1
|
Huang WQ, Lin Q, Tzeng CM. Leukoaraiosis: Epidemiology, Imaging, Risk Factors, and Management of Age-Related Cerebral White Matter Hyperintensities. J Stroke 2024; 26:131-163. [PMID: 38836265 PMCID: PMC11164597 DOI: 10.5853/jos.2023.02719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 01/15/2024] [Indexed: 06/06/2024] Open
Abstract
Leukoaraiosis (LA) manifests as cerebral white matter hyperintensities on T2-weighted magnetic resonance imaging scans and corresponds to white matter lesions or abnormalities in brain tissue. Clinically, it is generally detected in the early 40s and is highly prevalent globally in individuals aged >60 years. From the imaging perspective, LA can present as several heterogeneous forms, including punctate and patchy lesions in deep or subcortical white matter; lesions with periventricular caps, a pencil-thin lining, and smooth halo; as well as irregular lesions, which are not always benign. Given its potential of having deleterious effects on normal brain function and the resulting increase in public health burden, considerable effort has been focused on investigating the associations between various risk factors and LA risk, and developing its associated clinical interventions. However, study results have been inconsistent, most likely due to potential differences in study designs, neuroimaging methods, and sample sizes as well as the inherent neuroimaging heterogeneity and multi-factorial nature of LA. In this article, we provided an overview of LA and summarized the current knowledge regarding its epidemiology, neuroimaging classification, pathological characteristics, risk factors, and potential intervention strategies.
Collapse
Affiliation(s)
- Wen-Qing Huang
- Department of Central Laboratory, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qing Lin
- Department of Neurology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
- Xiamen Clinical Research Center for Neurological Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
- Fujian Provincial Clinical Research Center for Brain Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
- The Third Clinical College, Fujian Medical University, Fuzhou, Fujian, China
| | - Chi-Meng Tzeng
- Translational Medicine Research Center, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
2
|
Askew KE, Beverley J, Sigfridsson E, Szymkowiak S, Emelianova K, Dando O, Hardingham GE, Duncombe J, Hennessy E, Koudelka J, Samarasekera N, Salman RA, Smith C, Tavares AAS, Gomez‐Nicola D, Kalaria RN, McColl BW, Horsburgh K. Inhibiting CSF1R alleviates cerebrovascular white matter disease and cognitive impairment. Glia 2024; 72:375-395. [PMID: 37909242 PMCID: PMC10952452 DOI: 10.1002/glia.24481] [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: 06/02/2023] [Revised: 09/27/2023] [Accepted: 10/04/2023] [Indexed: 11/02/2023]
Abstract
White matter abnormalities, related to poor cerebral perfusion, are a core feature of small vessel cerebrovascular disease, and critical determinants of vascular cognitive impairment and dementia. Despite this importance there is a lack of treatment options. Proliferation of microglia producing an expanded, reactive population and associated neuroinflammatory alterations have been implicated in the onset and progression of cerebrovascular white matter disease, in patients and in animal models, suggesting that targeting microglial proliferation may exert protection. Colony-stimulating factor-1 receptor (CSF1R) is a key regulator of microglial proliferation. We found that the expression of CSF1R/Csf1r and other markers indicative of increased microglial abundance are significantly elevated in damaged white matter in human cerebrovascular disease and in a clinically relevant mouse model of chronic cerebral hypoperfusion and vascular cognitive impairment. Using the mouse model, we investigated long-term pharmacological CSF1R inhibition, via GW2580, and demonstrated that the expansion of microglial numbers in chronic hypoperfused white matter is prevented. Transcriptomic analysis of hypoperfused white matter tissue showed enrichment of microglial and inflammatory gene sets, including phagocytic genes that were the predominant expression modules modified by CSF1R inhibition. Further, CSF1R inhibition attenuated hypoperfusion-induced white matter pathology and rescued spatial learning impairments and to a lesser extent cognitive flexibility. Overall, this work suggests that inhibition of CSF1R and microglial proliferation mediates protection against chronic cerebrovascular white matter pathology and cognitive deficits. Our study nominates CSF1R as a target for the treatment of vascular cognitive disorders with broader implications for treatment of other chronic white matter diseases.
Collapse
Affiliation(s)
| | - Joshua Beverley
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | - Emma Sigfridsson
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | - Stefan Szymkowiak
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
- UK Dementia Research InstituteUniversity of EdinburghEdinburghUK
| | - Katherine Emelianova
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
- UK Dementia Research InstituteUniversity of EdinburghEdinburghUK
| | - Owen Dando
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
- UK Dementia Research InstituteUniversity of EdinburghEdinburghUK
| | - Giles E. Hardingham
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
- UK Dementia Research InstituteUniversity of EdinburghEdinburghUK
| | - Jessica Duncombe
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | - Edel Hennessy
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | - Juraj Koudelka
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
- UK Dementia Research InstituteUniversity of EdinburghEdinburghUK
| | - Neshika Samarasekera
- Centre for Clinical Brain Sciences and Sudden Death Brain BankUniversity of EdinburghEdinburghUK
| | - Rustam Al‐Shahi Salman
- Centre for Clinical Brain Sciences and Sudden Death Brain BankUniversity of EdinburghEdinburghUK
| | - Colin Smith
- Centre for Clinical Brain Sciences and Sudden Death Brain BankUniversity of EdinburghEdinburghUK
| | - Adriana A. S. Tavares
- British Heart Foundation Centre for Cardiovascular ScienceUniversity of EdinburghEdinburghUK
| | | | - Raj N. Kalaria
- Clinical and Translational Research InstituteNewcastle UniversityNewcastleUK
| | - Barry W. McColl
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
- UK Dementia Research InstituteUniversity of EdinburghEdinburghUK
| | - Karen Horsburgh
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| |
Collapse
|
3
|
Wharton SB, Simpson JE, Ince PG, Richardson CD, Merrick R, Matthews FE, Brayne C. Insights into the pathological basis of dementia from population-based neuropathology studies. Neuropathol Appl Neurobiol 2023; 49:e12923. [PMID: 37462105 PMCID: PMC10946587 DOI: 10.1111/nan.12923] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/13/2023] [Accepted: 06/29/2023] [Indexed: 08/17/2023]
Abstract
The epidemiological neuropathology perspective of population and community-based studies allows unbiased assessment of the prevalence of various pathologies and their relationships to late-life dementia. In addition, this approach provides complementary insights to conventional case-control studies, which tend to be more representative of a younger clinical cohort. The Cognitive Function and Ageing Study (CFAS) is a longitudinal study of cognitive impairment and frailty in the general United Kingdom population. In this review, we provide an overview of the major findings from CFAS, alongside other studies, which have demonstrated a high prevalence of pathology in the ageing brain, particularly Alzheimer's disease neuropathological change and vascular pathology. Increasing burdens of these pathologies are the major correlates of dementia, especially neurofibrillary tangles, but there is substantial overlap in pathology between those with and without dementia, particularly at intermediate burdens of pathology and also at the oldest ages. Furthermore, additional pathologies such as limbic-predominant age-related TDP-43 encephalopathy, ageing-related tau astrogliopathy and primary age-related tauopathies contribute to late-life dementia. Findings from ageing population-representative studies have implications for the understanding of dementia pathology in the community. The high prevalence of pathology and variable relationship to dementia status has implications for disease definition and indicate a role for modulating factors on cognitive outcome. The complexity of late-life dementia, with mixed pathologies, indicates a need for a better understanding of these processes across the life-course to direct the best research for reducing risk in later life of avoidable clinical dementia syndromes.
Collapse
Affiliation(s)
- Stephen B. Wharton
- Sheffield Institute for Translational NeuroscienceUniversity of SheffieldSheffieldUK
| | - Julie E. Simpson
- Sheffield Institute for Translational NeuroscienceUniversity of SheffieldSheffieldUK
| | - Paul G. Ince
- Sheffield Institute for Translational NeuroscienceUniversity of SheffieldSheffieldUK
| | | | - Richard Merrick
- Cambridge Public Health, School of Clinical MedicineUniversity of CambridgeSheffieldUK
| | | | - Carol Brayne
- Cambridge Public Health, School of Clinical MedicineUniversity of CambridgeSheffieldUK
| | | |
Collapse
|
4
|
Zhang X, An H, Chen Y, Shu N. Neurobiological Mechanisms of Cognitive Decline Correlated with Brain Aging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1419:127-146. [PMID: 37418211 DOI: 10.1007/978-981-99-1627-6_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Cognitive decline has emerged as one of the greatest health threats of old age. Meanwhile, aging is the primary risk factor for Alzheimer's disease (AD) and other prevalent neurodegenerative disorders. Developing therapeutic interventions for such conditions demands a greater understanding of the processes underlying normal and pathological brain aging. Despite playing an important role in the pathogenesis and incidence of disease, brain aging has not been well understood at a molecular level. Recent advances in the biology of aging in model organisms, together with molecular- and systems-level studies of the brain, are beginning to shed light on these mechanisms and their potential roles in cognitive decline. This chapter seeks to integrate the knowledge about the neurological mechanisms of age-related cognitive changes that underlie aging.
Collapse
Affiliation(s)
- Xiaxia Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, Faculty of Psychology, Beijing Normal University, Beijing, China
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
| | - Haiting An
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
- Beijing Neurosurgical Institute, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China
| | - Yuan Chen
- State Key Laboratory of Cognitive Neuroscience and Learning, Faculty of Psychology, Beijing Normal University, Beijing, China
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
| | - Ni Shu
- State Key Laboratory of Cognitive Neuroscience and Learning, Faculty of Psychology, Beijing Normal University, Beijing, China.
| |
Collapse
|
5
|
Ruchoux MM, Kalaria RN, Román GC. The pericyte: A critical cell in the pathogenesis of CADASIL. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2021; 2:100031. [PMID: 34950895 PMCID: PMC8661128 DOI: 10.1016/j.cccb.2021.100031] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 10/12/2021] [Accepted: 10/12/2021] [Indexed: 12/22/2022]
Abstract
CADASIL is the most common hereditary small vessel disease presenting with strokes and subcortical vascular dementia caused by mutations in the NOTCH3 gene. CADASIL is a vasculopathy primarily involving vascular smooth-muscle cells. Arteriolar and capillary pericyte damage or deficiency is a key feature in disease pathogenesis. Pericyte-mediated cerebral venous insufficiency may explain white matter lesions and increased perivascular spaces. Central role of the pericyte offers novel approaches to the treatment of CADASIL.
Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) is a hereditary small vessel disease presenting with migraine, mood and cognitive disorders, focal neurological deficits, recurrent ischemic attacks, lacunar infarcts and brain white matter changes. As they age, CADASIL patients invariably develop cognitive impairment and subcortical dementia. CADASIL is caused by missense mutations in the NOTCH3 gene resulting in a profound cerebral vasculopathy affecting primarily arterial vascular smooth muscle cells, which target the microcirculation and perfusion. Based on a thorough review of morphological lesions in arteries, veins, and capillaries in CADASIL, we surmise that arteriolar and capillary pericyte damage or deficiency appears a key feature in the pathogenesis of the disease. This may affect critical pericyte-endothelial interactions causing stroke injury and vasomotor disturbances. Changes in microvascular permeability due to perhaps localized blood-brain barrier alterations and pericyte secretory dysfunction likely contribute to delayed neuronal as well as glial cell death. Moreover, pericyte-mediated cerebral venous insufficiency may explain white matter lesions and the dilatation of Virchow-Robin perivascular spaces typical of CADASIL. The postulated central role of the pericyte offers some novel approaches to the study and treatment of CADASIL and enable elucidation of other forms of cerebral small vessel diseases and subcortical vascular dementia.
Collapse
Affiliation(s)
- Marie-Magdeleine Ruchoux
- Former researcher, Université d'Artois, Blood-Brain-Barrier Laboratory Lens France, Former advisor, Alzheimer's Clinic Methodist Neurological Institute, Houston TX, USA
| | - Raj N Kalaria
- Translational and Clinical Research Institute, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Gustavo C Román
- Methodist Neurological Institute, Department of Neurology, Houston Methodist Hospital Houston TX 77030, USA, Weill Cornell Medical College, New York NY, USA and Texas A&M Medical School, Bryan TX, USA
| |
Collapse
|
6
|
Huang J, Yang J, Zou X, Zuo S, Wang J, Cheng J, Zhu H, Li W, Shi M, Zhao G, Liu Z. Ginkgolide B promotes oligodendrocyte precursor cell differentiation and survival via Akt/CREB/bcl-2 signaling pathway after white matter lesion. Exp Biol Med (Maywood) 2021; 246:1198-1209. [PMID: 33557607 PMCID: PMC8142115 DOI: 10.1177/1535370221989955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/03/2021] [Indexed: 12/12/2022] Open
Abstract
White matter lesion (WML) is caused by chronic cerebral hypoperfusion, which are usually associated with cognitive impairment. Evidence from recent studies has shown that ginkgolide B has a neuroprotective effect that could be beneficial for the treatment of ischemia; however, it is not clear whether ginkgolide B has a protective effect on WML. Our data show that ginkgolide B can promote the differentiation of oligodendrocyte precursor cell (OPC) into oligodendrocytes and promote oligodendrocyte survival following a WML. Ginkgolide B (5, 10, 20 mg/kg) or saline is administered intraperitoneally every day after WML. After 4 weeks, the data of Morris water maze suggested that rats' memory and learning abilities were impaired, and the administration of ginkgolide B enhanced behavioral achievement. Also, treatment with ginkgolide B significantly attenuated this loss of myelin. Our result suggests that ginkgolide B promotes the differentiation of OPC into oligodendrocytes. We also found that ginkgolide B ameliorates oligodendrocytes apoptosis. Furthermore, ginkgolide B enhanced the expression of phosphorylated Akt and CREB. In conclusion, our data firstly show that ginkgolide B promotes oligodendrocyte genesis and oligodendrocyte myelin following a WML, possibly involving the Akt and CREB pathways.
Collapse
Affiliation(s)
- Jian Huang
- Department of Neurology, Xijing Hospital, Airforce Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jun Yang
- Department of Nephrology, Xijing Hospital, Airforce Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xingju Zou
- Department of Neurology, Xijing Hospital, Airforce Military Medical University, Xi'an, Shaanxi 710032, China
| | - Shilun Zuo
- Department of Neurology, Second Affiliated Hospital of Army Military Medical University, Chongqing 400038, China
| | - Jing Wang
- Department of Neurology, Xijing Hospital, Airforce Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jing Cheng
- Department of Neurology, Xijing Hospital, Airforce Military Medical University, Xi'an, Shaanxi 710032, China
| | - Hao Zhu
- Department of Neurology, Xijing Hospital, Airforce Military Medical University, Xi'an, Shaanxi 710032, China
| | - Weiwang Li
- Department of Neurology, Xijing Hospital, Airforce Military Medical University, Xi'an, Shaanxi 710032, China
| | - Ming Shi
- Department of Neurology, Xijing Hospital, Airforce Military Medical University, Xi'an, Shaanxi 710032, China
| | - Gang Zhao
- Department of Neurology, Xijing Hospital, Airforce Military Medical University, Xi'an, Shaanxi 710032, China
| | - Zhirong Liu
- Department of Neurology, Xijing Hospital, Airforce Military Medical University, Xi'an, Shaanxi 710032, China
| |
Collapse
|
7
|
Waller R, Narramore R, Simpson JE, Heath PR, Verma N, Tinsley M, Barnes JR, Haris HT, Henderson FE, Matthews FE, Richardson CD, Brayne C, Ince PG, Kalaria RN, Wharton SB. Heterogeneity of cellular inflammatory responses in ageing white matter and relationship to Alzheimer's and small vessel disease pathologies. Brain Pathol 2021; 31:e12928. [PMID: 33336479 PMCID: PMC8412112 DOI: 10.1111/bpa.12928] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 12/18/2022] Open
Abstract
White matter lesions (WML) are common in the ageing brain, often arising in a field effect of diffuse white matter abnormality. Although WML are associated with cerebral small vessel disease (SVD) and Alzheimer’s disease (AD), their cause and pathogenesis remain unclear. The current study tested the hypothesis that different patterns of neuroinflammation are associated with SVD compared to AD neuropathology by assessing the immunoreactive profile of the microglial (CD68, IBA1 and MHC‐II) and astrocyte (GFAP) markers in ageing parietal white matter (PARWM) obtained from the Cognitive Function and Ageing Study (CFAS), an ageing population‐representative neuropathology cohort. Glial responses varied extensively across the PARWM with microglial markers significantly higher in the subventricular region compared to either the middle‐zone (CD68 p = 0.028, IBA1 p < 0.001, MHC‐II p < 0.001) or subcortical region (CD68 p = 0.002, IBA1 p < 0.001, MHC‐II p < 0.001). Clasmatodendritic (CD) GFAP+ astrocytes significantly increased from the subcortical to the subventricular region (p < 0.001), whilst GFAP+ stellate astrocytes significantly decreased (p < 0.001). Cellular reactions could be grouped into two distinct patterns: an immune response associated with MHC‐II/IBA1 expression and CD astrocytes; and a more innate response characterised by CD68 expression associated with WML. White matter neuroinflammation showed weak relationships to the measures of SVD, but not to the measures of AD neuropathology. In conclusion, glial responses vary extensively across the PARWM with diverse patterns of white matter neuroinflammation. Although these findings support a role for vascular factors in the pathogenesis of age‐related white matter neuroinflammation, additional factors other than SVD and AD pathology may drive this. Understanding the heterogeneity in white matter neuroinflammation will be important for the therapeutic targeting of age‐associated white matter damage.
Collapse
Affiliation(s)
- Rachel Waller
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Ruth Narramore
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Julie E Simpson
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Paul R Heath
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Nikita Verma
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Megan Tinsley
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Jordan R Barnes
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Hanna T Haris
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Frances E Henderson
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Fiona E Matthews
- Translational and Clinical Research Institute, University of Newcastle, Newcastle upon Tyne, UK
| | - Connor D Richardson
- Translational and Clinical Research Institute, University of Newcastle, Newcastle upon Tyne, UK
| | - Carol Brayne
- Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Paul G Ince
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Raj N Kalaria
- Translational and Clinical Research Institute, University of Newcastle, Newcastle upon Tyne, UK
| | - Stephen B Wharton
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | | |
Collapse
|
8
|
Huang WQ, Lin Q, Chen S, Sun L, Chen Q, Yi K, Li Z, Ma Q, Tzeng CM. Integrated analysis of microRNA and mRNA expression profiling identifies BAIAP3 as a novel target of dysregulated hsa-miR-1972 in age-related white matter lesions. Aging (Albany NY) 2021; 13:4674-4695. [PMID: 33561007 PMCID: PMC7906144 DOI: 10.18632/aging.202562] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/18/2020] [Indexed: 11/25/2022]
Abstract
White matter lesions known as leukoaraiosis (LA) are cerebral white matter hyperintensities observed in elderly individuals. Currently, no reliable molecular biomarkers are available for monitoring their progression over time. To identify biomarkers for the onset and progression of LA, we analyzed whole blood-based, microRNA expression profiles of leukoaraiosis, validated those exhibiting significant microRNA changes in clinical subjects by means of quantitative real-time polymerase chain reactions and determined the function of miRNA in cell lines by means of microRNA mimic transfection assays. A total of seven microRNAs were found to be significantly down-regulated in leukoaraiosis. Among the microRNAs, hsa-miR-1972 was downregulated during the early onset phase of leukoaraiosis, as confirmed in independent patients, and it was found to target leukoaraiosis-dependent BAIAP3, decreasing its expression in 293T cell lines. Functional enrichment analysis revealed that significantly dysregulated miRNAs-mRNAs changes associated with the onset of leukoaraiosis were involved in neurogenesis, neuronal development, and differentiation. Taken together, the study identified a set of candidate microRNA biomarkers that may usefully monitor the onset and progression of leukoaraiosis. Given the enrichment of leukoaraiosis-associated microRNAs and mRNAs in neuron part and membrane system, BAIAP3 could potentially represent a novel target of hsa-miR-1972 in leukoaraiosis through which microRNAs are involved in the pathogenesis of white matter lesions.
