1
|
Le Grand Q, Tsuchida A, Koch A, Imtiaz MA, Aziz NA, Vigneron C, Zago L, Lathrop M, Dubrac A, Couffinhal T, Crivello F, Matthews PM, Mishra A, Breteler MMB, Tzourio C, Debette S. Diffusion imaging genomics provides novel insight into early mechanisms of cerebral small vessel disease. Mol Psychiatry 2024; 29:3567-3579. [PMID: 38811690 PMCID: PMC11541005 DOI: 10.1038/s41380-024-02604-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 05/06/2024] [Accepted: 05/09/2024] [Indexed: 05/31/2024]
Abstract
Cerebral small vessel disease (cSVD) is a leading cause of stroke and dementia. Genetic risk loci for white matter hyperintensities (WMH), the most common MRI-marker of cSVD in older age, were recently shown to be significantly associated with white matter (WM) microstructure on diffusion tensor imaging (signal-based) in young adults. To provide new insights into these early changes in WM microstructure and their relation with cSVD, we sought to explore the genetic underpinnings of cutting-edge tissue-based diffusion imaging markers across the adult lifespan. We conducted a genome-wide association study of neurite orientation dispersion and density imaging (NODDI) markers in young adults (i-Share study: N = 1 758, (mean[range]) 22.1[18-35] years), with follow-up in young middle-aged (Rhineland Study: N = 714, 35.2[30-40] years) and late middle-aged to older individuals (UK Biobank: N = 33 224, 64.3[45-82] years). We identified 21 loci associated with NODDI markers across brain regions in young adults. The most robust association, replicated in both follow-up cohorts, was with Neurite Density Index (NDI) at chr5q14.3, a known WMH locus in VCAN. Two additional loci were replicated in UK Biobank, at chr17q21.2 with NDI, and chr19q13.12 with Orientation Dispersion Index (ODI). Transcriptome-wide association studies showed associations of STAT3 expression in arterial and adipose tissue (chr17q21.2) with NDI, and of several genes at chr19q13.12 with ODI. Genetic susceptibility to larger WMH volume, but not to vascular risk factors, was significantly associated with decreased NDI in young adults, especially in regions known to harbor WMH in older age. Individually, seven of 25 known WMH risk loci were associated with NDI in young adults. In conclusion, we identified multiple novel genetic risk loci associated with NODDI markers, particularly NDI, in early adulthood. These point to possible early-life mechanisms underlying cSVD and to processes involving remyelination, neurodevelopment and neurodegeneration, with a potential for novel approaches to prevention.
Collapse
Affiliation(s)
- Quentin Le Grand
- University of Bordeaux, INSERM, Bordeaux Population Health research center, UMR1219, F-33000, Bordeaux, France
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Ami Tsuchida
- University of Bordeaux, INSERM, Bordeaux Population Health research center, UMR1219, F-33000, Bordeaux, France
- University of Bordeaux, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional Imaging Group, F-33000, Bordeaux, France
- CNRS, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional Imaging Group, F-33000, Bordeaux, France
- CEA, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional Imaging Group, F-33000, Bordeaux, France
| | - Alexandra Koch
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Mohammed-Aslam Imtiaz
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - N Ahmad Aziz
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurology, Faculty of Medicine, University of Bonn, Bonn, Germany
| | - Chloé Vigneron
- University of Bordeaux, INSERM, Bordeaux Population Health research center, UMR1219, F-33000, Bordeaux, France
| | - Laure Zago
- University of Bordeaux, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional Imaging Group, F-33000, Bordeaux, France
- CNRS, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional Imaging Group, F-33000, Bordeaux, France
- CEA, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional Imaging Group, F-33000, Bordeaux, France
| | - Mark Lathrop
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada; Victor Phillip Dahdaleh Institute of Genomic Medicine at McGill University, Montreal, QC, H3A 0G1, Canada
| | - Alexandre Dubrac
- Centre de Recherche, CHU Sainte-Justine, Montréal, QC, Canada
- Département de Pathologie et Biologie Cellulaire, Université de Montréal, Montréal, QC, Canada
- Département d'Ophtalmologie, Université de Montréal, Montréal, QC, Canada
| | - Thierry Couffinhal
- University of Bordeaux, INSERM, Biologie des maladies cardiovasculaires, U1034, F-33600, Pessac, France
| | - Fabrice Crivello
- University of Bordeaux, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional Imaging Group, F-33000, Bordeaux, France
- CNRS, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional Imaging Group, F-33000, Bordeaux, France
- CEA, Institute of Neurodegenerative Diseases, UMR5293, Neurofunctional Imaging Group, F-33000, Bordeaux, France
| | - Paul M Matthews
- UK Dementia Research Institute and Department of Brain Sciences, Imperial College, London, UK
| | - Aniket Mishra
- University of Bordeaux, INSERM, Bordeaux Population Health research center, UMR1219, F-33000, Bordeaux, France
| | - Monique M B Breteler
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Institute for Medical Biometry, Informatics and Epidemiology (IMBIE), Faculty of Medicine, University of Bonn, Bonn, Germany
| | - Christophe Tzourio
- University of Bordeaux, INSERM, Bordeaux Population Health research center, UMR1219, F-33000, Bordeaux, France
- Bordeaux University Hospital, Department of Medical Informatics, F-33000, Bordeaux, France
| | - Stéphanie Debette
- University of Bordeaux, INSERM, Bordeaux Population Health research center, UMR1219, F-33000, Bordeaux, France.
- Bordeaux University Hospital, Department of Neurology, Institute for Neurodegenerative Diseases, F-33000, Bordeaux, France.
| |
Collapse
|
2
|
Diany R, Gagliano Taliun SA. Systematic Review and Phenome-Wide Scans of Genetic Associations with Vascular Cognitive Impairment. Adv Biol (Weinh) 2024; 8:e2300692. [PMID: 38935518 DOI: 10.1002/adbi.202300692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 03/12/2024] [Indexed: 06/29/2024]
Abstract
Vascular cognitive impairment (VCI) is a heterogenous form of cognitive impairment that results from cerebrovascular disease. It is a result of both genetic and non-genetic factors. Although much research has been conducted on the genetic contributors to other forms of cognitive impairment (e.g. Alzheimer's disease), knowledge is lacking on the genetic factors associated with VCI. A better understanding of the genetics of VCI will be critical for prevention and treatment. To begin to fill this gap, the genetic contributors are reviewed with VCI from the literature. Phenome-wide scans of the identified genes are conducted and genetic variants identified in the review in large-scale resources displaying genetic variant-trait association information. Gene set are also carried out enrichment analysis using the genes identified from the review. Thirty one articles are identified meeting the search criteria and filters, from which 107 unique protein-coding genes are noted related to VCI. The phenome-wide scans and gene set enrichment analysis identify pathways associated with a diverse set of biological systems. This results indicate that genes with evidence of involvement in VCI are involved in a diverse set of biological functions. This information can facilitate downstream research to better dissect possible shared biological mechanisms for future therapies.
Collapse
Affiliation(s)
- Rime Diany
- Faculty of Medicine, Université de Montréal, 2900 Boulevard Edouard-Montpetit, Montréal, Québec, H3C 3J7, Canada
| | - Sarah A Gagliano Taliun
- Department of Medicine & Department of Neurosciences, Faculty of Medicine, Université de Montréal, 2900 Boulevard Edouard-Montpetit, Montréal, Québec, H3C 3J7, Canada
- Montreal Heart Institute, 5000 rue Bélanger, Montréal, Québec, H1T 1C8, Canada
| |
Collapse
|
3
|
Fleischman DA, Arfanakis K, Leurgans SE, Arvanitakis Z, Lamar M, Han SD, Poole VN, Bennett DA, Barnes LL. Cerebral arteriolosclerosis, lacunar infarcts, and cognition in older Black adults. Alzheimers Dement 2024; 20:5375-5384. [PMID: 38988020 PMCID: PMC11350059 DOI: 10.1002/alz.13917] [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: 02/12/2024] [Revised: 04/05/2024] [Accepted: 05/01/2024] [Indexed: 07/12/2024]
Abstract
INTRODUCTION Older Black adults are at risk of cerebral small vessel disease (CSVD), which contributes to dementia risk. Two subtypes of CSVD, arteriolosclerosis and ischemic lacunar infarcts, have been independently linked to lower cognition and higher dementia risk, but their combined effects on cognition in older Black adults are unclear. METHODS Mixed models were used to examine the associations of in vivo measures of arteriolosclerosis (ARTS) and ischemic lacunar infarcts to cognitive level and change in 370 older Black adults without dementia. RESULTS: Modeled together, higher ARTS load accounted for lower levels of global cognition, episodic memory, semantic memory, and perceptual speed, whereas higher infarct load accounted for lower levels of working memory. There were no associations with rate of cognitive change. DISCUSSION Both arteriolosclerosis and ischemic infarcts impact the cognitive health of older Black adults, but arteriolosclerosis affects cognition more broadly and offers promise as an in vivo biomarker of dementia risk. HIGHLIGHTS Older Black adults are at risk of cerebral small vessel disease (CSVD) and dementia. Examined magnetic resonance imaging-derived measure of arteriolosclerosis (ARTS), infarcts, and cognition. ARTS load was widely associated with lower cognition after adjusting for infarct load. Infarct load was specifically associated with lower complex attention. More within-Black in vivo studies of CSVD subtypes and cognition are needed.
Collapse
Affiliation(s)
- Debra A. Fleischman
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
- Department of Psychiatry and Behavioral SciencesRush University Medical CenterChicagoIllinoisUSA
| | - Konstantinos Arfanakis
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Diagnostic Radiology and Nuclear MedicineRush University Medical CenterChicagoIllinoisUSA
- Department of Biomedical EngineeringIllinois Institute of TechnologyChicagoIllinoisUSA
| | - Sue E. Leurgans
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
- Department of Family & Preventive MedicineRush University Medical CenterChicagoIllinoisUSA
| | - Zoe Arvanitakis
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
| | - Melissa Lamar
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Psychiatry and Behavioral SciencesRush University Medical CenterChicagoIllinoisUSA
| | - S. Duke Han
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of PsychologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Victoria N. Poole
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Orthopedic SurgeryRush University Medical CenterChicagoIllinoisUSA
| | - David A. Bennett
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
| | - Lisa L. Barnes
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
- Department of Neurological SciencesRush University Medical CenterChicagoIllinoisUSA
- Department of Psychiatry and Behavioral SciencesRush University Medical CenterChicagoIllinoisUSA
| |
Collapse
|
4
|
Hu X, Liu L, Xiong M, Lu J. Application of artificial intelligence-based magnetic resonance imaging in diagnosis of cerebral small vessel disease. CNS Neurosci Ther 2024; 30:e14841. [PMID: 39045778 PMCID: PMC11267174 DOI: 10.1111/cns.14841] [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: 04/23/2024] [Revised: 06/15/2024] [Accepted: 06/21/2024] [Indexed: 07/25/2024] Open
Abstract
Cerebral small vessel disease (CSVD) is an important cause of stroke, cognitive impairment, and other diseases, and its early quantitative evaluation can significantly improve patient prognosis. Magnetic resonance imaging (MRI) is an important method to evaluate the occurrence, development, and severity of CSVD. However, the diagnostic process lacks quantitative evaluation criteria and is limited by experience, which may easily lead to missed diagnoses and misdiagnoses. With the development of artificial intelligence technology based on deep learning, the extraction of high-dimensional features in imaging can assist doctors in clinical decision-making, and it has been widely used in brain function and mental disorders, and cardiovascular and cerebrovascular diseases. This paper summarizes the global research results in recent years and briefly describes the application of deep learning in evaluating CSVD signs in MRI imaging, including recent small subcortical infarcts, lacunes of presumed vascular origin, vascular white matter hyperintensity, enlarged perivascular spaces, cerebral microbleeds, brain atrophy, cortical superficial siderosis, and cortical cerebral microinfarct.
Collapse
Affiliation(s)
- Xiaofei Hu
- Xuanwu HospitalCapital Medical UniversityBeijingChina
- Department of Nuclear Medicine, Southwest HospitalThird Military Medical University (Army Medical University)ChongqingChina
| | - Li Liu
- Department of Digital Medicine, School of Biomedical Engineering and Medical ImagingThird Military Medical University (Army Medical University)ChongqingChina
| | - Ming Xiong
- Department of Digital Medicine, School of Biomedical Engineering and Medical ImagingThird Military Medical University (Army Medical University)ChongqingChina
| | - Jie Lu
- Xuanwu HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
5
|
Li S, Xiao Z. Recent Research Progress on the Use of Transcranial Magnetic Stimulation in the Treatment of Vascular Cognitive Impairment. Neuropsychiatr Dis Treat 2024; 20:1235-1246. [PMID: 38883416 PMCID: PMC11179638 DOI: 10.2147/ndt.s467357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 06/01/2024] [Indexed: 06/18/2024] Open
Abstract
Vascular Cognitive Impairment (VCI) is a condition where problems with brain blood vessels lead to a decline in cognitive abilities, commonly affecting the elderly and placing a significant burden on both patients and their families. Compared to medication and surgery, Transcranial Magnetic Stimulation (TMS) is a non-invasive treatment option with fewer risks and side effects, making it particularly suitable for elderly patients. TMS not only assesses the excitability and plasticity of the cerebral cortex, but its effectiveness in treating Vascular Cognitive Impairment (VCI) and its subtypes has also been validated in numerous clinical trials worldwide. However, there is still a lack of review on the physiological mechanisms of TMS treatment for VCI and its specific clinical application parameters. Therefore, this article initially provided a brief overview of the risk factors, pathological mechanisms, and classification of VCI. Next, the article explained the potential physiological mechanisms of TMS in treating VCI, particularly its role in promoting synaptic plasticity, regulating neurotransmitter balance, and improving the function of the default mode network. Additionally, The article also summarizes the application of rTMS in treating VCI and its subtypes, VCI-related sleep disorders, and the use of TMS in follow-up studies of VCI patients, providing empirical evidence for the clinical application of TMS and rTMS technologies.
Collapse
Affiliation(s)
- Sijing Li
- Department of Neurology, Multi-Omics Research Center for Brain Disorders, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People's Republic of China
- Clinical Research Center for Immune‑Related Encephalopathy of Hunan Province (The First Affiliated Hospital), Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People's Republic of China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, People's Republic of China
| | - Zijian Xiao
- Department of Neurology, Multi-Omics Research Center for Brain Disorders, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People's Republic of China
- Clinical Research Center for Immune‑Related Encephalopathy of Hunan Province (The First Affiliated Hospital), Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, People's Republic of China
| |
Collapse
|
6
|
Valančienė J, Melaika K, Šliachtenko A, Šiaurytė-Jurgelėnė K, Ekkert A, Jatužis D. Stroke genetics and how it Informs novel drug discovery. Expert Opin Drug Discov 2024; 19:553-564. [PMID: 38494780 DOI: 10.1080/17460441.2024.2324916] [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: 12/05/2023] [Accepted: 02/26/2024] [Indexed: 03/19/2024]
Abstract
INTRODUCTION Stroke is one of the main causes of death and disability worldwide. Nevertheless, despite the global burden of this disease, our understanding is limited and there is still a lack of highly efficient etiopathology-based treatment. It is partly due to the complexity and heterogenicity of the disease. It is estimated that around one-third of ischemic stroke is heritable, emphasizing the importance of genetic factors identification and targeting for therapeutic purposes. AREAS COVERED In this review, the authors provide an overview of the current knowledge of stroke genetics and its value in diagnostics, personalized treatment, and prognostication. EXPERT OPINION As the scale of genetic testing increases and the cost decreases, integration of genetic data into clinical practice is inevitable, enabling assessing individual risk, providing personalized prognostic models and identifying new therapeutic targets and biomarkers. Although expanding stroke genetics data provides different diagnostics and treatment perspectives, there are some limitations and challenges to face. One of them is the threat of health disparities as non-European populations are underrepresented in genetic datasets. Finally, a deeper understanding of underlying mechanisms of potential targets is still lacking, delaying the application of novel therapies into routine clinical practice.
