1
|
Yang S, Webb AJS. Reduced neurovascular coupling is associated with increased cardiovascular risk without established cerebrovascular disease: A cross-sectional analysis in UK biobank. J Cereb Blood Flow Metab 2024:271678X241302172. [PMID: 39576882 PMCID: PMC11585009 DOI: 10.1177/0271678x241302172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 10/31/2024] [Accepted: 11/02/2024] [Indexed: 11/24/2024]
Abstract
Mid-life vascular risk factors predict late-life cerebrovascular diseases and poor global brain health. Although endothelial dysfunction is hypothesized to contribute to this process, evidence of impaired neurovascular function in early stages remains limited. In this cross-sectional study of 31,934 middle-aged individuals from UK Biobank without established cerebrovascular disease, the overall 10-year risk of cardiovascular events was associated with reduced neurovascular coupling (p < 2 × 10-16) during a visual task with functional MRI, including in participants with no clinically apparent brain injury on MRI. Diabetes, smoking, waist-hip ratio, and hypertension were each strongly associated with decreased neurovascular coupling with the strongest relationships for diabetes and smoking, whilst in older adults there was an inverted U-shaped relationship with DBP, peaking at 70-80 mmHg DBP. These findings indicate that mid-life vascular risk factors are associated with impaired cerebral endothelial-dependent neurovascular function in the absence of overt brain injury. Neurovascular dysfunction, measured by neurovascular coupling, may play a role in the development of late-life cerebrovascular disease, underscoring the need for further longitudinal studies to explore its potential as a mediator of long-term cerebrovascular risk.
Collapse
Affiliation(s)
- Sheng Yang
- Wolfson Centre for Prevention of Stroke and Dementia, Nuffield, Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Alastair John Stewart Webb
- Wolfson Centre for Prevention of Stroke and Dementia, Nuffield, Department of Clinical Neurosciences, University of Oxford, Oxford, UK
- Department of Brain Sciences, Hammersmith Hospital, Imperial College London, London, UK
| |
Collapse
|
2
|
Schweitzer N, Son SJ, Thurston RC, Li J, Chen CL, Aizenstein H, Yang S, Iordanova B, Hong CH, Roh HW, Cho YH, Hong S, Nam YJ, Lee DY, Park B, Kim NR, Choi JW, Cheong J, Seo SW, An YS, Moon SY, Han SJ, Wu M. Sex-specific risk factors and clinical dementia outcomes for white matter hyperintensities in a large South Korean cohort. Alzheimers Res Ther 2024; 16:243. [PMID: 39482724 PMCID: PMC11529246 DOI: 10.1186/s13195-024-01598-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 10/08/2024] [Indexed: 11/03/2024]
Abstract
OBJECTIVE White matter hyperintensities (WMH) on brain MRI images are the most common feature of cerebral small vessel disease (CSVD). Studies have yielded divergent findings on the modifiable risk factors for WMH and WMH's impact on cognitive decline. Mounting evidence suggests sex differences in WMH burden and subsequent effects on cognition. Thus, we aimed to identify sex-specific modifiable risk factors for WMH. We then explored whether there were sex-specific associations of WMH to longitudinal clinical dementia outcomes. METHODS Participants aged 49-89 years were recruited at memory clinics and underwent a T2-weighted fluid-attenuated inversion recovery (FLAIR) 3T MRI scan to measure WMH volume. Participants were then recruited for two additional follow-up visits, 1-2 years apart, where clinical dementia rating sum of boxes (CDR-SB) scores were measured. We first explored which known modifiable risk factors for WMH were significant when tested for a sex-interaction effect. We additionally tested which risk factors were significant when stratified by sex. We then tested to see whether WMH is longitudinally associated with clinical dementia that is sex-specific. RESULTS The study utilized data from 713 participants (241 males, 472 females) with a mean age of 72.3 years and 72.8 years for males and females, respectively. 57.3% and 59.5% of participants were diagnosed with mild cognitive impairment (MCI) for males and females, respectively. 40.7% and 39.4% were diagnosed with dementia for males and females, respectively. Of the 713 participants, 181 participants had CDR-SB scores available for three longitudinal time points. Compared to males, females showed stronger association of age to WMH volume. Type 2 Diabetes was associated with greater WMH burden in females but not males. Finally, baseline WMH burden was associated with worse clinical dementia outcomes longitudinally in females but not in males. DISCUSSION Older females have an accelerated increase in cerebrovascular burden as they age, and subsequently are more vulnerable to clinical dementia decline due to CSVD. Additionally, females are more susceptible to the cerebrovascular consequences of diabetes. These findings emphasize the importance of considering sex when examining the consequences of CSVD. Future research should explore the underlying mechanisms driving these sex differences and personalized prevention and treatment strategies. CLINICAL TRIAL REGISTRATION The BICWALZS is registered in the Korean National Clinical Trial Registry (Clinical Research Information Service; identifier, KCT0003391). Registration Date 2018/12/14.
Collapse
Affiliation(s)
- Noah Schweitzer
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sang Joon Son
- Department of Psychiatry, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Rebecca C Thurston
- Department of Psychiatry, University of Pittsburgh School of Medicine, UPMC Oxford Building, Office 520.13 3501 Forbes Ave, Pittsburgh, PA, 15213, USA
| | - Jinghang Li
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chang-Le Chen
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Howard Aizenstein
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh School of Medicine, UPMC Oxford Building, Office 520.13 3501 Forbes Ave, Pittsburgh, PA, 15213, USA
| | - Shaolin Yang
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry, University of Pittsburgh School of Medicine, UPMC Oxford Building, Office 520.13 3501 Forbes Ave, Pittsburgh, PA, 15213, USA
| | - Bistra Iordanova
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Chang Hyung Hong
- Department of Psychiatry, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Hyun Woong Roh
- Department of Psychiatry, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Yong Hyuk Cho
- Department of Psychiatry, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Sunhwa Hong
- Department of Psychiatry, Ajou University School of Medicine, Suwon, Republic of Korea
| | - You Jin Nam
- Department of Psychiatry, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Dong Yun Lee
- Department of Biomedical Informatics, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Bumhee Park
- Department of Biomedical Informatics, Ajou University School of Medicine, Suwon, Republic of Korea
- Office of Biostatistics, Medical Research Collaborating Centre, Ajou Research Institute for Innovative Medicine, Ajou University Medical Centre, Suwon, Republic of Korea
| | - Na-Rae Kim
- Department of Biomedical Informatics, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Jin Wook Choi
- Department of Radiology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Jaeyoun Cheong
- Department of Gastroenterology, Ajou University School of Medicine, Suwon, Republic of Korea
- Human Genome Research and Bio-Resource Centre, Ajou University Medical Centre, Suwon, Republic of Korea
| | - Sang Woon Seo
- Department of Neurology, Samsung Medical Centre, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Young-Sil An
- Department of Nuclear Medicine and Molecular Imaging, Ajou University School of Medicine, Suwon, Republic of Korea
| | - So Young Moon
- Department of Neurology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Seung Jin Han
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Minjie Wu
- Department of Psychiatry, University of Pittsburgh School of Medicine, UPMC Oxford Building, Office 520.13 3501 Forbes Ave, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
3
|
Manco C, Cortese R, Leoncini M, Plantone D, Gentile G, Luchetti L, Zhang J, Di Donato I, Salvadori E, Poggesi A, Cosottini M, Mascalchi M, Federico A, Dotti MT, Battaglini M, Inzitari D, Pantoni L, De Stefano N. Hippocampal atrophy and white matter lesions characteristics can predict evolution to dementia in patients with vascular mild cognitive impairment. J Neurol Sci 2024; 464:123163. [PMID: 39128160 DOI: 10.1016/j.jns.2024.123163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/01/2024] [Accepted: 08/04/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND Vascular mild cognitive impairment (VMCI) is a transitional condition that may evolve into Vascular Dementia(VaD). Hippocampal volume (HV) is suggested as an early marker for VaD, the role of white matter lesions (WMLs) in neurodegeneration remains debated. OBJECTIVES Evaluate HV and WMLs as predictive markers of VaD in VMCI patients by assessing: (i)baseline differences in HV and WMLs between converters to VaD and non-converters, (ii) predictive power of HV and WMLs for VaD, (iii) associations between HV, WMLs, and cognitive decline, (iv)the role of WMLs on HV. METHODS This longitudinal multicenter study included 110 VMCI subjects (mean age:74.33 ± 6.63 years, 60males/50females) from the VMCI-Tuscany Study database. Subjects underwent brain MRI and cognitive testing, with 2-year follow-up data on VaD progression. HV and WMLs were semi-automatically segmented and measured. ANCOVA assessed group differences, while linear and logistic regression models evaluated predictive power. RESULTS After 2 years, 32/110 VMCI patients progressed to VaD. Converting patients had lower HV(p = 0.015) and higher lesion volumes in the posterior thalamic radiation (p = 0.046), splenium of the corpus callosum (p = 0.016), cingulate gyrus (p = 0.041), and cingulum hippocampus(p = 0.038). HV alone did not fully explain progression (p = 0.059), but combined with WMLs volume, the model was significant (p = 0.035). The best prediction model (p = 0.001) included total HV (p = 0.004) and total WMLs volume of the posterior thalamic radiation (p = 0.005) and cingulate gyrus (p = 0.005), achieving 80% precision, 81% specificity, and 74% sensitivity. Lower HV were linked to poorer performance on the Rey Auditory-Verbal Learning Test delayed recall (RAVLT) and Mini Mental State Examination (MMSE). CONCLUSIONS HV and WMLs are significant predictors of progression from VMCI to VaD. Lower HV correlate with worse cognitive performance on RAVLT and MMSE tests.
Collapse
Affiliation(s)
- Carlo Manco
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
| | - Rosa Cortese
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy.
| | | | - Domenico Plantone
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
| | - Giordano Gentile
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy; Siena Imaging SRL, 53100 Siena, Italy
| | - Ludovico Luchetti
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy; Siena Imaging SRL, 53100 Siena, Italy
| | | | | | - Emilia Salvadori
- Department of Biomedical and Clinical Sciences, University of Milano, Italy
| | - Anna Poggesi
- NEUROFARBA Department, Neuroscience Section, University of Florence, Florence, Italy
| | - Mirco Cosottini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Mario Mascalchi
- Department of Clinical and Experimental Biomedical Sciences -"Mario Serio", University of Florence, Florence, Italy
| | - Antonio Federico
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
| | - Maria Teresa Dotti
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
| | - Marco Battaglini
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy; Siena Imaging SRL, 53100 Siena, Italy
| | - Domenico Inzitari
- NEUROFARBA Department, Neuroscience Section, University of Florence, Florence, Italy
| | - Leonardo Pantoni
- Department of Biomedical and Clinical Sciences, University of Milano, Italy
| | - Nicola De Stefano
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
| |
Collapse
|
4
|
Misiura M, Munkombwe C, Igwe K, Verble DD, Likos KDS, Minto L, Bartlett A, Zetterberg H, Turner JA, Dotson VM, Brickman AM, Hu WT, Wharton W. Neuroimaging correlates of Alzheimer's disease biomarker concentrations in a racially diverse high-risk cohort of middle-aged adults. Alzheimers Dement 2024; 20:5961-5972. [PMID: 39136298 PMCID: PMC11497767 DOI: 10.1002/alz.14051] [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: 02/02/2024] [Revised: 04/24/2024] [Accepted: 05/15/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION In this study, we investigated biomarkers in a midlife, racially diverse, at-risk cohort to facilitate early identification and intervention. We examined neuroimaging measures, including resting state functional magnetic resonance imaging (fMRI), white matter hyperintensity vo (WMH), and hippocampal volumes, alongside cerebrospinal fluid (CSF) markers. METHODS Our data set included 76 cognitively unimpaired, middle-aged, Black Americans (N = 29, F/M = 17/12) and Non-Hispanic White (N = 47, F/M = 27/20) individuals. We compared cerebrospinal fluid phosphorylated tau141 and amyloid beta (Aβ)42 to fMRI default mode network (DMN) subnetwork connectivity, WMH volumes, and hippocampal volumes. RESULTS Results revealed a significant race × Aβ42 interaction in Black Americans: lower Aβ42 was associated with reduced DMN connectivity and increased WMH volumes regions but not in non-Hispanic White individuals. DISCUSSION Our findings suggest that precuneus DMN connectivity and temporal WMHs may be linked to Alzheimer's disease risk pathology during middle age, particularly in Black Americans. HIGHLIGHTS Cerebrospinal fluid (CSF) amyloid beta (Aβ)42 relates to precuneus functional connectivity in Black, but not White, Americans. Higher white matter hyperintensity volume relates to lower CSF Aβ42 in Black Americans. Precuneus may be a hub for early Alzheimer's disease pathology changes detected by functional connectivity.
Collapse
Affiliation(s)
- Maria Misiura
- Department of PsychologyGeorgia State UniversityAtlantaGeorgiaUSA
- Tri‐Institutional Center for Translational Research in Neuroimaging & Data Science, Georgia State University, Georgia Institute of TechnologyEmory UniversityAtlantaGeorgiaUSA
| | | | - Kay Igwe
- Taub Institute for Research in Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, and Department of Neurology, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
| | - Danielle D. Verble
- Nell Hodgson Woodruff School of NursingEmory UniversityAtlantaGeorgiaUSA
| | - Kelly D. S. Likos
- Nell Hodgson Woodruff School of NursingEmory UniversityAtlantaGeorgiaUSA
| | - Lex Minto
- Department of PsychologyGeorgia State UniversityAtlantaGeorgiaUSA
| | | | - Henrik Zetterberg
- The Sahlgrenska Academy, Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, Mölndal and GothenburgUniversity of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalMölndalSweden
- Department of Neurodegenerative Disease, UCL Institute of NeurologyUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCL, Maple HouseLondonUK
- Hong Kong Center for Neurodegenerative DiseasesHong KongChina
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public HealthUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Jessica A. Turner
- Department of Psychiatry and Mental Health, College of MedicineOhio State UniversityColumbusOhioUSA
| | - Vonetta M. Dotson
- Department of PsychologyGeorgia State UniversityAtlantaGeorgiaUSA
- Gerontology DepartmentGeorgia State UniversityAtlantaGeorgiaUSA
| | - Adam M. Brickman
- Taub Institute for Research in Alzheimer's Disease and the Aging Brain, Gertrude H. Sergievsky Center, and Department of Neurology, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- Department of Neurology, Vagelos College of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
| | - William T. Hu
- Institute for Health, Health Care Policy, and Aging ResearchRutgers UniversityNew BrunswickNew JerseyUSA
| | - Whitney Wharton
- Nell Hodgson Woodruff School of NursingEmory UniversityAtlantaGeorgiaUSA
| |
Collapse
|
5
|
Morrison C, Oliver MD, Kamal F, Dadar M. Beyond Hypertension: Examining Variable Blood Pressure's Role in Cognition and Brain Structure. J Gerontol B Psychol Sci Soc Sci 2024; 79:gbae121. [PMID: 39012223 PMCID: PMC11308164 DOI: 10.1093/geronb/gbae121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Indexed: 07/17/2024] Open
Abstract
OBJECTIVES Hypertension or high blood pressure (BP) is one of the 12 modifiable risk factors that contribute to 40% of dementia cases that could be delayed or prevented. Although hypertension is associated with cognitive decline and structural brain changes, less is known about the long-term association between variable BP and cognitive/brain changes. This study examined the relationship between variable BP and longitudinal cognitive, white matter hyperintensity (WMH), gray matter (GM), and white matter (WM) volume change over time and postmortem neuropathology. METHODS A total of 4,606 participants (32,776 follow-ups) from RADC Research Resource Sharing Hub (RUSH) and 2,114 participants (9,827 follow-ups) from the Alzheimer's Disease Neuroimaging Initiative (ADNI) were included. Participants were divided into 1 of 3 groups: normal, high, or variable BP. Linear-mixed models investigated the relationship between BP and cognition, brain structure, and neuropathology. RESULTS Older adults with variable BP exhibited the highest rate of cognitive decline followed by high and then normal BP. Increased GM volume loss and WMH burden were also observed in variable compared to high and normal BP. In postmortem neuropathology, both variable and high BP had increased rates compared to normal BP. Results were consistent across the RUSH and ADNI participants, supporting the generalizability of the findings. DISCUSSION Damages potentially associated with variable BP may reduce resilience to future dementia-related pathology and increased the risk of dementia more than that caused by high BP. Improved treatment and management of variable BP may help reduce cognitive decline in the older adult population.