Collapse
Affiliation(s)
- Wen-Qing Huang
- Shanghai Institute of Precision Medicine (SHIPM), Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Translational Medicine Research Center (TMRC), School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Qing Lin
- Department of Neurology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China.,Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China.,School of Medicine, Xiamen University, Xiamen, Fujian, China.,The First Clinical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Shuai Chen
- Department of Otolaryngology-Head and Neck Surgery, Xiamen Key Laboratory of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China.,Chen Zhi-nan Academician Workstation, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shanxi, China
| | - Lixiang Sun
- Translational Medicine Research Center (TMRC), School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Qingjie Chen
- Department of Nuclear Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Kehui Yi
- Department of Neurology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China.,Department of Neurology, Zhongshan Xiamen Hospital, Fudan University, Xiamen, Fujian, China
| | - Zhi Li
- Translational Medicine Research Center (TMRC), School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China
| | - Qilin Ma
- Department of Neurology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China.,Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China.,School of Medicine, Xiamen University, Xiamen, Fujian, China.,The First Clinical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Chi-Meng Tzeng
- Translational Medicine Research Center (TMRC), School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China.,College of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, Jiangsu, China
| |
Collapse
|
9
|
Bury JJ, Chambers A, Heath PR, Ince PG, Shaw PJ, Matthews FE, Brayne C, Simpson JE, Wharton SB. Type 2 diabetes mellitus-associated transcriptome alterations in cortical neurones and associated neurovascular unit cells in the ageing brain. Acta Neuropathol Commun 2021; 9:5. [PMID: 33407907 PMCID: PMC7788898 DOI: 10.1186/s40478-020-01109-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 12/15/2020] [Indexed: 12/15/2022] Open
Abstract
Type 2 diabetes mellitus (T2D), characterised by peripheral insulin resistance, is a risk factor for dementia. In addition to its contribution to small and large vessel disease, T2D may directly damage cells of the brain neurovascular unit. In this study, we investigated the transcriptomic changes in cortical neurones, and associated astrocytes and endothelial cells of the neurovascular unit, in the ageing brain. Neurone, astrocyte, and endothelial cell-enriched mRNA, obtained by immuno-laser capture microdissection of temporal cortex (Brodmann area 21/22) from 6 cases with self-reported T2D in the Cognitive Function and Ageing Study neuropathology cohort, and an equal number of age and sex-matched controls, was assessed by microarray analysis. Integrated Molecular Pathway Level Analysis was performed using the Kyoto Encyclopaedia of Genes and Genomes database on significantly differentially expressed genes, defined as P < 0.05 and fold-change ± 1.2. Hub genes identified from Weighted Gene Co-expression Network Analysis were validated in neurones using the NanoString nCounter platform. The expression and cellular localisation of proteins encoded by selected candidate genes were confirmed by immunohistochemistry. 912, 2202, and 1227 genes were significantly differentially expressed between cases with self-reported T2D and controls in neurones, astrocytes, and endothelial cells respectively. Changes in cortical neurones included alterations in insulin and other signalling pathways, cell cycle, cellular senescence, inflammatory mediators, and components of the mitochondrial respiratory electron transport chain. Impaired insulin signalling was shared by neurovascular unit cells with, additionally, apoptotic pathway changes in astrocytes and dysregulation of advanced glycation end-product signalling in endothelial cells. Transcriptomic analysis identified changes in key cellular pathways associated with T2D that may contribute to neuronal damage and dysfunction. These effects on brain cells potentially contribute to a diabetic dementia, and may provide novel approaches for therapeutic intervention.
Collapse
Affiliation(s)
- Joanna J Bury
- Sheffield Institute for Translational Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK
| | - Annabelle Chambers
- Sheffield Institute for Translational Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK
| | - Paul R Heath
- Sheffield Institute for Translational Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK
| | - Paul G Ince
- Sheffield Institute for Translational Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK
| | - Pamela J Shaw
- Sheffield Institute for Translational Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK
| | - Fiona E Matthews
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Carol Brayne
- Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Julie E Simpson
- Sheffield Institute for Translational Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK
| | - Stephen B Wharton
- Sheffield Institute for Translational Neuroscience, University of Sheffield, 385a Glossop Road, Sheffield, S10 2HQ, UK.
| |
Collapse
|
10
|
Transcriptomic Analysis of Age-Associated Periventricular Lesions Reveals Dysregulation of the Immune Response. Int J Mol Sci 2020; 21:ijms21217924. [PMID: 33113879 PMCID: PMC7663268 DOI: 10.3390/ijms21217924] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/23/2020] [Accepted: 10/23/2020] [Indexed: 12/22/2022] Open
Abstract
White matter lesions (WML) are a common feature of the ageing brain associated with cognitive impairment. The gene expression profiles of periventricular lesions (PVL, n = 7) and radiologically-normal-appearing (control) periventricular white matter cases (n = 11) obtained from the Cognitive Function and Ageing Study (CFAS) neuropathology cohort were interrogated using microarray analysis and NanoString to identify novel mechanisms potentially underlying their formation. Histological characterisation of control white matter cases identified a subgroup (n = 4) which contained high levels of MHC-II immunoreactive microglia, and were classified as “pre-lesional.” Microarray analysis identified 2256 significantly differentially-expressed genes (p ≤ 0.05, FC ≥ 1.2) in PVL compared to non-lesional control white matter (1378 upregulated and 878 downregulated); 2649 significantly differentially-expressed genes in “pre-lesional” cases compared to PVL (1390 upregulated and 1259 downregulated); and 2398 significantly differentially-expressed genes in “pre-lesional” versus non-lesional control cases (1527 upregulated and 871 downregulated). Whilst histological evaluation of a single marker (MHC-II) implicates immune-activated microglia in lesion pathology, transcriptomic analysis indicates significant downregulation of a number of activated microglial markers and suggests established PVL are part of a continuous spectrum of white matter injury. The gene expression profile of “pre-lesional” periventricular white matter suggests upregulation of several signalling pathways may be a neuroprotective response to prevent the pathogenesis of PVL.
Collapse
|
11
|
Association between Osteoporosis and Cognitive Impairment during the Acute and Recovery Phases of Ischemic Stroke. ACTA ACUST UNITED AC 2020; 56:medicina56060307. [PMID: 32585806 PMCID: PMC7353884 DOI: 10.3390/medicina56060307] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/08/2020] [Accepted: 06/19/2020] [Indexed: 12/15/2022]
Abstract
Background and objectives: Little is known about the effect of osteoporosis on cognitive function in the acute and recovery phases of stroke. Early bone mineral density assessments during acute stroke may be a useful marker of cognitive function. We evaluated the effect of osteoporosis on cognitive function at the early and recovery phase of ischemic stroke in patients aged >50 years. Materials and Methods: We retrospectively examined consecutive patients with acute stroke hospitalized between 2016 and 2018. Osteoporosis was defined as a T-score <–2.5 for the femoral neck or lumbar spine bone mineral density. The primary outcome was cognitive impairment measured by the Korean Mini-Mental State Examination in the acute phase and recovery phase of ischemic stroke. Results: Of the 260 included subjects (107 men and 153 women), 70 (26.9%) had osteoporosis. Cognitive impairment was more severe in the osteoporosis group than in the non-osteoporosis group (30.5% versus 47.1%, p = 0.001). After the recovery phase of stroke, the proportion of patients with cognitive impairment remained higher in the osteoporosis group. The multivariate analysis revealed a correlation between a low femoral neck bone mineral density and severe cognitive impairment in the acute and recovery phases of stroke (adjusted odds ratio (OR) 4.09, 95% confidence interval (CI) 1.11–15.14 in the acute phase, and adjusted OR 11.17, 95% CI 1.12–110.98 in the recovery phase). Conclusions: Low bone mineral density is associated with poor cognitive function in the acute and recovery phases of stroke.
Collapse
|
12
|
Waller R, Baxter L, Fillingham DJ, Coelho S, Pozo JM, Mozumder M, Frangi AF, Ince PG, Simpson JE, Highley JR. Iba-1-/CD68+ microglia are a prominent feature of age-associated deep subcortical white matter lesions. PLoS One 2019; 14:e0210888. [PMID: 30682074 PMCID: PMC6347230 DOI: 10.1371/journal.pone.0210888] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 01/03/2019] [Indexed: 11/23/2022] Open
Abstract
Deep subcortical lesions (DSCL) of the brain, are present in ~60% of the ageing population, and are linked to cognitive decline and depression. DSCL are associated with demyelination, blood brain barrier (BBB) dysfunction, and microgliosis. Microglia are the main immune cell of the brain. Under physiological conditions microglia have a ramified morphology, and react to pathology with a change to a more rounded morphology as well as showing protein expression alterations. This study builds on previous characterisations of DSCL and radiologically ‘normal-appearing’ white matter (NAWM) by performing a detailed characterisation of a range of microglial markers in addition to markers of vascular integrity. The Cognitive Function and Ageing Study (CFAS) provided control white matter (WM), NAWM and DSCL human post mortem tissue for immunohistochemistry using microglial markers (Iba-1, CD68 and MHCII), a vascular basement membrane marker (collagen IV) and markers of BBB integrity (fibrinogen and aquaporin 4). The immunoreactive profile of CD68 increased in a stepwise manner from control WM to NAWM to DSCL. This correlated with a shift from small, ramified cells, to larger, more rounded microglia. While there was greater Iba-1 immunoreactivity in NAWM compared to controls, in DSCL, Iba-1 levels were reduced to control levels. A prominent feature of these DSCL was a population of Iba-1-/CD68+ microglia. There were increases in collagen IV, but no change in BBB integrity. Overall the study shows significant differences in the immunoreactive profile of microglial markers. Whether this is a cause or effect of lesion development remains to be elucidated. Identifying microglia subpopulations based on their morphology and molecular markers may ultimately help decipher their function and role in neurodegeneration. Furthermore, this study demonstrates that Iba-1 is not a pan-microglial marker, and that a combination of several microglial markers is required to fully characterise the microglial phenotype.
