquerque, NM 87106, USA
| | - Tim D Fryer
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Hugh S Markus
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| |
Collapse
|
43
|
Lu T, Wang Z, Cui Y, Zhou J, Wang Y, Ju S. Disrupted Structural Brain Connectome Is Related to Cognitive Impairment in Patients With Ischemic Leukoaraiosis. Front Hum Neurosci 2021; 15:654750. [PMID: 34177491 PMCID: PMC8223255 DOI: 10.3389/fnhum.2021.654750] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 04/30/2021] [Indexed: 11/13/2022] Open
Abstract
Ischemic leukoaraiosis (ILA) is related to cognitive impairment and vascular dementia in the elderly. One possible mechanism could be the disruption of white matter (WM) tracts and network function that connect distributed brain regions involved in cognition. The purpose of this study was to investigate the relationship between structural connectome and cognitive functions in ILA patients. A total of 89 patients with ILA (Fazekas score ≥ 3) and 90 healthy controls (HCs) underwent comprehensive neuropsychological examinations and diffusion tensor imaging scans. The tract-based spatial statistics approach was employed to investigate the WM integrity. Graph theoretical analysis was further applied to construct the topological architecture of the structural connectome in ILA patients. Partial correlation analysis was used to investigate the relationships between network measures and cognitive performances in the ILA group. Compared with HCs, the ILA patients showed widespread WM integrity disruptions. The ILA group displayed increased characteristic path length (L p) and decreased global network efficiency at the level of the whole brain relative to HCs, and reduced nodal efficiencies, predominantly in the frontal-subcortical and limbic system regions. Furthermore, these structural connectomic alterations were associated with cognitive impairment in ILA patients. The association between WM changes (i.e., fractional anisotropy and mean diffusivity measures) and cognitive function was mediated by the structural connectivity measures (i.e., local network efficiency and L p). In conclusion, cognitive impairment in ILA patients is related to microstructural disruption of multiple WM fibers and topological disorganization of structural networks, which have implications in understanding the relationship between ILA and the possible attendant cognitive impairment.
Collapse
Affiliation(s)
- Tong Lu
- Nanjing Medical University, Nanjing, China.,Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Zan Wang
- Department of Neurology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Ying Cui
- Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Jiaying Zhou
- Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Yuancheng Wang
- Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Shenghong Ju
- Nanjing Medical University, Nanjing, China.,Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
44
|
Bir SC, Khan MW, Javalkar V, Toledo EG, Kelley RE. Emerging Concepts in Vascular Dementia: A Review. J Stroke Cerebrovasc Dis 2021; 30:105864. [PMID: 34062312 DOI: 10.1016/j.jstrokecerebrovasdis.2021.105864] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/29/2021] [Accepted: 04/28/2021] [Indexed: 01/18/2023] Open
Abstract
OBJECTIVE Vascular dementia (VaD) is the second most common cause of dementia and a major health concern worldwide. A comprehensive review on VaD is warranted for better understanding and guidance for the practitioner. We provide an updated overview of the epidemiology, pathophysiological mechanisms, neuroimaging patterns as well as current diagnostic and therapeutic approaches. MATERIALS AND METHODS A narrative review of current literature in VaD was performed based on publications from the database of PubMed, Scopus and Google Scholar up to January, 2021. RESULTS VaD can be the result of ischemic or hemorrhagic tissue injury in a particular region of the brain which translates into clinically significant cognitive impairment. For example, a cerebral infarct in the speech area of the dominant hemisphere would translate into clinically significant impairment as would involvement of projection pathways such as the arcuate fasciculus. Specific involvement of the angular gyrus of the dominant hemisphere, with resultant Gerstman's syndrome, could have a pronounced effect on functional ability despite being termed a "minor stroke". Small vessel cerebrovascular disease can have a cumulate effect on cognitive function over time. It is unfortunately well recognized that "good" functional recovery in acute ischemic or haemorrhagic stroke, including subarachnoid haemorrhage, does not necessarily translate into good cognitive recovery. The victim may often be left unable to have gainful employment, drive a car safely or handle their affairs independently. CONCLUSIONS This review should serve as a compendium of updated information on VaD and provide guidance in terms of newer diagnostic and potential therapeutic approaches.