Collapse
Affiliation(s)
| | | | | | - Kamilė Šiaurytė-Jurgelėnė
- Department of Human and Medical Genetics, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | | | - Dalius Jatužis
- Center of Neurology, Vilnius University, Vilnius, Lithuania
| |
Collapse
|
7
|
Shang B, Jia S, Zhang T, Gao F, Lu M, Chen K, Jiao J, Dai Z, Zeng Q, Xu B, Lei H. Study on the chemical constituents and mechanism of Kai-Xin-San based on UPLC-Q-Exactive MS and network pharmacology. JOURNAL OF ETHNOPHARMACOLOGY 2024; 322:117652. [PMID: 38151178 DOI: 10.1016/j.jep.2023.117652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 12/13/2023] [Accepted: 12/20/2023] [Indexed: 12/29/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Vascular disease (VD) is a kind of common disease harmful to the health of the middle-aged and elderly, which has the characteristics of long treatment cycle and high recurrence rate, and without effective method to treat so far. Traditional Chinese medicine (TCM) has the characteristics of multi-components and multi-targets to treat diseases. Kai-Xin-San is a TCM formula applied for treating psychiatric diseases such as depression in China for thousands of years, and it has been used in clinical treatment of VD. But up to now, its active composition and mechanism are not clear. AIM OF THE STUDY To explore the effective components of Kai-Xin-San, investigate the effect of Kai-Xin-San on angiogenesis, screen and verify the related targets and possible mechanisms of Kai-Xin-San against VD. MATERIALS AND METHODS UPLC-Q-Exactive Orbitrap MS was performed to identify the chemical components of Kai-Xin-San. The mechanism of multi-components, multi-targets, and multi-pathways of Kai-Xin-San in the treatment of VD were explored by network pharmacology. And then, quail chick chorioallantoic membrane (qCAM) assays were used to evaluate the vascular protective activity of Kai-Xin-San. Evaluation of angiogenesis by calculating the relative vessels area. The levels of VEGFA and Akt1 in qCAM were measured by RT-PCR. Twenty-five male SD rats were randomly divided into the sham group, model group, Donepezil (0.45 mg/kg) group, Kai-Xin-San low dose group (0.1575 g/kg), Kai-Xin-San high dose group (0.63 g/kg). Two-vessel occlusion (2-VO) rat model is established to evaluate the therapeutic effect of Kai-Xin-San pretreatment. Hematoxylin-eosin (HE) staining is conducted to detect the morphological changes of neurons in the hippocampus. RESULTS Data showed that 62 compounds were identified in Kai-Xin-San. The network pharmacology results showed 73 compounds in Kai-Xin-San play a role in the treatment of VD, such as Ginsenoside Rh4, kaempferol, and Poricoic acid C. A total of 7 main targets are predicted, including Akt1, TNF and so on. Kai-Xin-San could increase VEGFA and Akt1 expression, promote angiogenesis and regulate the PI3K-Akt signaling pathway. The results depict that Kai-Xin-San has dose-dependently improved the cognitive function in 2-VO model rats. It has also been showed that Kai-Xin-San can rescue neuron damage in the hippocampus. CONCLUSION The complex chemical components of Kai-Xin-San play a synergistic role in the treatment of VD, and involve multiple pathways and targets. To protect blood vessels by promoting angiogenesis is one of the potential mechanisms of Kai-Xin-San in the treatment of VD. This study reveals that Kai-Xin-San protects the 2-VO model rats from ischemic injury by alleviating neuron damage in the hippocampus.
Collapse
Affiliation(s)
- Bingxian Shang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Shuhe Jia
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Tong Zhang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Feng Gao
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Mingjun Lu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Kedian Chen
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Jingyi Jiao
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Ziqi Dai
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Qi Zeng
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China
| | - Bing Xu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China.
| | - Haimin Lei
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 102400, China.
| |
Collapse
|
8
|
Meng Y, Wang S, Zhu W, Wang T, Liu D, Wang M, Pi J, Liu Y, Zhuo Z, Pan Y, Wang Y. Association of Mean Upper Cervical Spinal Cord Cross-Sectional Area With Cerebral Small Vessel Disease: A Community-Based Cohort Study. Stroke 2024; 55:687-695. [PMID: 38269540 DOI: 10.1161/strokeaha.123.044666] [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: 07/29/2023] [Accepted: 12/13/2023] [Indexed: 01/26/2024]
Abstract
BACKGROUND The purpose of this study was to investigate the association between the mean upper cervical spinal cord cross-sectional area (MUCCA) and the risk and severity of cerebral small vessel disease (CSVD). METHODS Community-dwelling residents in Lishui City, China, from the cross-sectional survey in the PRECISE cohort study (Polyvascular Evaluation for Cognitive Impairment and Vascular Events) conducted from 2017 to 2019. We included 1644 of 3067 community-dwelling adults in the PRECISE study after excluding those with incorrect, incomplete, insufficient, or missing clinical or imaging data. Total and modified total CSVD scores, as well as magnetic resonance imaging features, including white matter hyperintensity, lacunes, cerebral microbleeds, enlarged perivascular spaces, and brain atrophy, were assessed at the baseline. The Spinal Cord Toolbox was used to measure the upper cervical spinal cord cross-sectional area of the C1 to C3 segments of the spinal cord and its average value was taken as MUCCA. Participants were divided into 4 groups according to quartiles of MUCCA. Associations were analyzed using linear regression models adjusted for age, sex, current smoking and drinking, medical history, intracranial volume, and total cortical volume. RESULTS The means±SD age of the participants was 61.4±6.5 years, and 635 of 1644 participants (38.6%) were men. The MUCCA was smaller in patients with CSVD than those without CSVD. Using the total CSVD score as a criterion, the MUCCA was 61.78±6.12 cm2 in 504 of 1644 participants with CSVD and 62.74±5.94 cm2 in 1140 of 1644 participants without CSVD. Using the modified total CSVD score, the MUCCA was 61.81±6.04 cm2 in 699 of 1644 participants with CSVD and 62.91±5.94 cm2 in 945 of 1644 without CSVD. There were statistical differences between the 2 groups after adjusting for covariates in 3 models. The MUCCA was negatively associated with the total and modified total CSVD scores (adjusted β value, -0.009 [95% CI, -0.01 to -0.003] and -0.007 [95% CI, -0.01 to -0.0006]) after adjustment for covariates. Furthermore, the MUCCA was negatively associated with the white matter hyperintensity burden (adjusted β value, -0.01 [95% CI, -0.02 to -0.003]), enlarged perivascular spaces in the basal ganglia (adjusted β value, -0.005 [95% CI, -0.009 to -0.001]), lacunes (adjusted β value, -0.004 [95% CI, -0.007 to -0.0007]), and brain atrophy (adjusted β value, -0.009 [95% CI, -0.01 to -0.004]). CONCLUSIONS The MUCCA and CSVD were correlated. Spinal cord atrophy may serve as an imaging marker for CSVD; thus, small vessel disease may involve the spinal cord in addition to being intracranial.
Collapse
Affiliation(s)
- Yufei Meng
- Department of Neurology, Beijing Tiantan Hospital (Y.M., W.Z., T.W., D.L., M.W., J.P., Y.L., Z.Z., Y.P., Y.W.), Capital Medical University, China
- Dongzhimen Hospital, Beijing University of Chinese Medicine, China (Y.M.)
| | - Suying Wang
- Department of Neurology and Cerebrovascular Research Laboratory, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical University, Zhejiang, China (S.W.)
| | - Wanlin Zhu
- Department of Neurology, Beijing Tiantan Hospital (Y.M., W.Z., T.W., D.L., M.W., J.P., Y.L., Z.Z., Y.P., Y.W.), Capital Medical University, China
| | - Tingting Wang
- Department of Neurology, Beijing Tiantan Hospital (Y.M., W.Z., T.W., D.L., M.W., J.P., Y.L., Z.Z., Y.P., Y.W.), Capital Medical University, China
- China National Clinical Research Center for Neurological Diseases, Beijing (T.W., D.L., M.W., Y.P., Y.W.)
| | - Dandan Liu
- Department of Neurology, Beijing Tiantan Hospital (Y.M., W.Z., T.W., D.L., M.W., J.P., Y.L., Z.Z., Y.P., Y.W.), Capital Medical University, China
- China National Clinical Research Center for Neurological Diseases, Beijing (T.W., D.L., M.W., Y.P., Y.W.)
| | - Mengxing Wang
- Department of Neurology, Beijing Tiantan Hospital (Y.M., W.Z., T.W., D.L., M.W., J.P., Y.L., Z.Z., Y.P., Y.W.), Capital Medical University, China
- China National Clinical Research Center for Neurological Diseases, Beijing (T.W., D.L., M.W., Y.P., Y.W.)
| | - Jingtao Pi
- Department of Neurology, Beijing Tiantan Hospital (Y.M., W.Z., T.W., D.L., M.W., J.P., Y.L., Z.Z., Y.P., Y.W.), Capital Medical University, China
| | - Yaou Liu
- Department of Neurology, Beijing Tiantan Hospital (Y.M., W.Z., T.W., D.L., M.W., J.P., Y.L., Z.Z., Y.P., Y.W.), Capital Medical University, China
| | - Zhizheng Zhuo
- Department of Neurology, Beijing Tiantan Hospital (Y.M., W.Z., T.W., D.L., M.W., J.P., Y.L., Z.Z., Y.P., Y.W.), Capital Medical University, China
| | - Yuesong Pan
- Department of Neurology, Beijing Tiantan Hospital (Y.M., W.Z., T.W., D.L., M.W., J.P., Y.L., Z.Z., Y.P., Y.W.), Capital Medical University, China
- China National Clinical Research Center for Neurological Diseases, Beijing (T.W., D.L., M.W., Y.P., Y.W.)
| | - Yilong Wang
- Department of Neurology, Beijing Tiantan Hospital (Y.M., W.Z., T.W., D.L., M.W., J.P., Y.L., Z.Z., Y.P., Y.W.), Capital Medical University, China
- Advanced Innovation Center for Human Brain Protection (Y.W.), Capital Medical University, China
- Beijing Laboratory of Oral Health (Y.W.), Capital Medical University, China
- Chinese Institute for Brain Research, Beijing, China (Y.W.)
- National Center for Neurological Diseases, Beijing, China (Y.W.)
- China National Clinical Research Center for Neurological Diseases, Beijing (T.W., D.L., M.W., Y.P., Y.W.)
| |
Collapse
|
9
|
Vuic B, Milos T, Tudor L, Nikolac Perkovic M, Konjevod M, Nedic Erjavec G, Farkas V, Uzun S, Mimica N, Svob Strac D. Pharmacogenomics of Dementia: Personalizing the Treatment of Cognitive and Neuropsychiatric Symptoms. Genes (Basel) 2023; 14:2048. [PMID: 38002991 PMCID: PMC10671071 DOI: 10.3390/genes14112048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Dementia is a syndrome of global and progressive deterioration of cognitive skills, especially memory, learning, abstract thinking, and orientation, usually affecting the elderly. The most common forms are Alzheimer's disease, vascular dementia, and other (frontotemporal, Lewy body disease) dementias. The etiology of these multifactorial disorders involves complex interactions of various environmental and (epi)genetic factors and requires multiple forms of pharmacological intervention, including anti-dementia drugs for cognitive impairment, antidepressants, antipsychotics, anxiolytics and sedatives for behavioral and psychological symptoms of dementia, and other drugs for comorbid disorders. The pharmacotherapy of dementia patients has been characterized by a significant interindividual variability in drug response and the development of adverse drug effects. The therapeutic response to currently available drugs is partially effective in only some individuals, with side effects, drug interactions, intolerance, and non-compliance occurring in the majority of dementia patients. Therefore, understanding the genetic basis of a patient's response to pharmacotherapy might help clinicians select the most effective treatment for dementia while minimizing the likelihood of adverse reactions and drug interactions. Recent advances in pharmacogenomics may contribute to the individualization and optimization of dementia pharmacotherapy by increasing its efficacy and safety via a prediction of clinical outcomes. Thus, it can significantly improve the quality of life in dementia patients.
Collapse
Affiliation(s)
- Barbara Vuic
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| | - Tina Milos
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| | - Lucija Tudor
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| | - Matea Nikolac Perkovic
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| | - Marcela Konjevod
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| | - Gordana Nedic Erjavec
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| | - Vladimir Farkas
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| | - Suzana Uzun
- Department for Biological Psychiatry and Psychogeriatry, University Hospital Vrapce, 10000 Zagreb, Croatia; (S.U.); (N.M.)
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Ninoslav Mimica
- Department for Biological Psychiatry and Psychogeriatry, University Hospital Vrapce, 10000 Zagreb, Croatia; (S.U.); (N.M.)
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Dubravka Svob Strac
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| |
Collapse
|
10
|
Rajeev V, Chai YL, Poh L, Selvaraji S, Fann DY, Jo DG, De Silva TM, Drummond GR, Sobey CG, Arumugam TV, Chen CP, Lai MKP. Chronic cerebral hypoperfusion: a critical feature in unravelling the etiology of vascular cognitive impairment. Acta Neuropathol Commun 2023; 11:93. [PMID: 37309012 PMCID: PMC10259064 DOI: 10.1186/s40478-023-01590-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/14/2023] Open
Abstract
Vascular cognitive impairment (VCI) describes a wide spectrum of cognitive deficits related to cerebrovascular diseases. Although the loss of blood flow to cortical regions critically involved in cognitive processes must feature as the main driver of VCI, the underlying mechanisms and interactions with related disease processes remain to be fully elucidated. Recent clinical studies of cerebral blood flow measurements have supported the role of chronic cerebral hypoperfusion (CCH) as a major driver of the vascular pathology and clinical manifestations of VCI. Here we review the pathophysiological mechanisms as well as neuropathological changes of CCH. Potential interventional strategies for VCI are also reviewed. A deeper understanding of how CCH can lead to accumulation of VCI-associated pathology could potentially pave the way for early detection and development of disease-modifying therapies, thus allowing preventive interventions instead of symptomatic treatments.
Collapse
Affiliation(s)
- Vismitha Rajeev
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
| | - Yuek Ling Chai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
| | - Luting Poh
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
| | - Sharmelee Selvaraji
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore, Singapore, Singapore
| | - David Y Fann
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - T Michael De Silva
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Grant R Drummond
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Christopher G Sobey
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Thiruma V Arumugam
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Christopher P Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
- NUS Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore.
- NUS Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
11
|
Granata A. Functional genomics in stroke: current and future applications of iPSCs and gene editing to dissect the function of risk variants. BMC Cardiovasc Disord 2023; 23:223. [PMID: 37120540 PMCID: PMC10148993 DOI: 10.1186/s12872-023-03227-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 04/04/2023] [Indexed: 05/01/2023] Open
Abstract
Stroke is an important disease with unmet clinical need. To uncover novel paths for treatment, it is of critical importance to develop relevant laboratory models that may help to shed light on the pathophysiological mechanisms of stroke. Induced pluripotent stem cells (iPSCs) technology has enormous potential to advance our knowledge into stroke by creating novel human models for research and therapeutic testing. iPSCs models generated from patients with specific stroke types and specific genetic predisposition in combination with other state of art technologies including genome editing, multi-omics, 3D system, libraries screening, offer the opportunity to investigate disease-related pathways and identify potential novel therapeutic targets that can then be tested in these models. Thus, iPSCs offer an unprecedented opportunity to make rapid progress in the field of stroke and vascular dementia research leading to clinical translation. This review paper summarizes some of the key areas in which patient-derived iPSCs technology has been applied to disease modelling and discusses the ongoing challenges and the future directions for the application of this technology in the field of stroke research.
Collapse
Affiliation(s)
- Alessandra Granata
- Department of Clinical Neurosciences, Victor Phillip Dahdaleh Heart & Lung Research Institute, Papworth Road, Cambridge Biomedical Campus, University of Cambridge, Cambridge, CB2 0BB, UK.
| |
Collapse
|
12
|
Myosin light chain phosphorylation exhibits a gradient across the wall of cerebellar arteries under sustained ex vivo vascular tone. Sci Rep 2023; 13:909. [PMID: 36650375 PMCID: PMC9845333 DOI: 10.1038/s41598-023-28092-3] [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: 06/14/2022] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Small blood vessel diseases are often associated with impaired regulation of vascular tone. The current understanding of resistance arteries often focuses on how a level of vascular tone is achieved in the acute phase, while less emphasis is placed on mechanisms that maintain vascular tone. In this study, cannulated rat superior cerebellar arteries (SCA) developed spontaneous myogenic tone and showed a marked and sustained constriction in the presence of diluted serum (10%), a stimulus relevant to cerebrovascular disease. Both phosphorylated myosin light chain (MLC-p) and smooth muscle alpha actin (SM-α-actin) aligned with phalloidin-stained actin filaments in the vessel wall, while exhibiting a 'high to low' gradient across the layers of vascular smooth muscle cells (VSMC), peaking in the outer layer. The MLC-p distribution profile shifted towards the adventitia in serum treated vessels, while removal of the serum reversed it. Furthermore, a positive correlation between the MLC-p signal and vessel wall tension was also evident. The gradients of phosphorylated MLC and SM-α-actin are consistent with a spatial regulation of the myosin-actin apparatus in the vessel wall during the maintenance of vascular tone. Further, the changing profiles of MLC-p and SM-α-actin are consistent with SCA vasoconstriction being accompanied by VSMC cytoskeletal reorganization.
Collapse
|
13
|
Branyan K, Labelle-Dumais C, Wang X, Hayashi G, Lee B, Peltz Z, Gorman S, Li BQ, Mao M, Gould DB. Elevated TGFβ signaling contributes to cerebral small vessel disease in mouse models of Gould syndrome. Matrix Biol 2023; 115:48-70. [PMID: 36435425 PMCID: PMC10393528 DOI: 10.1016/j.matbio.2022.11.007] [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: 06/26/2022] [Revised: 11/21/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022]
Abstract
Cerebral small vessel disease (CSVD) is a leading cause of stroke and vascular cognitive impairment and dementia. Studying monogenic CSVD can reveal pathways that are dysregulated in common sporadic forms of the disease and may represent therapeutic targets. Mutations in collagen type IV alpha 1 (COL4A1) and alpha 2 (COL4A2) cause highly penetrant CSVD as part of a multisystem disorder referred to as Gould syndrome. COL4A1 and COL4A2 form heterotrimers [a1α1α2(IV)] that are fundamental constituents of basement membranes. However, their functions are poorly understood and the mechanism(s) by which COL4A1 and COL4A2 mutations cause CSVD are unknown. We used histological, molecular, genetic, pharmacological, and in vivo imaging approaches to characterize central nervous system (CNS) vascular pathologies in Col4a1 mutant mouse models of monogenic CSVD to provide insight into underlying pathogenic mechanisms. We describe developmental CNS angiogenesis abnormalities characterized by impaired retinal vascular outgrowth and patterning, increased numbers of mural cells with abnormal morphologies, altered contractile protein expression in vascular smooth muscle cells (VSMCs) and age-related loss of arteriolar VSMCs in Col4a1 mutant mice. Importantly, we identified elevated TGFβ signaling as a pathogenic consequence of Col4a1 mutations and show that genetically suppressing TGFβ signaling ameliorated CNS vascular pathologies, including partial rescue of retinal vascular patterning defects, prevention of VSMC loss, and significant reduction of intracerebral hemorrhages in Col4a1 mutant mice aged up to 8 months. This study identifies a novel biological role for collagen α1α1α2(IV) as a regulator of TGFβ signaling and demonstrates that elevated TGFβ signaling contributes to CNS vascular pathologies caused by Col4a1 mutations. Our findings suggest that pharmacologically suppressing TGFβ signaling could reduce the severity of CSVD, and potentially other manifestations associated with Gould syndrome and have important translational implications that could extend to idiopathic forms of CSVD.