Collapse
Affiliation(s)
- Rachel Waller
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield, South Yorkshire, United Kingdom
- * E-mail:
| | - Lynne Baxter
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield, South Yorkshire, United Kingdom
| | - Daniel J. Fillingham
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield, South Yorkshire, United Kingdom
| | - Santiago Coelho
- School of Computing, Center for Computational Imaging & Simulation Technologies in Biomedicine, The University of Leeds, Leeds, West Yorkshire, United Kingdom
| | - Jose M. Pozo
- School of Computing, Center for Computational Imaging & Simulation Technologies in Biomedicine, The University of Leeds, Leeds, West Yorkshire, United Kingdom
| | - Meghdoot Mozumder
- Department of Engineering, Center for Computational Imaging & Simulation Technologies in Biomedicine, The University of Sheffield, Sheffield, South Yorkshire, United Kingdom
| | - Alejandro F. Frangi
- School of Computing, Center for Computational Imaging & Simulation Technologies in Biomedicine, The University of Leeds, Leeds, West Yorkshire, United Kingdom
| | - Paul G. Ince
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield, South Yorkshire, United Kingdom
| | - Julie E. Simpson
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield, South Yorkshire, United Kingdom
| | - J. Robin Highley
- Department of Neuroscience, Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield, South Yorkshire, United Kingdom
| |
Collapse
|
13
|
Shaaban CE, Jorgensen DR, Gianaros PJ, Mettenburg J, Rosano C. Cerebrovascular disease: Neuroimaging of cerebral small vessel disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 165:225-255. [DOI: 10.1016/bs.pmbts.2019.07.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
14
|
Jorgensen DR, Shaaban CE, Wiley CA, Gianaros PJ, Mettenburg J, Rosano C. A population neuroscience approach to the study of cerebral small vessel disease in midlife and late life: an invited review. Am J Physiol Heart Circ Physiol 2018; 314:H1117-H1136. [PMID: 29393657 PMCID: PMC6032084 DOI: 10.1152/ajpheart.00535.2017] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 01/09/2018] [Accepted: 01/22/2018] [Indexed: 12/28/2022]
Abstract
Aging in later life engenders numerous changes to the cerebral microvasculature. Such changes can remain clinically silent but are associated with greater risk for negative health outcomes over time. Knowledge is limited about the pathogenesis, prevention, and treatment of potentially detrimental changes in the cerebral microvasculature that occur with advancing age. In this review, we summarize literature on aging of the cerebral microvasculature, and we propose a conceptual framework to fill existing research gaps and advance future work on this heterogeneous phenomenon. We propose that the major gaps in this area are attributable to an incomplete characterization of cerebrovascular pathology, the populations being studied, and the temporality of exposure to risk factors. Specifically, currently available measures of age-related cerebral microvasculature changes are indirect, primarily related to parenchymal damage rather than direct quantification of small vessel damage, limiting the understanding of cerebral small vessel disease (cSVD) itself. Moreover, studies seldom account for variability in the health-related conditions or interactions with risk factors, which are likely determinants of cSVD pathogenesis. Finally, study designs are predominantly cross-sectional and/or have relied on single time point measures, leaving no clear evidence of time trajectories of risk factors or of change in cerebral microvasculature. We argue that more resources should be invested in 1) developing methodological approaches and basic science models to better understand the pathogenic and etiological nature of age-related brain microvascular diseases and 2) implementing state-of-the-science population study designs that account for the temporal evolution of cerebral microvascular changes in diverse populations across the lifespan.
Collapse
Affiliation(s)
- Dana R Jorgensen
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - C Elizabeth Shaaban
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Clayton A Wiley
- Department of Pathology, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Peter J Gianaros
- Departments of Psychology and Psychiatry, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Joseph Mettenburg
- Department of Radiology, University of Pittsburgh, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Caterina Rosano
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh , Pittsburgh, Pennsylvania
| |
Collapse
|
15
|
Huang WQ, Yi KH, Li Z, Wang H, Li ML, Cai LL, Lin HN, Lin Q, Tzeng CM. DNA Methylation Profiling Reveals the Change of Inflammation-Associated ZC3H12D in Leukoaraiosis. Front Aging Neurosci 2018; 10:143. [PMID: 29875652 PMCID: PMC5974056 DOI: 10.3389/fnagi.2018.00143] [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: 01/13/2018] [Accepted: 04/27/2018] [Indexed: 01/04/2023] Open
Abstract
Leukoaraiosis (LA) is neuroimaging abnormalities of the cerebral white matter in elderly people. However, the molecular mechanisms underlying the cerebral white matter lesions remain unclear. Here, we reported an epigenetic basis and potential pathogenesis for this complex illness. 317 differentially methylated genes were identified to distinguish the mechanism of occurrence and progression of LA. Gene-Ontology pathway analysis highlighted that those genes with epigenetic changes are mostly involved in four major signaling pathways including inflammation and immune response-associated processes (antigen processing and presentation, T cell costimulation and interferon-γ-mediated signaling pathway), synapse assembly, synaptic transmission and cell adhesion. Moreover, immune response seems to be specific to LA occurrence and subsequent disruption of nervous system functions could drive the progression of LA. The significant change of inflammation-associated ZC3H12D in promoter methylation and mRNA expression was implicated in the occurrence of LA, suggesting its potential functions in the molecular mechanism of LA. Our results suggested that inflammation-associated signaling pathways were involved in the pathogenesis of LA and ZC3H12D may contribute to such inflammatory process underlying LA, and further echoed it as a neuroinflammatory disorder in central nervous system (CNS).
Collapse
Affiliation(s)
- Wen-Qing Huang
- Translational Medicine Research Center, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China.,Key Laboratory for Cancer T-Cell Theranostics and Clinical Translation, Xiamen University, Fujian, China.,Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Ke-Hui Yi
- Department of Neurology, The First Affiliated Hospital of Xiamen University, Xiamen, China.,Department of Neurology, The First Clinical College of Fujian Medical University, Fuzhou, China
| | - Zhi Li
- Translational Medicine Research Center, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China.,Key Laboratory for Cancer T-Cell Theranostics and Clinical Translation, Xiamen University, Fujian, China
| | - Han Wang
- Translational Medicine Research Center, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China.,Key Laboratory for Cancer T-Cell Theranostics and Clinical Translation, Xiamen University, Fujian, China
| | - Ming-Li Li
- Translational Medicine Research Center, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China.,Key Laboratory for Cancer T-Cell Theranostics and Clinical Translation, Xiamen University, Fujian, China
| | - Liang-Liang Cai
- Translational Medicine Research Center, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China.,Key Laboratory for Cancer T-Cell Theranostics and Clinical Translation, Xiamen University, Fujian, China
| | - Hui-Nuan Lin
- Translational Medicine Research Center, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China.,Key Laboratory for Cancer T-Cell Theranostics and Clinical Translation, Xiamen University, Fujian, China
| | - Qing Lin
- Translational Medicine Research Center, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China.,Department of Neurology, The First Affiliated Hospital of Xiamen University, Xiamen, China.,Department of Neurology, The First Clinical College of Fujian Medical University, Fuzhou, China
| | - Chi-Meng Tzeng
- Translational Medicine Research Center, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China.,Key Laboratory for Cancer T-Cell Theranostics and Clinical Translation, Xiamen University, Fujian, China.,INNOVA Cell: TDx/Clinics and TRANSLA Health Group, Yangzhou, China.,College of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, China.,Jiansu Provincial Institute of Translation Medicine and Women-Child Health Care Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
16
|
Vijayan M, Reddy PH. Stroke, Vascular Dementia, and Alzheimer's Disease: Molecular Links. J Alzheimers Dis 2018; 54:427-43. [PMID: 27567871 DOI: 10.3233/jad-160527] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Stroke is a brain disease that occurs when blood flow stops, resulting in reduced oxygen supply to neurons. Stroke occurs at any time and at any age, but increases after the age of 55. It is the second leading cause of death and the third leading cause of disability-adjusted, life-years. The pathophysiology of ischemic stroke is complex and recent molecular, cellular, and animal models and postmortem brain studies have revealed that multiple cellular changes have been implicated, including oxidative stress/mitochondrial dysfunction, inflammatory responses, micro RNA alterations, and marked changes in brain proteins. These cellular changes provide new information for developing therapeutic strategies for ischemic stroke treatment. Research also revealed that stroke increases with a number of modifiable factors and most strokes can be prevented and/or controlled through pharmacological or surgical interventions and lifestyle changes. Ischemic stroke is the major risk factor for vascular dementia and Alzheimer's disease. This review summarizes the latest research findings on stroke, including causal factors and molecular links between stroke and vascular disease/Alzheimer's disease.
Collapse
Affiliation(s)
- Murali Vijayan
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - P Hemachandra Reddy
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Neuroscience and Pharmacology, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Department of Speech, Language and Hearing Sciences Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Garrison Institute on Aging, South West Campus, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| |
Collapse
|
17
|
Hase Y, Horsburgh K, Ihara M, Kalaria RN. White matter degeneration in vascular and other ageing-related dementias. J Neurochem 2018; 144:617-633. [DOI: 10.1111/jnc.14271] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 10/20/2017] [Accepted: 11/20/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Yoshiki Hase
- Neurovascular Research Group; Institute of Neuroscience; Newcastle University; Newcastle Upon Tyne UK
| | - Karen Horsburgh
- Centre for Neuroregeneration; University of Edinburgh; Edinburgh UK
| | - Masafumi Ihara
- Department of Neurology; National Cerebral and Cardiovascular Center; Suita Osaka Japan
| | - Raj N. Kalaria
- Neurovascular Research Group; Institute of Neuroscience; Newcastle University; Newcastle Upon Tyne UK
| |
Collapse
|
18
|
Kalaria RN. The pathology and pathophysiology of vascular dementia. Neuropharmacology 2017; 134:226-239. [PMID: 29273521 DOI: 10.1016/j.neuropharm.2017.12.030] [Citation(s) in RCA: 194] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 12/13/2017] [Accepted: 12/18/2017] [Indexed: 02/07/2023]
Abstract
Vascular dementia (VaD) is widely recognised as the second most common type of dementia. Consensus and accurate diagnosis of clinically suspected VaD relies on wide-ranging clinical, neuropsychological and neuroimaging measures in life but more importantly pathological confirmation. Factors defining subtypes of VaD include the nature and extent of vascular pathologies, degree of involvement of extra and intracranial vessels and the anatomical location of tissue changes as well as time after the initial vascular event. Atherosclerotic and cardioembolic diseases combined appear the most common subtypes of vascular brain injury. In recent years, cerebral small vessel disease (SVD) has gained prominence worldwide as an important substrate of cognitive impairment. SVD is characterised by arteriolosclerosis, lacunar infarcts and cortical and subcortical microinfarcts and diffuse white matter changes, which involve myelin loss and axonal abnormalities. Global brain atrophy and focal degeneration of the cerebrum including medial temporal lobe atrophy are also features of VaD similar to Alzheimer's disease. Hereditary arteriopathies have provided insights into the mechanisms of dementia particularly how arteriolosclerosis, a major contributor of SVD promotes cognitive impairment. Recently developed and validated neuropathology guidelines indicated that the best predictors of vascular cognitive impairment were small or lacunar infarcts, microinfarcts, perivascular space dilation, myelin loss, arteriolosclerosis and leptomeningeal cerebral amyloid angiopathy. While these substrates do not suggest high specificity, VaD is likely defined by key neuronal and dendro-synaptic changes resulting in executive dysfunction and related cognitive deficits. Greater understanding of the molecular pathology is needed to clearly define microvascular disease and vascular substrates of dementia. This article is part of the Special Issue entitled 'Cerebral Ischemia'.