Collapse
Affiliation(s)
| | - Michael D Oliver
- Department of Psychological Science and Neuroscience, Belmont University, Nashville, Tennessee, USA
- Belmont Data Collaborative, Belmont University, Nashville, Tennessee, USA
| | - Farooq Kamal
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
- Douglas Mental Health University Institute, Verdun, Quebec, Canada
| | - Mahsa Dadar
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
- Douglas Mental Health University Institute, Verdun, Quebec, Canada
| |
Collapse
|
6
|
Affuso F, Micillo F, Fazio S. Insulin Resistance, a Risk Factor for Alzheimer's Disease: Pathological Mechanisms and a New Proposal for a Preventive Therapeutic Approach. Biomedicines 2024; 12:1888. [PMID: 39200352 PMCID: PMC11351221 DOI: 10.3390/biomedicines12081888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Peripheral insulin resistance (IR) is a well-documented, independent risk factor for the development of type 2 diabetes, cardiovascular disease, cancer and cellular senescence. Recently, the brain has also been identified as an insulin-responsive region, where insulin acts as regulator of the brain metabolism. Despite the clear link between IR and the brain, the exact mechanisms underlying this relationship remain unclear. Therapeutic intervention in patients showing symptoms of neurodegenerative diseases has produced little or no results. It has been demonstrated that insulin resistance plays a significant role in the pathogenesis of neurodegenerative diseases, particularly cognitive decline. Peripheral and brain IR may represent a modifiable state that could be used to prevent major brain disorders. In this review, we will analyse the scientific literature supporting IR as a risk factor for Alzheimer's disease and suggest some therapeutic strategies to provide a new proposal for the prevention of brain IR and its consequences.
Collapse
Affiliation(s)
- Flora Affuso
- Independent Researcher, Viale Raffaello, 74, 80129 Napoli, Italy
| | - Filomena Micillo
- UOC of Geriatric Medicine AORN S.G. Moscati, 83100 Avellino, Italy
| | - Serafino Fazio
- Department of Internal Medicine, School of Medicine, Federico II University of Naples, 80138 Naples, Italy;
| |
Collapse
|
7
|
Axelsson Andrén E, Safi D, Wallin A, Svensson J. Low serum HDL-cholesterol is associated with increased risk of the subcortical small vessel type of dementia. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2024; 6:100229. [PMID: 38974908 PMCID: PMC11225847 DOI: 10.1016/j.cccb.2024.100229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/09/2024] [Accepted: 06/04/2024] [Indexed: 07/09/2024]
Abstract
Background There are conflicting results whether serum lipid pattern is related to the amount of white matter hyperintensities (WMHs) on magnetic resonance imaging. Little is known of the associations between lipid concentrations and the subsequent risk of the subcortical small vessel type of dementia (SSVD), in which WMHs are a prominent manifestation. Here, we determined whether lipid levels are associated with the risk of SSVD, Alzheimer's disease (AD), or mixed dementia (combined AD and SSVD). Methods This was a longitudinal, prospective study of 329 patients with subjective or objective mild cognitive impairment at baseline. The statistical analyses included Cox proportional hazards regression with adjustments for age, gender, education, body mass index, current smoking, hypertension, diabetes mellitus, and APOE ε4 genotype. Results During the follow-up (mean 4.1 years), 80 patients converted to dementia [SSVD, n = 15 (5 %); AD, n = 39 (12 %); and mixed dementia, n = 26 (8 %)]. Serum high-density lipoprotein cholesterol (HDL, per SD increase) was inversely associated with the risk of SSVD, whereas triglycerides (TG), low-density lipoprotein cholesterol (LDL)/HDL ratio, and TG/HDL ratio were positively associated with SSVD risk. Furthermore, the lowest HDL tertile was associated with a sevenfold, and the highest tertile of TG/HDL ratio with a threefold, increase in SSVD risk. There were no associations with the risk of AD or mixed dementia after adjustment for covariates. Conclusion In a memory clinic population, low HDL and high TG/HDL ratio were independent risk factors of SSVD, but not of AD or mixed dementia.
Collapse
Affiliation(s)
- Elin Axelsson Andrén
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Dewa Safi
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anders Wallin
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Johan Svensson
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Region Västra Götaland, Department of Internal Medicine, Skaraborg Central Hospital, Skövde, Sweden
| |
Collapse
|
8
|
Dallaire-Théroux C, Smith C, Duchesne S. Clinical Predictors of Postmortem Amyloid and Nonamyloid Cerebral Small Vessel Disease in Middle-Aged to Older Adults. Neurol Clin Pract 2024; 14:e200271. [PMID: 38525067 PMCID: PMC10959170 DOI: 10.1212/cpj.0000000000200271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 01/09/2024] [Indexed: 03/26/2024]
Abstract
Background and Objectives Sporadic cerebral small vessel disease (CSVD) is a class of important pathologic processes known to affect the aging brain and to contribute to cognitive impairment. We aimed to identify clinical risk factors associated with postmortem CSVD in middle-aged to older adults. Methods We developed and tested risk models for their predictive accuracy of a pathologic diagnosis of nonamyloid CSVD and cerebral amyloid angiopathy (CAA) in a retrospective sample of 160 autopsied cases from the Edinburgh Brain Bank. Individuals aged 40 years and older covering the spectrum of healthy aging and common forms of dementia (i.e., highly-prevalent etiologies such as Alzheimer disease (AD), vascular cognitive impairment (VCI), and mixed dementia) were included. We performed binomial logistic regression models using sample splitting and cross-validation methods. Demographics, lifestyle habits, traditional vascular risk factors, chronic medical conditions, APOE4, and cognitive status were assessed as potential predictors. Results Forty percent of our sample had a clinical diagnosis of dementia (AD = 33, VCI = 26 and mixed = 5) while others were cognitively healthy (n = 96). The mean age at death was 73.8 (SD 14.1) years, and 40% were female. The presence of none-to-mild vs moderate-to-severe nonamyloid CSVD was predicted by our model with good accuracy (area under the curve [AUC] = 0.84, sensitivity [SEN] = 72%, specificity [SPE] = 95%), with the most significant clinical predictors being age, history of cerebrovascular events, and cognitive impairment. The presence of CAA pathology was also predicted with high accuracy (AUC = 0.86, SEN = 93%, SPE = 79%). Significant predictors included alcohol intake, history of cerebrovascular events, and cognitive impairment. In a subset of atypical dementias (n = 24), our models provided poor predictive performance for both nonamyloid CSVD (AUC = 0.50) and CAA (AUC = 0.43). Discussion CSVD pathology can be predicted with high accuracy based on clinical factors in patients within the spectrum of AD, VCI, and normal aging. Whether this prediction can be enhanced by the addition of fluid and neuroimaging biomarkers warrants additional study. Improving our understanding of clinical determinants of vascular brain health may lead to novel strategies in the prevention and treatment of vascular etiologies contributing to cognitive decline. Classification of Evidence This study provides Class II evidence that selected clinical factors accurately distinguish between middle-aged to older adults with and without cerebrovascular small vessel disease (amyloid and nonamyloid) pathology.
Collapse
Affiliation(s)
- Caroline Dallaire-Théroux
- CERVO Brain Research Center (CD-T, SD); Faculty of Medicine (CD-T), Université Laval; Department of Neurological Sciences (CD-T), Centre Hospitalier Universitaire de Québec, Canada; Academic Neuropathology (CS), Centre for Clinical Brain Sciences, University of Edinburgh, United Kingdom; and Department of Radiology and Nuclear Medicine (SD), Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Colin Smith
- CERVO Brain Research Center (CD-T, SD); Faculty of Medicine (CD-T), Université Laval; Department of Neurological Sciences (CD-T), Centre Hospitalier Universitaire de Québec, Canada; Academic Neuropathology (CS), Centre for Clinical Brain Sciences, University of Edinburgh, United Kingdom; and Department of Radiology and Nuclear Medicine (SD), Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Simon Duchesne
- CERVO Brain Research Center (CD-T, SD); Faculty of Medicine (CD-T), Université Laval; Department of Neurological Sciences (CD-T), Centre Hospitalier Universitaire de Québec, Canada; Academic Neuropathology (CS), Centre for Clinical Brain Sciences, University of Edinburgh, United Kingdom; and Department of Radiology and Nuclear Medicine (SD), Faculty of Medicine, Université Laval, Quebec City, Canada
| |
Collapse
|
9
|
Jia R, Solé-Guardia G, Kiliaan AJ. Blood-brain barrier pathology in cerebral small vessel disease. Neural Regen Res 2024; 19:1233-1240. [PMID: 37905869 PMCID: PMC11467932 DOI: 10.4103/1673-5374.385864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/27/2023] [Accepted: 08/22/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT Cerebral small vessel disease is a neurological disease that affects the brain microvasculature and which is commonly observed among the elderly. Although at first it was considered innocuous, small vessel disease is nowadays regarded as one of the major vascular causes of dementia. Radiological signs of small vessel disease include small subcortical infarcts, white matter magnetic resonance imaging hyperintensities, lacunes, enlarged perivascular spaces, cerebral microbleeds, and brain atrophy; however, great heterogeneity in clinical symptoms is observed in small vessel disease patients. The pathophysiology of these lesions has been linked to multiple processes, such as hypoperfusion, defective cerebrovascular reactivity, and blood-brain barrier dysfunction. Notably, studies on small vessel disease suggest that blood-brain barrier dysfunction is among the earliest mechanisms in small vessel disease and might contribute to the development of the hallmarks of small vessel disease. Therefore, the purpose of this review is to provide a new foundation in the study of small vessel disease pathology. First, we discuss the main structural domains and functions of the blood-brain barrier. Secondly, we review the most recent evidence on blood-brain barrier dysfunction linked to small vessel disease. Finally, we conclude with a discussion on future perspectives and propose potential treatment targets and interventions.
Collapse
Affiliation(s)
- Ruxue Jia
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, the Netherlands
| | - Gemma Solé-Guardia
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, the Netherlands
| | - Amanda J. Kiliaan
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, the Netherlands
| |
Collapse
|
10
|
Morrison C, Oliver MD, Kamal F, Dadar M. Beyond Hypertension: Examining Variable Blood Pressure's Role in Cognition and Brain Structure. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.15.24301335. [PMID: 38293179 PMCID: PMC10827268 DOI: 10.1101/2024.01.15.24301335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Importance Hypertension is a known risk factor for cognitive decline and structural brain changes in aging and dementia. In addition to high blood pressure (BP), individuals may also experience variable BP, meaning that their BP fluctuates between normal and high. It is currently unclear what the effects of variable BP are on cognition and brain structure. Objective To investigate the influence of BP on cognition and brain structure in older adults. Design Setting and Participants This longitudinal cohort study included data from the Rush Alzheimer's Disease Center Research Resource Sharing Hub (RUSH) and the Alzheimer's Disease Neuroimaging Initiative (ADNI). Participants from the two studies were included if they had BP measurements and either cognitive scores or MRI scans from at least one visit. Main Outcomes and Measures Longitudinal gray matter, white matter, white matter hyperintensity volumes, postmortem neuropathology information, as well as cognitive test scores. Results A total of 4606 participants (3429 females, mean age = 76.8) with 32776 follow-ups (mean = 7 years) from RUSH and 2114 participants (1132 females, mean age = 73.3) with 9827 follow-ups (mean = 3 years) from ADNI were included in this study. Participants were divided into one of three groups: 1) normal BP, high BP, or variable BP. Older adults with variable BP exhibited the highest rate of cognitive decline followed by high BP and then normal BP. Increased GM volume loss and WMH burden was also observed in variable BP compared to high and normal BP. With respect to post-mortem neuropathology, both variable and high BP had increased severities compared to normal BP. Importantly, results were consistent across the RUSH and ADNI participants, supporting the generalizability of the findings. Conclusion and Relevance Limited research has examined the long-term impact of variable BP on cognition and brain structure. These findings show the importance that both high and variable BP have on cognitive decline and structural brain changes. Structural damages caused by variable BP may reduce resilience to future dementia-related pathology and increased risk of dementia. Improved treatment and management of variable BP may help reduce cognitive decline in the older adult population.
Collapse
Affiliation(s)
| | - Michael D Oliver
- Department of Psychological Science and Neuroscience, Belmont University, Nashville, Tennessee, United States
- Belmont Data Collaborative, Belmont University, Nashville, Tennessee, United States
| | - Farooq Kamal
- Department of Psychiatry, McGill University, Montreal, Quebec, H3A 1A1, Canada
- Douglas Mental Health University Institute, Montreal, Quebec, H4H 1R3, Canada
| | - Mahsa Dadar
- Department of Psychiatry, McGill University, Montreal, Quebec, H3A 1A1, Canada
- Douglas Mental Health University Institute, Montreal, Quebec, H4H 1R3, Canada
| |
Collapse
|
11
|
Morrison C, Dadar M, Collins DL. Sex differences in risk factors, burden, and outcomes of cerebrovascular disease in Alzheimer's disease populations. Alzheimers Dement 2024; 20:34-46. [PMID: 37735954 PMCID: PMC10916959 DOI: 10.1002/alz.13452] [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: 04/19/2023] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 09/23/2023]
Abstract
BACKGROUND White matter hyperintensities (WMHs) are associated with cognitive decline and progression to mild cognitive impairment (MCI) and dementia. It remains unclear if sex differences influence WMH progression or the relationship between WMH and cognition. METHODS Linear mixed models examined the relationship between risk factors, WMHs, and cognition in males and females. RESULTS Males exhibited increased WMH progression in occipital, but lower progression in frontal, total, and deep than females. For males, history of hypertension was the strongest contributor, while in females, the vascular composite was the strongest contributor to WMH burden. WMH burden was more strongly associated with decreases in global cognition, executive functioning, memory, and functional activities in females than males. DISCUSSION Controlling vascular risk factors may reduce WMH in both males and females. For males, targeting hypertension may be most important to reduce WMHs. The results have implications for therapies/interventions targeting cerebrovascular pathology and subsequent cognitive decline. HIGHLIGHTS Hypertension is the main vascular risk factor associated with WMH in males A combination of vascular risk factors contributes to WMH burden in females Only small WMH burden differences were observed between sexes Females' cognition was more negatively impacted by WMH burden than males Females with WMHs may have less resilience to future pathology.
Collapse
Affiliation(s)
- Cassandra Morrison
- McConnell Brain Imaging CentreMontreal Neurological InstituteMcGill UniversityMontrealQuebecCanada
- Department of Neurology and NeurosurgeryMcGill UniversityMontrealQuebecCanada
| | - Mahsa Dadar
- Department of PsychiatryMcGill UniversityMontrealQuebecCanada
- Douglas Mental Health University Institute, McGill UniversityMontrealQuebecCanada
| | - Donald Louis Collins
- McConnell Brain Imaging CentreMontreal Neurological InstituteMcGill UniversityMontrealQuebecCanada
- Department of Neurology and NeurosurgeryMcGill UniversityMontrealQuebecCanada
| | | |
Collapse
|
12
|
Wang R, Deng Y, Zhang W, Ning J, Li H, Feng J, Cheng W, Yu J. Associations between adiposity and white matter hyperintensities: Cross-sectional and longitudinal analyses of 34,653 participants. Hum Brain Mapp 2024; 45:e26560. [PMID: 38224536 PMCID: PMC10789203 DOI: 10.1002/hbm.26560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 11/15/2023] [Accepted: 11/28/2023] [Indexed: 01/17/2024] Open
Abstract
OBJECTIVES White matter hyperintensities (WMH) increase the risk of stroke and cognitive impairment. This study aims to determine the cross-sectional and longitudinal associations between adiposity and WMH. METHODS Participants were enrolled from the UK Biobank cohort. Associations of concurrent, past, and changes in overall and central adiposity with WMH were investigated by linear and nonlinear regression models. The association of longitudinal adiposity and WMH volume changes was determined by a linear mixed model. Mediation analysis investigated the potential mediating effect of blood pressure. RESULTS In 34,653 participants with available adiposity measures and imaging data, the concurrent obese group had a 25.3% (β [95% CI] = 0.253 [0.222-0.284]) higher WMH volume than the ideal weight group. Increment in all adiposity measures was associated with a higher WMH volume. Among them, waist circumference demonstrated the strongest effect (β [95% CI] = 0.113 [0.101-0.125]). Past adiposity also demonstrated similar effects. Among the subset of 2664 participants with available WMH follow-up data, adiposity measures were predictive of WMH change. Regarding changes of adiposity, compared with ideal weight stable group, those who turned from ideal weight to overweight/obese had a 8.1% higher WMH volume (β [95% CI] = 0.081 [0.039-0.123]), while participants who turned from overweight/obese to ideal weight demonstrated no significant WMH volume change. Blood pressure partly meditates the associations between adiposity and WMH. CONCLUSIONS Both concurrent and past adiposity were associated with a higher WMH volume. The detrimental effects of adiposity on WMH occurred throughout midlife and in the elderly and may still exist after changes in obesity status.