Collapse
Affiliation(s)
- Shyamal C Bir
- Department of Neurology Ocshner/LSU Health Sciences Center-Sheveport, Shreveport, LA, USA
| | - Muhammad W Khan
- Department of Neurology Ocshner/LSU Health Sciences Center-Sheveport, Shreveport, LA, USA
| | - Vijayakumar Javalkar
- Department of Neurology Ocshner/LSU Health Sciences Center-Sheveport, Shreveport, LA, USA
| | | | - Roger E Kelley
- Department of Neurology Ocshner/LSU Health Sciences Center-Sheveport, Shreveport, LA, USA.
| |
Collapse
|
45
|
Markus HS, Egle M, Croall ID, Sari H, Khan U, Hassan A, Harkness K, MacKinnon A, O'Brien JT, Morris RG, Barrick TR, Blamire AM, Tozer DJ, Ford GA. PRESERVE: Randomized Trial of Intensive Versus Standard Blood Pressure Control in Small Vessel Disease. Stroke 2021; 52:2484-2493. [PMID: 34044580 DOI: 10.1161/strokeaha.120.032054] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Hugh S Markus
- Stroke Research Group, Department of Clinical Neuroscience, University of Cambridge (H.S.M., M.E., I.D.C., H.S., D.J.T.)
| | - Marco Egle
- Stroke Research Group, Department of Clinical Neuroscience, University of Cambridge (H.S.M., M.E., I.D.C., H.S., D.J.T.)
| | - Iain D Croall
- Stroke Research Group, Department of Clinical Neuroscience, University of Cambridge (H.S.M., M.E., I.D.C., H.S., D.J.T.)
| | - Hasan Sari
- Stroke Research Group, Department of Clinical Neuroscience, University of Cambridge (H.S.M., M.E., I.D.C., H.S., D.J.T.)
| | - Usman Khan
- Atkinson Morley Neuroscience Centre, St. Georges NHS Healthcare Trust (U.K., A.M.)
| | | | | | - Andrew MacKinnon
- Atkinson Morley Neuroscience Centre, St. Georges NHS Healthcare Trust (U.K., A.M.)
| | - John T O'Brien
- Department of Psychiatry, University of Cambridge (J.T.O.)
| | - Robin G Morris
- Kings College Institute of Psychiatry, Psychology and Neurosciences, London, United Kingdom (R.G.M.)
| | - Thomas R Barrick
- Neurosciences Research Centre, Molecular and Clinical Science Research Institute, St George's University of London, United Kingdom (T.R.B.)
| | - Andrew M Blamire
- Magnetic Resonance Centre, Institute of Cellular Medicine, Newcastle University, United Kingdom (A.M.B.)
| | - Daniel J Tozer
- Stroke Research Group, Department of Clinical Neuroscience, University of Cambridge (H.S.M., M.E., I.D.C., H.S., D.J.T.)
| | - Gary A Ford
- Oxford University Hospitals NHS Foundation Trust, University of Oxford (G.A.F.)