Collapse
Affiliation(s)
- Raj N Kalaria
- Institute of Neuroscience, Newcastle University, Campus for Ageing & Vitality, Newcastle Upon Tyne NE4 5PL, United Kingdom.
| |
Collapse
|
19
|
Chronic cerebral hypoperfusion: a key mechanism leading to vascular cognitive impairment and dementia. Closing the translational gap between rodent models and human vascular cognitive impairment and dementia. Clin Sci (Lond) 2017; 131:2451-2468. [PMID: 28963120 DOI: 10.1042/cs20160727] [Citation(s) in RCA: 231] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 08/28/2017] [Accepted: 09/04/2017] [Indexed: 12/15/2022]
Abstract
Increasing evidence suggests that vascular risk factors contribute to neurodegeneration, cognitive impairment and dementia. While there is considerable overlap between features of vascular cognitive impairment and dementia (VCID) and Alzheimer's disease (AD), it appears that cerebral hypoperfusion is the common underlying pathophysiological mechanism which is a major contributor to cognitive decline and degenerative processes leading to dementia. Sustained cerebral hypoperfusion is suggested to be the cause of white matter attenuation, a key feature common to both AD and dementia associated with cerebral small vessel disease (SVD). White matter changes increase the risk for stroke, dementia and disability. A major gap has been the lack of mechanistic insights into the evolution and progress of VCID. However, this gap is closing with the recent refinement of rodent models which replicate chronic cerebral hypoperfusion. In this review, we discuss the relevance and advantages of these models in elucidating the pathogenesis of VCID and explore the interplay between hypoperfusion and the deposition of amyloid β (Aβ) protein, as it relates to AD. We use examples of our recent investigations to illustrate the utility of the model in preclinical testing of candidate drugs and lifestyle factors. We propose that the use of such models is necessary for tackling the urgently needed translational gap from preclinical models to clinical treatments.
Collapse
|
20
|
Buchman AS, Leurgans SE, Nag S, VanderHorst VGJM, Kapasi A, Schneider JA, Bennett DA. Spinal Arteriolosclerosis Is Common in Older Adults and Associated With Parkinsonism. Stroke 2017; 48:2792-2798. [PMID: 28931619 DOI: 10.1161/strokeaha.117.017643] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 08/09/2017] [Accepted: 08/14/2017] [Indexed: 01/23/2023]
Abstract
BACKGROUND AND PURPOSE There are few studies of spinal microvascular pathologies in older adults. We characterized spinal cord microvascular pathologies and examined their associations with other spinal and brain postmortem indices and parkinsonism in older adults. METHODS We documented 3 features of microvascular pathologies in spinal cord and brain specimens from 165 deceased older participants. We also measured spinal white matter pallor. Parkinsonian signs were assessed with a modified version of the motor section of the Unified Parkinson's Disease Rating Scale. We examined the associations of spinal arteriolosclerosis with other spinal and brain postmortem indices and parkinsonism proximate to death using regression models which controlled for age and sex. RESULTS Microinfarcts and cerebral amyloid angiopathy were not observed within the spinal cord parenchyma. Spinal arteriolosclerosis was observed at all spinal levels (C7, T7, L4, S4) examined and was more severe posteriorly than anteriorly (posterior: 4.3, SD=0.72 versus anterior: 3.9, SD=0.74; t=14.58; P<0.001). Arteriolosclerosis was more severe in the spinal cord than in the brain (cord: 4.10, SD=0.70; brain: 3.5, SD=0.98; t=10.39; P<0.001). The severity of spinal arteriolosclerosis was associated with spinal white matter pallor (r=0.47; P<0.001). Spinal arteriolosclerosis accounted for ≈3% of the variation in parkinsonism in models controlling for age, sex, brain arteriolosclerosis, and cerebrovascular disease pathologies. Further models showed that the association of spinal arteriolosclerosis and parkinsonism was not mediated via spinal white matter pallor. CONCLUSIONS Although the regional distribution of microvascular pathologies varies within the central nervous system, spinal arteriolosclerosis is common and may contribute to the severity of spinal white matter pallor and parkinsonism in older adults.
Collapse
Affiliation(s)
- Aron S Buchman
- From the Rush Alzheimer's Disease Center (A.S.B., S.E.L., S.N., A.K., J.A.S., D.A.B.), Department of Neurological Sciences (A.S.B., S.E.L., J.A.S., D.A.B.), and Department of Pathology (Neuropathology) (S.N., J.A.S.), Rush University Medical Center, Chicago, IL; Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (V.G.J.M.V.); and Harvard Medical School, Boston, MA (V.G.J.M.V.).
| | - Sue E Leurgans
- From the Rush Alzheimer's Disease Center (A.S.B., S.E.L., S.N., A.K., J.A.S., D.A.B.), Department of Neurological Sciences (A.S.B., S.E.L., J.A.S., D.A.B.), and Department of Pathology (Neuropathology) (S.N., J.A.S.), Rush University Medical Center, Chicago, IL; Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (V.G.J.M.V.); and Harvard Medical School, Boston, MA (V.G.J.M.V.)
| | - Sukriti Nag
- From the Rush Alzheimer's Disease Center (A.S.B., S.E.L., S.N., A.K., J.A.S., D.A.B.), Department of Neurological Sciences (A.S.B., S.E.L., J.A.S., D.A.B.), and Department of Pathology (Neuropathology) (S.N., J.A.S.), Rush University Medical Center, Chicago, IL; Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (V.G.J.M.V.); and Harvard Medical School, Boston, MA (V.G.J.M.V.)
| | - Veronique G J M VanderHorst
- From the Rush Alzheimer's Disease Center (A.S.B., S.E.L., S.N., A.K., J.A.S., D.A.B.), Department of Neurological Sciences (A.S.B., S.E.L., J.A.S., D.A.B.), and Department of Pathology (Neuropathology) (S.N., J.A.S.), Rush University Medical Center, Chicago, IL; Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (V.G.J.M.V.); and Harvard Medical School, Boston, MA (V.G.J.M.V.)
| | - Alifiya Kapasi
- From the Rush Alzheimer's Disease Center (A.S.B., S.E.L., S.N., A.K., J.A.S., D.A.B.), Department of Neurological Sciences (A.S.B., S.E.L., J.A.S., D.A.B.), and Department of Pathology (Neuropathology) (S.N., J.A.S.), Rush University Medical Center, Chicago, IL; Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (V.G.J.M.V.); and Harvard Medical School, Boston, MA (V.G.J.M.V.)
| | - Julie A Schneider
- From the Rush Alzheimer's Disease Center (A.S.B., S.E.L., S.N., A.K., J.A.S., D.A.B.), Department of Neurological Sciences (A.S.B., S.E.L., J.A.S., D.A.B.), and Department of Pathology (Neuropathology) (S.N., J.A.S.), Rush University Medical Center, Chicago, IL; Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (V.G.J.M.V.); and Harvard Medical School, Boston, MA (V.G.J.M.V.)
| | - David A Bennett
- From the Rush Alzheimer's Disease Center (A.S.B., S.E.L., S.N., A.K., J.A.S., D.A.B.), Department of Neurological Sciences (A.S.B., S.E.L., J.A.S., D.A.B.), and Department of Pathology (Neuropathology) (S.N., J.A.S.), Rush University Medical Center, Chicago, IL; Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA (V.G.J.M.V.); and Harvard Medical School, Boston, MA (V.G.J.M.V.)
| |
Collapse
|
21
|
Lin H, Satizabal C, Xie Z, Yang Q, Huan T, Joehanes R, Wen C, Munson PJ, Beiser A, Levy D, Seshadri S. Whole blood gene expression and white matter Hyperintensities. Mol Neurodegener 2017; 12:67. [PMID: 28923099 PMCID: PMC5604498 DOI: 10.1186/s13024-017-0209-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 09/12/2017] [Indexed: 11/10/2022] Open
Abstract
Background White matter hyperintensities (WMH) are an important biomarker of cumulative vascular brain injury and have been associated with cognitive decline and an increased risk of dementia, stroke, depression, and gait impairments. The pathogenesis of white matter lesions however, remains uncertain. The characterization of gene expression profiles associated with WMH might help uncover molecular mechanisms underlying WMH. Methods We performed a transcriptome-wide association study of gene expression profiles with WMH in 3248 participants from the Framingham Heart Study using the Affymetrix Human Exon 1.0 ST Array. Results We identified 13 genes that were significantly associated with WMH (FDR < 0.05) after adjusting for age, sex and blood cell components. Many of these genes are involved in inflammation-related pathways. Conclusion Thirteen genes were significantly associated with WMH. Our study confirms the hypothesis that inflammation might be an important factor contributing to white matter lesions. Future work is needed to explore if these gene products might serve as potential therapeutic targets. Electronic supplementary material The online version of this article (10.1186/s13024-017-0209-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Honghuang Lin
- National Heart Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA. .,Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, 72 East Concord Street, B-616, Boston, MA, 02118, USA.
| | - Claudia Satizabal
- National Heart Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA.,Department of Neurology, Boston University School of Medicine, 72 East Concord Street, B-602, Boston, MA, 02118, USA
| | - Zhijun Xie
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Qiong Yang
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Tianxiao Huan
- National Heart Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA.,Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Roby Joehanes
- Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institute of Health, Bethesda, MD, USA.,Hebrew Senior Life, 1200 Centre Street Room #609, Boston, MA, 02131, USA
| | - Chengping Wen
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Peter J Munson
- Mathematical and Statistical Computing Laboratory, Center for Information Technology, National Institute of Health, Bethesda, MD, USA
| | - Alexa Beiser
- National Heart Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA.,Department of Neurology, Boston University School of Medicine, 72 East Concord Street, B-602, Boston, MA, 02118, USA.,Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Daniel Levy
- National Heart Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA.,Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Sudha Seshadri
- National Heart Lung and Blood Institute's and Boston University's Framingham Heart Study, Framingham, MA, USA. .,Department of Neurology, Boston University School of Medicine, 72 East Concord Street, B-602, Boston, MA, 02118, USA.