Collapse
Affiliation(s)
- Rong‐Ze Wang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Yue‐Ting Deng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Wei Zhang
- Institute of Science and Technology for Brain‐Inspired IntelligenceFudan UniversityShanghaiChina
- Key Laboratory of Computational Neuroscience and Brain Inspired IntelligenceFudan University, Ministry of EducationShanghaiChina
| | - Jing Ning
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Hong‐Qi Li
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Jian‐Feng Feng
- Institute of Science and Technology for Brain‐Inspired IntelligenceFudan UniversityShanghaiChina
- Key Laboratory of Computational Neuroscience and Brain Inspired IntelligenceFudan University, Ministry of EducationShanghaiChina
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Institute of Science and Technology for Brain‐Inspired IntelligenceFudan UniversityShanghaiChina
- Key Laboratory of Computational Neuroscience and Brain Inspired IntelligenceFudan University, Ministry of EducationShanghaiChina
| | - Jin‐Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontier Center for Brain Science, Shanghai Medical CollegeFudan UniversityShanghaiChina
| |
Collapse
|
13
|
Horvath A, Quinlan P, Eckerström C, Åberg ND, Wallin A, Svensson J. The Associations Between Serum Insulin-like Growth Factor-I, Brain White Matter Volumes, and Cognition in Mild Cognitive Impairment and Alzheimer's Disease. J Alzheimers Dis 2024; 99:609-622. [PMID: 38701139 PMCID: PMC11191442 DOI: 10.3233/jad-231026] [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] [Accepted: 03/21/2024] [Indexed: 05/05/2024]
Abstract
Background Insulin-like growth factor-I (IGF-I) regulates myelin, but little is known whether IGF-I associates with white matter functions in subjective and objective mild cognitive impairment (SCI/MCI) or Alzheimer's disease (AD). Objective To explore whether serum IGF-I is associated with magnetic resonance imaging - estimated brain white matter volumes or cognitive functions. Methods In a prospective study of SCI/MCI (n = 106) and AD (n = 59), we evaluated the volumes of the total white matter, corpus callosum (CC), and white matter hyperintensities (WMHs) as well as Mini-Mental State Examination (MMSE), Trail Making Test A and B (TMT-A/B), and Stroop tests I-III at baseline, and after 2 years. Results IGF-I was comparable in SCI/MCI and AD (113 versus 118 ng/mL, p = 0.44). In SCI/MCI patients, the correlations between higher baseline IGF-I and greater baseline and 2-year volumes of the total white matter and total CC lost statistical significance after adjustment for intracranial volume and other covariates. However, after adjustment for covariates, higher baseline IGF-I correlated with better baseline scores of MMSE and Stroop test II in SCI/MCI and with better baseline results of TMT-B and Stroop test I in AD. IGF-I did not correlate with WMH volumes or changes in any of the variables. Conclusions Both in SCI/MCI and AD, higher IGF-I was associated with better attention/executive functions at baseline after adjustment for covariates. Furthermore, the baseline associations between IGF-I and neuropsychological test results in AD may argue against significant IGF-I resistance in the AD brain.
Collapse
Affiliation(s)
- Alexandra Horvath
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Patrick Quinlan
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Carl Eckerström
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - N. David Åberg
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Acute Medicine and Geriatrics, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anders Wallin
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Johan Svensson
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Internal Medicine, Skaraborg Central Hospital, Skövde, Sweden
| |
Collapse
|
14
|
Kamal F, Morrison C, Dadar M. Investigating the relationship between sleep disturbances and white matter hyperintensities in older adults on the Alzheimer's disease spectrum. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2024; 16:e12553. [PMID: 38476639 PMCID: PMC10927930 DOI: 10.1002/dad2.12553] [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: 05/01/2023] [Revised: 12/15/2023] [Accepted: 01/19/2024] [Indexed: 03/14/2024]
Abstract
INTRODUCTION While studies report that sleep disturbance can have negative effects on brain vasculature, its impact on cerebrovascular diseases such as white matter hyperintensities (WMHs) in beta-amyloid-positive older adults remains unexplored. METHODS Sleep disturbance, WMH burden, and cognition in normal controls (NCs), and individuals with mild cognitive impairment (MCI) and Alzheimer's disease (AD), were examined at baseline and longitudinally. A total of 912 amyloid-positive participants were included (198 NC, 504 MCI, and 210 AD). RESULTS Individuals with AD reported more sleep disturbances than NC and MCI participants. Those with sleep disturbances had more WMHs than those without sleep disturbances in the AD group. Mediation analysis revealed an effect of regional WMH burden on the relationship between sleep disturbance and future cognition. DISCUSSION These results suggest that WMH burden and sleep disturbance increase from aging to AD. Sleep disturbance decreases cognition through increases in WMH burden. Improved sleep could mitigate the impact of WMH accumulation and cognitive decline.
Collapse
Affiliation(s)
- Farooq Kamal
- Department of PsychiatryMcGill UniversityMontrealQuebecCanada
- Douglas Mental Health University InstituteMontrealQuebecCanada
| | | | - Mahsa Dadar
- Department of PsychiatryMcGill UniversityMontrealQuebecCanada
- Douglas Mental Health University InstituteMontrealQuebecCanada
| |
Collapse
|
15
|
Parent O, Bussy A, Devenyi GA, Dai A, Costantino M, Tullo S, Salaciak A, Bedford S, Farzin S, Béland ML, Valiquette V, Villeneuve S, Poirier J, Tardif CL, Dadar M, Chakravarty MM. Assessment of white matter hyperintensity severity using multimodal magnetic resonance imaging. Brain Commun 2023; 5:fcad279. [PMID: 37953840 PMCID: PMC10636521 DOI: 10.1093/braincomms/fcad279] [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: 05/04/2023] [Revised: 09/05/2023] [Accepted: 10/17/2023] [Indexed: 11/14/2023] Open
Abstract
White matter hyperintensities are radiological abnormalities reflecting cerebrovascular dysfunction detectable using MRI. White matter hyperintensities are often present in individuals at the later stages of the lifespan and in prodromal stages in the Alzheimer's disease spectrum. Tissue alterations underlying white matter hyperintensities may include demyelination, inflammation and oedema, but these are highly variable by neuroanatomical location and between individuals. There is a crucial need to characterize these white matter hyperintensity tissue alterations in vivo to improve prognosis and, potentially, treatment outcomes. How different MRI measure(s) of tissue microstructure capture clinically-relevant white matter hyperintensity tissue damage is currently unknown. Here, we compared six MRI signal measures sampled within white matter hyperintensities and their associations with multiple clinically-relevant outcomes, consisting of global and cortical brain morphometry, cognitive function, diagnostic and demographic differences and cardiovascular risk factors. We used cross-sectional data from 118 participants: healthy controls (n = 30), individuals at high risk for Alzheimer's disease due to familial history (n = 47), mild cognitive impairment (n = 32) and clinical Alzheimer's disease dementia (n = 9). We sampled the median signal within white matter hyperintensities on weighted MRI images [T1-weighted (T1w), T2-weighted (T2w), T1w/T2w ratio, fluid-attenuated inversion recovery (FLAIR)] as well as the relaxation times from quantitative T1 (qT1) and T2* (qT2*) images. qT2* and fluid-attenuated inversion recovery signals within white matter hyperintensities displayed different age- and disease-related trends compared to normal-appearing white matter signals, suggesting sensitivity to white matter hyperintensity-specific tissue deterioration. Further, white matter hyperintensity qT2*, particularly in periventricular and occipital white matter regions, was consistently associated with all types of clinically-relevant outcomes in both univariate and multivariate analyses and across two parcellation schemes. qT1 and fluid-attenuated inversion recovery measures showed consistent clinical relationships in multivariate but not univariate analyses, while T1w, T2w and T1w/T2w ratio measures were not consistently associated with clinical variables. We observed that the qT2* signal was sensitive to clinically-relevant microstructural tissue alterations specific to white matter hyperintensities. Our results suggest that combining volumetric and signal measures of white matter hyperintensity should be considered to fully characterize the severity of white matter hyperintensities in vivo. These findings may have implications in determining the reversibility of white matter hyperintensities and the potential efficacy of cardio- and cerebrovascular treatments.
Collapse
Affiliation(s)
- Olivier Parent
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - Aurélie Bussy
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - Gabriel Allan Devenyi
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - Alyssa Dai
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec H3A 1A1, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Quebec H3A 2B4, Canada
| | - Manuela Costantino
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
| | - Stephanie Tullo
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - Alyssa Salaciak
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
| | - Saashi Bedford
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - Sarah Farzin
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
| | - Marie-Lise Béland
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
| | - Vanessa Valiquette
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - Sylvia Villeneuve
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
- Center for the Studies in the Prevention of Alzheimer's Disease, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Quebec H3A 2B4, Canada
| | - Judes Poirier
- Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
- Center for the Studies in the Prevention of Alzheimer's Disease, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
- Molecular Neurobiology Unit, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
- Department of Medicine, McGill University, Montreal, Quebec H4A 3J1, Canada
| | - Christine Lucas Tardif
- McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Quebec H3A 2B4, Canada
- Department of Biomedical Engineering, McGill University, Montreal, Quebec H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - Mahsa Dadar
- Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - M Mallar Chakravarty
- Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, Quebec H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec H3A 1A1, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
- Department of Biomedical Engineering, McGill University, Montreal, Quebec H3A 2B4, Canada
| |
Collapse
|
16
|
Kamal F, Morrison C, Maranzano J, Zeighami Y, Dadar M. White Matter Hyperintensity Trajectories in Patients With Progressive and Stable Mild Cognitive Impairment. Neurology 2023; 101:e815-e824. [PMID: 37407262 PMCID: PMC10449435 DOI: 10.1212/wnl.0000000000207514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/25/2023] [Indexed: 07/07/2023] Open
Abstract
BACKGROUND AND OBJECTIVES White matter hyperintensities (WMH) are pathologic brain changes that are associated with increased age and cognitive decline. However, the association of WMH burden with amyloid positivity and conversion to dementia in people with mild cognitive impairment (MCI) is unclear. The aim of this study was to expand on this research by examining whether change in WMH burden over time differs in amyloid-negative (Aβ-) and amyloid-positive (Aβ+) people with MCI who either remain stable or convert to dementia. To examine this question, we compared regional WMH burden in 4 groups: Aβ+ progressor, Aβ- progressor, Aβ+ stable, and Aβ- stable. METHODS Participants with MCI from the Alzheimer Disease Neuroimaging Initiative were included if they had APOE ɛ4 status and if amyloid measures were available to determine amyloid status (i.e., Aβ+, or Aβ-). Participants with a baseline diagnosis of MCI and who had APOE ɛ4 information and amyloid measures were included. An average of 5.7 follow-up time points per participant were included, with a total of 5,054 follow-up time points with a maximum follow-up duration of 13 years. Differences in total and regional WMH burden were examined using linear mixed-effects models. RESULTS A total of 820 participants (55-90 years of age) were included in the study (Aβ+ progressor, n = 239; Aβ- progressor, n = 22; Aβ+ stable, n = 343; Aβ- stable, n = 216). People who were Aβ- stable exhibited reduced baseline WMH compared with Aβ+ progressors and people who were Aβ+ stable at all regions of interest (β belongs to 0.20-0.33, CI belongs to 0.03-0.49, p < 0.02), except deep WMH. When examining longitudinal results, compared with people who were Aβ- stable, all groups had steeper accumulation in WMH burden with Aβ+ progressors (β belongs to -0.03 to 0.06, CI belongs to -0.05 to 0.09, p < 0.01) having the largest increase (i.e., largest increase in WMH accumulation over time). DISCUSSION These results indicate that WMH accumulation contributes to conversion to dementia in older adults with MCI who are Aβ+ and Aβ-.
Collapse
Affiliation(s)
- Farooq Kamal
- From the Department of Psychiatry (F.K., Y.Z., M.D.), McGill University; Douglas Mental Health University Institute (F.K., Y.Z., M.D.); Department of Neurology and Neurosurgery (C.M., J.M.), Faculty of Medicine, and McConnell Brain Imaging Centre (C.M.), Montreal Neurological Institute, McGill University; and Department of Anatomy (J.M.), University of Quebec in Trois-Rivières, Canada.
| | - Cassandra Morrison
- From the Department of Psychiatry (F.K., Y.Z., M.D.), McGill University; Douglas Mental Health University Institute (F.K., Y.Z., M.D.); Department of Neurology and Neurosurgery (C.M., J.M.), Faculty of Medicine, and McConnell Brain Imaging Centre (C.M.), Montreal Neurological Institute, McGill University; and Department of Anatomy (J.M.), University of Quebec in Trois-Rivières, Canada
| | - Josefina Maranzano
- From the Department of Psychiatry (F.K., Y.Z., M.D.), McGill University; Douglas Mental Health University Institute (F.K., Y.Z., M.D.); Department of Neurology and Neurosurgery (C.M., J.M.), Faculty of Medicine, and McConnell Brain Imaging Centre (C.M.), Montreal Neurological Institute, McGill University; and Department of Anatomy (J.M.), University of Quebec in Trois-Rivières, Canada
| | - Yashar Zeighami
- From the Department of Psychiatry (F.K., Y.Z., M.D.), McGill University; Douglas Mental Health University Institute (F.K., Y.Z., M.D.); Department of Neurology and Neurosurgery (C.M., J.M.), Faculty of Medicine, and McConnell Brain Imaging Centre (C.M.), Montreal Neurological Institute, McGill University; and Department of Anatomy (J.M.), University of Quebec in Trois-Rivières, Canada
| | - Mahsa Dadar
- From the Department of Psychiatry (F.K., Y.Z., M.D.), McGill University; Douglas Mental Health University Institute (F.K., Y.Z., M.D.); Department of Neurology and Neurosurgery (C.M., J.M.), Faculty of Medicine, and McConnell Brain Imaging Centre (C.M.), Montreal Neurological Institute, McGill University; and Department of Anatomy (J.M.), University of Quebec in Trois-Rivières, Canada
| |
Collapse
|
17
|
Jia X, Li Y, Ying Y, Jia X, Tang W, Bian Y, Zhang J, Wang DJJ, Cheng X, Yang Q. Effect of corticosubcortical iron deposition on dysfunction in CADASIL is mediated by white matter microstructural damage. Neuroimage Clin 2023; 39:103485. [PMID: 37542975 PMCID: PMC10407949 DOI: 10.1016/j.nicl.2023.103485] [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: 03/30/2023] [Revised: 07/26/2023] [Accepted: 07/26/2023] [Indexed: 08/07/2023]
Abstract
Iron dysregulation may attenuate cognitive performance in patients with CADASIL. However, the underlying pathophysiological mechanisms remain incompletely understood. Whether white matter microstructural changes mediate these processes is largely unclear. In the present study, 30 cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) patients were confirmed via genetic analysis and 30 sex- and age-matched healthy controls underwent multimodal MRI examinations and neuropsychological assessments. Quantitative susceptibility mapping and peak width of skeletonized mean diffusivity (PSMD) were analyzed. Mediation effect analysis was performed to explore the interrelationship between iron deposition, white matter microstructural changes and cognitive deficits in CADASIL. Cognitive deterioration was most affected in memory and executive function, followed by attention and working memory in CADASIL. Excessive iron in the temporal-precuneus pathway and deep gray matter specific to CADASIL were identified. Mediation analysis further revealed that PSMD mediated the relationship between iron concentration and cognitive profile in CADASIL. The present findings provide a new perspective on iron deposition in the corticosubcortical circuit and its contribution to disease-related selective cognitive decline, in which iron concentration may affect cognition by white matter microstructural changes in CADASIL.
Collapse
Affiliation(s)
- Xiuqin Jia
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China; Key Lab of Medical Engineering for Cardiovascular Disease, Ministry of Education, Beijing 100020, China
| | - Yingying Li
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Yunqing Ying
- Department of Neurology, National Center for Neurological Disorders, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xuejia Jia
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Weijun Tang
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yueyan Bian
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Jiajia Zhang
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Danny J J Wang
- Laboratory of FMRI Technology (LOFT), USC Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90007, United States
| | - Xin Cheng
- Department of Neurology, National Center for Neurological Disorders, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China.
| | - Qi Yang
- Department of Radiology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China; Key Lab of Medical Engineering for Cardiovascular Disease, Ministry of Education, Beijing 100020, China.
| |
Collapse
|
18
|
Verovnik B, Khachatryan E, Šuput D, Van Hulle MM. Effects of risk factors on longitudinal changes in brain structure and function in the progression of AD. Alzheimers Dement 2023; 19:2666-2676. [PMID: 36807765 DOI: 10.1002/alz.12991] [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: 10/21/2022] [Revised: 01/03/2023] [Accepted: 01/05/2023] [Indexed: 02/20/2023]
Abstract
INTRODUCTION Past research on Alzheimer's disease (AD) has focused on biomarkers, cognition, and neuroimaging as primary predictors of its progression, albeit additional ones have recently gained attention. When turning to the prediction of the progression from one stage to another, one could benefit from the joint assessment of imaging-based biomarkers and risk/protective factors. METHODS We included 86 studies that fulfilled our inclusion criteria. RESULTS Our review summarizes and discusses the results of 30 years of longitudinal research on brain changes assessed with neuroimaging and the risk/protective factors and their effect on AD progression. We group results into four sections: genetic, demographic, cognitive and cardiovascular, and lifestyle factors. DISCUSSION Given the complex nature of AD, including risk factors could prove invaluable for a better understanding of AD progression. Some of these risk factors are modifiable and could be targeted by potential future treatments.