| | | |
Collapse
|
46
|
Kapoor A, Bartha R, Black SE, Borrie M, Freedman M, Gao F, Herrmann N, Mandzia J, Ozzoude M, Ramirez J, Scott CJM, Symons S, Fischer CE, Frank A, Seitz D, Wolf MU, Verhoeff NPLG, Naglie G, Reichman W, Masellis M, Mitchell SB, Tang-Wai DF, Tartaglia MC, Kumar S, Pollock BG, Rajji TK, Finger E, Pasternak SH, Swartz RH. Structural Brain Magnetic Resonance Imaging to Rule Out Comorbid Pathology in the Assessment of Alzheimer's Disease Dementia: Findings from the Ontario Neurodegenerative Disease Research Initiative (ONDRI) Study and Clinical Trials Over the Past 10 Years. J Alzheimers Dis 2021; 74:747-757. [PMID: 32116253 PMCID: PMC7242844 DOI: 10.3233/jad-191097] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND/OBJECTIVE Structural brain magnetic resonance imaging (MRI) is not mandatory in Alzheimer's disease (AD) research or clinical guidelines. We aimed to explore the use of structural brain MRI in AD/mild cognitive impairment (MCI) trials over the past 10 years and determine the frequency with which inclusion of standardized structural MRI acquisitions detects comorbid vascular and non-vascular pathologies. METHODS We systematically searched ClinicalTrials.gov for AD clinical trials to determine their neuroimaging criteria and then used data from an AD/MCI cohort who underwent standardized MRI protocols, to determine type and incidence of clinically relevant comorbid pathologies. RESULTS Of 210 AD clinical trials, 105 (50%) included structural brain imaging in their eligibility criteria. Only 58 (27.6%) required MRI. 16,479 of 53,755 (30.7%) AD participants were in trials requiring MRI. In the observational AD/MCI cohort, 141 patients met clinical criteria; 22 (15.6%) had relevant MRI findings, of which 15 (10.6%) were exclusionary for the study. DISCUSSION In AD clinical trials over the last 10 years, over two-thirds of participants could have been enrolled without brain MRI and half without even a brain CT. In a study sample, relevant comorbid pathology was found in 15% of participants, despite careful screening. Standardized structural MRI should be incorporated into NIA-AA diagnostic guidelines (when available) and research frameworks routinely to reduce diagnostic heterogeneity.
Collapse
Affiliation(s)
| | - Robert Bartha
- Robarts Research Institute and the Department of Medical Biophysics, the University of Western Ontario, London, ON, Canada
| | - Sandra E Black
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada.,University of Toronto, Toronto, ON, Canada
| | - Michael Borrie
- Parkwood Institute, St. Joseph's Health Care Center, London, ON, Canada
| | - Morris Freedman
- University of Toronto, Toronto, ON, Canada.,Rotman Research Institute of Baycrest Health Sciences, Toronto, ON, Canada.,Baycrest Health Sciences, Toronto, ON, Canada
| | - Fuqiang Gao
- Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Nathan Herrmann
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada.,University of Toronto, Toronto, ON, Canada
| | - Jennifer Mandzia
- Western University, London, ON, Canada.,London Health Sciences Centre, London, ON, Canada
| | - Miracle Ozzoude
- Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Joel Ramirez
- Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | | | - Sean Symons
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Corinne E Fischer
- Keenan Research Centre for Biomedical Research, the Li Ka Shing Knowledge Institute, St. Michaels Hospital, Toronto, ON, Canada
| | | | - Dallas Seitz
- Department of Psychiatry and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Michael Uri Wolf
- University of Toronto, Toronto, ON, Canada.,Baycrest Health Sciences, Toronto, ON, Canada
| | | | - Gary Naglie
- University of Toronto, Toronto, ON, Canada.,Rotman Research Institute of Baycrest Health Sciences, Toronto, ON, Canada.,Baycrest Health Sciences, Toronto, ON, Canada
| | - William Reichman
- University of Toronto, Toronto, ON, Canada.,Baycrest Health Sciences, Toronto, ON, Canada
| | - Mario Masellis
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada.,University of Toronto, Toronto, ON, Canada
| | - Sara B Mitchell
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,University of Toronto, Toronto, ON, Canada
| | - David F Tang-Wai
- University of Toronto, Toronto, ON, Canada.,University Health Network Memory Clinic, University of Toronto, Division of Neurology & Geriatric Medicine, Toronto, ON, Canada
| | - Maria Carmela Tartaglia