| |
Collapse
|
22
|
Starr JM. Giant steps forward. Age Ageing 2017. [PMID: 28633430 DOI: 10.1093/ageing/afx097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- John M Starr
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, EH8 9JZ, UK
| |
Collapse
|
23
|
Barzilay JI, Bůžková P, Fink HA, Cauley JA, Robbins JA, Garimella PS, Jalal DI, Mukamal KJ. Systemic markers of microvascular disease and bone mineral density in older adults : The cardiovascular health study. Osteoporos Int 2016; 27:3217-3225. [PMID: 27250972 DOI: 10.1007/s00198-016-3649-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Accepted: 05/23/2016] [Indexed: 10/21/2022]
Abstract
UNLABELLED Here we report that abnormal brain white matter and, to a lesser extent, albuminuria are associated with reduced bone mineral density in the hip, spine, and total body in men and women. These findings may explain the increased hip fracture risk reported in some studies in association with microvascular disorders. INTRODUCTION Markers of microvascular disease have been individually associated with increased risk of osteoporotic fractures in some studies. Here, we examine whether these markers are associated with reduced bone mineral density (BMD) individually and together. METHODS BMD testing using dual x-ray absorptiometry of the hip, lumbar spine, and total body was performed in 1473 participants from the Cardiovascular Health Study (mean age ~ 78 years): 1215 were assessed for urinary albumin-creatinine ratio, 944 for abnormal white matter disease (AWMD) by brain MRI, and 541 for retinal vascular disease with fundus photographs. Linear regression models were used to evaluate the cross-sectional association of each marker with BMD accounting for potentially confounding factors. RESULTS AWMD was associated with lower hip, spine, and total body BMD in women (β -3.08 to -4.53; p < 0.01 for all) and lower hip and total body BMD in men (β -2.90 to -4.24; p = 0.01-0.03). Albuminuria was associated with lower hip (β -3.37; p = .05) and total body (β -3.21; p = .02) BMD in men, but not in women. The associations of AWMD and albuminuria with BMD persisted with mutual adjustment and appeared to be additive to each other. Retinal vascular disease was not associated with BMD in men or women. CONCLUSION AWMD and, to a lesser extent, albuminuria were independently associated with lower BMD, suggesting that microvascular disease may play a role in the pathogenesis of reduced BMD. These findings need to be confirmed by longitudinal studies.
Collapse
Affiliation(s)
- J I Barzilay
- Kaiser Permanente of Georgia, Division of Endocrinology and the Division of Endocrinology, Emory University School of Medicine, Atlanta, GA, USA.
- Kaiser Permanente of Georgia, 3650 Steve Reynolds Blvd, Duluth, GA, 30096, USA.
| | - P Bůžková
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - H A Fink
- Geriatric Research Education and Clinical Center, VA Health Care System, Minneapolis, MN, USA
| | - J A Cauley
- Department of Epidemiology, Graduate School, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - J A Robbins
- Department of Medicine, University of California at Davis, Modesto, CA, USA
| | - P S Garimella
- Division of Nephrology, Tufts Medical Center, Boston, MA, USA
| | - D I Jalal
- Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Center, Aurora, CO, USA
| | - K J Mukamal
- Department of Medicine, Beth Israel Deaconess Medical Center, Brookline, MA, USA
| |
Collapse
|
24
|
Huang WQ, Ye HM, Li FF, Yi KH, Zhang Y, Cai LL, Lin HN, Lin Q, Tzeng CM. Analysis of genetic polymorphisms associated with leukoaraiosis in the southern Chinese population: A case-control study. Medicine (Baltimore) 2016; 95:e3857. [PMID: 27583843 PMCID: PMC5008527 DOI: 10.1097/md.0000000000003857] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Leukoaraiosis (LA) is a frequent neuroimaging finding commonly observed on brain MRIs of elderly people with prevalence ranging from 50% to 100%. Multiple susceptibility genes or genetic risk factors for LA have been identified in subjects of European descent. Here, we report the first replication study on several common and novel genetic variations in the Chinese population. In this study, a total of 244 subjects (201 LA patients and 43 controls) were enrolled according to our new and strict definition for LA. Subsequently, 6 genetic variants at 5 genes, rs3744028 in TRIM65, rs1055129 in TRIM47, rs1135889 in FBF1, rs1052053 in PMF1, and rs1801133 (C677T) and rs1801131(A1298C) in MTHFR, were selected for genotyping using polymerase chain reaction (PCR)-based pyrosequencing and restriction fragment length polymorphism (RFLP) together with capillary electrophoresis (CE) and agarose gel electrophoresis. Finally, Pearson's χ and multivariate logistic regression tests were used to examine the associations between the genotypes and LA. Among these candidate polymorphisms, except for rs1052053 and rs1801131, rs1135889 (P = 0.012) showed significant associations with LA in the dominant model, and the other 3 SNPs, rs3744028 (P = 0.043), rs1055129 (P = 0.038), and rs1801133 (P = 0.027), showed significant associations with LA in the recessive model. However, these differences no longer remained significant after adjusting for age, gender, hypertension, and diabetes mellitus and applying Bonferroni correction or Sidak correction for multiple testing. These results suggest that the above-mentioned genetic variants are not associated with LA risk. In summary, the study did not replicate the susceptibility of rs3744028, rs1055129, and rs1135889 at the Chr17q25 locus for LA nor did it find any other significant results for rs1052053, rs1801133, and rs1801131 in the Chinese population. It strongly indicated the ethnic differences in the genetics of LA. However, the associations of rs3744028 (TRIM65), rs1055129 (TRIM47), rs1135889 (FBF1), and rs1801133 (MTHFR) with LA before Bonferroni correction and Sidak correction for multiple testing are worth highlighting. Thus, we believe that a genome-wide association study and candidate gene association studies are needed to reassess the previous findings and screen novel risk genes for LA in China.
Collapse
Affiliation(s)
- Wen-Qing Huang
- Translational Medicine Research Center
- Key Laboratory for Cancer T-Cell Theranostics and Clinical Translation
| | - Hui-Ming Ye
- Translational Medicine Research Center
- Key Laboratory for Cancer T-Cell Theranostics and Clinical Translation
- Maternity and Child Health Hospital, Xiamen, Fujian, China
| | - Fang-Fang Li
- Translational Medicine Research Center
- Key Laboratory for Cancer T-Cell Theranostics and Clinical Translation
| | - Ke-Hui Yi
- Department of Neurology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian
| | - Ya Zhang
- Translational Medicine Research Center
- Key Laboratory for Cancer T-Cell Theranostics and Clinical Translation
| | - Liang-Liang Cai
- Translational Medicine Research Center
- Key Laboratory for Cancer T-Cell Theranostics and Clinical Translation
| | - Hui-Nuan Lin
- Translational Medicine Research Center
- Key Laboratory for Cancer T-Cell Theranostics and Clinical Translation
- INNOVA Cell: TDx Clinics and TRANSLA Health Group, China
| | - Qing Lin
- Translational Medicine Research Center
- Department of Neurology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian
- Correspondence: Chi-Meng Tzeng, Translational Medicine Research Center, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China (e-mail: ); Qing Lin, Department of Neurology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China (e-mail: )
| | - Chi-Meng Tzeng
- Translational Medicine Research Center
- Key Laboratory for Cancer T-Cell Theranostics and Clinical Translation
- INNOVA Cell: TDx Clinics and TRANSLA Health Group, China
- Correspondence: Chi-Meng Tzeng, Translational Medicine Research Center, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, China (e-mail: ); Qing Lin, Department of Neurology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China (e-mail: )
| |
Collapse
|
25
|
Neuropathological diagnosis of vascular cognitive impairment and vascular dementia with implications for Alzheimer's disease. Acta Neuropathol 2016; 131:659-85. [PMID: 27062261 PMCID: PMC4835512 DOI: 10.1007/s00401-016-1571-z] [Citation(s) in RCA: 260] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 03/23/2016] [Accepted: 03/24/2016] [Indexed: 12/16/2022]
Abstract
Vascular dementia (VaD) is recognised as a neurocognitive disorder, which is explained by numerous vascular causes in the general absence of other pathologies. The heterogeneity of cerebrovascular disease makes it challenging to elucidate the neuropathological substrates and mechanisms of VaD as well as vascular cognitive impairment (VCI). Consensus and accurate diagnosis of VaD relies on wide-ranging clinical, neuropsychometric and neuroimaging measures with subsequent pathological confirmation. Pathological diagnosis of suspected clinical VaD requires adequate postmortem brain sampling and rigorous assessment methods to identify important substrates. Factors that define the subtypes of VaD include the nature and extent of vascular pathologies, degree of involvement of extra and intracranial vessels and the anatomical location of tissue changes. Atherosclerotic and cardioembolic diseases appear the most common substrates of vascular brain injury or infarction. Small vessel disease characterised by arteriolosclerosis and lacunar infarcts also causes cortical and subcortical microinfarcts, which appear to be the most robust substrates of cognitive impairment. Diffuse WM changes with loss of myelin and axonal abnormalities are common to almost all subtypes of VaD. Medial temporal lobe and hippocampal atrophy accompanied by variable hippocampal sclerosis are also features of VaD as they are of Alzheimer’s disease. Recent observations suggest that there is a vascular basis for neuronal atrophy in both the temporal and frontal lobes in VaD that is entirely independent of any Alzheimer pathology. Further knowledge on specific neuronal and dendro-synaptic changes in key regions resulting in executive dysfunction and other cognitive deficits, which define VCI and VaD, needs to be gathered. Hereditary arteriopathies such as cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy or CADASIL have provided insights into the mechanisms of dementia associated with cerebral small vessel disease. Greater understanding of the neurochemical and molecular investigations is needed to better define microvascular disease and vascular substrates of dementia. The investigation of relevant animal models would be valuable in exploring the pathogenesis as well as prevention of the vascular causes of cognitive impairment.