Collapse
Affiliation(s)
- Barbara Verovnik
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Laboratory for Neuro- and Psychophysiology, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Elvira Khachatryan
- Laboratory for Neuro- and Psychophysiology, Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Dušan Šuput
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Center for Clinical Physiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Marc M Van Hulle
- Laboratory for Neuro- and Psychophysiology, Department of Neurosciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
19
|
Ding J, Zhang H, Hua B, Feng C, Yang M, Ding X, Yang C. Frequency specificity in the amplitude of low frequency oscillations in patients with white matter lesions. J Clin Neurosci 2023; 113:86-92. [PMID: 37229795 DOI: 10.1016/j.jocn.2023.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/12/2023] [Accepted: 05/16/2023] [Indexed: 05/27/2023]
Abstract
Previous studies have reported that patients with white matter lesions (WMLs) have abnormal spontaneous brain activity in the resting state. However, the spontaneous neuronal activity of specific frequency bands in WMLs patients is unknown. Here, we included 16 WMLs patients and 13 gender and age-matched healthy controls (HCs) underwent resting-state magnetic resonance imaging (rs-fMRI) scan and studied the specificity of the amplitude of low-frequency fluctuations (ALFF) in WMLs patients in the slow-5 (0.01-0.027 Hz), slow-4 (0.027-0.073 Hz), and typical (0.01-0.08 Hz) frequency bands. In addition, ALFF values of different frequency bands were extracted as classification features and support vector machines (SVM) were used to classify WMLs patients. In all three frequency bands, significant increases in ALFF values in WMLs patients were observed in the cerebellum. In the slow-5 band, the ALFF values of the left anterior cingulate and paracingulate gyri (ACG), and the right precentral gyrus, rolandic operculum and inferior temporal gyrus in WMLs patients were lower than those in HCs. In the slow-4 band, ALFF values were lower in WMLs patients than in HCs at the left ACG, the right median cingulate and paracingulate gyri, parahippocampal gyrus, caudate nucleus, and the bilateral lenticular nucleus, putamen. In the SVM classification model, the classification accuracy of slow-5, slow-4 and typical frequency bands is 75.86%, 86.21% and 72.41%, respectively. The results indicate that the ALFF abnormalities in WMLs patients have frequency specificity, and the ALFF abnormalities in the slow-4 frequency band may serve as imaging markers for WMLs.
Collapse
Affiliation(s)
- Jurong Ding
- School of Automation and Information Engineering, Sichuan University of Science and Engineering, Zigong, PR China; Artificial Intelligence Key Laboratory of Sichuan Province, Sichuan University of Science & Engineering, Zigong, PR China.
| | - Hui Zhang
- School of Automation and Information Engineering, Sichuan University of Science and Engineering, Zigong, PR China; Artificial Intelligence Key Laboratory of Sichuan Province, Sichuan University of Science & Engineering, Zigong, PR China
| | - Bo Hua
- School of Automation and Information Engineering, Sichuan University of Science and Engineering, Zigong, PR China; Artificial Intelligence Key Laboratory of Sichuan Province, Sichuan University of Science & Engineering, Zigong, PR China
| | - Chenyu Feng
- School of Automation and Information Engineering, Sichuan University of Science and Engineering, Zigong, PR China; Artificial Intelligence Key Laboratory of Sichuan Province, Sichuan University of Science & Engineering, Zigong, PR China
| | - Mei Yang
- School of Automation and Information Engineering, Sichuan University of Science and Engineering, Zigong, PR China; Artificial Intelligence Key Laboratory of Sichuan Province, Sichuan University of Science & Engineering, Zigong, PR China
| | - Xin Ding
- Department of Neurology, Chengdu Second People's Hospital, Chengdu, PR China.
| | - Chenghao Yang
- Department of Neurosurgery, Zigong Fourth People's Hospital, Zigong, PR China
| |
Collapse
|
20
|
Sadeghzadeh J, Jafarzadeh J, Hadinezhad P, Nazari A, Sohrabi S, Musazadeh V, Barzegar A, Shahabi P. Profiling inflammatory mechanisms, hyperphosphorylated tau of hippocampal tissue and spatial memory following vitamin D3 treatment in the mice model of vascular dementia. Int Immunopharmacol 2023; 120:110314. [PMID: 37220695 DOI: 10.1016/j.intimp.2023.110314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/21/2023] [Accepted: 05/07/2023] [Indexed: 05/25/2023]
Abstract
BACKGROUND The aim of this study was to investigate the effect of vitamin D3 (VitD3) on inflammatory mechanisms, hyperphosphorylated tau (p-tau) in the hippocampus, and cognitive impairment of the mouse model of vascular dementia (VaD). METHODS In this study, 32 male mice were randomly assigned to the control, VaD, VitD3 (300 IU/Kg/day), and VitD3 (500 IU/Kg/day) groups. VaD and VitD3 groups were gavaged daily for 4 weeks with a gastric needle. For biochemical assessments, blood samples and the hippocampus were isolated. IL-1β and TNF-α were analyzed by ELISA, and p-tau and other inflammatory molecules were measured by western blot. RESULTS VitD3 supplements significantly (P < 0.05) decreased the level of inflammatory factors in the hippocampus and prevented apoptosis. However, regarding p-tau in hippocampal tissue, this decrease was not statistically significant (P > 0.05). The results of behavioral assessments showed that VitD3 significantly improved the spatial memory of treated mice. CONCLUSION These results suggest that the neuroprotective effects of VitD3 are mainly associated with their anti-inflammatory effects.
Collapse
Affiliation(s)
- Jafar Sadeghzadeh
- Department of Neuroscience and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jaber Jafarzadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Community Nutrition, Faculty of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pezhman Hadinezhad
- Department of Psychiatry, Mazandaran University of Medical Sciences, Sari, Iran; Psychiatry and Behavioral Sciences Research Center, Addiction Institute, Mazandaran University of Medical Sciences,Sari, Iran
| | - Ahmad Nazari
- Tehran University of Medical Sciences, Tehran, Iran
| | - Saeedeh Sohrabi
- School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Vali Musazadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Barzegar
- Department of Community Nutrition, Faculty of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Parviz Shahabi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Departments of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
21
|
Cao Q, Chen J, Zhang Z, Shu S, Qian Y, Yang L, Xu L, Zhang Y, Bao X, Xia S, Yang H, Xu Y, Qiu S. Astrocytic CXCL5 hinders microglial phagocytosis of myelin debris and aggravates white matter injury in chronic cerebral ischemia. J Neuroinflammation 2023; 20:105. [PMID: 37138312 PMCID: PMC10155379 DOI: 10.1186/s12974-023-02780-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/17/2023] [Indexed: 05/05/2023] Open
Abstract
BACKGROUND Chronic cerebral ischemia induces white matter injury (WMI) contributing to cognitive decline. Both astrocytes and microglia play vital roles in the demyelination and remyelination processes, but the underlying mechanism remains unclear. This study aimed to explore the influence of the chemokine CXCL5 on WMI and cognitive decline in chronic cerebral ischemia and the underlying mechanism. METHODS Bilateral carotid artery stenosis (BCAS) model was constructed to mimic chronic cerebral ischemia in 7-10 weeks old male mice. Astrocytic Cxcl5 conditional knockout (cKO) mice were constructed and mice with Cxcl5 overexpressing in astrocytes were generated by stereotactic injection of adeno-associated virus (AAV). WMI was evaluated by magnetic resonance imaging (MRI), electron microscopy, histological staining and western blotting. Cognitive function was examined by a series of neurobehavioral tests. The proliferation and differentiation of oligodendrocyte progenitor cells (OPCs), phagocytosis of microglia were analyzed via immunofluorescence staining, western blotting or flow cytometry. RESULTS CXCL5 was significantly elevated in the corpus callosum (CC) and serum in BCAS model, mainly expressed in astrocytes, and Cxcl5 cKO mice displayed improved WMI and cognitive performance. Recombinant CXCL5 (rCXCL5) had no direct effect on the proliferation and differentiation of OPCs in vitro. Astrocytic specific Cxcl5 overexpression aggravated WMI and cognitive decline induced by chronic cerebral ischemia, while microglia depletion counteracted this effect. Recombinant CXCL5 remarkably hindered microglial phagocytosis of myelin debris, which was rescued by inhibition of CXCL5 receptor C-X-C motif chemokine receptor 2 (CXCR2). CONCLUSION Our study revealed that astrocyte-derived CXCL5 aggravated WMI and cognitive decline by inhibiting microglial phagocytosis of myelin debris, suggesting a novel astrocyte-microglia circuit mediated by CXCL5-CXCR2 signaling in chronic cerebral ischemia.
Collapse
Affiliation(s)
- Qian Cao
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, China
| | - Jian Chen
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Zhi Zhang
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, China
| | - Shu Shu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Yi Qian
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Lixuan Yang
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Lushan Xu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, China
| | - Yuxin Zhang
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, China
| | - Xinyu Bao
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Shengnan Xia
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Haiyan Yang
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China
| | - Yun Xu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China.
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China.
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, 210008, China.
| | - Shuwei Qiu
- Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, 210008, China.
| |
Collapse
|
22
|
Kamal F, Morrison C, Dadar M. Investigating the relationship between sleep disturbances and white matter hyperintensities in older adults on the Alzheimer's disease spectrum. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.13.23288544. [PMID: 37131746 PMCID: PMC10153314 DOI: 10.1101/2023.04.13.23288544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Background While studies report that sleep disturbance can have negative effects on brain vasculature, its impact on cerebrovascular disease such as white matter hyperintensities (WMHs) in beta-amyloid positive older adults remains unexplored. Methods Linear regressions, mixed effects models, and mediation analysis examined the crosssectional and longitudinal associations between sleep disturbance, cognition, and WMH burden, and cognition in normal controls (NCs), mild cognitive impairment (MCI), and Alzheimer's disease (AD) at baseline and longitudinally. Results People with AD reported more sleep disturbance than NC and MCI. AD with sleep disturbance had more WMHs than AD without sleep disturbances. Mediation analysis revealed an effect of regional WMH burden on the relationship between sleep disturbance and future cognition. Conclusion These results suggest that WMH burden and sleep disturbance increases from aging to AD. Sleep disturbance decreases cognition through increases in WMH burden. Improved sleep could mitigate the impact of WMH accumulation and cognitive decline.
Collapse
Affiliation(s)
- Farooq Kamal
- Department of Psychiatry, McGill University, Montreal, Quebec, H3A 1A1, Canada
- Douglas Mental Health University Institute, Montreal, Quebec, H4H 1R3, Canada
| | - Cassandra Morrison
- Department of Neurology and Neurosurgery, Faculty of Medicine, McGill University, Montreal, Quebec, H3A 2B4, Canada
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, H3A 2B4, Canada
| | - Mahsa Dadar
- Department of Psychiatry, McGill University, Montreal, Quebec, H3A 1A1, Canada
- Douglas Mental Health University Institute, Montreal, Quebec, H4H 1R3, Canada
| |
Collapse
|
23
|
Al-Chalabi M, Hegde P, Asghar F, Aladamat N, Delcimmuto N, Gharaibeh K, Samara M, Esengul Y, Mahfooz N, Sheikh A. Transient headache and neurological deficits with cerebrospinal fluid lymphocytosis syndrome: A comprehensive systematic review of 93 patients from 57 studies. Cephalalgia 2023; 43:3331024231157694. [PMID: 36856002 DOI: 10.1177/03331024231157694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
BACKGROUND Headache with neurologic deficits and cerebrospinal fluid lymphocytosis, previously also termed pseudomigraine with temporary neurologic symptoms and lymphocytic pleocytosis, is a self-limiting syndrome characterized by moderate to severe headache associated with focal neurological deficits occurring in the context of lymphocytosis in the cerebrospinal fluid. As a consequence of its rarity, data regarding headache with neurologic deficits and cerebrospinal fluid lymphocytosis is sparse. Therefore, we conducted this review to analyze data related to 93 patients of headache with neurologic deficits and cerebrospinal fluid lymphocytosis, to characterize their demographics, clinical manifestations, investigations and treatment options. METHODS We performed a systematic review of cases reported through PubMed and Google scholar database, using Preferred Reporting Items for Systematic Reviews and Meta-Analyses protocol. Keywords used were 'Headache with Neurologic Deficits and cerebrospinal fluid lymphocytosis', 'Headache with neurologic deficits and cerebrospinal fluid lymphocytosis syndrome'. The quality of the included studies was assessed using the Joanna Briggs Institute Critical Appraisal Tool. RESULTS We analyzed a total of 93 cases of headache with neurologic deficits and cerebrospinal fluid lymphocytosis with a mean age of 28.8 years at onset. Seventy patients (75.2%) were adults, while 23 (24.7%) belonged to the pediatric age group. Comparing these groups, mean age at onset was 32.5 years and 14.3 years, respectively. The average duration of follow-up was 11.08 months. Thirty percent of patients experienced relapsing episodes of headache with neurologic deficits and cerebrospinal fluid lymphocytosis symptoms. The most common type of headache reported was unilateral severe throbbing episodic headache. Other associated symptoms included sensory deficit (60%) and motor deficits (54.8%). The least common symptoms were nystagmus and agraphia, which were reported in one patient each. Antiviral agents were a common treatment option in the acute phase (n = 23 patients [23.6%]), while Flunarizine was the most commonly used agent in the chronic setting (n = 3 patients [3.2%]). While most of the patients had normal brain magnetic resonance imaging, 20 patients had magnetic resonance imaging abnormalities, including (but not limited to) non-specific white matter lesions (eight patients) and meningeal enhancement (six patients). The most common electroencephalographic findings included diffuse and focal slowing. The mean cerebrospinal fluid opening-pressure was 240.5 mmH2O. Cerebrospinal fluid protein was elevated in 59 (63.4%) patients, with a mean value of 114 mg/dL. Two patients in our cohort were found to have cerebrospinal fluid oligoclonal bands. CONCLUSION Headache with neurologic deficits and cerebrospinal fluid lymphocytosis tends to affect young individuals with a slight male predominance. Unilateral severe throbbing episodic headache with associated hemi-paresthesia and hemiparesis were the most common symptoms based on our review. Elevated cerebrospinal fluid opening-pressure can be seen in headache with neurologic deficits and cerebrospinal fluid lymphocytosis syndrome. Early recognition of the syndrome is paramount. Antivirals were found to be among the most widely used treatments in the acute setting. Magnetic resonance imaging of the brain is mostly normal. Diffuse and focal slowing were among the most common electroencephalographic findings. Cerebral flow abnormalities on perfusion scans are not uncommon in headache with neurologic deficits and cerebrospinal fluid lymphocytosis. Prospective studies with a larger sample size are needed to validate our findings and guide the clinical care of these patients.
Collapse
Affiliation(s)
| | - Prajwal Hegde
- College of Medicine and Life Sciences, University of Toledo, OH, USA
| | - Fahham Asghar
- Department of Neurology, University of Toledo, Toledo, OH, USA
| | - Nameer Aladamat
- Department of Neurology, University of Toledo, Toledo, OH, USA
| | | | | | - Mohammad Samara
- Department of Neurology, University of Toledo, Toledo, OH, USA
| | - Yasar Esengul
- Department of Neurology, University of Toledo, Toledo, OH, USA
| | - Naeem Mahfooz
- Department of Neurology, University of Toledo, Toledo, OH, USA.,College of Medicine and Life Sciences, University of Toledo, OH, USA
| | - Ajaz Sheikh
- College of Medicine and Life Sciences, University of Toledo, OH, USA
| |
Collapse
|
24
|
Foddis M, Blumenau S, Holtgrewe M, Paquette K, Westra K, Alonso I, Macario MDC, Morgadinho AS, Velon AG, Santo G, Santana I, Mönkäre S, Kuuluvainen L, Schleutker J, Pöyhönen M, Myllykangas L, Pavlovic A, Kostic V, Dobricic V, Lohmann E, Hanagasi H, Santos M, Guven G, Bilgic B, Bras J, Beule D, Dirnagl U, Guerreiro R, Sassi C. TREX1 p.A129fs and p.Y305C variants in a large multi-ethnic cohort of CADASIL-like unrelated patients. Neurobiol Aging 2023; 123:208-215. [PMID: 36586737 DOI: 10.1016/j.neurobiolaging.2022.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 11/16/2022] [Accepted: 11/22/2022] [Indexed: 11/27/2022]
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) and retinal vasculopathy with cerebral leukodystrophy and systemic manifestations (RVCL-S) are the most common forms of rare monogenic early-onset cerebral small vessel disease and share clinical, and, to different extents, neuroradiological and neuropathological features. However, whether CADASIL and RVCL-S overlapping phenotype may be explained by shared genetic risk or causative factors such as TREX1 coding variants remains poorly understood. To investigate this intriguing hypothesis, we used exome sequencing to screen TREX1 protein-coding variability in a large multi-ethnic cohort of 180 early-onset independent familial and apparently sporadic CADASIL-like Caucasian patients from the USA, Portugal, Finland, Serbia and Turkey. We report 2 very rare and likely pathogenic TREX1 mutations: a loss of function mutation (p.Ala129fs) clustering in the catalytic domain, in an apparently sporadic 46-year-old patient from the USA and a missense mutation (p.Tyr305Cys) in the well conserved C-terminal region, in a 57-year-old patient with positive family history from Serbia. In concert with recent findings, our study expands the clinical spectrum of diseases associated with TREX1 mutations.