- University of Toronto, Toronto, ON, Canada.,Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Sanjeev Kumar
- University of Toronto, Toronto, ON, Canada.,Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Bruce G Pollock
- University of Toronto, Toronto, ON, Canada.,Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Tarek K Rajji
- University of Toronto, Toronto, ON, Canada.,Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Elizabeth Finger
- Parkwood Institute, St. Joseph's Health Care Center, London, ON, Canada.,Western University, London, ON, Canada
| | - Stephen H Pasternak
- Robarts Research Institute and the Department of Medical Biophysics, the University of Western Ontario, London, ON, Canada.,Parkwood Institute, St. Joseph's Health Care Center, London, ON, Canada.,Western University, London, ON, Canada
| | | | - Richard H Swartz
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada.,Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada.,University of Toronto, Toronto, ON, Canada
| |
Collapse
|
47
|
Hamilton OKL, Backhouse EV, Janssen E, Jochems ACC, Maher C, Ritakari TE, Stevenson AJ, Xia L, Deary IJ, Wardlaw JM. Cognitive impairment in sporadic cerebral small vessel disease: A systematic review and meta-analysis. Alzheimers Dement 2021; 17:665-685. [PMID: 33185327 PMCID: PMC8593445 DOI: 10.1002/alz.12221] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 02/08/2020] [Accepted: 05/10/2020] [Indexed: 01/09/2023]
Abstract
This paper is a proposal for an update on the characterization of cognitive impairments associated with sporadic cerebral small vessel disease (SVD). We pose a series of questions about the nature of SVD-related cognitive impairments and provide answers based on a comprehensive review and meta-analysis of published data from 69 studies. Although SVD is thought primarily to affect executive function and processing speed, we hypothesize that SVD affects all major domains of cognitive ability. We also identify low levels of education as a potentially modifiable risk factor for SVD-related cognitive impairment. Therefore, we propose the use of comprehensive cognitive assessments and the measurement of educational level both in clinics and research settings, and suggest several recommendations for future research.
Collapse
Affiliation(s)
- Olivia KL Hamilton
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, UK, EH16 4SB
- Dementia Research Institute, University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, UK, EH16 4SB
- Lothian Birth Cohorts, University of Edinburgh, 7 George Square, Edinburgh, UK, EH8 9JZ
| | - Ellen V Backhouse
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, UK, EH16 4SB
- Dementia Research Institute, University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, UK, EH16 4SB
| | - Esther Janssen
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, UK, EH16 4SB
- Dementia Research Institute, University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, UK, EH16 4SB
| | - Angela CC Jochems
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, UK, EH16 4SB
- Dementia Research Institute, University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, UK, EH16 4SB
| | - Caragh Maher
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, UK, EH16 4SB
- Dementia Research Institute, University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, UK, EH16 4SB
| | - Tuula E Ritakari
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, UK, EH16 4SB
- Dementia Research Institute, University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, UK, EH16 4SB
| | - Anna J Stevenson
- Dementia Research Institute, University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, UK, EH16 4SB
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital Campus, Crewe Road, Edinburgh, UK, EH4 2XU
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, 15 George Square, Edinburgh, UK, EH8 9XD
| | - Lihua Xia
- Department of Psychology, University of Edinburgh, 7 George Square, Edinburgh, UK, EH8 9JZ
| | - Ian J Deary
- Lothian Birth Cohorts, University of Edinburgh, 7 George Square, Edinburgh, UK, EH8 9JZ
- Department of Psychology, University of Edinburgh, 7 George Square, Edinburgh, UK, EH8 9JZ
| | - Joanna M Wardlaw
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, UK, EH16 4SB
- Dementia Research Institute, University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, UK, EH16 4SB