Collapse
|
26
|
Sfera A, Osorio C, Inderias L, Cummings M. The Ticking of the Epigenetic Clock: Antipsychotic Drugs in Old Age. Front Endocrinol (Lausanne) 2016; 7:122. [PMID: 27630617 PMCID: PMC5005952 DOI: 10.3389/fendo.2016.00122] [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: 05/02/2016] [Accepted: 08/23/2016] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Exposed to antipsychotic drugs (APDs), older individuals with dementing illness are at risk of cerebrovascular adverse effects (CVAE), including sudden death. Transient microvascular dysfunctions are known to occur in younger persons exposed to APDs; however, they seldom progress to CVAE, suggesting that APDs alone are insufficient for engendering this untoward effect. It is, therefore, believed that a preexistent microvascular damage is necessary for CVAE to take place, but the exact nature of this lesion remains unclear. CNS small vessel disease (SVD) is a well-known age-related risk factor for strokes, dementia, and sudden death, which may constitute the initial CVAE-predisposing pathology. Therefore, we propose the two strikes CVAE paradigm, in which SVD represents the first strike, while exposure to APDs, the second. In this model, both strikes must be present for CVAE to take place, and the neuroimaging load of white matter hyperintensities may be directly proportional with the CVAE risk. To investigate this hypothesis at the molecular level, we focused on a seemingly unrelated phenomenon: both APDs and SVD were found protective against a similar repertoire of cancers and their spread to the brain (1-4). Since microRNA-29 has shown efficacy against the same malignancies and has been associated with small vessels pathology, we narrowed our search down to this miR, hypothesizing that the APDs mechanism of action includes miR-29 upregulation, which in turn facilitates the development of SVD. AIM To assess whether miR-29 can be utilized as a peripheral blood biomarker for SVD and CVAE risk. METHOD We conducted a search of experimentally verified miR-29 target genes utilizing the public domain tools miRanda, RNA22 and Weizemann Institute of Science miRNA Analysis. We identified in total 67 experimentally verified target genes for miR-29 family, 18 of which correlate with microvascular integrity and may be relevant for CVAE. CONCLUSION Upregulated microRNA-29 silences the expression of 18 genes connected with capillary stability, engendering a major vulnerability for SVD (first strike) which in turn increases the risk for CVAE after exposure to APDs (second strike).
Collapse
Affiliation(s)
- Adonis Sfera
- Psychiatry, Patton State Hospital, Patton, CA, USA
- *Correspondence: Adonis Sfera,
| | | | | | | |
Collapse
|
27
|
Lin Q, Huang WQ, Tzeng CM. Genetic associations of leukoaraiosis indicate pathophysiological mechanisms in white matter lesions etiology. Rev Neurosci 2015; 26:343-58. [PMID: 25781674 DOI: 10.1515/revneuro-2014-0082] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 12/18/2014] [Indexed: 11/15/2022]
Abstract
Leukoaraiosis (LA), also called white matter lesions (WMLs) and white matter hyperintensities (WMHs), is a frequent neuroimaging finding commonly seen on magnetic resonance imaging brain scans of elderly people with prevalence ranging from 50% to 100%. Although it remains asymptomatic, LA is not considered to be benign, and it is showed to be related to a host of poor clinical outcomes and increases the risk of disability, dementia, depression, stroke, and the overall morbidity and mortality. Pathologically, LA is characterized by loss of myelin and axons, patchy demyelination, and denudation of ependyma in regions of WMH. Age and hypertension are the most importantly established risk factors for LA. However, the precise pathogenic mechanisms remain unclear. Together with the previous findings, our recent genetic results strongly supported that LA is associated with immune response and neuroinflammation. Therefore, we confidently hypothesized that LA was not only a common neuroimaging phenomenon in the elderly but also an emerging neuroinflammatory disorder in the central nervous system. This article focusing on neuroimaging classification, genetics basis, and putative molecular mechanism introduced the basic knowledge and current status of LA and put forward some of our research ideas and results from our molecular genetics research, which may pave the way for deciphering the putative pathogenic mechanism, risk factor, epigenetic index, and its application in diagnostic agents or drug target for prevention and treatment. Thus, it could provide clinicians and researchers with a specific and modern overview of LA to enable the understanding of recent progress and future directions in this illness.
Collapse
|
28
|
Bailey EL, McBride MW, Beattie W, McClure JD, Graham D, Dominiczak AF, Sudlow CLM, Smith C, Wardlaw JM. Differential gene expression in multiple neurological, inflammatory and connective tissue pathways in a spontaneous model of human small vessel stroke. Neuropathol Appl Neurobiol 2015; 40:855-72. [PMID: 24417612 PMCID: PMC4260148 DOI: 10.1111/nan.12116] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 01/08/2014] [Indexed: 02/03/2023]
Abstract
AIMS Cerebral small vessel disease (SVD) causes a fifth of all strokes plus diffuse brain damage leading to cognitive decline, physical disabilities and dementia. The aetiology and pathogenesis of SVD are unknown, but largely attributed to hypertension or microatheroma. METHODS We used the spontaneously hypertensive stroke-prone rat (SHRSP), the closest spontaneous experimental model of human SVD, and age-matched control rats kept under identical, non-salt-loaded conditions, to perform a blinded analysis of mRNA microarray, qRT-PCR and pathway analysis in two brain regions (frontal and mid-coronal) commonly affected by SVD in the SHRSP at age five, 16 and 21 weeks. RESULTS We found gene expression abnormalities, with fold changes ranging from 2.5 to 59 for the 10 most differentially expressed genes, related to endothelial tight junctions (reduced), nitric oxide bioavailability (reduced), myelination (impaired), glial and microglial activity (increased), matrix proteins (impaired), vascular reactivity (impaired) and albumin (reduced), consistent with protein expression defects in the same rats. All were present at age 5 weeks thus predating blood pressure elevation. 'Neurological' and 'inflammatory' pathways were more affected than 'vascular' functional pathways. CONCLUSIONS This set of defects, although individually modest, when acting in combination could explain the SHRSP's susceptibility to microvascular and brain injury, compared with control rats. Similar combined, individually modest, but multiple neurovascular unit defects, could explain susceptibility to spontaneous human SVD.
Collapse
Affiliation(s)
- Emma L Bailey
- Centre for Clinical Brain Sciences, University of Edinburgh, Western General Hospital, Edinburgh; Department of Bioengineering, Imperial College London, London
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Wharton SB, Simpson JE, Brayne C, Ince PG. Age-associated white matter lesions: the MRC Cognitive Function and Ageing Study. Brain Pathol 2015; 25:35-43. [PMID: 25521175 DOI: 10.1111/bpa.12219] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 10/08/2014] [Indexed: 12/11/2022] Open
Abstract
Cerebral white matter lesions (WML) are common in the aging brain and are associated with dementia and depression. They are associated with vascular risk factors and small vessel disease, suggesting an ischemic origin, but recent pathology studies suggest a more complex pathogenesis. Studies using samples from the population-representative Medical Research Council Cognitive Function and Ageing Study neuropathology cohort used post-mortem magnetic resonance imaging to identify WML for further study. Expression of hypoxia-related molecules and other injury and protective cellular pathways in candidate immunohistochemical and gene expression microarray studies support a role for hypoxia/ischemia. However, these approaches also suggest that immune activation, blood-brain barrier dysfunction, altered cell metabolic pathways and glial cell injury contribute to pathogenesis. These abnormalities are not confined to WML, but are also found in apparently normal white matter in brains with lesions, suggesting a field effect of white matter abnormality within which lesions arise. WML are an active pathology with a complex pathogenesis that may potentially offer a number of primary and secondary intervention targets.
Collapse
Affiliation(s)
- Stephen B Wharton
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | | | | | | |
Collapse
|
30
|
Mok V, Kim JS. Prevention and Management of Cerebral Small Vessel Disease. J Stroke 2015; 17:111-22. [PMID: 26060798 PMCID: PMC4460330 DOI: 10.5853/jos.2015.17.2.111] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Revised: 04/07/2015] [Accepted: 04/07/2015] [Indexed: 01/22/2023] Open
Abstract
Lacunar infarcts/lacunes, white matter hyperintensities (WMH), and cerebral microbleeds (CMBs) are considered various manifestations of cerebral small vessel disease (SVD). Since the exact mechanisms of these manifestations differ, their associated risk factors differ. High blood pressure is the most consistent risk factor for all of these manifestations. However, a "J curve" phenomenon in terms of blood pressure probably exists for WMH. The association between cholesterol levels and lacunar infarcts/lacunes or WMH was less consistent and sometimes conflicting; a low cholesterol level probably increases the risk of CMBs. Homocysteinemia appears to be associated with WMH. It is noteworthy that the risk factors profile may also differ between different lacunar patterns and CMBs located at different parts of the brain. Thrombolysis, antihypertensives, and statins are used to treat patients with symptomatic lacunar infarction, just as in those with other stroke subtypes. However, it should be remembered that bleeding risks increase in patients with extensive WMH and CMBs after thrombolysis therapy. According to the Secondary Prevention of Small Subcortical Strokes trial results, a blood pressure reduction to <130 mmHg is recommended in patients with symptomatic lacunar infarction. However, an excessive blood pressure decrease may induce cognitive decline in older patients with extensive WMH. Dual antiplatelet therapy (aspirin plus clopidogrel) should be avoided because of the excessive risk of intracerebral hemorrhage. Although no particular antiplatelet is recommended, drugs such as cilostazol or triflusal may have advantages for patients with SVD since they are associated with less frequent bleeding complications than aspirin.
Collapse
Affiliation(s)
- Vincent Mok
- Division of Neurology, Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Jong S Kim
- Stroke Center and Department of Neurology, University of Ulsan, Asan Medical Center, Seoul, Korea
| |
Collapse
|
31
|
Al-Mashhadi S, Simpson JE, Heath PR, Dickman M, Forster G, Matthews FE, Brayne C, Ince PG, Wharton SB. Oxidative Glial Cell Damage Associated with White Matter Lesions in the Aging Human Brain. Brain Pathol 2014; 25:565-74. [PMID: 25311358 PMCID: PMC4861214 DOI: 10.1111/bpa.12216] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 10/07/2014] [Indexed: 01/04/2023] Open
Abstract
White matter lesions (WML) are common in brain aging and are associated with dementia. We aimed to investigate whether oxidative DNA damage and occur in WML and in apparently normal white matter in cases with lesions. Tissue from WML and control white matter from brains with lesions (controls lesional) and without lesions (controls non‐lesional) were obtained, using post‐mortem magnetic resonance imaging‐guided sampling, from the Medical Research Council Cognitive Function and Ageing Study. Oxidative damage was assessed by immunohistochemistry to 8‐hydroxy‐2′‐deoxoguanosine (8‐OHdG) and Western blotting for malondialdehyde. DNA response was assessed by phosphorylated histone H2AX (γH2AX), p53, senescence markers and by quantitative Reverse transcription polymerase chain reaction (RT‐PCR) panel for candidate DNA damage‐associated genes. 8‐OHdG was expressed in glia and endothelium, with increased expression in both WML and controls lesional compared with controls non‐lesional (P < 0.001). γH2Ax showed a similar, although attenuated difference among groups (P = 0.03). Expression of senescence‐associated β‐galactosidase and p16 suggested induction of senescence mechanisms in glia. Oxidative DNA damage and a DNA damage response are features of WML pathogenesis and suggest candidate mechanisms for glial dysfunction. Their expression in apparently normal white matter in cases with WML suggests that white matter dysfunction is not restricted to lesions. The role of this field‐effect lesion pathogenesis and cognitive impairment are areas to be defined.