Collapse
Affiliation(s)
- Marco Foddis
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Sonja Blumenau
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Manuel Holtgrewe
- Berlin Institute of Health, BIH, Core Unit Bioinformatics and Charité - Universitätsmedizin Berlin, Berlin Germany
| | - Kimberly Paquette
- Department for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan
| | - Kaitlyn Westra
- Department for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan
| | - Isabel Alonso
- CGPP and UnIGENe, Instituto Biologia Molecular Celular, Instituto de Investigação e Inovação em Saúde, Porto, Portugal
| | - Maria do Carmo Macario
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Ana Sofia Morgadinho
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Ana Graça Velon
- Department of Neurology, Centro Hospitalar Trás-os-Montes e Alto Douro, Portugal
| | - Gustavo Santo
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal; Centro de Neurociências e Biologia Celular da Universidade de Coimbra, Coimbra, Portugal
| | - Isabel Santana
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal; Faculdade de Medicina da Universidade de Coimbra, Coimbra, Portugal; Centro de Neurociências e Biologia Celular da Universidade de Coimbra, Coimbra, Portugal
| | - Saana Mönkäre
- Department of Medical Genetics, University of Helsinki, Helsinki, Finland; Turku University Hospital, Laboratory Division, Genomics, Department of Medical Genetics, Turku, Finland
| | - Liina Kuuluvainen
- Department of Clinical Genetics, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland; Department of Medical Genetics, University of Helsinki, Helsinki, Finland
| | - Johanna Schleutker
- Turku University Hospital, Laboratory Division, Genomics, Department of Medical Genetics, Turku, Finland
| | - Minna Pöyhönen
- Department of Medical Genetics, University of Helsinki, Helsinki, Finland; Department of Clinical Genetics, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Liisa Myllykangas
- Department of Pathology, University of Helsinki and HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Aleksandra Pavlovic
- Clinic of Neurology, University of Belgrade, Belgrade, Serbia; Faculty for Special Education and Rehabilitation, University of Belgrade, Belgrade
| | - Vladimir Kostic
- Clinic of Neurology, University of Belgrade, Belgrade, Serbia
| | | | - Ebba Lohmann
- Behavioural Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey; Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany; DZNE, German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Hasmet Hanagasi
- Behavioural Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Mariana Santos
- UnIGENe, IBMC-Institute for Molecular and Cell Biology, i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Gamze Guven
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Basar Bilgic
- Behavioural Neurology and Movement Disorders Unit, Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Jose Bras
- Department for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan; Division of Psychiatry and Behavioral Medicine, Michigan State University College of Human Medicine, Grand Rapids, MI, USA
| | - Dieter Beule
- Berlin Institute of Health, BIH, Core Unit Bioinformatics and Charité - Universitätsmedizin Berlin, Berlin Germany
| | - Ulrich Dirnagl
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Rita Guerreiro
- Department for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, Michigan; Division of Psychiatry and Behavioral Medicine, Michigan State University College of Human Medicine, Grand Rapids, MI, USA
| | - Celeste Sassi
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
| |
Collapse
|
25
|
Morrison C, Dadar M, Manera AL, Collins DL. Racial differences in white matter hyperintensity burden in older adults. Neurobiol Aging 2023; 122:112-119. [PMID: 36543016 DOI: 10.1016/j.neurobiolaging.2022.11.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 11/17/2022] [Accepted: 11/19/2022] [Indexed: 11/27/2022]
Abstract
White matter hyperintensities (WMHs) may be one of the earliest pathological changes in aging. Race differences in WMH burden has been conflicting. This study examined if race influences WMHs and whether these differences are influenced by vascular risk factors. Alzheimer's Disease Neuroimaging Initiative participants were included if they had a baseline MRI, diagnosis, and WMH measurements. Ninety-one Blacks and 1937 Whites were included. Using bootstrap re-sampling, 91 Whites were randomly sampled and matched to Blacks based on age, sex, education, and diagnosis 1000 times. Linear models examined the influence of race on baseline WMHs, and change of WMHs over time, with and without vascular factors. Vascular risk factors had higher prevalence in Blacks than Whites. When not including vascular factors, Blacks had greater frontal, parietal, deep, and total WMH burden compared to Whites. There were no race differences in longitudinal progression of WMH accumulation. After controlling for vascular factors, only overall longitudinal parietal WMH group differences remained significant, suggesting that vascular factors contribute to racial group differences observed in WMHs.
Collapse
Affiliation(s)
- Cassandra Morrison
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada.
| | - Mahsa Dadar
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Ana L Manera
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - D Louis Collins
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
26
|
Solé-Guardia G, Custers E, de Lange A, Clijncke E, Geenen B, Gutierrez J, Küsters B, Claassen JAHR, de Leeuw FE, Wiesmann M, Kiliaan AJ. Association between hypertension and neurovascular inflammation in both normal-appearing white matter and white matter hyperintensities. Acta Neuropathol Commun 2023; 11:2. [PMID: 36600303 PMCID: PMC9811756 DOI: 10.1186/s40478-022-01497-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/19/2022] [Indexed: 01/05/2023] Open
Abstract
The major vascular cause of dementia is cerebral small vessel disease (SVD), including white matter hyperintensities (WMH) amongst others. While the underlying pathology of SVD remains unclear, chronic hypertension and neuroinflammation are recognized as important risk factors for SVD and for the conversion of normal-appearing white matter (NAWM) to WMH. Unfortunately, most studies investigating the role of neuroinflammation in WMH relied on peripheral blood markers, e.g., markers for systemic or vascular inflammation, as a proxy for inflammation in the brain itself. However, it is unknown whether such markers accurately capture inflammatory changes within the cerebral white matter. Therefore, we aimed to comprehensively investigate the impact of hypertension on perivascular- and neuroinflammation in both WMH and NAWM. We conducted high field brain magnetic resonance imaging (MRI), followed by (immuno-)histopathological staining of neuroinflammatory markers on human post-mortem brains of elderly people with a history of hypertension (n = 17) and age-matched normotensive individuals (n = 5). MRI images were co-registered to (immuno-)histopathological data including stainings for microglia and astroglia to assess changes in MRI-based WMH at microscopic resolution. Perivascular inflammation was carefully assessed based on the severity of perivascular astrogliosis of the smallest vessels throughout white matter regions. Hypertension was associated with a larger inflammatory response in both WMH and NAWM. Notably, the presence of close-range perivascular inflammation was twice as common among those with hypertension than in controls both in WMH and NAWM, suggesting that neurovascular inflammation is critical in the etiology of WMH. Moreover, a higher degree of microglial activation was related to a higher burden of WMH. Our results indicate that neuro(vascular)inflammation at the level of the brain itself is involved in the etiology of WMH. Future therapeutic strategies focusing on multitarget interventions including antihypertensive treatment as well as neuroinflammation may ameliorate WMH progression.
Collapse
Affiliation(s)
- Gemma Solé-Guardia
- grid.10417.330000 0004 0444 9382Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands
| | - Emma Custers
- grid.10417.330000 0004 0444 9382Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands
| | - Arthur de Lange
- grid.10417.330000 0004 0444 9382Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands
| | - Elyne Clijncke
- grid.10417.330000 0004 0444 9382Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands
| | - Bram Geenen
- grid.10417.330000 0004 0444 9382Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands
| | - Jose Gutierrez
- grid.239585.00000 0001 2285 2675Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University Medical Center, New York, NY USA
| | - Benno Küsters
- grid.10417.330000 0004 0444 9382Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jurgen A. H. R. Claassen
- grid.10417.330000 0004 0444 9382Department of Geriatrics, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Center for Medical Neuroscience, Radboud Alzheimer Center, Nijmegen, The Netherlands
| | - Frank-Erik de Leeuw
- grid.10417.330000 0004 0444 9382Department of Neurology, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Center for Medical Neuroscience, Nijmegen, The Netherlands
| | - Maximilian Wiesmann
- grid.10417.330000 0004 0444 9382Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands
| | - Amanda J. Kiliaan
- grid.10417.330000 0004 0444 9382Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, The Netherlands
| |
Collapse
|
27
|
Zhang X, An H, Chen Y, Shu N. Neurobiological Mechanisms of Cognitive Decline Correlated with Brain Aging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1419:127-146. [PMID: 37418211 DOI: 10.1007/978-981-99-1627-6_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Cognitive decline has emerged as one of the greatest health threats of old age. Meanwhile, aging is the primary risk factor for Alzheimer's disease (AD) and other prevalent neurodegenerative disorders. Developing therapeutic interventions for such conditions demands a greater understanding of the processes underlying normal and pathological brain aging. Despite playing an important role in the pathogenesis and incidence of disease, brain aging has not been well understood at a molecular level. Recent advances in the biology of aging in model organisms, together with molecular- and systems-level studies of the brain, are beginning to shed light on these mechanisms and their potential roles in cognitive decline. This chapter seeks to integrate the knowledge about the neurological mechanisms of age-related cognitive changes that underlie aging.
Collapse
Affiliation(s)
- Xiaxia Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, Faculty of Psychology, Beijing Normal University, Beijing, China
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
| | - Haiting An
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
- Beijing Neurosurgical Institute, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China
| | - Yuan Chen
- State Key Laboratory of Cognitive Neuroscience and Learning, Faculty of Psychology, Beijing Normal University, Beijing, China
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
| | - Ni Shu
- State Key Laboratory of Cognitive Neuroscience and Learning, Faculty of Psychology, Beijing Normal University, Beijing, China.
| |
Collapse
|
28
|
Zou X, Dong Z, Chen X, Yu Q, Yin H, Yi L, Zuo H, Xu J, Du X, Han Y, Zou D, Peng J, Cheng O. White matter hyperintensities burden in the frontal regions is positively correlated to the freezing of gait in Parkinson's disease. Front Aging Neurosci 2023; 15:1156648. [PMID: 37181626 PMCID: PMC10172504 DOI: 10.3389/fnagi.2023.1156648] [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: 02/07/2023] [Accepted: 04/11/2023] [Indexed: 05/16/2023] Open
Abstract
Objective Previous studies have reported that white matter hyperintensities (WMHs) are associated with freezing of gait (FOG), but it is not clear whether their distribution areas have correlations with FOG in Parkinson's disease (PD) and the potential influencing factors about WMHs. Methods Two hundred and forty-six patients with PD who underwent brain MRI were included. Participants were divided into PD with FOG (n = 111) and PD without FOG (n = 135) groups. Scheltens score was used to assess the WMHs burden in the areas of deep white matter hyperintensities (DWMHs), periventricular hyperintensities (PVHs), basal ganglia hyperintensities (BGHs), and infratentorial foci of hyperintensities (ITF). Whole brain WMHs volume was evaluated by automatic segmentation. Binary logistic regression was used to evaluate relationships between WMHs and FOG. The common cerebrovascular risk factors that may affect WMHs were evaluated by mediation analysis. Results There were no statistical differences between PD with and without FOG groups in whole brain WMHs volume, total Scheltens score, BGHs, and ITF. Binary logistic regression showed that the total scores of DWMHs (OR = 1.094; 95% CI, 1.001, 1.195; p = 0.047), sum scores of PVHs and DWMHs (OR = 1.080; 95% CI, 1.003, 1.164; p = 0.042), especially the DWMHs in frontal (OR = 1.263; 95% CI, 1.060, 1.505 p = 0.009), and PVHs in frontal caps (OR = 2.699; 95% CI, 1.337, 5.450; p = 0.006) were associated with FOG. Age, hypertension, and serum alkaline phosphatase (ALP) are positively correlated with scores of DWMHs in frontal and PVHs in frontal caps. Conclusion These results indicate that WMHs distribution areas especially in the frontal of DWMHs and PVHs play a role in PD patients with FOG.
Collapse
Affiliation(s)
- Xiaoya Zou
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhaoying Dong
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xinwei Chen
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qian Yu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Huimei Yin
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Yi
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongzhou Zuo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiaman Xu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xinyi Du
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yu Han
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dezhi Zou
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Juan Peng
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- *Correspondence: Juan Peng,
| | - Oumei Cheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Oumei Cheng,
| |
Collapse
|
29
|
Zhu R, Li Y, Chen L, Wang Y, Cai G, Chen X, Ye Q, Chen Y. Total Burden of Cerebral Small Vessel Disease on MRI May Predict Cognitive Impairment in Parkinson’s Disease. J Clin Med 2022; 11:jcm11185381. [PMID: 36143028 PMCID: PMC9501874 DOI: 10.3390/jcm11185381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/17/2022] [Accepted: 08/25/2022] [Indexed: 11/25/2022] Open
Abstract
(1) Objective: to investigate the association between the total burden of cerebral small vessel disease (CSVD) and cognitive function in Parkinson’s disease (PD). (2) Methods: this retrospective study compared clinical and neuroimaging characteristics of 122 PD patients to determine the association between cognitive decline and total burden of CSVD in PD. All patients underwent brain MRI examinations, and their total CSVD burden scores were evaluated by silent lacunar infarction (SLI), cerebral microbleeds (CMB), white matter hyperintensities (WMH), and enlarged perivascular spaces (EPVS). The cognitive function was assessed by administering Mini-Mental State Examination (MMSE). Receiver-operating characteristic (ROC) curve and the area under the ROC curve (AUC) were performed to quantify the accuracy of the total burden of CSVD and PVH in discriminating PD patients with or without cognitive impairment. (3) Results: the PD patients with cognitive impairment had a significantly higher SLI, CMB, periventricular hyperintensities (PVH), deep white matter hyperintensities (DWMH), enlarged perivascular spaces of basal ganglia (BG-EPVS), and the total CSVD score compared with no cognitive impairment. Total CSVD score and MMSE had a significant negative correlation (r = −0. 483). Furthermore, total burden of CSVD and PVH were the independent risk factors of cognitive impairment in PD, and their good accuracy in discriminating PD patients with cognitive impairment from those with no cognitive impairment was confirmed by the results of ROC curves. (4) Conclusions: total burden of CSVD tightly linked to cognitive impairment in PD patients. The total burden of CSVD or PVH may predict the cognitive impairment in PD.
Collapse
Affiliation(s)
- Ruihan Zhu
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Department of Neurology, The Second Affiliated Hospital, Xiamen Medical College, Xiamen 361021, China
- Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou 350004, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou 350004, China
| | - Yunjing Li
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou 350004, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou 350004, China
| | - Lina Chen
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou 350004, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou 350004, China
| | - Yingqing Wang
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou 350004, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou 350004, China
| | - Guoen Cai
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou 350004, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou 350004, China
| | - Xiaochun Chen
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou 350004, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou 350004, China
| | - Qinyong Ye
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou 350004, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou 350004, China
- Correspondence: (Q.Y.); (Y.C.)
| | - Ying Chen
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou 350004, China
- Institute of Clinical Neurology, Fujian Medical University, Fuzhou 350004, China
- Correspondence: (Q.Y.); (Y.C.)
| |
Collapse
|
30
|
Dose-response association between plasma homocysteine and white matter lesions in patients with hypertension: a case-control study. Hypertens Res 2022; 45:1794-1801. [PMID: 35999281 DOI: 10.1038/s41440-022-00999-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 06/05/2022] [Accepted: 07/07/2022] [Indexed: 11/08/2022]
Abstract
White matter lesions (WMLs) are common MRI changes that are indicative of cerebral small vessel disease (CSVD). Elevated plasma homocysteine (Hcy) levels are related to an increased risk of vascular disease. We aimed to analyze the relationship between Hcy levels and WMLs in patients with hypertension. A total of 1961 patients with WMLs and 15,463 patients without WMLs were matched at a 1:1 ratio by age and sex. Hyperhomocysteinemia (HHcy) was defined as an abnormally high level (>15 µmol/l) of Hcy in a plasma sample. In total, 1888 (WML group) and 1888 (No-WMLs group) patients were enrolled, with 51.6% of the sample being male and a mean age of 63 years. Multivariate logistic regression analysis showed a significant association between a higher level of plasma Hcy and a higher prevalence of WMLs (OR 1.03 95% CI, 1.02-1.04) when the Hcy level was used as a continuous variable. Patients with Hcy levels of 15-20 µmol/l (OR 1.54, 95% CI 1.31-1.81) and >20 µmol/l (OR 1.51, 95% CI 1.26-1.82) also had a significantly higher risk of WMLs than patients with Hcy levels <15 µmol/l. Multivariable-adjusted spline regression models showed that the risk of WMLs started to increase only in patients with Hcy levels above 13.85 µmol/l (P < 0.001). In subgroup analyses of WMLs, there was no significant interaction between the Hcy group and subgroup heterogeneity for the prevalence of WMLs (P > 0.05). Our study found a dose-response association between plasma homocysteine levels, especially a Hcy level >13.85 µmol/l, and the prevalence of WMLs, implying that lowering Hcy levels might be a target for prevention.