- Lothian Birth Cohorts, University of Edinburgh, 7 George Square, Edinburgh, UK, EH8 9JZ
| |
Collapse
|
48
|
Nikolopoulos D, Fanouriakis A, Bertsias G. Treatment of neuropsychiatric systemic lupus erythematosus: clinical challenges and future perspectives. Expert Rev Clin Immunol 2021; 17:317-330. [PMID: 33682602 DOI: 10.1080/1744666x.2021.1899810] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Introduction: Neuropsychiatric (NP) involvement represents an emerging frontier in systemic lupus erythematosus (SLE), posing significant challenges due to its clinical diversity and obscure pathophysiology. The authors herein discuss selected aspects in the management of NPSLE based on existing literature and our experience, aiming to facilitate routine medical care.Areas covered: Research related to diagnosis, neuroimaging, treatment and outcome is discussed, focusing on data published in PubMed during the last 5 years. Selected translational studies of clinical relevance are included.Expert opinion: Identification of NPSLE patients who may benefit from appropriate treatment can be facilitated by attribution algorithms. Immunosuppressants are typically indicated in recurrent seizures, optic neuritis, myelopathy, psychosis and peripheral nerve disease, although a low threshold is recommended for cerebrovascular disease and other NP manifestations, especially when SLE is active. With the exception of stroke with positive antiphospholipid antibodies, anti-coagulation is rarely indicated in other syndromes. Refractory NPSLE can be treated with rituximab, whereas the role of other biologics remains unknown. Advances in the fields of biomarkers, neuroimaging for brain structural, perfusion or functional abnormalities, and design of novel compounds targeting not only systemic autoimmunity but also inflammatory and regenerative pathways within the nervous system, hold promise for optimizing NPSLE management.
Collapse
Affiliation(s)
- Dionysis Nikolopoulos
- 4th Department of Internal Medicine, Joint Rheumatology Program, National and Kapodistrian University of Athens, Athens, Greece.,Laboratory of Immune Regulation and Tolerance, Autoimmunity and Inflammation, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | | | - George Bertsias
- Department of Rheumatology, Clinical Immunology, University of Crete Medical School and University Hospital of Heraklion, Heraklion, Greece.,Laboratory of Rheumatology, Autoimmunity and Inflammation, Infections & Immunity Division, Institute of Molecular Biology and Biotechnology (FORTH), Heraklion, Greece
| |
Collapse
|
49
|
Egle M, Loubiere L, Maceski A, Kuhle J, Peters N, Markus HS. Neurofilament light chain predicts future dementia risk in cerebral small vessel disease. J Neurol Neurosurg Psychiatry 2021; 92:jnnp-2020-325681. [PMID: 33558370 PMCID: PMC8142459 DOI: 10.1136/jnnp-2020-325681] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVES Serum neurofilament light chain (NfL) has been proposed as prognostic markers in neurogenerative disease. A cross-sectional study in cerebral small vessel disease (SVD) reported an association with cognition and disability. If NfL is to be used to predict outcome, studies are required to demonstrate baseline NfL predicts future dementia risk. Furthermore, if it is to be used as a surrogate marker in clinical trials, change in NfL over time periods typical of a clinical trial must be linked to clinical progression. In a longitudinal study of patients with lacunar stroke and confluent white matter hyperintensities, we determined whether both baseline, and change, in NfL levels were linked to changes in MRI markers, cognitive decline and dementia risk. METHODS Patients underwent MRI, cognitive testing and blood taking at baseline and annually for 3 years. Clinical and cognitive follow-up continued for 5 years. RESULTS NfL data were available for 113 subjects for baseline analysis, and 90 patients for the longitudinal analysis. Baseline NfL predicted cognitive decline (global cognition β=-0.335, SE=0.094, p=0.001) and risk of converting to dementia (HR=1.676 (95% CI 1.183 to 2.373), p=0.004). In contrast to imaging, there was no change in NfL values over the follow-up period. CONCLUSIONS Baseline NfL predicts changes in MRI markers, cognitive decline and dementia rate over a 5 years follow-up period in SVD, suggesting NfL may be a useful prognostic marker. However, change in NfL values was not detected, and therefore NfL may not be a useful surrogate marker in clinical trials in SVD.