Collapse
Affiliation(s)
- Kayla Branyan
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States
| | - Cassandre Labelle-Dumais
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States
| | - Xiaowei Wang
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States
| | - Genki Hayashi
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States
| | - Bryson Lee
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States
| | - Zoe Peltz
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States
| | - Seán Gorman
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States
| | - Bo Qiao Li
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States
| | - Mao Mao
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States
| | - Douglas B Gould
- Department of Ophthalmology, University of California, 555 Mission Bay Boulevard South, San Francisco, CA 94158, United States; Department of Anatomy, Cardiovascular Research Institute, Bakar Aging Research Institute, and Institute for Human Genetics, University of California, San Francisco, United States.
| |
Collapse
|
14
|
Ferguson AC, Thrippleton S, Henshall D, Whittaker E, Conway B, MacLeod M, Malik R, Rawlik K, Tenesa A, Sudlow C, Rannikmae K. Frequency and Phenotype Associations of Rare Variants in 5 Monogenic Cerebral Small Vessel Disease Genes in 200,000 UK Biobank Participants. Neurol Genet 2022; 8:e200015. [PMID: 36035235 PMCID: PMC9403885 DOI: 10.1212/nxg.0000000000200015] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 06/17/2022] [Indexed: 04/14/2023]
Abstract
Background and Objectives Based on previous case reports and disease-based cohorts, a minority of patients with cerebral small vessel disease (cSVD) have a monogenic cause, with many also manifesting extracerebral phenotypes. We investigated the frequency, penetrance, and phenotype associations of putative pathogenic variants in cSVD genes in the UK Biobank (UKB), a large population-based study. Methods We used a systematic review of previous literature and ClinVar to identify putative pathogenic rare variants in CTSA, TREX1, HTRA1, and COL4A1/2. We mapped phenotypes previously attributed to these variants (phenotypes-of-interest) to disease coding systems used in the UKB's linked health data from UK hospital admissions, death records, and primary care. Among 199,313 exome-sequenced UKB participants, we assessed the following: the proportion of participants carrying ≥1 variant(s); phenotype-of-interest penetrance; and the association between variant carrier status and phenotypes-of-interest using a binary (any phenotype present/absent) and phenotype burden (linear score of the number of phenotypes a participant possessed) approach. Results Among UKB participants, 0.5% had ≥1 variant(s) in studied genes. Using hospital admission and death records, 4%-20% of variant carriers per gene had an associated phenotype. This increased to 7%-55% when including primary care records. Only COL4A1 variant carrier status was significantly associated with having ≥1 phenotype-of-interest and a higher phenotype score (OR = 1.29, p = 0.006). Discussion While putative pathogenic rare variants in monogenic cSVD genes occur in 1:200 people in the UKB population, only approximately half of variant carriers have a relevant disease phenotype recorded in their linked health data. We could not replicate most previously reported gene-phenotype associations, suggesting lower penetrance rates, overestimated pathogenicity, and/or limited statistical power.
Collapse
Affiliation(s)
- Amy Christina Ferguson
- From the Centre for Medical Informatics (A.C.F., D.H., A.T., K.Rannikmae), Usher Institute, University of Edinburgh; Edinburgh Medical School (S.T., E.W.), University of Edinburgh; Centre for Cardiovascular Science (B.C.), The Queen's Medical Research Institute, University of Edinburgh; Centre for Clinical Brain Sciences (M.M.), University of Edinburgh, United Kingdom; Institute for Stroke and Dementia Research (ISD) (R.M.), University Hospital, LMU Munich, Germany; The Roslin Institute (K. Rawlik, A.T.), University of Edinburgh; MRC Human Genetics Unit (A.T.), Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital; and BHF Data Science Centre (C.S.), Health Death Research UK, London, United Kingdom
| | - Sophie Thrippleton
- From the Centre for Medical Informatics (A.C.F., D.H., A.T., K.Rannikmae), Usher Institute, University of Edinburgh; Edinburgh Medical School (S.T., E.W.), University of Edinburgh; Centre for Cardiovascular Science (B.C.), The Queen's Medical Research Institute, University of Edinburgh; Centre for Clinical Brain Sciences (M.M.), University of Edinburgh, United Kingdom; Institute for Stroke and Dementia Research (ISD) (R.M.), University Hospital, LMU Munich, Germany; The Roslin Institute (K. Rawlik, A.T.), University of Edinburgh; MRC Human Genetics Unit (A.T.), Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital; and BHF Data Science Centre (C.S.), Health Death Research UK, London, United Kingdom
| | - David Henshall
- From the Centre for Medical Informatics (A.C.F., D.H., A.T., K.Rannikmae), Usher Institute, University of Edinburgh; Edinburgh Medical School (S.T., E.W.), University of Edinburgh; Centre for Cardiovascular Science (B.C.), The Queen's Medical Research Institute, University of Edinburgh; Centre for Clinical Brain Sciences (M.M.), University of Edinburgh, United Kingdom; Institute for Stroke and Dementia Research (ISD) (R.M.), University Hospital, LMU Munich, Germany; The Roslin Institute (K. Rawlik, A.T.), University of Edinburgh; MRC Human Genetics Unit (A.T.), Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital; and BHF Data Science Centre (C.S.), Health Death Research UK, London, United Kingdom
| | - Ed Whittaker
- From the Centre for Medical Informatics (A.C.F., D.H., A.T., K.Rannikmae), Usher Institute, University of Edinburgh; Edinburgh Medical School (S.T., E.W.), University of Edinburgh; Centre for Cardiovascular Science (B.C.), The Queen's Medical Research Institute, University of Edinburgh; Centre for Clinical Brain Sciences (M.M.), University of Edinburgh, United Kingdom; Institute for Stroke and Dementia Research (ISD) (R.M.), University Hospital, LMU Munich, Germany; The Roslin Institute (K. Rawlik, A.T.), University of Edinburgh; MRC Human Genetics Unit (A.T.), Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital; and BHF Data Science Centre (C.S.), Health Death Research UK, London, United Kingdom
| | - Bryan Conway
- From the Centre for Medical Informatics (A.C.F., D.H., A.T., K.Rannikmae), Usher Institute, University of Edinburgh; Edinburgh Medical School (S.T., E.W.), University of Edinburgh; Centre for Cardiovascular Science (B.C.), The Queen's Medical Research Institute, University of Edinburgh; Centre for Clinical Brain Sciences (M.M.), University of Edinburgh, United Kingdom; Institute for Stroke and Dementia Research (ISD) (R.M.), University Hospital, LMU Munich, Germany; The Roslin Institute (K. Rawlik, A.T.), University of Edinburgh; MRC Human Genetics Unit (A.T.), Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital; and BHF Data Science Centre (C.S.), Health Death Research UK, London, United Kingdom
| | - Malcolm MacLeod
- From the Centre for Medical Informatics (A.C.F., D.H., A.T., K.Rannikmae), Usher Institute, University of Edinburgh; Edinburgh Medical School (S.T., E.W.), University of Edinburgh; Centre for Cardiovascular Science (B.C.), The Queen's Medical Research Institute, University of Edinburgh; Centre for Clinical Brain Sciences (M.M.), University of Edinburgh, United Kingdom; Institute for Stroke and Dementia Research (ISD) (R.M.), University Hospital, LMU Munich, Germany; The Roslin Institute (K. Rawlik, A.T.), University of Edinburgh; MRC Human Genetics Unit (A.T.), Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital; and BHF Data Science Centre (C.S.), Health Death Research UK, London, United Kingdom
| | - Rainer Malik
- From the Centre for Medical Informatics (A.C.F., D.H., A.T., K.Rannikmae), Usher Institute, University of Edinburgh; Edinburgh Medical School (S.T., E.W.), University of Edinburgh; Centre for Cardiovascular Science (B.C.), The Queen's Medical Research Institute, University of Edinburgh; Centre for Clinical Brain Sciences (M.M.), University of Edinburgh, United Kingdom; Institute for Stroke and Dementia Research (ISD) (R.M.), University Hospital, LMU Munich, Germany; The Roslin Institute (K. Rawlik, A.T.), University of Edinburgh; MRC Human Genetics Unit (A.T.), Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital; and BHF Data Science Centre (C.S.), Health Death Research UK, London, United Kingdom
| | - Konrad Rawlik
- From the Centre for Medical Informatics (A.C.F., D.H., A.T., K.Rannikmae), Usher Institute, University of Edinburgh; Edinburgh Medical School (S.T., E.W.), University of Edinburgh; Centre for Cardiovascular Science (B.C.), The Queen's Medical Research Institute, University of Edinburgh; Centre for Clinical Brain Sciences (M.M.), University of Edinburgh, United Kingdom; Institute for Stroke and Dementia Research (ISD) (R.M.), University Hospital, LMU Munich, Germany; The Roslin Institute (K. Rawlik, A.T.), University of Edinburgh; MRC Human Genetics Unit (A.T.), Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital; and BHF Data Science Centre (C.S.), Health Death Research UK, London, United Kingdom
| | - Albert Tenesa
- From the Centre for Medical Informatics (A.C.F., D.H., A.T., K.Rannikmae), Usher Institute, University of Edinburgh; Edinburgh Medical School (S.T., E.W.), University of Edinburgh; Centre for Cardiovascular Science (B.C.), The Queen's Medical Research Institute, University of Edinburgh; Centre for Clinical Brain Sciences (M.M.), University of Edinburgh, United Kingdom; Institute for Stroke and Dementia Research (ISD) (R.M.), University Hospital, LMU Munich, Germany; The Roslin Institute (K. Rawlik, A.T.), University of Edinburgh; MRC Human Genetics Unit (A.T.), Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital; and BHF Data Science Centre (C.S.), Health Death Research UK, London, United Kingdom
| | - Cathie Sudlow
- From the Centre for Medical Informatics (A.C.F., D.H., A.T., K.Rannikmae), Usher Institute, University of Edinburgh; Edinburgh Medical School (S.T., E.W.), University of Edinburgh; Centre for Cardiovascular Science (B.C.), The Queen's Medical Research Institute, University of Edinburgh; Centre for Clinical Brain Sciences (M.M.), University of Edinburgh, United Kingdom; Institute for Stroke and Dementia Research (ISD) (R.M.), University Hospital, LMU Munich, Germany; The Roslin Institute (K. Rawlik, A.T.), University of Edinburgh; MRC Human Genetics Unit (A.T.), Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital; and BHF Data Science Centre (C.S.), Health Death Research UK, London, United Kingdom
| | - Kristiina Rannikmae
- From the Centre for Medical Informatics (A.C.F., D.H., A.T., K.Rannikmae), Usher Institute, University of Edinburgh; Edinburgh Medical School (S.T., E.W.), University of Edinburgh; Centre for Cardiovascular Science (B.C.), The Queen's Medical Research Institute, University of Edinburgh; Centre for Clinical Brain Sciences (M.M.), University of Edinburgh, United Kingdom; Institute for Stroke and Dementia Research (ISD) (R.M.), University Hospital, LMU Munich, Germany; The Roslin Institute (K. Rawlik, A.T.), University of Edinburgh; MRC Human Genetics Unit (A.T.), Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital; and BHF Data Science Centre (C.S.), Health Death Research UK, London, United Kingdom
| |
Collapse
|
15
|
Cheng YW, Chao CC, Chen CH, Yeh TY, Jeng JS, Tang SC, Hsieh ST. Small Fiber Pathology in CADASIL: Clinical Correlation With Cognitive Impairment. Neurology 2022; 99:e583-e593. [PMID: 35584924 PMCID: PMC9442619 DOI: 10.1212/wnl.0000000000200672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/16/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES This study investigated the cutaneous small fiber pathology of cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) and its clinical significance, that is, the NOTCH3 deposition in cutaneous vasculatures and CNS neurodegeneration focusing on cognitive impairment. METHODS Thirty-seven patients with CADASIL and 59 age-matched healthy controls were enrolled to evaluate cutaneous small fiber pathology by quantitative measures of intraepidermal nerve fiber density (IENFD), sweat gland innervation, and vascular innervation. Cognitive performance of patients with CADASIL was evaluated by a comprehensive neuropsychological assessment, and its association with small fiber pathology was tested using multivariable linear regression analysis adjusted for age and diabetes mellitus. We further assessed the relationships of IENFD with cutaneous vascular NOTCH3 ectodomain (NOTCH3ECD) deposition and biomarkers of neurodegeneration including structural brain MRI measures, serum neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP), tau, and ubiquitin carboxy-terminal hydrolase L1. RESULTS Patients with CADASIL showed reduced IENFD (5.22 ± 2.42 vs 7.88 ± 2.89 fibers/mm, p = 0.0001) and reduced sweat gland (p < 0.0001) and vascular (p < 0.0001) innervations compared with age-matched controls. Reduced IENFD was associated with impaired global cognition measured by Mini-Mental State Examination (B = 1.062, 95% CI = 0.370-1.753, p = 0.004), and this association remained after adjustment for age and diabetes mellitus (p = 0.043). In addition, IENFD in patients with CADASIL was associated with mean cortical thickness (Pearson r = 0.565, p = 0.0023) but not white matter hyperintensity volume, total lacune count, or total microbleed count. Reduced IENFD was associated with cutaneous vascular NOTCH3ECD deposition amount among patients harboring pathogenic variants in exon 11 (mainly p.R544C) (B = -0.092, 95% CI = -0.175 to -0.009, p = 0.031). Compared with those with normal cognition, patients with CADASIL with cognitive impairment had an elevated plasma NfL level regardless of concurrent small fiber denervation, whereas only patients with both cognitive impairment and small fiber denervation showed an elevated plasma GFAP level. DISCUSSION Cutaneous small fiber pathology correlates with cognitive impairment and CNS neurodegeneration in patients with CADASIL, indicating a peripheral neurodegenerative process related to NOTCH3ECD aggregation.
Collapse
Affiliation(s)
- Yu-Wen Cheng
- From the Department of Neurology (Y.-W.C.), National Taiwan University Hospital, Hsin-Chu Branch; Graduate Institute of Clinical Medicine (Y.-W.C.), College of Medicine, National Taiwan University; Department of Neurology (C.-C.C., C.-H.C., J.-S.J., S.-C.T., S.-T.H.), National Taiwan University Hospital; Department of Anatomy and Cell Biology (T.-Y.Y., S.-T.H.), College of Medicine, National Taiwan University; Graduate Institute of Brain and Mind Sciences (S.-T.H.), College of Medicine, National Taiwan University; and Center of Precision Medicine (S.-T.H.), National Taiwan University College of Medicine, Taipei
| | - Chi-Chao Chao
- From the Department of Neurology (Y.-W.C.), National Taiwan University Hospital, Hsin-Chu Branch; Graduate Institute of Clinical Medicine (Y.-W.C.), College of Medicine, National Taiwan University; Department of Neurology (C.-C.C., C.-H.C., J.-S.J., S.-C.T., S.-T.H.), National Taiwan University Hospital; Department of Anatomy and Cell Biology (T.-Y.Y., S.-T.H.), College of Medicine, National Taiwan University; Graduate Institute of Brain and Mind Sciences (S.-T.H.), College of Medicine, National Taiwan University; and Center of Precision Medicine (S.-T.H.), National Taiwan University College of Medicine, Taipei
| | - Chih-Hao Chen
- From the Department of Neurology (Y.-W.C.), National Taiwan University Hospital, Hsin-Chu Branch; Graduate Institute of Clinical Medicine (Y.-W.C.), College of Medicine, National Taiwan University; Department of Neurology (C.-C.C., C.-H.C., J.-S.J., S.-C.T., S.-T.H.), National Taiwan University Hospital; Department of Anatomy and Cell Biology (T.-Y.Y., S.-T.H.), College of Medicine, National Taiwan University; Graduate Institute of Brain and Mind Sciences (S.-T.H.), College of Medicine, National Taiwan University; and Center of Precision Medicine (S.-T.H.), National Taiwan University College of Medicine, Taipei
| | - Ti-Yen Yeh
- From the Department of Neurology (Y.-W.C.), National Taiwan University Hospital, Hsin-Chu Branch; Graduate Institute of Clinical Medicine (Y.-W.C.), College of Medicine, National Taiwan University; Department of Neurology (C.-C.C., C.-H.C., J.-S.J., S.-C.T., S.-T.H.), National Taiwan University Hospital; Department of Anatomy and Cell Biology (T.-Y.Y., S.-T.H.), College of Medicine, National Taiwan University; Graduate Institute of Brain and Mind Sciences (S.-T.H.), College of Medicine, National Taiwan University; and Center of Precision Medicine (S.-T.H.), National Taiwan University College of Medicine, Taipei
| | - Jiann-Shing Jeng
- From the Department of Neurology (Y.-W.C.), National Taiwan University Hospital, Hsin-Chu Branch; Graduate Institute of Clinical Medicine (Y.-W.C.), College of Medicine, National Taiwan University; Department of Neurology (C.-C.C., C.-H.C., J.-S.J., S.-C.T., S.-T.H.), National Taiwan University Hospital; Department of Anatomy and Cell Biology (T.-Y.Y., S.-T.H.), College of Medicine, National Taiwan University; Graduate Institute of Brain and Mind Sciences (S.-T.H.), College of Medicine, National Taiwan University; and Center of Precision Medicine (S.-T.H.), National Taiwan University College of Medicine, Taipei
| | - Sung-Chun Tang
- From the Department of Neurology (Y.-W.C.), National Taiwan University Hospital, Hsin-Chu Branch; Graduate Institute of Clinical Medicine (Y.-W.C.), College of Medicine, National Taiwan University; Department of Neurology (C.-C.C., C.-H.C., J.-S.J., S.-C.T., S.-T.H.), National Taiwan University Hospital; Department of Anatomy and Cell Biology (T.-Y.Y., S.-T.H.), College of Medicine, National Taiwan University; Graduate Institute of Brain and Mind Sciences (S.-T.H.), College of Medicine, National Taiwan University; and Center of Precision Medicine (S.-T.H.), National Taiwan University College of Medicine, Taipei
| | - Sung-Tsang Hsieh
- From the Department of Neurology (Y.-W.C.), National Taiwan University Hospital, Hsin-Chu Branch; Graduate Institute of Clinical Medicine (Y.-W.C.), College of Medicine, National Taiwan University; Department of Neurology (C.-C.C., C.-H.C., J.-S.J., S.-C.T., S.-T.H.), National Taiwan University Hospital; Department of Anatomy and Cell Biology (T.-Y.Y., S.-T.H.), College of Medicine, National Taiwan University; Graduate Institute of Brain and Mind Sciences (S.-T.H.), College of Medicine, National Taiwan University; and Center of Precision Medicine (S.-T.H.), National Taiwan University College of Medicine, Taipei.