Collapse
|
31
|
Chi NF, Chung CP, Cheng HM, Liu CH, Lin CJ, Hsu LC, Tang SC, Lee JT, Po HL, Jeng JS, Wang TD, Lee IH. 2021 Taiwan Stroke Society Guidelines of blood pressure control for ischemic stroke prevention. J Chin Med Assoc 2022; 85:651-664. [PMID: 35507097 DOI: 10.1097/jcma.0000000000000738] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Since the publication of the 2015 Taiwan Stroke Society Blood Pressure for Treatment and Prevention of Stroke Guideline (2015 TSS BP Guideline), several new clinical studies have addressed whether a stricter blood pressure (BP) target would be effective for stroke prevention. METHODS TSS guideline consensus group provides recommendations on BP targets for stroke prevention based on updated evidences. RESULTS The present guideline covers five topics: (1) diagnosis of hypertension; (2) BP control and primary prevention of ischemic stroke; (3) BP control and secondary prevention of ischemic stroke; (4) BP control and secondary prevention of large artery atherosclerosis ischemic stroke; and (5) BP control and secondary prevention of small vessel occlusion ischemic stroke. CONCLUSION The BP target for most stroke patients with hypertension is <130/80 mm Hg.
Collapse
Affiliation(s)
- Nai-Fang Chi
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Neurology in School of Medicine, National Yang Ming Chiao Tung University College of Medicine, Taipei, Taiwan, ROC
| | - Chih-Ping Chung
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Neurology in School of Medicine, National Yang Ming Chiao Tung University College of Medicine, Taipei, Taiwan, ROC
| | - Hao-Ming Cheng
- Center for Evidence-based Medicine & Division of Cardiology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Medicine, National Yang Ming Chiao Tung University College of Medicine, Taipei, Taiwan, ROC
- Institute of Public Health, National Yang Ming Chiao Tung University College of Medicine, Taipei, Taiwan, ROC
| | - Chi-Hung Liu
- Department of Neurology, Linkou Chang Gung Memorial Hospital, Taoyuan City, Taiwan, ROC
| | - Chun-Jen Lin
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Neurology in School of Medicine, National Yang Ming Chiao Tung University College of Medicine, Taipei, Taiwan, ROC
| | - Li-Chi Hsu
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Neurology in School of Medicine, National Yang Ming Chiao Tung University College of Medicine, Taipei, Taiwan, ROC
| | - Sung-Chun Tang
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan, ROC
| | - Jiunn-Tay Lee
- Department of Neurology, Tri-Service General Hospital, Taipei, Taiwan, ROC
| | - Helen L Po
- Department of Neurology, Mackay Memorial Hospital, Taipei, Taiwan, ROC
| | - Jiann-Shing Jeng
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan, ROC
| | - Tzung-Dau Wang
- Department of Cardiology, National Taiwan University, Taipei, Taiwan, ROC
| | - I-Hui Lee
- Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Neurology in School of Medicine, National Yang Ming Chiao Tung University College of Medicine, Taipei, Taiwan, ROC
| |
Collapse
|
32
|
Ye Q, Zhu H, Chen H, Liu R, Huang L, Chen H, Cheng Y, Qin R, Shao P, Xu H, Ma J, Xu Y. Effects of cognitive reserve proxies on cognitive function and frontoparietal control network in subjects with white matter hyperintensities: A cross-sectional functional magnetic resonance imaging study. CNS Neurosci Ther 2022; 28:932-941. [PMID: 35274485 PMCID: PMC9062549 DOI: 10.1111/cns.13824] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 02/16/2022] [Accepted: 02/21/2022] [Indexed: 12/01/2022] Open
Abstract
AIMS This study aimed to analyze the potential association between cognition reserve (CR) components, including education, working activity, and leisure time activity, and cognitive function in subjects with white matter hyperintensities (WMH). The study also explored the role of the frontoparietal control network (FPCN) in such association. METHODS White matter hyperintensities subjects with and without cognitive impairment (CI) were evaluated with multimodal magnetic resonance imaging, neuropsychological testing, and CR survey. FPCN patterns were assessed with dorsolateral prefrontal cortex seed-based functional connectivity analysis. RESULTS Education was positively associated with cognitive function in WMH subjects with or without CI, whereas working activity and leisure time activity were positively associated with cognitive function only in those without CI. Similarly, education was associated with bilateral FPCN in both WMH groups, whereas working activity and leisure time activity were associated with bilateral FPCN mainly in the group without CI. Furthermore, FPCN partially mediated the association between education and cognitive function in both WMH groups. CONCLUSION Education showed a positive impact on cognitive function in WMH subjects regardless of their cognitive status, whereas working activity and leisure time activity exhibited beneficial effects only in those without CI. The FPCN mediated the beneficial effect of education on cognitive function.
Collapse
|
33
|
Wang J, Zhou Y, He Y, Li Q, Zhang W, Luo Z, Xue R, Lou M. Impact of different white matter hyperintensities patterns on cognition: A cross-sectional and longitudinal study. Neuroimage Clin 2022; 34:102978. [PMID: 35255417 PMCID: PMC8897653 DOI: 10.1016/j.nicl.2022.102978] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 02/27/2022] [Accepted: 02/28/2022] [Indexed: 11/03/2022]
Abstract
OBJECTIVES White matter hyperintensities (WMH) are highly prevalent in older adults and considered to be a contributor to cognition impairment. However, the strategic WMH lesion distribution related to cognitive impairment is still debated. The aim of this study was to characterize the spatial patterns of WMH associated with cognitive impairment and explore its risk factors. METHODS We retrospectively analyzed patients who underwent T2 fluid attenuated inversion recovery (FLAIR) and mini-mental state examination (MMSE) in two centers. WHM was classified into four patterns based on T2 FLAIR as follows: (1) multiple subcortical spots (multi-spots); (2) peri-basal ganglia (peri-BG); (3) anterior subcortical patches (anterior SC patches); and (4) posterior subcortical patches (posterior SC patches). We cross-sectionally and longitudinally estimated associations between different WMH patterns and all-cause dementia and cognitive decline. Multivariable logistic regression analysis was followed to identify risk factors of WMH patterns related to cognitive impairment. RESULTS A total of 442 patients with WMH were enrolled, with average age of 71.6 ± 11.3 years, and MMSE score of 24.1 ± 5.4. Among them, 281 (63.6%), 66 (14.9%), 163 (36.9%) and 197 (44.6%) patients presented multi-spots, peri-BG, anterior SC patches and posterior SC patches, respectively. Patients with anterior SC patches were more likely to have all-cause dementia in cross-sectional study (OR 2.002; 95% CI 1.098-3.649; p = 0.024), and have cognitive decline in longitudinal analysis (OR 3.029; 95% CI 1.270-7.223; p = 0.012). Four patterns of WMH referred to different cognitive domains, and anterior SC patches had the most significant and extensive impact on cognition after Bonferroni multiple comparison correction (all p < 0.0125). In addition, older age (OR 1.054; 95% CI 1.027-1.082; p < 0.001), hypertension (OR 1.956; 95% CI 1.145-3.341; p = 0.014), higher percentage of neutrophils (OR 1.046; 95% CI 1.014-1.080; p = 0.005) and lower concentration of hemoglobin (OR 0.983; 95% CI 0.967-1.000; p = 0.044) were risk factors for the presence of anterior SC patches. CONCLUSIONS Different patterns of subcortical leukoaraiosis visually identified on MRI might have different impacts on cognitive impairment. Further studies should be undertaken to validate this simple visual classification of WMH in different population.
Collapse
Affiliation(s)
- Junjun Wang
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine. 88# Jiefang Road, Hangzhou, China; Department of Neurology, Zhejiang Hospital, #12 Lingyin Road, Hangzhou, China
| | - Ying Zhou
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine. 88# Jiefang Road, Hangzhou, China
| | - Yaode He
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine. 88# Jiefang Road, Hangzhou, China
| | - Qingqing Li
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine. 88# Jiefang Road, Hangzhou, China
| | - Wenhua Zhang
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine. 88# Jiefang Road, Hangzhou, China
| | - Zhongyu Luo
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine. 88# Jiefang Road, Hangzhou, China
| | - Rui Xue
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine. 88# Jiefang Road, Hangzhou, China
| | - Min Lou
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine. 88# Jiefang Road, Hangzhou, China.
| |
Collapse
|
34
|
Hicks CW, Wang D, Schneider ALC, Johansen MC, Gottesman RF, Matsushita K, Coresh J, Windham BG, Selvin E. Associations of Peripheral Neuropathy Defined by Monofilament Insensitivity with Mild Cognitive Impairment and Dementia in Older Adults. Dement Geriatr Cogn Disord 2022; 51:150-158. [PMID: 35344962 PMCID: PMC9167718 DOI: 10.1159/000523762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 02/17/2022] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION The aim of this study was to assess the association of peripheral neuropathy (PN) as defined by monofilament insensitivity with mild cognitive impairment (MCI) and dementia in older adults with and without diabetes. METHODS We conducted a cross-sectional analysis of 3,362 Black and White participants in the Atherosclerosis Risk in Communities Neurocognitive Study (ARIC-NCS) who underwent monofilament testing at visit 6 (2016-2017, age 71-94 years). Participants' cognitive status was classified by an adjudication committee as cognitively normal, MCI, or dementia after completing a comprehensive battery of neurocognitive assessments. We used logistic regression to evaluate the association of PN with MCI or dementia overall and stratified by diabetes status after adjusting for traditional dementia risk factors. We also compared age-adjusted brain MRI measures among a subset (N = 1,095) of participants with versus without PN. RESULTS Overall, the prevalence of MCI (21.9% vs. 16.7%) and dementia (7.8% vs. 3.9%) were higher among participants with versus without PN (both p < 0.05). After adjustment, PN was positively associated with MCI or dementia in the overall study population (OR 1.45, 95% CI 1.23, 1.73). Results were similar by diabetes status (diabetes: OR 1.38, 95% CI 1.03-1.87; no diabetes: OR 1.48, 95% CI 1.20-1.83; p-for-interaction = 0.46). Age-adjusted total and lobar brain volumes were significantly lower in participants with versus without PN (both, p < 0.05). DISCUSSION/CONCLUSIONS PN as defined by monofilament insensitivity was associated with cognitive status independent of vascular risk factors and regardless of diabetes status. Our findings support a connection between PN and cognitive impairment, even in the absence of diabetes.
Collapse
Affiliation(s)
- Caitlin W Hicks
- Division of Vascular Surgery and Endovascular Therapy, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA,
| | - Dan Wang
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Andrea L C Schneider
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michelle C Johansen
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rebecca F Gottesman
- National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, Maryland, USA
| | - Kunihiro Matsushita
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - B Gwen Windham
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Elizabeth Selvin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
35
|
Jeppsson A, Bjerke M, Hellström P, Blennow K, Zetterberg H, Kettunen P, Wikkelsø C, Wallin A, Tullberg M. Shared CSF Biomarker Profile in Idiopathic Normal Pressure Hydrocephalus and Subcortical Small Vessel Disease. Front Neurol 2022; 13:839307. [PMID: 35309577 PMCID: PMC8927666 DOI: 10.3389/fneur.2022.839307] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 01/27/2022] [Indexed: 11/13/2022] Open
Abstract
IntroductionIn this study, we examine similarities and differences between 52 patients with idiopathic normal pressure hydrocephalus (iNPH) and 17 patients with subcortical small vessel disease (SSVD), in comparison to 28 healthy controls (HCs) by a panel of cerebrospinal fluid (CSF) biomarkers.MethodsWe analyzed soluble amyloid precursor protein alpha (sAPPα) and beta (sAPPβ), Aβ isoforms −38, −40, and −42, neurofilament light protein (NFL), glial fibrillary acidic protein (GFAP), myelin basic protein (MBP), matrix metalloproteinases (MMP −1, −2, −3, −9, and −10), and tissue inhibitors of metalloproteinase 1 (TIMP1). Radiological signs of white matter damage were scored using the age-related white matter changes (ARWMC) scale.ResultsAll amyloid fragments were reduced in iNPH and SSVD (p < 0.05), although more in iNPH than in SSVD in comparison to HC. iNPH and SSVD showed comparable elevations of NFL, MBP, and GFAP (p < 0.05). MMPs were similar in all three groups except for MMP-10, which was increased in iNPH and SSVD. Patients with iNPH had larger ventricles and fewer WMCs than patients with SSVD.ConclusionThe results indicate that patients with iNPH and SSVD share common features of subcortical neuronal degeneration, demyelination, and astroglial response. The reduction in all APP-derived proteins characterizing iNPH patients is also present, indicating that SSVD encompasses similar pathophysiological phenomena as iNPH.
Collapse
Affiliation(s)
- Anna Jeppsson
- Hydrocephalus Research Unit, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- *Correspondence: Anna Jeppsson
| | - Maria Bjerke
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Department of Clinical Chemistry, Universitair Ziekenhuis Brussel and Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
- Department of Biomedical Sciences, Institute Born-Bunge, University of Antwerp, Antwerp, Belgium
| | - Per Hellström
- Hydrocephalus Research Unit, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, Hong Kong SAR, China
| | - Petronella Kettunen
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Carsten Wikkelsø
- Hydrocephalus Research Unit, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anders Wallin
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Mölndal, Sweden
| | - Mats Tullberg
- Hydrocephalus Research Unit, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
36
|
Filho ABV, Puccioni-Sohler M. Cardiovascular risk factors and brain white matter lesions in HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). J Neurovirol 2022; 28:168-171. [PMID: 35235153 DOI: 10.1007/s13365-022-01063-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/19/2021] [Accepted: 02/08/2022] [Indexed: 11/25/2022]
Abstract
The origin of brain white matter lesion found in HTLV-1-associated myelopathy (HAM/TSP) remains undefined. We investigated the association between white matter lesions in HAM/TSP with cardiovascular risk factors. The group of 40 patients with HAM/TSP included 60% females and mean age of 58.6 ± 8 years old. The probability of 10-year cardiovascular disease was low in 53%, moderate in 38%, and high in 10% of the patients. There was no difference between the cardiovascular risk factors in HAM/TSP patients with and without brain lesions (p > 0.05). Our data suggest that the brain white matter abnormalities are not associated to increased cardiovascular risk in HAM/TSP.
Collapse
Affiliation(s)
- Andre Bertola Vanzan Filho
- Escola de Medicina e Cirurgia, Hospital Universitário Gaffrée e Guinle, Universidade Federal do Estado do Rio de Janeiro, Rua Mariz e Barros 775, Tijuca, Rio de Janeiro, 20270-004, Brazil
| | - Marzia Puccioni-Sohler
- Escola de Medicina e Cirurgia, Hospital Universitário Gaffrée e Guinle, Universidade Federal do Estado do Rio de Janeiro, Rua Mariz e Barros 775, Tijuca, Rio de Janeiro, 20270-004, Brazil.
| |
Collapse
|
37
|
Leung DYL, Tham CC. Normal-tension glaucoma: Current concepts and approaches-A review. Clin Exp Ophthalmol 2022; 50:247-259. [PMID: 35040248 DOI: 10.1111/ceo.14043] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/09/2022] [Accepted: 01/11/2022] [Indexed: 12/19/2022]
Abstract
Normal tension glaucoma (NTG) has remained a challenging disease. We review, from an epidemiological perspective, why we should redefine normality, act earlier at lower pre-treatment intraocular pressure (IOP) level, and the role of ocular perfusion pressures, noting that perfusion is affected by defective vascular bed autoregulation and endothelial dysfunction. The correlation of silent cerebral infarcts (SCI) and NTG may indicate that NTG belongs to a wider spectrum of small vessel diseases (SVD), with its main pathology being also on vascular endothelium. Epidemiological studies also suggested that vascular geometry, such as fractal dimension, may affect perfusion efficiency, occurrence of SCI, SVD and glaucoma. Artificial intelligence with deep learning, may help predicting NTG progression from vascular geometry. Finally, we review latest evidence on the role of minimally-invasive glaucoma surgery, lasers, and newer drugs. We conclude that IOP is not the only modifiable risk factors as, many vascular risk factors are readily modifiable.