Collapse
Affiliation(s)
- Marco Egle
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Laurence Loubiere
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Aleksandra Maceski
- Neurologic Clinic and Policlinic, Departments of Medicine, Biomedicine and Clinical Research, University Hospital and University of Basel, Basel, Switzerland
| | - Jens Kuhle
- Neurologic Clinic and Policlinic, Departments of Medicine, Biomedicine and Clinical Research, University Hospital and University of Basel, Basel, Switzerland
| | - Nils Peters
- Stroke Center, Klinik Hirslanden, Zürich, Switzerland
- Stroke Center and Department of Neurology, University Hospital and University of Basel, Basel, Switzerland
| | - Hugh S Markus
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| |
Collapse
|
50
|
Xing Y, Yang J, Zhou A, Wang F, Wei C, Tang Y, Jia J. White Matter Fractional Anisotropy Is a Superior Predictor for Cognitive Impairment Than Brain Volumes in Older Adults With Confluent White Matter Hyperintensities. Front Psychiatry 2021; 12:633811. [PMID: 34025467 PMCID: PMC8131652 DOI: 10.3389/fpsyt.2021.633811] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 04/08/2021] [Indexed: 11/13/2022] Open
Abstract
Older patients with confluent white matter hyperintensities (WMHs) on magnetic resonance imaging have an increased risk for the onset of vascular cognitive impairment (VCI). This study investigates the predictive effects of the white matter (WM) fractional anisotropy (FA) and brain volumes on cognitive impairment for those with confluent WMHs. This study enrolled 77 participants with confluent WMHs (Fazekas grade 2 or 3), including 44 with VCI-no dementia (VCIND) and 33 with normal cognition (NC). The mean FA of 20 WM tracts was calculated to evaluate the global WM microstructural integrity, and major WM tracts were reconstructed using probabilistic tractography. Voxel-based morphometry was used to calculate brain volumes for the total gray matter (GM), the hippocampus, and the nucleus basalis of Meynert (NbM). All volumetric assays were corrected for total intracranial volume. All regression analyses were adjusted for age, gender, education, and apolipoprotein E (ApoE) gene ε4 status. Logistic regression analysis revealed that the mean FA value for global WM was the only independent risk factor for VCI (z score of FA: OR = 4.649, 95%CI 1.576-13.712, p = 0.005). The tract-specific FAs were not associated with the risk of cognitive impairment after controlling the mean FA for global WM. The mean FA value was significantly associated with scores of Mini-Mental State Examination (MMSE) and Auditory Verbal Learning Test. A lower FA was also associated with smaller volumes of total GM, hippocampus, and NbM. However, brain volumes were not found to be directly related to cognitive performances, except for an association between the hippocampal volume and MMSE. In conclusion, the mean FA for global WM microstructural integrity is a superior predictor for cognitive impairment than tract-specific FA and brain volumes in people with confluent WMHs.
Collapse
Affiliation(s)
- Yi Xing
- Department of Neurology, Innovation Center for Neurological Disorders, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Neurodegenerative Diseases, Ministry of Education of the People's Republic of China, Beijing, China
| | - Jianwei Yang
- Department of Neurology, Innovation Center for Neurological Disorders, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Neurodegenerative Diseases, Ministry of Education of the People's Republic of China, Beijing, China
| | - Aihong Zhou
- Department of Neurology, Innovation Center for Neurological Disorders, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Neurodegenerative Diseases, Ministry of Education of the People's Republic of China, Beijing, China
| | - Fen Wang
- Department of Neurology, Innovation Center for Neurological Disorders, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Neurodegenerative Diseases, Ministry of Education of the People's Republic of China, Beijing, China
| | - Cuibai Wei
- Department of Neurology, Innovation Center for Neurological Disorders, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Neurodegenerative Diseases, Ministry of Education of the People's Republic of China, Beijing, China
| | - Yi Tang
- Department of Neurology, Innovation Center for Neurological Disorders, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Neurodegenerative Diseases, Ministry of Education of the People's Republic of China, Beijing, China
| | - Jianping Jia
- Department of Neurology, Innovation Center for Neurological Disorders, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.,Key Laboratory of Neurodegenerative Diseases, Ministry of Education of the People's Republic of China, Beijing, China
| |
Collapse
|