| |
Collapse
|
16
|
Ren B, Tan L, Song Y, Li D, Xue B, Lai X, Gao Y. Cerebral Small Vessel Disease: Neuroimaging Features, Biochemical Markers, Influencing Factors, Pathological Mechanism and Treatment. Front Neurol 2022; 13:843953. [PMID: 35775047 PMCID: PMC9237477 DOI: 10.3389/fneur.2022.843953] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 05/12/2022] [Indexed: 01/15/2023] Open
Abstract
Cerebral small vessel disease (CSVD) is the most common chronic vascular disease involving the whole brain. Great progress has been made in clinical imaging, pathological mechanism, and treatment of CSVD, but many problems remain. Clarifying the current research dilemmas and future development direction of CSVD can provide new ideas for both basic and clinical research. In this review, the risk factors, biological markers, pathological mechanisms, and the treatment of CSVD will be systematically illustrated to provide the current research status of CSVD. The future development direction of CSVD will be elucidated by summarizing the research difficulties.
Collapse
Affiliation(s)
- Beida Ren
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
- Chinese Medicine Key Research Room of Brain Disorders Syndrome and Treatment of the National Administration of Traditonal Chinese Medicine, Beijing, China
| | - Ling Tan
- Department of Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuebo Song
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Danxi Li
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
- Chinese Medicine Key Research Room of Brain Disorders Syndrome and Treatment of the National Administration of Traditonal Chinese Medicine, Beijing, China
| | - Bingjie Xue
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
- Chinese Medicine Key Research Room of Brain Disorders Syndrome and Treatment of the National Administration of Traditonal Chinese Medicine, Beijing, China
| | - Xinxing Lai
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
| | - Ying Gao
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
17
|
Abstract
Stroke is the second leading cause of death worldwide and a complex, heterogeneous condition. In this review, we provide an overview of the current knowledge on monogenic and multifactorial forms of stroke, highlighting recent insight into the continuum between these. We describe how, in recent years, large-scale genome-wide association studies have enabled major progress in deciphering the genetic basis for stroke and its subtypes, although more research is needed to interpret these findings. We cover the potential of stroke genetics to reveal novel pathophysiological processes underlying stroke, to accelerate the discovery of new therapeutic approaches, and to identify individuals in the population who are at high risk of stroke and could be targeted for tailored preventative interventions.
Collapse
Affiliation(s)
- Stéphanie Debette
- Bordeaux Population Health Research Center, Inserm U1219, University of Bordeaux, France (S.D.).,Department of Neurology, Bordeaux University Hospital, Institute for Neurodegenerative Diseases, France (S.D.)
| | - Hugh S Markus
- Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, United Kingdom (H.S.M.)
| |
Collapse
|
18
|
Connectomic-genetic signatures in the cerebral small vessel disease. Neurobiol Dis 2022; 167:105671. [DOI: 10.1016/j.nbd.2022.105671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 01/31/2022] [Accepted: 02/21/2022] [Indexed: 11/19/2022] Open
|
19
|
Abstract
Inflammation and its myriad pathways are now recognized to play both causal and consequential roles in vascular brain health. From acting as a trigger for vascular brain injury, as evidenced by the coronavirus disease 2019 (COVID-19) pandemic, to steadily increasing the risk for chronic cerebrovascular disease, distinct inflammatory cascades play differential roles in varying states of cerebrovascular injury. New evidence is regularly emerging that characterizes the role of specific inflammatory pathways in these varying states including those at risk for stroke and chronic cerebrovascular injury as well as during the acute, subacute, and repair phases of stroke. Here, we aim to highlight recent basic science and clinical evidence for many distinct inflammatory cascades active in these varying states of cerebrovascular injury. The role of cerebrovascular infections, spotlighted by the severe acute respiratory syndrome coronavirus 2 pandemic, and its association with increased stroke risk is also reviewed. Rather than converging on a shared mechanism, these emerging studies implicate varied and distinct inflammatory processes in vascular brain injury and repair. Recognition of the phasic nature of inflammatory cascades on varying states of cerebrovascular disease is likely essential to the development and implementation of an anti-inflammatory strategy in the prevention, treatment, and repair of vascular brain injury. Although advances in revascularization have taught us that time is brain, targeting inflammation for the treatment of cerebrovascular disease will undoubtedly show us that timing is brain.
Collapse
Affiliation(s)
- Katherine T Mun
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles
| | - Jason D Hinman
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles
| |
Collapse
|
20
|
Lecordier S, Manrique-Castano D, El Moghrabi Y, ElAli A. Neurovascular Alterations in Vascular Dementia: Emphasis on Risk Factors. Front Aging Neurosci 2021; 13:727590. [PMID: 34566627 PMCID: PMC8461067 DOI: 10.3389/fnagi.2021.727590] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 08/05/2021] [Indexed: 12/25/2022] Open
Abstract
Vascular dementia (VaD) constitutes the second most prevalent cause of dementia in the world after Alzheimer’s disease (AD). VaD regroups heterogeneous neurological conditions in which the decline of cognitive functions, including executive functions, is associated with structural and functional alterations in the cerebral vasculature. Among these cerebrovascular disorders, major stroke, and cerebral small vessel disease (cSVD) constitute the major risk factors for VaD. These conditions alter neurovascular functions leading to blood-brain barrier (BBB) deregulation, neurovascular coupling dysfunction, and inflammation. Accumulation of neurovascular impairments over time underlies the cognitive function decline associated with VaD. Furthermore, several vascular risk factors, such as hypertension, obesity, and diabetes have been shown to exacerbate neurovascular impairments and thus increase VaD prevalence. Importantly, air pollution constitutes an underestimated risk factor that triggers vascular dysfunction via inflammation and oxidative stress. The review summarizes the current knowledge related to the pathological mechanisms linking neurovascular impairments associated with stroke, cSVD, and vascular risk factors with a particular emphasis on air pollution, to VaD etiology and progression. Furthermore, the review discusses the major challenges to fully elucidate the pathobiology of VaD, as well as research directions to outline new therapeutic interventions.
Collapse
Affiliation(s)
- Sarah Lecordier
- Neuroscience Axis, Research Center of CHU de Québec-Université Laval, Québec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Daniel Manrique-Castano
- Neuroscience Axis, Research Center of CHU de Québec-Université Laval, Québec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Yara El Moghrabi
- Neuroscience Axis, Research Center of CHU de Québec-Université Laval, Québec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| | - Ayman ElAli
- Neuroscience Axis, Research Center of CHU de Québec-Université Laval, Québec City, QC, Canada.,Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada
| |
Collapse
|
21
|
Pokhilko A, Brezzo G, Handunnetthi L, Heilig R, Lennon R, Smith C, Allan SM, Granata A, Sinha S, Wang T, Markus HS, Naba A, Fischer R, Van Agtmael T, Horsburgh K, Cader MZ. Global proteomic analysis of extracellular matrix in mouse and human brain highlights relevance to cerebrovascular disease. J Cereb Blood Flow Metab 2021; 41:2423-2438. [PMID: 33730931 PMCID: PMC8392779 DOI: 10.1177/0271678x211004307] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The extracellular matrix (ECM) is a key interface between the cerebrovasculature and adjacent brain tissues. Deregulation of the ECM contributes to a broad range of neurological disorders. However, despite this importance, our understanding of the ECM composition remains very limited mainly due to difficulties in its isolation. To address this, we developed an approach to extract the cerebrovascular ECM from mouse and human post-mortem normal brain tissues. We then used mass spectrometry with off-line high-pH reversed-phase fractionation to increase the protein detection. This identified more than 1000 proteins in the ECM-enriched fraction, with > 66% of the proteins being common between the species. We report 147 core ECM proteins of the human brain vascular matrisome, including collagens, laminins, fibronectin and nidogens. We next used network analysis to identify the connection between the brain ECM proteins and cerebrovascular diseases. We found that genes related to cerebrovascular diseases, such as COL4A1, COL4A2, VCAN and APOE were significantly enriched in the cerebrovascular ECM network. This provides unique mechanistic insight into cerebrovascular disease and potential drug targets. Overall, we provide a powerful resource to study the functions of brain ECM and highlight a specific role for brain vascular ECM in cerebral vascular disease.
Collapse
Affiliation(s)
- Alexandra Pokhilko
- Translational Molecular Neuroscience Group, Weatherall Institute of Molecular Medicine, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Gaia Brezzo
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK.,Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | | | - Raphael Heilig
- Discovery Proteomics Facility, Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Rachel Lennon
- Division of Cell-Matrix Biology and Regenerative Medicine, Wellcome Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK.,Department of Paediatric Nephrology, Royal Manchester Children's Hospital, Manchester University Hospitals National Health Service Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Colin Smith
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Stuart M Allan
- Lydia Becker Institute of Immunology and Inflammation, Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Alessandra Granata
- Clinical Neurosciences Department, University of Cambridge, Cambridge, UK
| | | | - Tao Wang
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Hugh S Markus
- Department of Neurology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Alexandra Naba
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Roman Fischer
- Discovery Proteomics Facility, Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Tom Van Agtmael
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Karen Horsburgh
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - M Zameel Cader
- Translational Molecular Neuroscience Group, Weatherall Institute of Molecular Medicine, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
22
|
Humphreys CA, Smith C, Wardlaw JM. Correlations in post-mortem imaging-histopathology studies of sporadic human cerebral small vessel disease: A systematic review. Neuropathol Appl Neurobiol 2021; 47:910-930. [PMID: 34037264 DOI: 10.1111/nan.12737] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/29/2021] [Accepted: 05/02/2021] [Indexed: 11/30/2022]
Abstract
AIMS Sporadic human cerebral small vessel disease (SVD) commonly causes stroke and dementia but its pathogenesis is poorly understood. There are recognised neuroimaging and histopathological features. However, relatively few studies have examined the relationship between the radiological and pathological correlates of SVD; better correlation would promote greater insight into the underlying biological changes. METHODS We performed a systematic review to collate and appraise the information derived from studies that correlated histological with neuroimaging-defined SVD lesions. We searched for studies describing post-mortem imaging and histological tissue examination in adults, extracted data from published studies, categorised the information and compiled this narrative. RESULTS We identified 38 relevant studies, including at least 1146 subjects, 342 of these with SVD: 29 studies focussed on neuroradiological white matter lesions (WML), six on microinfarcts and three on dilated perivascular spaces (PVS) and lacunes. The histopathology terminology was diverse with few robust definitions. Reporting and methodology varied widely between studies, precluding formal meta-analysis. PVS and 'oedema' were frequent findings in WML, being described in at least 94 and 18 radiological WML, respectively, in addition to myelin pallor. Histopathological changes extended beyond the radiological lesion margins in at least 33 radiological WML. At least 43 radiological lesions not seen pathologically and at least 178 histological lesions were not identified on imaging. CONCLUSIONS Histopathological assessment of human SVD is hindered by inconsistent methodological approaches and unstandardised definitions. The data from this systematic review will help to develop standardised definitions to promote consistency in human SVD research.
Collapse
Affiliation(s)
| | - Colin Smith
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Joanna M Wardlaw
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.,UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK.,Row Fogo Centre for Research into Ageing and the Brain, Edinburgh, UK
| |
Collapse
|
23
|
Traylor M, Persyn E, Tomppo L, Klasson S, Abedi V, Bakker MK, Torres N, Li L, Bell S, Rutten-Jacobs L, Tozer DJ, Griessenauer CJ, Zhang Y, Pedersen A, Sharma P, Jimenez-Conde J, Rundek T, Grewal RP, Lindgren A, Meschia JF, Salomaa V, Havulinna A, Kourkoulis C, Crawford K, Marini S, Mitchell BD, Kittner SJ, Rosand J, Dichgans M, Jern C, Strbian D, Fernandez-Cadenas I, Zand R, Ruigrok Y, Rost N, Lemmens R, Rothwell PM, Anderson CD, Wardlaw J, Lewis CM, Markus HS. Genetic basis of lacunar stroke: a pooled analysis of individual patient data and genome-wide association studies. Lancet Neurol 2021; 20:351-361. [PMID: 33773637 PMCID: PMC8062914 DOI: 10.1016/s1474-4422(21)00031-4] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 11/06/2020] [Accepted: 01/15/2021] [Indexed: 01/07/2023]
Abstract
BACKGROUND The genetic basis of lacunar stroke is poorly understood, with a single locus on 16q24 identified to date. We sought to identify novel associations and provide mechanistic insights into the disease. METHODS We did a pooled analysis of data from newly recruited patients with an MRI-confirmed diagnosis of lacunar stroke and existing genome-wide association studies (GWAS). Patients were recruited from hospitals in the UK as part of the UK DNA Lacunar Stroke studies 1 and 2 and from collaborators within the International Stroke Genetics Consortium. Cases and controls were stratified by ancestry and two meta-analyses were done: a European ancestry analysis, and a transethnic analysis that included all ancestry groups. We also did a multi-trait analysis of GWAS, in a joint analysis with a study of cerebral white matter hyperintensities (an aetiologically related radiological trait), to find additional genetic associations. We did a transcriptome-wide association study (TWAS) to detect genes for which expression is associated with lacunar stroke; identified significantly enriched pathways using multi-marker analysis of genomic annotation; and evaluated cardiovascular risk factors causally associated with the disease using mendelian randomisation. FINDINGS Our meta-analysis comprised studies from Europe, the USA, and Australia, including 7338 cases and 254 798 controls, of which 2987 cases (matched with 29 540 controls) were confirmed using MRI. Five loci (ICA1L-WDR12-CARF-NBEAL1, ULK4, SPI1-SLC39A13-PSMC3-RAPSN, ZCCHC14, ZBTB14-EPB41L3) were found to be associated with lacunar stroke in the European or transethnic meta-analyses. A further seven loci (SLC25A44-PMF1-BGLAP, LOX-ZNF474-LOC100505841, FOXF2-FOXQ1, VTA1-GPR126, SH3PXD2A, HTRA1-ARMS2, COL4A2) were found to be associated in the multi-trait analysis with cerebral white matter hyperintensities (n=42 310). Two of the identified loci contain genes (COL4A2 and HTRA1) that are involved in monogenic lacunar stroke. The TWAS identified associations between the expression of six genes (SCL25A44, ULK4, CARF, FAM117B, ICA1L, NBEAL1) and lacunar stroke. Pathway analyses implicated disruption of the extracellular matrix, phosphatidylinositol 5 phosphate binding, and roundabout binding (false discovery rate <0·05). Mendelian randomisation analyses identified positive associations of elevated blood pressure, history of smoking, and type 2 diabetes with lacunar stroke. INTERPRETATION Lacunar stroke has a substantial heritable component, with 12 loci now identified that could represent future treatment targets. These loci provide insights into lacunar stroke pathogenesis, highlighting disruption of the vascular extracellular matrix (COL4A2, LOX, SH3PXD2A, GPR126, HTRA1), pericyte differentiation (FOXF2, GPR126), TGF-β signalling (HTRA1), and myelination (ULK4, GPR126) in disease risk. FUNDING British Heart Foundation.