Collapse
Affiliation(s)
- Dexter Y L Leung
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Kowloon, Hong Kong SAR, China
- Department of Ophthalmology, Hong Kong Sanatorium & Hospital, Happy Valley, Hong Kong SAR, China
| | - Clement C Tham
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Kowloon, Hong Kong SAR, China
- Hong Kong Eye Hospital, Kowloon, Hong Kong SAR, China
- Lam Kin Chung . Jet King-Shing Ho Glaucoma Treatment and Research Centre, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
38
|
Öchsner M, Mak E, Ersche KD. Detecting Small Vessel Pathology in Cocaine Use Disorder. Front Neurosci 2022; 15:827329. [PMID: 35221893 PMCID: PMC8867820 DOI: 10.3389/fnins.2021.827329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 12/20/2021] [Indexed: 11/13/2022] Open
Abstract
BackgroundCocaine use is associated with an increased risk of cerebrovascular accidents. Small vessel pathology has been linked to the risk of stroke in cocaine users, but can be challenging to detect on conventional magnetic resonance (MR) scans. Fluid-attenuated inversion recovery (FLAIR) scans permit better resolution of small vessel lesions.ObjectivesFLAIR scans are currently only acquired based on the subjective judgement of abnormalities on MR scans at face value. We sought to evaluate this practice and the added value of FLAIR scans for patients with cocaine use disorder (CUD), by comparing microbleeds detected by MR and FLAIR scans. We hypothesised that microbleeds are more pronounced in CUD patients, particularly so in participants who had been selected for a FLAIR scan by radiographers.MethodsSixty-four patients with CUD and 60 control participants underwent a brain scan. The MR of 20 CUD patients and 16 control participants showed indicators of cerebral infarction at face value and were followed up by a FLAIR scan. We determined the volume of microbleeds in both MR and FLAIR scans and examined associations with various risk factors.ResultsWhile MR lesion volumes were significantly increased in CUD patients, no significant differences in lesion volume were found in the subgroup of individuals who received a FLAIR.ConclusionThe current practice of subjectively evaluating MR scans as a basis for the follow-up FLAIR scans to detect vascular pathology may miss vulnerable individuals. Hence, FLAIR scans should be included as a routine part of research studies.
Collapse
Affiliation(s)
- Marco Öchsner
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Elijah Mak
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Karen D. Ersche
- Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
- Department of Systems Neuroscience, University Hospital Hamburg-Eppendorf, Hamburg, Germany
- *Correspondence: Karen D. Ersche,
| |
Collapse
|
39
|
Zhang D, He M, He Q, Li Z. Blood Pressure Rhythm and Blood Pressure Variability as Risk Factors for White Matter Lesions: A Cross-Sectional Study. MEDICAL SCIENCE MONITOR : INTERNATIONAL MEDICAL JOURNAL OF EXPERIMENTAL AND CLINICAL RESEARCH 2022; 28:e933880. [PMID: 35115481 PMCID: PMC8822846 DOI: 10.12659/msm.933880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background White matter lesions are common in the elderly. The aim of this study was to explore the correlation between blood pressure rhythm and blood pressure variability with white matter lesions. Material/Methods A total of 144 subjects aged 40 to 80 years underwent MRI scanning to assess the degree of white matter lesions using the Fazekas scale. The regional cerebral blood flow was detected by brain perfusion imaging, and an ambulatory blood pressure monitor was used to measure the circadian blood pressure rhythm. Odds ratio and the 95% confidence interval was computed using logistics regression analysis. The relationship between various factors and blood pressure was calculated by curve simulation. Results With the increase of white matter lesions, the regional cerebral blood flow at the lesion decreased gradually. Systolic blood pressure day/night difference ratio (OR=0.815, 95% CI 0.729–0.910), diastolic blood pressure day/night difference ratio (OR=0.895, 95% CI 0.831–0.964), systolic blood pressure coefficient of variation (OR=1.589, 95% CI 1.273–1.983), and diastolic blood pressure coefficient of variation (OR=1.363, 95% CI 1.150–1.616) were significantly associated with Fazekas score (P<0.05 for all). Conclusions Greater blood pressure variability and blood pressure rhythm disorders were associated with lower regional cerebral blood flow in patients with white matter lesions.
Collapse
Affiliation(s)
- Dong Zhang
- Department of Neurology, The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China (mainland)
| | - Mingli He
- Department of Neurology, The Lianyungang Hospital Affiliated to Xuzhou Medical University, Lianyungang, Jiangsu, China (mainland)
| | - Qing He
- Department of Neurology, The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China (mainland)
| | - Zeheng Li
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China (mainland)
| |
Collapse
|
40
|
Dobrynina L, Gadzhieva Z, Shamtieva K, Kremneva E, Filatov A, Bitsieva E, Mirokova E, Krotenkova M. Predictors and integrative index of severity of cognitive disorders in cerebral microangiopathy. Zh Nevrol Psikhiatr Im S S Korsakova 2022; 122:52-60. [DOI: 10.17116/jnevro202212204152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
41
|
Kuo YM, Lee YH. Epoxyeicosatrienoic acids and soluble epoxide hydrolase in physiology and diseases of the central nervous system. CHINESE J PHYSIOL 2022; 65:1-11. [PMID: 35229747 DOI: 10.4103/cjp.cjp_80_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Epoxyeicosatrienoic acids (EETs) are fatty acid signaling molecules synthesized by cytochrome P450 epoxygenases from arachidonic acid. The biological activity of EETs is terminated when being metabolized by soluble epoxide hydrolase (sEH), a process that serves as a key regulator of tissue EETs levels. EETs act through several signaling pathways to mediate various beneficial effects, including anti-inflammation, anti-apoptosis, and anti-oxidation with relieve of endoplasmic reticulum stress, thereby sEH has become a potential therapeutic target in cardiovascular disease and cancer therapy. Enzymes for EET biosynthesis and metabolism are both widely detected in both neuron and glial cells in the central nervous system (CNS). Recent studies discovered that astrocyte-derived EETs not only mediate neurovascular coupling and neuronal excitability by maintaining glutamate homeostasis but also glia-dependent neuroprotection. Genetic ablation as well as pharmacologic inhibition of sEH has greatly helped to elucidate the physiologic actions of EETs, and maintaining or elevating brain EETs level has been demonstrated beneficial effects in CNS disease models. Here, we review the literature regarding the studies on the bioactivity of EETs and their metabolic enzyme sEH with special attention paid to their action mechanisms in the CNS, including their modulation of neuronal activity, attenuation of neuroinflammation, regulation of cerebral blood flow, and improvement of neuronal and glial cells survival. We further reviewed the recent advance on the potential application of sEH inhibition for treating cerebrovascular disease, epilepsy, and pain disorder.
Collapse
Affiliation(s)
- Yi-Min Kuo
- Department of Anesthesiology, Taipei Veterans General Hospital; Department of Anesthesiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Hsuan Lee
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University; Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
42
|
Dadar M, Manera AL, Ducharme S, Collins DL. White matter hyperintensities are associated with grey matter atrophy and cognitive decline in Alzheimer's disease and frontotemporal dementia. Neurobiol Aging 2021; 111:54-63. [PMID: 34968832 DOI: 10.1016/j.neurobiolaging.2021.11.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/21/2021] [Accepted: 11/26/2021] [Indexed: 01/18/2023]
Abstract
White matter hyperintensities (WMHs) are commonly assumed to represent non-specific cerebrovascular disease comorbid to neurodegenerative processes, rather than playing a synergistic role. We compared the impact of WMHs on grey matter (GM) atrophy and cognition in normal aging (n = 571), mild cognitive impairment (MCI, n = 551), Alzheimer's dementia (AD, n = 212), fronto-temporal dementia (FTD, n = 125), and Parkinson's disease (PD, n = 271). Longitudinal data were obtained from ADNI, FTLDNI, and PPMI datasets. Mixed-effects models were used to compare WMHs and GM atrophy between patients and controls and assess the impact of WMHs on GM atrophy and cognition. MCI, AD, and FTD patients had significantly higher WMH loads than controls. WMHs were related to GM atrophy in insular and parieto-occipital regions in MCI/AD, and frontal regions and basal ganglia in FTD. In addition, WMHs contributed to more severe cognitive deficits in AD and FTD compared to controls, whereas their impact in MCI and PD was not significantly different from controls. These results suggest potential synergistic effects between WMHs and proteinopathies in the neurodegenerative process in MCI, AD and FTD.
Collapse
Affiliation(s)
- Mahsa Dadar
- NeuroImaging and Surgical Tools Laboratory, Montreal Neurological Institute, McGill University, Montreal, QC, Canada; McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada.
| | - Ana Laura Manera
- NeuroImaging and Surgical Tools Laboratory, Montreal Neurological Institute, McGill University, Montreal, QC, Canada; McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Simon Ducharme
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada; Department of Psychiatry, Douglas Mental Health University Institute and Douglas Research Centre, McGill University, Montreal, QC, Canada
| | - D Louis Collins
- NeuroImaging and Surgical Tools Laboratory, Montreal Neurological Institute, McGill University, Montreal, QC, Canada; McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
43
|
Liao FF, Lin G, Chen X, Chen L, Zheng W, Raghow R, Zhou FM, Shih AY, Tan XL. Endothelial Nitric Oxide Synthase-Deficient Mice: A Model of Spontaneous Cerebral Small-Vessel Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1932-1945. [PMID: 33711310 PMCID: PMC8647425 DOI: 10.1016/j.ajpath.2021.02.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 02/04/2021] [Accepted: 02/24/2021] [Indexed: 02/08/2023]
Abstract
Age-related cerebral small-vessel disease (CSVD) is a major cause of stroke and dementia. Despite a widespread acceptance of small-vessel arteriopathy, lacunar infarction, diffuse white matter injury, and cognitive impairment as four cardinal features of CSVD, a unifying pathologic mechanism of CSVD remains elusive. Herein, we introduce partial endothelial nitric oxide synthase (eNOS)-deficient mice as a model of age-dependent, spontaneous CSVD. These mice developed cerebral hypoperfusion and blood-brain barrier leakage at a young age, which progressively worsened with advanced age. Their brains exhibited elevated oxidative stress, astrogliosis, cerebral amyloid angiopathy, microbleeds, microinfarction, and white matter pathology. Partial eNOS-deficient mice developed gait disturbances at middle age, and hippocampus-dependent memory deficits at older ages. These mice also showed enhanced expression of bone morphogenetic protein 4 (BMP4) in brain pericytes before myelin loss and white matter pathology. Because BMP4 signaling not only promotes astrogliogenesis but also blocks oligodendrocyte differentiation, we posit that paracrine actions of BMP4, localized within the neurovascular unit, promote white matter disorganization and neurodegeneration. These observations point to BMP4 signaling pathway in the aging brain vasculature as a potential therapeutic target. Finally, because studies in partial eNOS-deficient mice corroborated recent clinical evidence that blood-brain barrier disruption is a primary cause of white matter pathology, the mechanism of impaired nitric oxide signaling-mediated CSVD warrants further investigation.
Collapse
Affiliation(s)
- Francesca-Fang Liao
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee.
| | - Geng Lin
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee; Department of Histology and Embryology, Basic Medical University, China Medical University, Shenyang, China
| | - Xingyong Chen
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee; Department of Neurology, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou, China
| | - Ling Chen
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee; Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Wei Zheng
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee; Department of Histology and Embryology, Basic Medical University, China Medical University, Shenyang, China
| | - Rajendra Raghow
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee
| | - Fu-Ming Zhou
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee
| | - Andy Y Shih
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington
| | - Xing-Lin Tan
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee; Department of Neurology, Nanhai Hospital of Southern Medical University, Foshan, China
| |
Collapse
|
44
|
MacDonald ME, Pike GB. MRI of healthy brain aging: A review. NMR IN BIOMEDICINE 2021; 34:e4564. [PMID: 34096114 DOI: 10.1002/nbm.4564] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 05/08/2021] [Accepted: 05/11/2021] [Indexed: 06/12/2023]
Abstract
We present a review of the characterization of healthy brain aging using MRI with an emphasis on morphology, lesions, and quantitative MR parameters. A scope review found 6612 articles encompassing the keywords "Brain Aging" and "Magnetic Resonance"; papers involving functional MRI or not involving imaging of healthy human brain aging were discarded, leaving 2246 articles. We first consider some of the biogerontological mechanisms of aging, and the consequences of aging in terms of cognition and onset of disease. Morphological changes with aging are reviewed for the whole brain, cerebral cortex, white matter, subcortical gray matter, and other individual structures. In general, volume and cortical thickness decline with age, beginning in mid-life. Prevalent silent lesions such as white matter hyperintensities, microbleeds, and lacunar infarcts are also observed with increasing frequency. The literature regarding quantitative MR parameter changes includes T1 , T2 , T2 *, magnetic susceptibility, spectroscopy, magnetization transfer, diffusion, and blood flow. We summarize the findings on how each of these parameters varies with aging. Finally, we examine how the aforementioned techniques have been used for age prediction. While relatively large in scope, we present a comprehensive review that should provide the reader with sound understanding of what MRI has been able to tell us about how the healthy brain ages.
Collapse
Affiliation(s)
- M Ethan MacDonald
- Department of Electrical and Software Engineering, University of Calgary, Calgary, Alberta, Canada
- Departments of Radiology and Clinical Neuroscience, University of Calgary, Calgary, Alberta, Canada
- Healthy Brain Aging Laboratory, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - G Bruce Pike
- Departments of Radiology and Clinical Neuroscience, University of Calgary, Calgary, Alberta, Canada
- Healthy Brain Aging Laboratory, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
45
|
Yildirim F, Foddis M, Blumenau S, Müller S, Kajetan B, Holtgrewe M, Kola V, Beule D, Sassi C. Shared and oppositely regulated transcriptomic signatures in Huntington's disease and brain ischemia confirm known and unveil novel potential neuroprotective genes. Neurobiol Aging 2021; 104:122.e1-122.e17. [PMID: 33875290 DOI: 10.1016/j.neurobiolaging.2021.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 02/13/2021] [Accepted: 03/02/2021] [Indexed: 11/20/2022]
Abstract
Huntington's disease and subcortical vascular dementia display similar dementing features, shaped by different degrees of striatal atrophy, deep white matter degeneration and tau pathology. To investigate the hypothesis that Huntington's disease transcriptomic hallmarks may provide a window into potential protective genes upregulated during brain acute and subacute ischemia, we compared RNA sequencing signatures in the most affected brain areas of 2 widely used experimental mouse models: Huntington's disease, (R6/2, striatum and cortex and Q175, hippocampus) and brain ischemia-subcortical vascular dementia (BCCAS, striatum, cortex and hippocampus). We identified a cluster of 55 shared genes significantly differentially regulated in both models and we screened these in 2 different mouse models of Alzheimer's disease, and 96 early-onset familial and apparently sporadic small vessel ischemic disease patients. Our data support the prevalent role of transcriptional regulation upon genetic coding variability of known neuroprotective genes (Egr2, Fos, Ptgs2, Itga5, Cdkn1a, Gsn, Npas4, Btg2, Cebpb) and provide a list of potential additional ones likely implicated in different dementing disorders and worth further investigation.