Collapse
Affiliation(s)
- Matthew Traylor
- Clinical Pharmacology and The Barts Heart Centre and NIHR Barts Biomedical Research Centre, Barts Health NHS Trust, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Elodie Persyn
- Department of Medical and Molecular Genetics, King's College London, London, UK
| | - Liisa Tomppo
- Department of Neurology, Helsinki University Hospital, Helsinki, Finland
| | - Sofia Klasson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Vida Abedi
- Department of Molecular and Functional Genomics, Weis Center for Research, Geisinger Health System, Danville, PA, USA
| | - Mark K Bakker
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands
| | - Nuria Torres
- Stroke Pharmacogenomics and Genetics, Sant Pau Institute of Research, Hospital de la Santa Creu I Sant Pau, Barcelona, Spain
| | - Linxin Li
- Centre for the Prevention of Stroke and Dementia, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK
| | - Steven Bell
- Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Loes Rutten-Jacobs
- Product Development Personalized Health Care, F Hoffmann-La Roche, Basel, Switzerland
| | - Daniel J Tozer
- Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Christoph J Griessenauer
- Neuroscience Institute, Geisinger Health System, Danville, PA, USA; Institute of Neurointervention, Paracelsus Medical University, Salzburg, Austria
| | - Yanfei Zhang
- Genomic Medicine Institute, Geisinger Health System, Danville, PA, USA
| | - Annie Pedersen
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Pankaj Sharma
- Institute of Cardiovascular Research, Royal Holloway University of London, London, UK
| | - Jordi Jimenez-Conde
- Neurovascular Research Group, Department of Neurology of Hospital del Mar-IMIM (Institut Hospital del Mar d'Investigacions Mediques), Universitat Autonoma de Barcelona/DCEXS-Universitat Pompeu Fabra, Barcelona, Spain
| | - Tatjana Rundek
- Evelyn F McKnight Brain Institute, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Raji P Grewal
- Neuroscience Institute, Saint Francis Medical Center, School of Health and Medical Sciences, Seton Hall University, South Orange, NJ, USA
| | - Arne Lindgren
- Department of Neurology, Skane University Hospital, Lund, Sweden; Department of Clinical Sciences Lund, Neurology, Lund University, Lund, Sweden
| | | | - Veikko Salomaa
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Aki Havulinna
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland; Institute for Molecular Medicine Finland (FIMM HiLIFE), Helsinki, Finland
| | - Christina Kourkoulis
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Henry and Allison McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA; Program in Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Katherine Crawford
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Program in Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Sandro Marini
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Program in Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Braxton D Mitchell
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA; Geriatrics Research and Education Clinical Center, Baltimore Veterans Administration Medical Center, Baltimore, MD, USA
| | - Steven J Kittner
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, USA; Geriatrics Research and Education Clinical Center, Baltimore Veterans Administration Medical Center, Baltimore, MD, USA
| | - Jonathan Rosand
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Henry and Allison McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA; Department of Neurology, Massachusetts General Hospital, Boston, MA, USA; Program in Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Martin Dichgans
- Institute for Stroke and Dementia Research (ISD), LMU Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Christina Jern
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Daniel Strbian
- Department of Neurology, Helsinki University Hospital, Helsinki, Finland; Clinical Neurosciences, University of Helsinki, Helsinki, Finland
| | - Israel Fernandez-Cadenas
- Stroke Pharmacogenomics and Genetics, Sant Pau Institute of Research, Hospital de la Santa Creu I Sant Pau, Barcelona, Spain; Neurovascular Research Laboratory and Neurovascular Unit, Institut de Recerca, Hospital Vall d'Hebron, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Ramin Zand
- Neuroscience Institute, Geisinger Health System, Danville, PA, USA
| | - Ynte Ruigrok
- Department of Neurology and Neurosurgery, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, Netherlands
| | - Natalia Rost
- J Philip Kistler Stroke Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Robin Lemmens
- Experimental Neurology, Department of Neurosciences, KU Leuven, Leuven, Belgium; VIB Center for Brain & Disease Research, Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Peter M Rothwell
- Centre for the Prevention of Stroke and Dementia, Nuffield Department of Clinical Neuroscience, University of Oxford, Oxford, UK
| | - Christopher D Anderson
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA; Henry and Allison McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA, USA; Department of Neurology, Massachusetts General Hospital, Boston, MA, USA; Program in Medical & Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Joanna Wardlaw
- Centre for Clinical Brain Sciences, UK Dementia Research Institute and Row Fogo Centre for Research into the Ageing Brain, University of Edinburgh, Edinburgh, UK
| | - Cathryn M Lewis
- Department of Medical and Molecular Genetics, King's College London, London, UK; Social, Genetic, and Developmental Psychiatry Centre, King's College London, London, UK
| | - Hugh S Markus
- Clinical Neurosciences, University of Cambridge, Cambridge, UK.
| |
Collapse
|
24
|
Wang HK, Huang CY, Chen YW, Sun YT. Hyperglycemia compromises the ischemia-provoked dedifferentiation of cerebral pericytes through p21-SOX2 signaling in high-fat diet-induced murine model. Diab Vasc Dis Res 2021; 18:1479164121990641. [PMID: 33557613 PMCID: PMC8482726 DOI: 10.1177/1479164121990641] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
AIM Diabetes-related cerebral small vessel disease (CSVD) causes neurological deficits. Patients with diabetes showed pericyte loss as a hallmark of retinopathy. Cerebral pericytes, which densely localize around brain capillaries, are quiescent stem cells regulating regeneration of brain and may have a role in CSVD development. This study investigated whether diabetes impairs ischemia-provoked dedifferentiation of pericytes. METHODS A murine high-fat diet (HFD)-induced diabetes model was used. After cerebral ischemia induction in the mice, pericytes were isolated and grown for a sphere formation assay. RESULTS The sphere counts from the HFD group were lower than those in the chow group. As the spheres formed, pericyte marker levels decreased and SOX2 levels increased gradually in the chow group, but not in the HFD group. Before sphere formation, pericytes from the HFD group showed high p21 levels. The use of a p21 inhibitor rescued the reduction of sphere counts in the HFD group. At cellular level, hyperglycemia-induced ROS increased the level of p21 in cerebral pericytes. The p21-SOX2 signaling was then activated after oxygen-glucose deprivation. CONCLUSION HFD-induced diabetes compromises the stemness of cerebral pericytes by altering p21-SOX2 signaling. These results provide evidence supporting the role of pericytes in diabetes-related CSVD and subsequent cerebral dysfunction.
Collapse
Affiliation(s)
- Hao-Kuang Wang
- School of Medicine for International Students, I-Shou University, Kaohsiung
- Department of Neurosurgery, E-Da Hospital, I-Shou University, Kaohsiung
| | - Chih-Yuan Huang
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan
| | - Yun-Wen Chen
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan
| | - Yuan-Ting Sun
- Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan
- Department of Genomic Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan
| |
Collapse
|
25
|
Maners J, Gill D, Pankratz N, Laffan MA, Wolberg AS, de Maat MPM, Ligthart S, Tang W, Ward-Caviness CK, Fornage M, Debette S, Dichgans M, McKnight B, Boerwinkle E, Smith NL, Morrison AC, Dehghan A, de Vries PS. A Mendelian randomization of γ' and total fibrinogen levels in relation to venous thromboembolism and ischemic stroke. Blood 2020; 136:3062-3069. [PMID: 33367543 PMCID: PMC7770565 DOI: 10.1182/blood.2019004781] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 07/12/2020] [Indexed: 12/23/2022] Open
Abstract
Fibrinogen is a key component of the coagulation cascade, and variation in its circulating levels may contribute to thrombotic diseases, such as venous thromboembolism (VTE) and ischemic stroke. Gamma prime (γ') fibrinogen is an isoform of fibrinogen that has anticoagulant properties. We applied 2-sample Mendelian randomization (MR) to estimate the causal effect of total circulating fibrinogen and its isoform, γ' fibrinogen, on risk of VTE and ischemic stroke subtypes using summary statistics from genome-wide association studies. Genetic instruments for γ' fibrinogen and total fibrinogen were selected, and the inverse-variance weighted MR approach was used to estimate causal effects in the main analysis, complemented by sensitivity analyses that are more robust to the inclusion of pleiotropic variants, including MR-Egger, weighted median MR, and weighted mode MR. The main inverse-variance weighted MR estimates based on a combination of 16 genetic instruments for γ' fibrinogen and 75 genetic instruments for total fibrinogen indicated a protective effect of higher γ' fibrinogen and higher total fibrinogen on VTE risk. There was also a protective effect of higher γ' fibrinogen levels on cardioembolic and large artery stroke risk. Effect estimates were consistent across sensitivity analyses. Our results provide evidence to support effects of genetically determined γ' fibrinogen on VTE and ischemic stroke risk. Further research is needed to explore mechanisms underlying these effects and their clinical applications.
Collapse
Affiliation(s)
- Jillian Maners
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
| | - Nathan Pankratz
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Minnesota, Minneapolis, MN
| | - Michael A Laffan
- Centre for Haematology, Imperial College London, London, United Kingdom
| | - Alisa S Wolberg
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC
| | | | - Symen Ligthart
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Weihong Tang
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN
| | - Cavin K Ward-Caviness
- National Health and Environmental Effects Laboratory, US Environmental Protection Agency, Chapel Hill, NC
| | - Myriam Fornage
- The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX
| | - Stephanie Debette
- Stroke Research Group, Division of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
- Department of Neurology, Institute for Neurodegenerative Disease, Bordeaux University Hospital, Bordeaux, France
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Barbara McKnight
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
- Department of Biostatistics, University of Washington, Seattle, WA
| | - Nicholas L Smith
- Kaiser Permanente Washington Research Institute, Kaiser Permanente Washington, Seattle, WA
- Department of Epidemiology, University of Washington, Seattle, WA
- Seattle Epidemiologic Research and Information Center, Office of Research and Development, Department of Veteran Affairs, Seattle, WA
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Abbas Dehghan
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- Medical Research Council-Public Health England (MRC-PHE) Centre for Environment and Health, School of Public Health, Imperial College London, London, United Kingdom; and
- UK Dementia Research Institute, Imperial College London, London, United Kingdom
| | - Paul S de Vries
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| |
Collapse
|
26
|
Sun T, Xie T, Zhang A, Fan L, Xu Z, Chen X, Fan Z, Wang C. Relation between left atrial structure and lacunar infarction in patients with hypertension. Aging (Albany NY) 2020; 12:17295-17304. [PMID: 32915163 PMCID: PMC7521509 DOI: 10.18632/aging.103697] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 07/06/2020] [Indexed: 01/24/2023]
Abstract
A lacunar infarction (LACI) can cause damage to the surrounding brain tissue and place an individual at greater risk for future major stroke. LACI is associated with hypertension and hypertension is associated with left atrial enlargement. It is important to identify a high-risk patient who is more vulnerable to suffering a LACI in hypertensive group. So, we studied whether left atrium size is an independent risk predictor for LACI in hypertensive patients. We performed cross-sectional analysis of 365 patients with hypertension at Shanghai Ninth People's Hospital from January 2016 to January 2017. The results showed that left atrial diameter(LAD), left atrial volume (LAV) and the ratio of left atrial diameter to left ventricular diameter (LAD/LVD) were significantly associated with LACI in hypertensive patients. Based on the ROC curve analysis, the area under the ROC curve (AUC) of LAV used to predict LACI was 0.737 (95% CI: 0.686 - 0.788), and the AUC of LAD/LVD was 0.784 (95% CI: 0.737 - 0.830). The optimal cut-off value for LAV was 30.14, and the sensitivity and specificity were 72% and 63%, respectively. The optimal cut-off value for LAD/LVD was 0.757, and the sensitivity and specificity were 77% and 70%, respectively. LAV or LAD/LVD played an important role in LACI with hypertension and could be an independent risk factor in hypertensive patients.
Collapse
Affiliation(s)
- Ting Sun
- Department of Cardiology, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
| | - Tong Xie
- Department of Cardiology, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China,Department of Intensive Care Unit, Shanghai Xuhui District Central Hospital, Shanghai 200031, China
| | - Alian Zhang
- Department of Cardiology, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
| | - Li Fan
- Department of Cardiology, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
| | - Zuojun Xu
- Department of Cardiology, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
| | - Xin Chen
- Department of Cardiology, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
| | - Zhicheng Fan
- Department of Cardiology, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
| | - Changqian Wang
- Department of Cardiology, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
27
|
Abstract
Objective: Whether cerebrovascular regulation is different in patients with controlled high blood pressure (HBP) with and without small vessel disease (SVD). Methods: Sixty-seven healthy controls (mean age ± SD, 45 ± 16 years; 30 women, 37 men) and 40 patients (mean age, 64 ± 13 years; 14 women, 26 men) with HBP and different stages of SVD, underwent simultaneous recordings of the spontaneous fluctuations of BP, blood flow velocity (CBFV) in both middle cerebral arteries (MCA), and of end-tidal CO2 (ETCO2). Coherence and transfer function gain and phase between BP and CBFV were assessed in the frequency ranges of VLF (0.02–0.07 Hz), low frequency (0.07–0.15), and high frequency (>0.15). BP SD indicated BP variability (BPV). Results: In controls (BP, 86 ± 13 mmHg; ETCO2, 39 ± 4 mmHg; BPV, 15 ± 6 mmHg), gain, phase and coherence were not age-dependent in simple or a multiple regression models. BPV correlated significantly in both MCAs with gain in low frequency and high frequency, and with phase in VLF and high frequency. In patients (BP, 91 ± 16 mmHg, ETCO2, 39 ± 4 mmHg, BPV 18 ± 5 mmHg), only gain showed some differences between different SVD groups. Comparing all patients with 25 controls of similar age and sex, patients exhibited significantly (P < 0.05–P < 0.005): increased coherence and gain in VLF, decreased phase in VLF and low frequency, correlations between BPV with phase in low frequency (left) and with gain in VLF (left) and in high frequency (left and right). Conclusion: Phase seems an age independent autoregulatory index. In controlled HBP, CBF regulation is degraded at longlasting CBF changes; BPV effects lose their physiological bilateral distribution.
Collapse
|
28
|
Locatelli M, Padovani A, Pezzini A. Pathophysiological Mechanisms and Potential Therapeutic Targets in Cerebral Autosomal Dominant Arteriopathy With Subcortical Infarcts and Leukoencephalopathy (CADASIL). Front Pharmacol 2020; 11:321. [PMID: 32231578 PMCID: PMC7082755 DOI: 10.3389/fphar.2020.00321] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 03/05/2020] [Indexed: 12/13/2022] Open
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), is a hereditary small-vessels angiopathy caused by mutations in the NOTCH 3 gene, located on chromosome 19, usually affecting middle-ages adults, whose clinical manifestations include migraine with aura, recurrent strokes, mood disorders, and cognitive impairment leading to dementia and disability. In this review, we provide an overview of the current knowledge on the pathogenic mechanisms underlying the disease, focus on the corresponding therapeutic targets, and discuss the most promising treatment strategies currently under investigations. The hypothesis that CADASIL is an appropriate model to explore the pathogenesis of sporadic cerebral small vessel disease is also reviewed.
Collapse
Affiliation(s)
- Martina Locatelli
- Department of Clinical and Experimental Sciences, Neurology Clinic, University of Brescia, Brescia, Italy
| | - Alessandro Padovani
- Department of Clinical and Experimental Sciences, Neurology Clinic, University of Brescia, Brescia, Italy
| | - Alessandro Pezzini
- Department of Clinical and Experimental Sciences, Neurology Clinic, University of Brescia, Brescia, Italy
| |
Collapse
|
29
|
Yuan H, Fu M, Yang X, Huang K, Ren X. Single nucleotide polymorphism of MTHFR rs1801133 associated with elevated Hcy levels affects susceptibility to cerebral small vessel disease. PeerJ 2020; 8:e8627. [PMID: 32117640 PMCID: PMC7036271 DOI: 10.7717/peerj.8627] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 01/23/2020] [Indexed: 11/20/2022] Open
Abstract
Background Methylenetetrahydrofolate reductase (MTHFR) is indispensable for the conversion of homocysteine (Hcy) to methionine. The single nucleotide polymorphism (SNP) of MTHFR gene (rs1801133, C667T) is correlated with decreased enzyme activity that eventually results in elevated plasma Hcy levels. Hyperhomocysteinemia has been confirmed to be involved in the pathogenesis of stroke, cerebral small vessel disease (CSVD), various metabolic disorders and so on. However, the relationship between the MTHFR gene polymorphisms, Hcy, and CSVD has not been investigated. In this study, the relationship between SNPs of MTHFR gene and CSVD was determined after adjusting for cardiovascular risk factors, and the potential mechanism based on Hcy levels was explored. Methods A total of 163 consecutive CSVD patients were collected as the case group. In the corresponding period, 326 healthy people were selected as the control group, who were matched to these cases according to age (±2 years) and gender at a ratio of 2:1. SNPs of MTHFR rs1801133, rs1801131, rs2274976, rs4846048, rs4846049, rs13306561 and rs3737964, were genotyped with TaqMan Pre-Designed SNP Genotyping Assays. Plasma Hcy levels were detected using Hcy reagent through enzymatic cycling assay. Multivariate analysis was used to identify the SNPs associated with CSVD susceptibility. Plasma Hcy levels were compared between different genotypes. Results The MTHFR rs1801133 TT and CT genotype had increased risk for CSVD, and the OR was higher in the TT genotype than in the CT genotype (2.307 vs 1.473). The plasma Hcy levels of different genotypes showed the tendency of the TT genotype > CT genotype > CC genotype (19.91 ± 8.73 pg/ml vs 17.04 ± 5.68 pg/ml vs 14.96 ± 4.85 pg/ml). Conclusions The SNP of MTHFR rs1801133 was correlated with CSVD, and the TT and CT genotypes had increased risk for CSVD compared to the CC genotype. The potential mechanism was associated with elevated Hcy levels.
Collapse
Affiliation(s)
- Hongyu Yuan
- The Third Department of Neurology, Heze Municipal Hospital, Heze, Shandong Province, China
| | - Man Fu
- Department of Neurology, The Third People's Hospital of Heze, Heze, Shandong Province, China
| | - Xianzhang Yang
- The Third Department of Neurology, Heze Municipal Hospital, Heze, Shandong Province, China
| | - Kun Huang
- The Third Department of Neurology, Heze Municipal Hospital, Heze, Shandong Province, China
| | - Xiaoyan Ren
- The Third Department of Neurology, Heze Municipal Hospital, Heze, Shandong Province, China
| |
Collapse
|
30
|
Tan RYY, Traylor M, Megy K, Duarte D, Deevi SVV, Shamardina O, Mapeta RP, Ouwehand WH, Gräf S, Downes K, Markus HS. How common are single gene mutations as a cause for lacunar stroke? A targeted gene panel study. Neurology 2019; 93:e2007-e2020. [PMID: 31719132 PMCID: PMC6913325 DOI: 10.1212/wnl.0000000000008544] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 06/10/2019] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVES To determine the frequency of rare and pertinent disease-causing variants in small vessel disease (SVD)-associated genes (such as NOTCH3, HTRA1, COL4A1, COL4A2, FOXC1, TREX1, and GLA) in cerebral SVD, we performed targeted gene sequencing in 950 patients with younger-onset apparently sporadic SVD stroke using a targeted sequencing panel. METHODS We designed a high-throughput sequencing panel to identify variants in 15 genes (7 known SVD genes, 8 SVD-related disorder genes). The panel was used to screen a population of 950 patients with younger-onset (≤70 years) MRI-confirmed SVD stroke, recruited from stroke centers across the United Kingdom. Variants were filtered according to their frequency in control databases, predicted effect, presence in curated variant lists, and combined annotation dependent depletion scores. Whole genome sequencing and genotyping were performed on a subset of patients to provide a direct comparison of techniques. The frequency of known disease-causing and pertinent variants of uncertain significance was calculated. RESULTS We identified previously reported variants in 14 patients (8 cysteine-changing NOTCH3 variants in 11 patients, 2 HTRA1 variants in 2 patients, and 1 missense COL4A1 variant in 1 patient). In addition, we identified 29 variants of uncertain significance in 32 patients. CONCLUSION Rare monogenic variants account for about 1.5% of younger onset lacunar stroke. Most are cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy variants, but the second most common gene affected is HTRA1. A high-throughput sequencing technology platform is an efficient, reliable method to screen for such mutations.
Collapse
Affiliation(s)
- Rhea Y Y Tan
- From the Stroke Research Group, Department of Clinical Neurosciences (R.Y.Y.T., M.T., H.S.M.), Department of Haematology (K.M., D.D., S.V.V.D., O.S., R.P.M., W.H.O., S.G., K.D.), and Division of Respiratory Medicine, Department of Medicine (S.G.), University of Cambridge; and NIHR BioResource: Rare Diseases (K.M., S.V.V.D., O.S., R.P.M., W.H.O., S.G., H.S.M.), Biomedical Campus, Cambridge, UK.
| | - Matthew Traylor
- From the Stroke Research Group, Department of Clinical Neurosciences (R.Y.Y.T., M.T., H.S.M.), Department of Haematology (K.M., D.D., S.V.V.D., O.S., R.P.M., W.H.O., S.G., K.D.), and Division of Respiratory Medicine, Department of Medicine (S.G.), University of Cambridge; and NIHR BioResource: Rare Diseases (K.M., S.V.V.D., O.S., R.P.M., W.H.O., S.G., H.S.M.), Biomedical Campus, Cambridge, UK
| | - Karyn Megy
- From the Stroke Research Group, Department of Clinical Neurosciences (R.Y.Y.T., M.T., H.S.M.), Department of Haematology (K.M., D.D., S.V.V.D., O.S., R.P.M., W.H.O., S.G., K.D.), and Division of Respiratory Medicine, Department of Medicine (S.G.), University of Cambridge; and NIHR BioResource: Rare Diseases (K.M., S.V.V.D., O.S., R.P.M., W.H.O., S.G., H.S.M.), Biomedical Campus, Cambridge, UK
| | - Daniel Duarte
- From the Stroke Research Group, Department of Clinical Neurosciences (R.Y.Y.T., M.T., H.S.M.), Department of Haematology (K.M., D.D., S.V.V.D., O.S., R.P.M., W.H.O., S.G., K.D.), and Division of Respiratory Medicine, Department of Medicine (S.G.), University of Cambridge; and NIHR BioResource: Rare Diseases (K.M., S.V.V.D., O.S., R.P.M., W.H.O., S.G., H.S.M.), Biomedical Campus, Cambridge, UK
| | - Sri V V Deevi
- From the Stroke Research Group, Department of Clinical Neurosciences (R.Y.Y.T., M.T., H.S.M.), Department of Haematology (K.M., D.D., S.V.V.D., O.S., R.P.M., W.H.O., S.G., K.D.), and Division of Respiratory Medicine, Department of Medicine (S.G.), University of Cambridge; and NIHR BioResource: Rare Diseases (K.M., S.V.V.D., O.S., R.P.M., W.H.O., S.G., H.S.M.), Biomedical Campus, Cambridge, UK
| | - Olga Shamardina
- From the Stroke Research Group, Department of Clinical Neurosciences (R.Y.Y.T., M.T., H.S.M.), Department of Haematology (K.M., D.D., S.V.V.D., O.S., R.P.M., W.H.O., S.G., K.D.), and Division of Respiratory Medicine, Department of Medicine (S.G.), University of Cambridge; and NIHR BioResource: Rare Diseases (K.M., S.V.V.D., O.S., R.P.M., W.H.O., S.G., H.S.M.), Biomedical Campus, Cambridge, UK
| | - Rutendo P Mapeta
- From the Stroke Research Group, Department of Clinical Neurosciences (R.Y.Y.T., M.T., H.S.M.), Department of Haematology (K.M., D.D., S.V.V.D., O.S., R.P.M., W.H.O., S.G., K.D.), and Division of Respiratory Medicine, Department of Medicine (S.G.), University of Cambridge; and NIHR BioResource: Rare Diseases (K.M., S.V.V.D., O.S., R.P.M., W.H.O., S.G., H.S.M.), Biomedical Campus, Cambridge, UK
| | - Willem H Ouwehand
- From the Stroke Research Group, Department of Clinical Neurosciences (R.Y.Y.T., M.T., H.S.M.), Department of Haematology (K.M., D.D., S.V.V.D., O.S., R.P.M., W.H.O., S.G., K.D.), and Division of Respiratory Medicine, Department of Medicine (S.G.), University of Cambridge; and NIHR BioResource: Rare Diseases (K.M., S.V.V.D., O.S., R.P.M., W.H.O., S.G., H.S.M.), Biomedical Campus, Cambridge, UK
| | - Stefan Gräf
- From the Stroke Research Group, Department of Clinical Neurosciences (R.Y.Y.T., M.T., H.S.M.), Department of Haematology (K.M., D.D., S.V.V.D., O.S., R.P.M., W.H.O., S.G., K.D.), and Division of Respiratory Medicine, Department of Medicine (S.G.), University of Cambridge; and NIHR BioResource: Rare Diseases (K.M., S.V.V.D., O.S., R.P.M., W.H.O., S.G., H.S.M.), Biomedical Campus, Cambridge, UK
| | - Kate Downes
- From the Stroke Research Group, Department of Clinical Neurosciences (R.Y.Y.T., M.T., H.S.M.), Department of Haematology (K.M., D.D., S.V.V.D., O.S., R.P.M., W.H.O., S.G., K.D.), and Division of Respiratory Medicine, Department of Medicine (S.G.), University of Cambridge; and NIHR BioResource: Rare Diseases (K.M., S.V.V.D., O.S., R.P.M., W.H.O., S.G., H.S.M.), Biomedical Campus, Cambridge, UK
| | - Hugh S Markus
- From the Stroke Research Group, Department of Clinical Neurosciences (R.Y.Y.T., M.T., H.S.M.), Department of Haematology (K.M., D.D., S.V.V.D., O.S., R.P.M., W.H.O., S.G., K.D.), and Division of Respiratory Medicine, Department of Medicine (S.G.), University of Cambridge; and NIHR BioResource: Rare Diseases (K.M., S.V.V.D., O.S., R.P.M., W.H.O., S.G., H.S.M.), Biomedical Campus, Cambridge, UK
| |
Collapse
|
31
|
Affiliation(s)
- Hugh S Markus
- From the Stroke Research Group, Department of Clinical Neurosciences, University of Cambridge, United Kingdom (H.S.M.)
| | - Reinhold Schmidt
- Department of Neurology, Medical University of Graz, Austria (R.S.)
| |
Collapse
|
32
|
Haffner C. Proteostasis in Cerebral Small Vessel Disease. Front Neurosci 2019; 13:1142. [PMID: 31798396 PMCID: PMC6874119 DOI: 10.3389/fnins.2019.01142] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 10/10/2019] [Indexed: 01/02/2023] Open
Abstract
Maintaining the homeostasis of proteins (proteostasis) by controlling their synthesis, folding and degradation is a central task of cells and tissues. The gradual decline of the capacity of the various proteostasis machineries, frequently in combination with their overload through mutated, aggregation-prone proteins, is increasingly recognized as an important catalyst of age-dependent pathologies in the brain, most prominently neurodegenerative disorders. A dysfunctional proteostasis might also contribute to neurovascular disease as indicated by the occurrence of excessive protein accumulation or massive extracellular matrix expansion within vessel walls in conditions such as cerebral small vessel disease (SVD), a major cause of ischemic stroke, and cerebral amyloid angiopathy. Recent advances in brain vessel isolation techniques and mass spectrometry methodology have facilitated the analysis of cerebrovascular proteomes and fueled efforts to determine the proteomic signatures associated with neurovascular disease. In several studies in humans and mice considerable differences between healthy and diseased vessel proteomes were observed, emphasizing the critical contribution of an impaired proteostasis to disease pathogenesis. These findings highlight the important role of a balanced proteostasis for cerebrovascular health.
Collapse
Affiliation(s)
- Christof Haffner
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
33
|
Batllori M, Casado M, Sierra C, Salgado MDC, Marti-Sanchez L, Maynou J, Fernandez G, Garcia-Cazorla A, Ormazabal A, Molero-Luis M, Artuch R. Effect of blood contamination of cerebrospinal fluid on amino acids, biogenic amines, pterins and vitamins. Fluids Barriers CNS 2019; 16:34. [PMID: 31727079 PMCID: PMC6857153 DOI: 10.1186/s12987-019-0154-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 11/02/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cerebrospinal fluid (CSF) metabolomic investigations are a powerful tool for studying neurometabolic diseases. We aimed to assess the effect of CSF contamination with blood on the concentrations of selected biomarkers. METHODS CSF samples were spiked in duplicate with increasing volumes of whole blood under two conditions: (A) pooled CSF spiked with fresh whole blood and frozen to cause red blood cell (RBC) lysis; (B) pooled CSF spiked with fresh blood and centrifuged (the supernatant with no RBCs was frozen until the moment of analysis). CSF concentrations of amino acids, biogenic amines, pterins, and vitamins were analysed by HPLC coupled with tandem mass spectrometry, electrochemical and fluorescence detection. RESULTS Aspartate, glutamate, taurine, ornithine, glycine, citrulline, pyridoxal 5´-phosphate, 5-methyltetrahydrofolate, and thiamine showed higher values when RBCs were lysed when compared with those of CSF with no RBC, while arginine, 5-hydroxyindoleacetic and homovanillic acids showed lower values. When RBCs were removed from CSF, only some amino acids, thiamine and pyridoxal 5´-phosphate showed moderately higher values when compared with the non-spiked CSF sample. CONCLUSIONS CSF-targeted metabolomic analysis is feasible even when substantial RBC contamination of CSF has occurred since CSF centrifugation to remove RBC prior to freezing eliminated most of the interferences observed.
Collapse
Affiliation(s)
- Marta Batllori
- Clinical Biochemistry Department, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Mercedes Casado
- Clinical Biochemistry Department, Hospital Sant Joan de Déu, Barcelona, Spain.,CIBERER-Instituto de Salud Carlos III, Barcelona, Spain
| | - Cristina Sierra
- Clinical Biochemistry Department, Hospital Sant Joan de Déu, Barcelona, Spain
| | | | - Laura Marti-Sanchez
- Clinical Biochemistry Department, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Joan Maynou
- Molecular Genetics, Hospital Sant Joan de Déu, Barcelona, Spain
| | | | - Angels Garcia-Cazorla
- Pediatric Neurology Department, Institut de Recerca Sant Joan de Déu, Barcelona, Spain.,CIBERER-Instituto de Salud Carlos III, Barcelona, Spain
| | - Aida Ormazabal
- Clinical Biochemistry Department, Hospital Sant Joan de Déu, Barcelona, Spain.,CIBERER-Instituto de Salud Carlos III, Barcelona, Spain
| | - Marta Molero-Luis
- Clinical Biochemistry Department, Hospital Sant Joan de Déu, Barcelona, Spain.
| | - Rafael Artuch
- Clinical Biochemistry Department, Hospital Sant Joan de Déu, Barcelona, Spain.,CIBERER-Instituto de Salud Carlos III, Barcelona, Spain
| |
Collapse
|
34
|
Mustapha M, Nassir CMNCM, Aminuddin N, Safri AA, Ghazali MM. Cerebral Small Vessel Disease (CSVD) - Lessons From the Animal Models. Front Physiol 2019; 10:1317. [PMID: 31708793 PMCID: PMC6822570 DOI: 10.3389/fphys.2019.01317] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 09/30/2019] [Indexed: 12/28/2022] Open
Abstract
Cerebral small vessel disease (CSVD) refers to a spectrum of clinical and imaging findings resulting from pathological processes of various etiologies affecting cerebral arterioles, perforating arteries, capillaries, and venules. Unlike large vessels, it is a challenge to visualize small vessels in vivo, hence the difficulty to directly monitor the natural progression of the disease. CSVD might progress for many years during the early stage of the disease as it remains asymptomatic. Prevalent among elderly individuals, CSVD has been alarmingly reported as an important precursor of full-blown stroke and vascular dementia. Growing evidence has also shown a significant association between CSVD's radiological manifestation with dementia and Alzheimer's disease (AD) pathology. Although it remains contentious as to whether CSVD is a cause or sequelae of AD, it is not far-fetched to posit that effective therapeutic measures of CSVD would mitigate the overall burden of dementia. Nevertheless, the unifying theory on the pathomechanism of the disease remains elusive, hence the lack of effective therapeutic approaches. Thus, this chapter consolidates the contemporary insights from numerous experimental animal models of CSVD, to date: from the available experimental animal models of CSVD and its translational research value; the pathomechanical aspects of the disease; relevant aspects on systems biology; opportunities for early disease biomarkers; and finally, converging approaches for future therapeutic directions of CSVD.
Collapse
Affiliation(s)
- Muzaimi Mustapha
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | | | - Niferiti Aminuddin
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
- Department of Basic Medical Sciences, Kulliyyah of Pharmacy, International Islamic University Malaysia, Kuantan, Malaysia
| | - Amanina Ahmad Safri
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Mazira Mohamad Ghazali
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| |
Collapse
|
35
|
Giau VV, Bagyinszky E, Youn YC, An SSA, Kim SY. Genetic Factors of Cerebral Small Vessel Disease and Their Potential Clinical Outcome. Int J Mol Sci 2019; 20:ijms20174298. [PMID: 31484286 PMCID: PMC6747336 DOI: 10.3390/ijms20174298] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 08/27/2019] [Accepted: 09/01/2019] [Indexed: 12/23/2022] Open
Abstract
Cerebral small vessel diseases (SVD) have been causally correlated with ischemic strokes, leading to cognitive decline and vascular dementia. Neuroimaging and molecular genetic tests could improve diagnostic accuracy in patients with potential SVD. Several types of monogenic, hereditary cerebral SVD have been identified: cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy (CARASIL), cerebral autosomal-dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), cathepsin A-related arteriopathy with strokes and leukoencephalopathy (CARASAL), hereditary diffuse leukoencephalopathy with spheroids (HDLS), COL4A1/2-related disorders, and Fabry disease. These disorders can be distinguished based on their genetics, pathological and imaging findings, clinical manifestation, and diagnosis. Genetic studies of sporadic cerebral SVD have demonstrated a high degree of heritability, particularly among patients with young-onset stroke. Common genetic variants in monogenic disease may contribute to pathological progress in several cerebral SVD subtypes, revealing distinct genetic mechanisms in different subtype of SVD. Hence, genetic molecular analysis should be used as the final gold standard of diagnosis. The purpose of this review was to summarize the recent discoveries made surrounding the genetics of cerebral SVD and their clinical significance, to provide new insights into the pathogenesis of cerebral SVD, and to highlight the possible convergence of disease mechanisms in monogenic and sporadic cerebral SVD.