Collapse
Affiliation(s)
- Ferah Yildirim
- Department of Neuropsychiatry, Department of Psychiatry and Psychotherapy, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marco Foddis
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Sonja Blumenau
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Susanne Müller
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Bentele Kajetan
- Berlin Institute of Health, BIH, Core Unit Bioinformatics, Berlin, Germany
| | - Manuel Holtgrewe
- Berlin Institute of Health, BIH, Core Unit Bioinformatics, Berlin, Germany
| | - Vasilis Kola
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Dieter Beule
- Berlin Institute of Health, BIH, Core Unit Bioinformatics, Berlin, Germany
| | - Celeste Sassi
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
| |
Collapse
|
46
|
Liu Y, Han Y, Guan M, Cai Y, Wang W, Chen H, Zhao X. Added value of femoral artery atherosclerosis for determining severity of white matter lesion by carotid atherosclerosis: a magnetic resonance imaging study. Acta Radiol 2021; 62:1112-1121. [PMID: 32811157 DOI: 10.1177/0284185120950106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Previous studies reported that single vascular atherosclerosis was an effective indicator for white matter lesions (WMLs). PURPOSE To investigate the added value of femoral atherosclerosis for determining severity of WMLs by carotid atherosclerosis using three-dimensional vessel wall magnetic resonance imaging (MRI). MATERIAL AND METHODS Elderly individuals without cardiovascular symptoms within the previous six months were recruited. The plaque features of carotid and femoral arteries were evaluated and compared between individuals with WML score ≤ 3 and those with WML score > 3. Logistic regression and receiver operating characteristic (ROC) analyses were used to determine the value of plaque features in discriminating WMLs with score > 3. RESULTS In total, 112 individuals (49 men, mean age 72.0±5.6 years) were included. Participants with a WML score > 3 showed a significantly greater carotid wall area and femoral artery stenosis and higher incidence of carotid calcification and femoral artery calcification and lipid-rich necrotic cores (LRNC) compared to those with a WML score ≤ 3 (all P < 0.05). Carotid artery calcification and femoral artery calcification, LRNC, and stenosis were found to be significantly associated with severe WMLs before and after adjustment for clinical factors (odds ratio 1.51-3.79, all P < 0.05). ROC analysis showed, in discriminating severe WMLs, the area under the curve increased from 0.615 to 0.754 after combining femoral artery LRNC and stenosis with carotid calcification compared to the carotid calcification alone. CONCLUSION Characteristics of femoral artery atherosclerosis determined by vessel wall MRI have added value for carotid atherosclerosis in determining the severity of WMLs.
Collapse
Affiliation(s)
- Yang Liu
- Department of Radiology, The Affiliated Hospital of Yangzhou University, Yangzhou, PR China
- Center for Biomedical Imaging Research, Department of Biomedical Engineering, Tsinghua University School of Medicine, Beijing, PR China
| | - Yongjun Han
- Center for Biomedical Imaging Research, Department of Biomedical Engineering, Tsinghua University School of Medicine, Beijing, PR China
- Center for Brain Disorders Research, Capital Medical University and Beijing Institute for Brain Disorders, Beijing, PR China
| | - Maobin Guan
- Department of Radiology, The Affiliated Hospital of Yangzhou University, Yangzhou, PR China
| | - Ying Cai
- Department of Radiology, Taizhou People’s Hospital, Taizhou, PR China
| | - Wei Wang
- Department of Radiology, The Affiliated Hospital of Yangzhou University, Yangzhou, PR China
| | - Huijun Chen
- Center for Biomedical Imaging Research, Department of Biomedical Engineering, Tsinghua University School of Medicine, Beijing, PR China
| | - Xihai Zhao
- Center for Biomedical Imaging Research, Department of Biomedical Engineering, Tsinghua University School of Medicine, Beijing, PR China
| |
Collapse
|
47
|
Schoemaker D, Velilla-Jimenez L, Zuluaga Y, Baena A, Ospina C, Bocanegra Y, Alvarez S, Ochoa-Escudero M, Guzmán-Vélez E, Martinez J, Lopera F, Arboleda-Velasquez JF, Quiroz YT. Global Cardiovascular Risk Profile and Cerebrovascular Abnormalities in Presymptomatic Individuals with CADASIL or Autosomal Dominant Alzheimer's Disease. J Alzheimers Dis 2021; 82:841-853. [PMID: 34092645 DOI: 10.3233/jad-210313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Cardiovascular risk factors increase the risk of developing dementia, including Alzheimer's disease and vascular dementia. OBJECTIVE Studying individuals with autosomal dominant mutations leading to the early onset of dementia, this study examines the effect of the global cardiovascular risk profile on early cognitive and neuroimaging features of Alzheimer's disease and vascular dementia. METHODS We studied 85 non-demented and stroke-free individuals, including 20 subjects with Presenilin1 (PSEN1) E280A mutation leading to the early onset of autosomal dominant Alzheimer's disease (ADAD), 20 subjects with NOTCH3 mutations leading to cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) and to the early onset of vascular dementia, and 45 non-affected family members (non-carriers). All subjects underwent clinical and neuropsychological evaluations and an MRI. The global cardiovascular risk profile was estimated using the office-based Framingham Cardiovascular Risk Profile (FCRP) score. RESULTS In individuals with CADASIL, a higher FCRP score was associated with a reduced hippocampal volume (B = -0.06, p < 0.05) and an increased severity of cerebral microbleeds (B = 0.13, p < 0.001), lacunes (B = 0.30, p < 0.001), and perivascular space enlargement in the basal ganglia (B = 0.50, p < 0.05). There was no significant association between the FCRP score and neuroimaging measures in ADAD or non-carrier subjects. While the FCRP score was related to performance in executive function in non-carrier subjects (B = 0.06, p < 0.05), it was not significantly associated with cognitive performance in individuals with CADASIL or ADAD. CONCLUSION Our results suggest that individuals with CADASIL and other forms of vascular cognitive impairment might particularly benefit from early interventions aimed at controlling cardiovascular risks.
Collapse
Affiliation(s)
- Dorothee Schoemaker
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | | | - Yesica Zuluaga
- Grupo de Neurociencias, Universidad de Antioquia, Medellín, Colombia
| | - Ana Baena
- Grupo de Neurociencias, Universidad de Antioquia, Medellín, Colombia
| | - Carolina Ospina
- Grupo de Neurociencias, Universidad de Antioquia, Medellín, Colombia
| | - Yamile Bocanegra
- Grupo de Neurociencias, Universidad de Antioquia, Medellín, Colombia
| | - Sergio Alvarez
- Department of Radiology, Hospital Pablo Tobon Uribe, Medellín, Colombia
| | | | - Edmarie Guzmán-Vélez
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jairo Martinez
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Francisco Lopera
- Grupo de Neurociencias, Universidad de Antioquia, Medellín, Colombia
| | - Joseph F Arboleda-Velasquez
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Yakeel T Quiroz
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Grupo de Neurociencias, Universidad de Antioquia, Medellín, Colombia.,Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
48
|
Du J, Zhu H, Yu L, Lu P, Qiu Y, Zhou Y, Cao W, Lu D, Zhao W, Yang J, Sun J, Xu Q. Multi-Dimensional Diffusion Tensor Imaging Biomarkers for Cognitive Decline From the Preclinical Stage: A Study of Post-stroke Small Vessel Disease. Front Neurol 2021; 12:687959. [PMID: 34322083 PMCID: PMC8311001 DOI: 10.3389/fneur.2021.687959] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 06/14/2021] [Indexed: 11/13/2022] Open
Abstract
Objectives: We aim to investigate whether multi-dimensional diffusion tensor imaging (DTI) measures can sensitively identify different cognitive status of cerebral small vessel disease (CSVD) and to explore the underlying pattern of white matter disruption in CSVD. Methods: Two hundred and two participants were recruited, composed of 99 CSVD patients with mild cognitive impairment (VaMCI) and 60 with no cognitive impairment (NCI) and 43 healthy subjects as normal controls (NC). Full domain neuropsychological tests and diffusion-weighted imaging were performed on each subject. DTI metrics such as fractional anisotropy (FA), mean diffusivity (MD), the skeletonized mean diffusivity (PSMD), and structural brain network measures including network strength, global efficiency (EGlobal), and local efficiency (ELocal) were calculated. Region of interest (ROI) analysis of 42 white matter tracts was performed to examine the regional anatomical white matter disruption for each group. Results: Significant differences of multiple cognitive test scores across all cognitive domains especially processing and executive function existed among the three groups. DTI measures (FA, MD, and PSMD) showed significant group difference with the cognitive status changing. FA and EGlobal showed significant correlation with processing speed, executive function, and memory. ROI analysis found that white matter integrity impairment occurred from the preclinical stage of vascular cognitive impairment (VCI) due to CSVD. These lesions in the NCI group mainly involved some longitudinal fibers such as right superior longitudinal fasciculus (SLF-R), right superior fronto-occipital fasciculus (SFO-R), and right uncinate fasciculus (UNC-R), which might be more vulnerable to the cerebrovascular aging and disease process. Conclusions: DTI measures are sensitive neuroimaging markers in detecting the early cognitive impairment and able to differentiate the different cognitive status due to CSVD. Subtle changes of some vulnerable white matter tracts may be observed from the preclinical stage of VCI and have a local to general spreading pattern during the disease progression.
Collapse
Affiliation(s)
- Jing Du
- Renji-UNSW CHeBA (Centre for Healthy Brain Ageing of University of New South Wales) Neurocognitive Center, Renji Hospital, Medical School of Shanghai Jiao Tong University, Shanghai, China.,Department of Neurology, Renji Hospital, Medical School of Shanghai Jiao Tong University, Shanghai, China.,Department of Health Management Center, Renji Hospital, Medical School of Shanghai Jiao Tong University, Shanghai, China
| | - Hong Zhu
- Shanghai Med-X Engineering Research Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Ling Yu
- Renji-UNSW CHeBA (Centre for Healthy Brain Ageing of University of New South Wales) Neurocognitive Center, Renji Hospital, Medical School of Shanghai Jiao Tong University, Shanghai, China.,Department of Neurology, Renji Hospital, Medical School of Shanghai Jiao Tong University, Shanghai, China
| | - Peiwen Lu
- Renji-UNSW CHeBA (Centre for Healthy Brain Ageing of University of New South Wales) Neurocognitive Center, Renji Hospital, Medical School of Shanghai Jiao Tong University, Shanghai, China.,Department of Neurology, Renji Hospital, Medical School of Shanghai Jiao Tong University, Shanghai, China.,Department of Health Management Center, Renji Hospital, Medical School of Shanghai Jiao Tong University, Shanghai, China
| | - Yage Qiu
- Department of Radiology, Renji Hospital, Medical School of Shanghai Jiao Tong University, Shanghai, China
| | - Yan Zhou
- Department of Radiology, Renji Hospital, Medical School of Shanghai Jiao Tong University, Shanghai, China
| | - Wenwei Cao
- Renji-UNSW CHeBA (Centre for Healthy Brain Ageing of University of New South Wales) Neurocognitive Center, Renji Hospital, Medical School of Shanghai Jiao Tong University, Shanghai, China.,Department of Neurology, Renji Hospital, Medical School of Shanghai Jiao Tong University, Shanghai, China
| | - Dong Lu
- Renji-UNSW CHeBA (Centre for Healthy Brain Ageing of University of New South Wales) Neurocognitive Center, Renji Hospital, Medical School of Shanghai Jiao Tong University, Shanghai, China.,Department of Neurology, Renji Hospital, Medical School of Shanghai Jiao Tong University, Shanghai, China
| | - Wei Zhao
- Renji-UNSW CHeBA (Centre for Healthy Brain Ageing of University of New South Wales) Neurocognitive Center, Renji Hospital, Medical School of Shanghai Jiao Tong University, Shanghai, China.,Department of Neurology, Renji Hospital, Medical School of Shanghai Jiao Tong University, Shanghai, China
| | - Jie Yang
- Renji-UNSW CHeBA (Centre for Healthy Brain Ageing of University of New South Wales) Neurocognitive Center, Renji Hospital, Medical School of Shanghai Jiao Tong University, Shanghai, China.,Department of Neurology, Renji Hospital, Medical School of Shanghai Jiao Tong University, Shanghai, China
| | - Junfeng Sun
- Shanghai Med-X Engineering Research Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Qun Xu
- Renji-UNSW CHeBA (Centre for Healthy Brain Ageing of University of New South Wales) Neurocognitive Center, Renji Hospital, Medical School of Shanghai Jiao Tong University, Shanghai, China.,Department of Neurology, Renji Hospital, Medical School of Shanghai Jiao Tong University, Shanghai, China.,Department of Health Management Center, Renji Hospital, Medical School of Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
49
|
Wu X, Wang H, Chen C, Xiong Y, Zhu L, Jia J, Yang T, Ma F. The association between cardiovascular risk burden and cognitive function amongst the old: a 9-year longitudinal cohort study. Eur J Neurol 2021; 28:2907-2912. [PMID: 34075662 DOI: 10.1111/ene.14947] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/01/2022]
Abstract
BACKGROUND AND PURPOSE Cardiovascular risk burden in midlife has been linked to cognitive decline in later life, but whether this association still exists in older cohorts is unclear. METHODS The association between the cardiovascular risk score and cognitive function was investigated using 9-year follow-up data. The risk score algorithms were from the Chinese guidelines on the prevention and treatment of dyslipidemia in adults (2016 revised), which were assessed at baseline and categorized into tertiles (low, middle and high). Full intelligence quotient (FIQ), verbal intelligence quotient (VIQ) and performance intelligence quotient (PIQ) were assessed at follow-ups with the Wechsler Adult Intelligence Scale-Chinese, revised (WAIS-RC). Data were analyzed using the linear mixed-effects model. RESULTS A total of 924 participants (mean age 78.06 ± 7.58 years) were included in our study. In all participants, the risk score ranged from 0.02 to 0.55 (mean score 0.16 ± 0.08). Compared with the low tertile, a higher risk score was associated with lower FIQ (β -0.094, 95% confidence interval [CI] -0.181, -0.007) and VIQ (β -0.100; 95% CI -0.192, -0.007) at the follow-up. There is a more significant association between higher risk score and lower FIQ amongst females (β -0.263; 95% CI -0.462, -0.065) and VIQ (β -0.268; 95% CI -0.478, -0.057). CONCLUSIONS A higher cardiovascular risk score was associated with lower FIQ and VIQ. Higher cardiovascular risk burden increased the risk of cognition impairment and accelerated its progression over time. This study has implications for early detection of cognition impairment.
Collapse
Affiliation(s)
- Xiaomin Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Environment, Nutrition and Public Health, Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, China
| | - Hualou Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Environment, Nutrition and Public Health, Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, China
| | - Chong Chen
- Department of Clinical Laboratory, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, National Human Genetic Resources Sharing Service Platform, Tianjin, China
| | - Ying Xiong
- Department of Community Service, Wangdingdi Hospital of Tianjin Nankai District, Tianjin, China
| | - Liping Zhu
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Environment, Nutrition and Public Health, Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, China
| | - Jingya Jia
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Environment, Nutrition and Public Health, Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, China
| | - Tong Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Environment, Nutrition and Public Health, Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, China
| | - Fei Ma
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China.,Tianjin Key Laboratory of Environment, Nutrition and Public Health, Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, China
| |
Collapse
|
50
|
van Dalen JW, Mutsaerts HJ, Petr J, Caan MW, van Charante EPM, MacIntosh BJ, van Gool WA, Nederveen AJ, Richard E. Longitudinal relation between blood pressure, antihypertensive use and cerebral blood flow, using arterial spin labelling MRI. J Cereb Blood Flow Metab 2021; 41:1756-1766. [PMID: 33325767 PMCID: PMC8217888 DOI: 10.1177/0271678x20966975] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Consistent cerebral blood flow (CBF) is fundamental to brain function. Cerebral autoregulation ensures CBF stability. Chronic hypertension can lead to disrupted cerebral autoregulation in older people, potentially leading to blood pressure levels interfering with CBF. This study tested whether low BP and AHD use are associated with contemporaneous low CBF, and whether longitudinal change in BP is associated with change in CBF, using arterial spin labelling (ASL) MRI, in a prospective longitudinal cohort of 186 community-dwelling older individuals with hypertension (77 ± 3 years, 53% female), 125 (67%) of whom with 3-year follow-up. Diastolic blood pressure, systolic blood pressure, mean arterial pressure, and pulse pressure were assessed as blood pressure parameters. As additional cerebrovascular marker, we evaluated the ASL signal spatial coefficient of variation (ASL SCoV), a measure of ASL signal heterogeneity that may reflect cerebrovascular health. We found no associations between any of the blood pressure measures and concurrent CBF nor between changes in blood pressure measures and CBF over three-year follow-up. Antihypertensive use was associated with lower grey matter CBF (-5.49 ml/100 g/min, 95%CI = -10.7|-0.27, p = 0.04) and higher ASL SCoV (0.32 SD, 95%CI = 0.12|0.52, p = 0.002). These results warrant future research on the potential relations between antihypertensive use and cerebral perfusion.
Collapse
Affiliation(s)
- Jan Willem van Dalen
- Department of Neurology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.,Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Henri Jmm Mutsaerts
- Department of Radiology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Jan Petr
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Matthan Wa Caan
- Department of Biomedical Engineering and Physics, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Eric P Moll van Charante
- Department of General Practice, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Bradley J MacIntosh
- Department of Medical Biophysics, University of Toronto, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Willem A van Gool
- Department of Neurology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Aart J Nederveen
- Department of Radiology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Edo Richard
- Department of Neurology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands.,Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|