Collapse
Affiliation(s)
- Vo Van Giau
- Department of Bionano Technology & Gachon Bionano Research Institute, Gachon University, Seongnam-si, Gyeonggi-do 461-701, Korea
| | - Eva Bagyinszky
- Department of Bionano Technology & Gachon Bionano Research Institute, Gachon University, Seongnam-si, Gyeonggi-do 461-701, Korea
| | - Young Chul Youn
- Department of Neurology, Chung-Ang University College of Medicine, Seoul 06973, Korea.
| | - Seong Soo A An
- Department of Bionano Technology & Gachon Bionano Research Institute, Gachon University, Seongnam-si, Gyeonggi-do 461-701, Korea.
| | - Sang Yun Kim
- Department of Neurology, Seoul National University College of Medicine & Neurocognitive Behavior Center, Seoul National University Bundang Hospital, Seoul 06973, Korea
| |
Collapse
|
36
|
Genome-Wide Association Study of Cerebral Microbleeds on MRI. Neurotox Res 2019; 37:146-155. [PMID: 31209788 DOI: 10.1007/s12640-019-00073-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/29/2019] [Accepted: 06/07/2019] [Indexed: 10/26/2022]
Abstract
Cerebral microbleeds are the presence of a group of pathological processes affecting the small arteries, arterioles, capillaries, and venules of the brain. Previous studies showed that cerebral microbleeds were associated with higher risk of dementia and stroke. We conducted a genome-wide association study of cerebral microbleeds to identify novel loci associated with the presence and progression of cerebral microbleeds. This study included 454 individuals composed by 176 subjects with cerebral microbleeds and 278 subjects without cerebral microbleeds in a non-Hispanic/Latino white population. Association of genetic variants with the presence and progression of cerebral microbleeds was assessed by logistic regression model. Potential genetic risk variants Apolipoprotein E (ApoE) polymorphisms were independently genotyped and checked the association with the presence and progression of cerebral microbleeds. No single-nucleotide polymorphisms (SNPs) associated with the presence or progression of cerebral microbleeds were identified at genome-wide significant level (P < 1 × 10-8). A total of 19 SNPs were associated with the presence of microbleeds at suggestive level (P < 1 × 10-5). One SNP was associated with lower progression risk for cerebral microbleeds with suggestive evidence (P < 1 × 10-5). ApoE ε4ε4 was independently associated with the presence and progression of cerebral microbleeds (odds ratio = 2.54, 95% confidence interval 1.08-6.00 and odds ratio = 5.1, 95% confidence interval 1.36-19.16). We highlighted 19 novel SNPs associated with the presence of cerebral microbleeds and one novel SNP associated with the progression of cerebral microbleeds for the first time. ApoE ε4ε4 was confirmed independently associated with the presence and progression of cerebral microbleeds.
Collapse
|
37
|
Abstract
Vascular dementia (VaD) is a common disorder that encompasses heterogeneous entities, which creates challenges in order to reach a global consensus for diagnostic criteria. While the genetic basis for sporadic VaD remains poorly understood, the identification of causal genes in monogenic forms of VaD sheds light on the pathophysiological mechanisms of VaD. This special report describes progress in genetic research on monogenic and sporadic VaD, as well as on associated phenotypes, such as cerebral small vessel disease, stroke and Alzheimer's disease. Methodological issues (e.g., small-size studies) and strategies to overcome these problems (e.g., collaborative consortiums, endophenotypes) are discussed. Lastly, future perspectives in the field and how such work could benefit patients and clinicians are mentioned.
Collapse
Affiliation(s)
- Raquel Manso-Calderón
- Department of Neurology, University Hospital of Salamanca, Salamanca 37007, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), University of Salamanca, Salamanca 37007, Spain
| |
Collapse
|
38
|
Bian Y, Wang JC, Sun F, Sun ZY, Lin YJ, Liu Y, Zhao B, Liu L, Luo XG. Assessment of cerebrovascular reserve impairment using the breath-holding index in patients with leukoaraiosis. Neural Regen Res 2019; 14:1412-1418. [PMID: 30964067 PMCID: PMC6524493 DOI: 10.4103/1673-5374.251332] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Many studies have demonstrated that leukoaraiosis is associated with impaired cerebrovascular reserve function. However, the definitive hemodynamic changes that occur in leukoaraiosis are not clear, and there are many controversies. This study aimed to investigate hemodynamic changes in symptomatic leukoaraiosis using transcranial Doppler ultrasonography and the breath-holding test in a Chinese Han population, from northern China. A total of 203 patients who were diagnosed with ischemic stroke or clinical chronic progressive ischemic symptoms were enrolled in this study, including 97 males and 106 females, with an age range of 43–93 years. The severity of leukoaraiosis was evaluated according to the Fazekas grading scale, and patients were divided into four groups accordingly. Grade 0 was no leukoaraiosis, and grades I, II, and III were mild, moderate, and severe leukoaraiosis, respectively, with 44, 79, 44, and 36 cases in each group. Transcranial Doppler ultrasonography and the breath-holding test were performed. The mean blood flow velocity of the bilateral middle cerebral artery was measured and the breath-holding index was calculated. The breath holding index was correlated with leukoaraiosis severity and cognitive impairment. Patients with a low breath holding index presented poor performance in the Montreal Cognitive Assessment (MoCA) and executive function tests. That is, the lower the breath holding index, the lower the scores for the MoCA and the higher for the trail-making test Parts A and B. These results indicate that the breath-holding index is a useful parameter for the evaluation of cerebrovascular reserve impairment in patients with leukoaraiosis. In addition, the breath-holding index can reflect cognitive dysfunction, providing a new insight into the pathophysiology of leukoaraiosis. This study was approved by the Ethics Committee of the Fifth People’s Hospital of Shenyang, China (approval No. 20160301) and registered in the Chinese Clinical Trial Registry (registration number: ChiCTR1800014421).
Collapse
Affiliation(s)
- Ying Bian
- Department of Neurology, the First Affiliated Hospital of China Medical University; Department of Neurology, the Fifth People's Hospital of Shenyang, Shenyang, Liaoning Province, China
| | - Jin-Chun Wang
- Department of Neurology, the Fifth People's Hospital of Shenyang, Shenyang, Liaoning Province, China
| | - Feng Sun
- Department of Neurology, the Fifth People's Hospital of Shenyang, Shenyang, Liaoning Province, China
| | - Zi-Yi Sun
- Department of Endocrinology, Shengjing Hospital, China Medical University, Shenyang, Liaoning Province, China
| | - Yu-Jiao Lin
- Department of Neurology, the Fifth People's Hospital of Shenyang, Shenyang, Liaoning Province, China
| | - Yang Liu
- Department of Neurology, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Bin Zhao
- Department of Neurology, the Fifth People's Hospital of Shenyang, Shenyang, Liaoning Province, China
| | - Li Liu
- Department of Neurology, the Fifth People's Hospital of Shenyang, Shenyang, Liaoning Province, China
| | - Xiao-Guang Luo
- Department of Neurology, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
39
|
Norrving B, Barrick J, Davalos A, Dichgans M, Cordonnier C, Guekht A, Kutluk K, Mikulik R, Wardlaw J, Richard E, Nabavi D, Molina C, Bath PM, Stibrant Sunnerhagen K, Rudd A, Drummond A, Planas A, Caso V. Action Plan for Stroke in Europe 2018-2030. Eur Stroke J 2018; 3:309-336. [PMID: 31236480 PMCID: PMC6571507 DOI: 10.1177/2396987318808719] [Citation(s) in RCA: 315] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 10/01/2018] [Indexed: 12/21/2022] Open
Abstract
Two previous pan-European consensus meetings, the 1995 and 2006 Helsingborg meetings, were convened to review the scientific evidence and the state of current services to identify priorities for research and development and to set targets for the development of stroke care for the decade to follow. Adhering to the same format, the European Stroke Organisation (ESO) prepared a European Stroke Action Plan (ESAP) for the years 2018 to 2030, in cooperation with the Stroke Alliance for Europe (SAFE). The ESAP included seven domains: primary prevention, organisation of stroke services, management of acute stroke, secondary prevention, rehabilitation, evaluation of stroke outcome and quality assessment and life after stroke. Research priorities for translational stroke research were also identified. Documents were prepared by a working group and were open to public comments. The final document was prepared after a workshop in Munich on 21-23 March 2018. Four overarching targets for 2030 were identified: (1) to reduce the absolute number of strokes in Europe by 10%, (2) to treat 90% or more of all patients with stroke in Europe in a dedicated stroke unit as the first level of care, (3) to have national plans for stroke encompassing the entire chain of care, (4) to fully implement national strategies for multisector public health interventions. Overall, 30 targets and 72 research priorities were identified for the seven domains. The ESAP provides a basic road map and sets targets for the implementation of evidence-based preventive actions and stroke services to 2030.
Collapse
Affiliation(s)
- Bo Norrving
- Department of Clinical Sciences Lund, Neurology, Skåne
University Hospital, Lund University, Lund, Sweden
| | | | - Antoni Davalos
- Department of Neurosciences, Hospital Universitari Germans Trias
i Pujol, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, University Hospital,
Ludwig-Maximilians University, Munich, and Munich Cluster of Systems Neurology
(SyNergy), Munich, Germany
| | | | - Alla Guekht
- Clinical Center for Neuropsychiatry, Russian National Research
Medical University, Moscow, Russia
| | - Kursad Kutluk
- Department of Neurology, Stroke Unit, University of Dokuz Eylul,
Izmir, Turkey
| | - Robert Mikulik
- International Clinical Research Center and Neurology Department,
St Anne's University Hospital Brno and Masaryk University Brno, Czech
Republic
| | - Joanna Wardlaw
- Centre for Clinical Neurosciences, Edinburgh Imaging and UK
Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Edo Richard
- Department of Neurology, Radboud University Medical Centre,
Nijmegen, and Department of Neurology, Academic Medical Centre, Amsterdam, the
Netherlands
| | - Darius Nabavi
- Department of Neurology with Stroke Unit, Vivantes Hospital
Neukölln, Berlin, Germany
| | - Carlos Molina
- Stroke Unit, Department of Neurology, Hospital Vall d´Hebron
Barcelona, Spain
| | - Philip M Bath
- Stroke Trials Unit, Division of Clinical Neuroscience,
University of Nottingham, Nottingham, UK
| | | | - Anthony Rudd
- Guy's and St Thomas' NHS Foundation Trust, Stroke NHS England
and Royal College of Physicians, London, UK
| | - Avril Drummond
- School of Health Sciences, University of Nottingham,
Nottingham, UK
| | - Anna Planas
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB),
Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona,
Spain
| | - Valeria Caso
- Stroke Unit, Department of Medicine and Cardiovascular
Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
40
|
Miyatake S, Schneeberger S, Koyama N, Yokochi K, Ohmura K, Shiina M, Mori H, Koshimizu E, Imagawa E, Uchiyama Y, Mitsuhashi S, Frith MC, Fujita A, Satoh M, Taguri M, Tomono Y, Takahashi K, Doi H, Takeuchi H, Nakashima M, Mizuguchi T, Takata A, Miyake N, Saitsu H, Tanaka F, Ogata K, Hennet T, Matsumoto N. Biallelic COLGALT1 variants are associated with cerebral small vessel disease. Ann Neurol 2018; 84:843-853. [PMID: 30412317 DOI: 10.1002/ana.25367] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 10/24/2018] [Accepted: 10/24/2018] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Approximately 5% of cerebral small vessel diseases are hereditary, which include COL4A1/COL4A2-related disorders. COL4A1/COL4A2 encode type IV collagen α1/2 chains in the basement membranes of cerebral vessels. COL4A1/COL4A2 mutations impair the secretion of collagen to the extracellular matrix, thereby resulting in vessel fragility. The diagnostic yield for COL4A1/COL4A2 variants is around 20 to 30%, suggesting other mutated genes might be associated with this disease. This study aimed to identify novel genes that cause COL4A1/COL4A2-related disorders. METHODS Whole exome sequencing was performed in 2 families with suspected COL4A1/COL4A2-related disorders. We validated the role of COLGALT1 variants by constructing a 3-dimensional structural model, evaluating collagen β (1-O) galactosyltransferase 1 (ColGalT1) protein expression and ColGalT activity by Western blotting and collagen galactosyltransferase assays, and performing in vitro RNA interference and rescue experiments. RESULTS Exome sequencing demonstrated biallelic variants in COLGALT1 encoding ColGalT1, which was involved in the post-translational modification of type IV collagen in 2 unrelated patients: c.452 T > G (p.Leu151Arg) and c.1096delG (p.Glu366Argfs*15) in Patient 1, and c.460G > C (p.Ala154Pro) and c.1129G > C (p.Gly377Arg) in Patient 2. Three-dimensional model analysis suggested that p.Leu151Arg and p.Ala154Pro destabilized protein folding, which impaired enzymatic activity. ColGalT1 protein expression and ColGalT activity in Patient 1 were undetectable. RNA interference studies demonstrated that reduced ColGalT1 altered COL4A1 secretion, and rescue experiments showed that mutant COLGALT1 insufficiently restored COL4A1 production in cells compared with wild type. INTERPRETATION Biallelic COLGALT1 variants cause cerebral small vessel abnormalities through a common molecular pathogenesis with COL4A1/COL4A2-related disorders. Ann Neurol 2018;84:843-853.
Collapse
Affiliation(s)
- Satoko Miyatake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan.,Clinical Genetics Department, Yokohama City University Hospital, Yokohama, Japan
| | | | - Norihisa Koyama
- Department of Pediatrics, Toyohashi Municipal Hospital, Toyohashi, Japan
| | - Kenji Yokochi
- Department of Pediatrics, Toyohashi Municipal Hospital, Toyohashi, Japan.,Department of Pediatrics, Seirei Mikatahara General Hospital, Shizuoka, Japan
| | - Kayo Ohmura
- Department of Pediatric Neurology, Morinomiya Hospital, Osaka, Japan
| | - Masaaki Shiina
- Department of Biochemistry, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Harushi Mori
- Department of Radiology, Graduate School and Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | - Eriko Koshimizu
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Eri Imagawa
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yuri Uchiyama
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Satomi Mitsuhashi
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Martin C Frith
- Artificial Intelligence Research Center, National Institute of Advanced Industrial Science and Technology, Tokyo, Japan.,Graduate School of Frontier Sciences, University of Tokyo, Chiba, Japan.,Computational Bio Big-Data Open Innovation Laboratory, National Institute of Advanced Industrial Science and Technology, Tokyo, Japan
| | - Atsushi Fujita
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Mai Satoh
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Masataka Taguri
- Department of Data Science, Yokohama City University School of Data Science, Yokohama, Japan
| | - Yasuko Tomono
- Division of Molecular and Cell Biology, Shigei Medical Research Institute, Okayama, Japan
| | - Keita Takahashi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hiroshi Doi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hideyuki Takeuchi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Mitsuko Nakashima
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan.,Department of Biochemistry, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Takeshi Mizuguchi
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Atsushi Takata
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Noriko Miyake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hirotomo Saitsu
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan.,Department of Biochemistry, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Fumiaki Tanaka
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kazuhiro Ogata
- Department of Biochemistry, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Thierry Hennet
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
41
|
Ilinca A, Samuelsson S, Piccinelli P, Soller M, Kristoffersson U, Lindgren AG. A stroke gene panel for whole-exome sequencing. Eur J Hum Genet 2018; 27:317-324. [PMID: 30356112 DOI: 10.1038/s41431-018-0274-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 08/19/2018] [Accepted: 08/28/2018] [Indexed: 12/21/2022] Open
Abstract
Extensive analyses of known monogenic causes of stroke by whole-exome/genome sequencing are technically possible today. We here aimed to compile a comprehensive panel of genes associated with monogenic causes of stroke for use in clinical and research situations. We systematically searched the publically available database Online Mendelian Inheritance in Man, and validated the entries against original peer-reviewed publications in PubMed. First, we selected known pathogenic or putatively pathogenic stroke genes reported in at least one person with stroke, and classified the stroke phenotype for each gene into eight subgroups: (1) large artery atherosclerotic, (2) large artery non-atherosclerotic (tortuosity, dolichoectasia, aneurysm, non-atherosclerotic dissection, occlusion), (3) cerebral small-vessel diseases, (4) cardioembolic (arrhythmia, heart defect, cardiomyopathy), (5) coagulation dysfunctions (venous thrombosis, arterial thrombosis, bleeding tendency), (6) intracerebral hemorrhage, (7) vascular malformations (cavernoma, arteriovenous malformations), and (8) metabolism disorders. Second, we selected other genes that may plausibly cause stroke through diseases related to stroke, but without any documented stroke patient description. A third section comprised SNPs associated with stroke in genome-wide association studies (GWAS). We identified in total 214 genes: 120 associated with stroke, 62 associated with diseases that may cause stroke, and 32 stroke-related genes from recent GWAS. We describe these 214 genes and the clinical stroke subtype(s) associated with each of them. The resulting gene panel can be used to interpret exome sequencing results regarding monogenic stroke. Based on the panel's clinical phenotype description, the pathogenicity of novel variants in these genes may be evaluated in specific situations.
Collapse
Affiliation(s)
- Andreea Ilinca
- Department of Clinical Sciences Lund, Neurology, Lund University, Lund, Sweden. .,Department of Neurology and Rehabilitation Medicine, Skåne University Hospital, Malmö, Sweden.
| | - Sofie Samuelsson
- Department of Clinical Genetics and Pathology, Laboratory Medicine, Region Skåne and Lund University, Lund, Sweden
| | - Paul Piccinelli
- Department of Clinical Genetics and Pathology, Laboratory Medicine, Region Skåne and Lund University, Lund, Sweden
| | - Maria Soller
- Department of Clinical Genetics and Pathology, Laboratory Medicine, Region Skåne and Lund University, Lund, Sweden.,Department of Clinical Genetics, Karolinska University Hospital, Solna, Sweden
| | - Ulf Kristoffersson
- Department of Clinical Genetics and Pathology, Laboratory Medicine, Region Skåne and Lund University, Lund, Sweden
| | - Arne G Lindgren
- Department of Clinical Sciences Lund, Neurology, Lund University, Lund, Sweden.,Department of Neurology and Rehabilitation Medicine, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
42
|
Abstract
Cerebral small vessel disease (CSVD) is composed of several diseases affecting the small arteries, arterioles, venules, and capillaries of the brain, and refers to several pathological processes and etiologies. Neuroimaging features of CSVD include recent small subcortical infarcts, lacunes, white matter hyperintensities, perivascular spaces, microbleeds, and brain atrophy. The main clinical manifestations of CSVD include stroke, cognitive decline, dementia, psychiatric disorders, abnormal gait, and urinary incontinence. Currently, there are no specific preventive or therapeutic measures to improve this condition. In this review, we will discuss the pathophysiology, clinical aspects, neuroimaging, progress of research to treat and prevent CSVD and current treatment of this disease.
Collapse
Affiliation(s)
- Qian Li
- 1 Department of Pediatrics, The Third Affiliated Hospital & Field Surgery Institution, Army Medical University, Chongqing, China.,Both the authors contributed equally as co-authors
| | - Yang Yang
- 2 Department of Neurology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.,Both the authors contributed equally as co-authors
| | - Cesar Reis
- 3 Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Tao Tao
- 2 Department of Neurology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Wanwei Li
- 1 Department of Pediatrics, The Third Affiliated Hospital & Field Surgery Institution, Army Medical University, Chongqing, China
| | - Xiaogang Li
- 2 Department of Neurology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - John H Zhang
- 3 Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, USA.,4 Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| |
Collapse
|
43
|
Brown R, Benveniste H, Black SE, Charpak S, Dichgans M, Joutel A, Nedergaard M, Smith KJ, Zlokovic BV, Wardlaw JM. Understanding the role of the perivascular space in cerebral small vessel disease. Cardiovasc Res 2018; 114:1462-1473. [PMID: 29726891 PMCID: PMC6455920 DOI: 10.1093/cvr/cvy113] [Citation(s) in RCA: 237] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/18/2018] [Accepted: 05/02/2018] [Indexed: 12/17/2022] Open
Abstract
Small vessel diseases (SVDs) are a group of disorders that result from pathological alteration of the small blood vessels in the brain, including the small arteries, capillaries and veins. Of the 35-36 million people that are estimated to suffer from dementia worldwide, up to 65% have an SVD component. Furthermore, SVD causes 20-25% of strokes, worsens outcome after stroke and is a leading cause of disability, cognitive impairment and poor mobility. Yet the underlying cause(s) of SVD are not fully understood. Magnetic resonance imaging has confirmed enlarged perivascular spaces (PVS) as a hallmark feature of SVD. In healthy tissue, these spaces are proposed to form part of a complex brain fluid drainage system which supports interstitial fluid exchange and may also facilitate clearance of waste products from the brain. The pathophysiological signature of PVS and what this infers about their function and interaction with cerebral microcirculation, plus subsequent downstream effects on lesion development in the brain has not been established. Here we discuss the potential of enlarged PVS to be a unique biomarker for SVD and related brain disorders with a vascular component. We propose that widening of PVS suggests presence of peri-vascular cell debris and other waste products that form part of a vicious cycle involving impaired cerebrovascular reactivity, blood-brain barrier dysfunction, perivascular inflammation and ultimately impaired clearance of waste proteins from the interstitial fluid space, leading to accumulation of toxins, hypoxia, and tissue damage. Here, we outline current knowledge, questions and hypotheses regarding understanding the brain fluid dynamics underpinning dementia and stroke through the common denominator of SVD.
Collapse
Affiliation(s)
- Rosalind Brown
- Centre for Clinical Brain Sciences, The University of Edinburgh, Chancellor's Building, Edinburgh, UK
| | - Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, USA
| | - Sandra E Black
- LC Campbell Cognitive Neurology Research Unit, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
- Department of Medicine (Neurology), Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
- Hurvitz Brain Sciences Program, Sunnybrook Health Sciences Center, University of Toronto, Toronto, Canada
- Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - Serge Charpak
- INSERM U1128, Laboratory of Neurophysiology and New Microscopies, Université Paris Descartes, Paris, France
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE, Munich), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Anne Joutel
- Genetics and Pathogenesis of Cerebrovascular Diseases, INSERM, Université Paris Diderot-Paris 7, Paris, France
- DHU NeuroVasc, Sorbonne Paris Cité, Paris, France
| | - Maiken Nedergaard
- Section for Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
- Division of Glia Disease and Therapeutics, Center for Translational Neuromedicine, University of Rochester Medical School, Rochester, USA
| | - Kenneth J Smith
- Department of Neuroinflammation, UCL Institute of Neurology, London, UK
| | - Berislav V Zlokovic
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Joanna M Wardlaw
- Centre for Clinical Brain Sciences, Chancellor's Building, Edinburgh, UK
- UK Dementia Research Institute at The University of Edinburgh, Chancellor's Building, Edinburgh, UK
- Row Fogo Centre for Research into Ageing and the Brain, The University of Edinburgh, Chancellor's Building, Edinburgh, UK
| |
Collapse
|
44
|
Khan I, Krishnaswamy S, Sabale M, Groth D, Wijaya L, Morici M, Berger I, Schaffitzel C, Fraser PE, Martins RN, Verdile G. Efficient production of a mature and functional gamma secretase protease. Sci Rep 2018; 8:12834. [PMID: 30150752 PMCID: PMC6110731 DOI: 10.1038/s41598-018-30788-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 08/06/2018] [Indexed: 12/27/2022] Open
Abstract
Baculoviral protein expression in insect cells has been previously used to generate large quantities of a protein of interest for subsequent use in biochemical and structural analyses. The MultiBac baculovirus protein expression system has enabled, the use of a single baculovirus to reconstitute a protein complex of interest, resulting in a larger protein yield. Using this system, we aimed to reconstruct the gamma (γ)-secretase complex, a multiprotein enzyme complex essential for the production of amyloid-β (Aβ) protein. A MultiBac vector containing all components of the γ-secretase complex was generated and expression was observed for all components. The complex was active in processing APP and Notch derived γ-secretase substrates and proteolysis could be inhibited with γ-secretase inhibitors, confirming specificity of the recombinant γ-secretase enzyme. Finally, affinity purification was used to purify an active recombinant γ-secretase complex. In this study we demonstrated that the MultiBac protein expression system can be used to generate an active γ-secretase complex and provides a new tool to study γ-secretase enzyme and its variants.
Collapse
Affiliation(s)
- Imran Khan
- School of Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia. .,School of Psychiatry and Clinical Neurosciences, University of Western Australia, Crawley, Western Australia, Australia. .,Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.
| | - Sudarsan Krishnaswamy
- Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Miheer Sabale
- School of Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - David Groth
- School of Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
| | - Linda Wijaya
- Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,School of Psychology and Exercise Sciences, Murdoch University, Murdoch, Western Australia, Australia
| | - Michael Morici
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, Crawley, Western Australia, Australia.,Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Imre Berger
- European Molecular Biology Laboratories, Grenoble, France.,School of Biochemistry, University of Bristol, Bristol, UK
| | - Christiane Schaffitzel
- European Molecular Biology Laboratories, Grenoble, France.,School of Biochemistry, University of Bristol, Bristol, UK
| | - Paul E Fraser
- Tanz Centre for Research in Neurodegenerative Diseases and Department of Medical Biophysics, Krembil Discovery Tower, University of Toronto, Toronto, Ontario, Canada
| | - Ralph N Martins
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, Crawley, Western Australia, Australia.,Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.,Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Giuseppe Verdile
- School of Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia. .,School of Psychiatry and Clinical Neurosciences, University of Western Australia, Crawley, Western Australia, Australia. .,Centre of Excellence for Alzheimer's Disease Research & Care, School of Medical Sciences, Edith Cowan University, Joondalup, Western Australia, Australia.
| |
Collapse
|
45
|
Zhang Q, Zhou J, Lei H, Zhu CY, Li FF, Zheng D, Liu SL. RBPJ polymorphisms associated with cerebral infarction diseases in Chinese Han population: A Clinical Trial/Experimental Study (CONSORT Compliant). Medicine (Baltimore) 2018; 97:e11420. [PMID: 30075508 PMCID: PMC6081149 DOI: 10.1097/md.0000000000011420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
TRIAL DESIGN Cerebral small vessel diseases (CSVDs) are a group of brain pathological processes involving cerebral small arteries, brain venules, and capillaries. The recombination signal-binding protein Jκ (RBPJ) is implicated in the pathogenesis of these diseases but its actual roles need confirmation. The aim of this work was to evaluate variations in RBPJ gene for their possible associations with the disease. METHODS The RBPJ gene was sequenced for 400 patients with cerebral infarction disease and 600 normal controls. The statistical analyses and Hardy-Weinberg equilibrium tests of the patients and control populations were conducted using the SPSS software (version 19.0) and Plink (version 1.9), Haploview software, and online software SNPSpD. RESULTS We characterized variants rs2871198, rs1397731, rs3822223, rs2077777, rs2270226, and rs2788861 within or near the RBPJ gene. The genetic heterozygosity of rs2871198, rs1397731, rs3822223, rs2077777, and rs2270226 was very high. Statistical analysis showed that the variants rs2270226 and rs2077777 in the gene were associated with the risk of cerebral infarction diseases in the Chinese Han population. CONCLUSIONS rs2270226 and rs2077777 in the RBPJ gene were associated with the risk of cerebral infarction diseases in the Chinese Han population.
Collapse
Affiliation(s)
- Qiong Zhang
- College of Wildlife Resources, Northeast Forestry University
- Department of Antibiotics, Heilongjiang Institute for Food and Drug Control
- Systemomics Center, College of Pharmacy, and Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin
| | - Jie Zhou
- Systemomics Center, College of Pharmacy, and Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang
| | - Hong Lei
- Systemomics Center, College of Pharmacy, and Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang
| | - Chun-Yu Zhu
- Department of Neurology, Daqing Oilfield General Hospital, Daqing, China
| | - Fei-Feng Li
- Systemomics Center, College of Pharmacy, and Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang
| | - Dong Zheng
- College of Wildlife Resources, Northeast Forestry University
| | - Shu-Lin Liu
- Systemomics Center, College of Pharmacy, and Genomics Research Center (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China), Harbin Medical University, Harbin
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Heilongjiang
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Canada
| |
Collapse
|
46
|
Zellner A, Scharrer E, Arzberger T, Oka C, Domenga-Denier V, Joutel A, Lichtenthaler SF, Müller SA, Dichgans M, Haffner C. CADASIL brain vessels show a HTRA1 loss-of-function profile. Acta Neuropathol 2018; 136:111-125. [PMID: 29725820 DOI: 10.1007/s00401-018-1853-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 03/29/2018] [Accepted: 04/24/2018] [Indexed: 01/06/2023]
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) and a phenotypically similar recessive condition (CARASIL) have emerged as important genetic model diseases for studying the molecular pathomechanisms of cerebral small vessel disease (SVD). CADASIL, the most frequent and intensely explored monogenic SVD, is characterized by a severe pathology in the cerebral vasculature including the mutation-induced aggregation of the Notch3 extracellular domain (Notch3ECD) and the formation of protein deposits of insufficiently determined composition in vessel walls. To identify key molecules and pathways involved in this process, we quantitatively determined the brain vessel proteome from CADASIL patient and control autopsy samples (n = 6 for each group), obtaining 95 proteins with significantly increased abundance. Intriguingly, high-temperature requirement protein A1 (HTRA1), the extracellular protease mutated in CARASIL, was found to be strongly enriched (4.9-fold, p = 1.6 × 10-3) and to colocalize with Notch3ECD deposits in patient vessels suggesting a sequestration process. Furthermore, the presence of increased levels of several HTRA1 substrates in the CADASIL proteome was compatible with their reduced degradation as consequence of a loss of HTRA1 activity. Indeed, a comparison with the brain vessel proteome of HTRA1 knockout mice (n = 5) revealed a highly significant overlap of 18 enriched proteins (p = 2.2 × 10-16), primarily representing secreted and extracellular matrix factors. Several of them were shown to be processed by HTRA1 in an in vitro proteolysis assay identifying them as novel substrates. Our study provides evidence for a loss of HTRA1 function as a critical step in the development of CADASIL pathology linking the molecular mechanisms of two distinct SVD forms.
Collapse
Affiliation(s)
- Andreas Zellner
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Eva Scharrer
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Thomas Arzberger
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-Universität München, Munich, Germany
- Department of Psychiatry and Psychotherapy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Chio Oka
- Laboratory of Gene Function in Animals, Nara Institute of Science and Technology, Takayama, Ikoma, Nara, Japan
| | - Valérie Domenga-Denier
- Department of Genetics and Pathogenesis of Cerebrovascular Diseases, INSERM, UMRS 1161, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- DHU NeuroVasc, Sorbonne Paris Cité, Paris, France
| | - Anne Joutel
- Department of Genetics and Pathogenesis of Cerebrovascular Diseases, INSERM, UMRS 1161, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
- DHU NeuroVasc, Sorbonne Paris Cité, Paris, France
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Institute for Advanced Study, Technische Universität München, Garching, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Stephan A Müller
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Christof Haffner
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität München, Feodor-Lynen-Straße 17, 81377, Munich, Germany.
| |
Collapse
|
47
|
Vinters HV, Zarow C, Borys E, Whitman JD, Tung S, Ellis WG, Zheng L, Chui HC. Review: Vascular dementia: clinicopathologic and genetic considerations. Neuropathol Appl Neurobiol 2018; 44:247-266. [DOI: 10.1111/nan.12472] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 01/13/2018] [Indexed: 12/21/2022]
Affiliation(s)
- H. V. Vinters
- Departments of Pathology & Laboratory Medicine (Neuropathology) and Neurology; David Geffen School of Medicine at UCLA; Los Angeles CA USA
| | - C. Zarow
- Department of Neurology; Keck School of Medicine at University of Southern California; Los Angeles CA USA
| | - E. Borys
- Department of Pathology; University of California Davis School of Medicine; Sacramento CA USA
- Department of Pathology; Loyola University Medical Center; Maywood IL USA
| | - J. D. Whitman
- Departments of Pathology & Laboratory Medicine (Neuropathology) and Neurology; David Geffen School of Medicine at UCLA; Los Angeles CA USA
- Departments of Pathology & Laboratory Medicine; UC San Francisco Medical Center; San Francisco CA USA
| | - S. Tung
- Departments of Pathology & Laboratory Medicine (Neuropathology) and Neurology; David Geffen School of Medicine at UCLA; Los Angeles CA USA
| | - W. G. Ellis
- Department of Pathology; University of California Davis School of Medicine; Sacramento CA USA
| | - L. Zheng
- Department of Neurology; Keck School of Medicine at University of Southern California; Los Angeles CA USA
| | - H. C. Chui
- Department of Neurology; Keck School of Medicine at University of Southern California; Los Angeles CA USA
| |
Collapse
|
48
|
Abstract
The term vascular cognitive impairment (VCI) was introduced around the start of the new millennium and refers to the contribution of vascular pathology to any severity of cognitive impairment, ranging from subjective cognitive decline and mild cognitive impairment to dementia. Although vascular pathology is common in elderly individuals with cognitive decline, pure vascular dementia (that is, dementia caused solely by vascular pathology) is uncommon. Indeed, most patients with vascular dementia also have other types of pathology, the most common of which is Alzheimer disease (specifically, the diffuse accumulation of amyloid-β plaques and neurofibrillary tangles composed of tau). At present, the main treatment for VCI is prevention by treating vascular diseases and other risk factors for VCI, such as hypertension and diabetes mellitus. Despite the current paucity of disease-modifying pharmacological treatments, we foresee that eventually, we might be able to target specific brain diseases to prevent cognitive decline and dementia.
Collapse
|
49
|
Neurovascular dysfunction in dementia - human cellular models and molecular mechanisms. Clin Sci (Lond) 2018; 132:399-418. [PMID: 29444850 DOI: 10.1042/cs20160720] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 01/15/2018] [Accepted: 01/19/2018] [Indexed: 02/08/2023]
Abstract
From the earliest stages of development, when cerebral angiogenesis and neurogenesis are entwined, to the end of life, the interplay between vascular and neural systems of the brain is critical in health and disease. Cerebral microvascular endothelial cells constitute the blood-brain barrier and in concert with pericytes or smooth muscle cells, glia and neurons, integrate into a functional neurovascular unit (NVU). This multicellular NVU maintains homoeostasis of the brain's microenvironment by restricting the entry of systemic pathogens and neurotoxins as well as meeting the metabolic demands of neural activity. Recent evidence of cerebral microvascular pathologies in vascular diseases and dementia, including Alzheimer's disease, has challenged the notion that vascular events are merely the consequence of neuronal pathology. This review focuses on molecular mechanisms of neurovascular dysfunction in dementia and outlines currently employed in vitro models to decode such mechanisms. Deciphering neurovascular crosstalk is likely to be more important in understanding the molecular mechanisms of disease than previously anticipated and may offer novel therapeutic opportunities for dementia and related conditions.
Collapse
|