Collapse
Affiliation(s)
- Sufana Al-Mashhadi
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK.,King Fahad Medical City, Riyadh, Saudi Arabia
| | - Julie E Simpson
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Paul R Heath
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Mark Dickman
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK
| | - Gillian Forster
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Fiona E Matthews
- Medical Research Council Biostatistics Unit, University of Cambridge, Cambridge, UK
| | - Carol Brayne
- Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Paul G Ince
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Stephen B Wharton
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | | |
Collapse
|
32
|
Craggs LJ, Yamamoto Y, Deramecourt V, Kalaria RN. Microvascular pathology and morphometrics of sporadic and hereditary small vessel diseases of the brain. Brain Pathol 2014; 24:495-509. [PMID: 25323665 PMCID: PMC4228759 DOI: 10.1111/bpa.12177] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 06/27/2014] [Indexed: 12/12/2022] Open
Abstract
Small vessel diseases (SVDs) of the brain are likely to become increasingly common in tandem with the rise in the aging population. In recent years, neuroimaging and pathological studies have informed on the pathogenesis of sporadic SVD and several single gene (monogenic) disorders predisposing to subcortical strokes and diffuse white matter disease. However, one of the limitations toward studying SVD lies in the lack of consistent assessment criteria and lesion burden for both clinical and pathological measures. Arteriolosclerosis and diffuse white matter changes are the hallmark features of both sporadic and hereditary SVDs. The pathogenesis of the arteriopathy is the key to understanding the differential progression of disease in various SVDs. Remarkably, quantification of microvascular abnormalities in sporadic and hereditary SVDs has shown that qualitatively the processes involved in arteriolar degeneration are largely similar in sporadic SVD compared with hereditary disorders such as cerebral autosomal arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL). Important significant regional differences in lesion location within the brain may enable one to distinguish SVDs, where frontal lobe involvement appears consistently with almost every SVD, but others bear specific pathologies in other lobes, such as the temporal pole in CADASIL and the pons in pontine autosomal dominant microangiopathy and leukoencephalopathy or PADMAL. Additionally, degenerative changes in the vascular smooth muscle cells, the cerebral endothelium and the basal lamina are often rapid and more aggressive in genetic disorders. Further quantification of other microvascular elements and even neuronal cells is needed to fully characterize SVD pathogenesis and to differentiate the usefulness of vascular interventions and treatments on the resulting pathology.
Collapse
Affiliation(s)
| | - Yumi Yamamoto
- Department of Regenerative Medicine and Tissue EngineeringNational Cerebral and Cardiovascular CenterNational Cerebral and Cardiovascular Center Research InstituteOsakaJapan
| | | | - Raj N. Kalaria
- Institute for Ageing and HealthNewcastle UniversityNewcastle upon TyneUK
| |
Collapse
|
33
|
Rincon F, Wright CB. Current pathophysiological concepts in cerebral small vessel disease. Front Aging Neurosci 2014; 6:24. [PMID: 24715862 PMCID: PMC3970024 DOI: 10.3389/fnagi.2014.00024] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 02/12/2014] [Indexed: 11/13/2022] Open
Abstract
The association between cerebral small vessel disease (SVD) - in the form of white matter lesions, infarctions, and hemorrhages - with vascular cognitive impairment (VCI), has mostly been deduced from observational studies. Pathological conditions affecting the small vessels of the brain and leading to SVD have suggested plausible molecular mechanisms involved in vascular damage and their impact on brain function. However, much still needs to be clarified in understanding the pathophysiology of VCI, the role of neurodegenerative processes such as Alzheimer's disease, and the impact of aging itself. In addition, both genetic predispositions and environmental exposures may potentiate the development of SVD and interact with normal aging to impact cognitive function and require further study. Advances in technology, in the analysis of genetic and epigenetic data, neuroimaging such as magnetic resonance imaging, and new biomarkers will help to clarify the complex factors leading to SVD and the expression of VCI.
Collapse
Affiliation(s)
- Fred Rincon
- Department of Neurology, Thomas Jefferson University Philadelphia, PA, USA
| | - Clinton B Wright
- Department of Neurosurgery, Thomas Jefferson University Philadelphia, PA, USA ; Evelyn F. McKnight Brain Institute, Department of Neurology, University of Miami Miami, FL, USA ; Department of Epidemiology and Public Health, University of Miami Miami, FL, USA ; Neuroscience Program, University of Miami Miami, FL, USA
| |
Collapse
|
34
|
Datta A, Qian J, Chong R, Kalaria RN, Francis P, Lai MKP, Chen CP, Sze SK. Novel pathophysiological markers are revealed by iTRAQ-based quantitative clinical proteomics approach in vascular dementia. J Proteomics 2014; 99:54-67. [PMID: 24448401 PMCID: PMC4024194 DOI: 10.1016/j.jprot.2014.01.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2013] [Revised: 12/10/2013] [Accepted: 01/08/2014] [Indexed: 12/14/2022]
Abstract
UNLABELLED Vascular dementia (VaD) is a leading cause of dementia in the elderly together with Alzheimer's disease with limited treatment options. Poor understanding of the pathophysiology underlying VaD is hindering the development of new therapies. Hence, to unravel its underlying molecular pathology, an iTRAQ-2D-LC-MS/MS strategy was used for quantitative analysis of pooled lysates from Brodmann area 21 of pathologically confirmed cases of VaD and matched non-neurological controls. A total of 144 differentially expressed proteins out of 2281 confidently identified proteins (false discovery rate=0.3%) were shortlisted for bioinformatics analysis. Western blot analysis of selected proteins using samples from individual patients (n=10 per group) showed statistically significant increases in the abundance of SOD1 and NCAM and reduced ATP5A in VaD. This suggested a state of hypometabolism and vascular insufficiency along with an inflammatory condition during VaD. Elevation of SOD1 and increasing trend for iron-storage proteins (FTL, FTH1) may be indicative of an oxidative imbalance that is accompanied by an aberrant iron metabolism. The synaptic proteins did not exhibit a generalized decrease in abundance (e.g. syntaxin) in the VaD subjects. This reported proteome offers a reference data set for future basic or translational studies on VaD. BIOLOGICAL SIGNIFICANCE Our study is the first quantitative clinical proteomic study where iTRAQ-2D-LC-MS/MS strategy has been used to identify the differential proteome in the VaD cortex by comparing VaD and matched control subjects. We generate testable hypothesis about the involvement of various proteins in the vascular and parenchymal events during the evolution of VaD that finally leads to malfunction and demise of brain cells. This study also establishes quantitative proteomics as a complementary approach and viable alternative to existing neurochemical, electron microscopic and neuroimaging techniques that are traditionally being used to understand the molecular pathology of VaD. Our study could inspire fellow researchers to initiate similar retrospective studies targeting various ethnicities, age-groups or sub-types of VaD using brain samples available from brain banks across the world. Meta-analysis of these studies in the future may be able to shortlist candidate proteins or pathways for rationale exploration of therapeutic targets or biomarkers for VaD.
Collapse
Affiliation(s)
- Arnab Datta
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jingru Qian
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Ruifen Chong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Raj N Kalaria
- Institute for Ageing Health, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne, UK
| | - Paul Francis
- Wolfson Centre for Age-related Diseases, King's College London, London, UK
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Memory, Aging and Cognition Centre, National University Health System, Singapore
| | - Christopher P Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Memory, Aging and Cognition Centre, National University Health System, Singapore.
| | - Siu Kwan Sze
- School of Biological Sciences, Nanyang Technological University, Singapore.
| |
Collapse
|
35
|
Deramecourt V. Vascular neuropathology and cognitive decline. Rev Neurol (Paris) 2013; 169:765-71. [DOI: 10.1016/j.neurol.2013.07.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 07/02/2013] [Accepted: 07/09/2013] [Indexed: 10/26/2022]
|
36
|
Adib-Samii P, Rost N, Traylor M, Devan W, Biffi A, Lanfranconi S, Fitzpatrick K, Bevan S, Kanakis A, Valant V, Gschwendtner A, Malik R, Richie A, Gamble D, Segal H, Parati EA, Ciusani E, Holliday EG, Maguire J, Wardlaw J, Worrall B, Bis J, Wiggins KL, Longstreth W, Kittner SJ, Cheng YC, Mosley T, Falcone GJ, Furie KL, Leiva-Salinas C, Lau BC, Saleem Khan M, Sharma P, Fornage M, Mitchell BD, Psaty BM, Sudlow C, Levi C, Boncoraglio GB, Rothwell PM, Meschia J, Dichgans M, Rosand J, Markus HS. 17q25 Locus is associated with white matter hyperintensity volume in ischemic stroke, but not with lacunar stroke status. Stroke 2013; 44:1609-15. [PMID: 23674528 DOI: 10.1161/strokeaha.113.679936] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND AND PURPOSE Recently, a novel locus at 17q25 was associated with white matter hyperintensities (WMH) on MRI in stroke-free individuals. We aimed to replicate the association with WMH volume (WMHV) in patients with ischemic stroke. If the association acts by promoting a small vessel arteriopathy, it might be expected to also associate with lacunar stroke. METHODS We quantified WMH on MRI in the stroke-free hemisphere of 2588 ischemic stroke cases. Association between WMHV and 6 single-nucleotide polymorphisms at chromosome 17q25 was assessed by linear regression. These single-nucleotide polymorphisms were also investigated for association with lacunar stroke in 1854 cases and 51 939 stroke-free controls from METASTROKE. Meta-analyses with previous reports and a genetic risk score approach were applied to identify other novel WMHV risk variants and uncover shared genetic contributions to WMHV in community participants without stroke and ischemic stroke. RESULTS Single-nucleotide polymorphisms at 17q25 were associated with WMHV in ischemic stroke, the most significant being rs9894383 (P=0.0006). In contrast, there was no association between any single-nucleotide polymorphism and lacunar stroke. A genetic risk score analysis revealed further genetic components to WMHV shared between community participants without stroke and ischemic stroke. CONCLUSIONS This study provides support for an association between the 17q25 locus and WMH. In contrast, it is not associated with lacunar stroke, suggesting that the association does not act by promoting small-vessel arteriopathy or the same arteriopathy responsible for lacunar infarction.
Collapse
Affiliation(s)
- Poneh Adib-Samii
- Stroke and Dementia Research Centre, St George’s University of London, London, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW Alzheimer's disease has long been primarily considered a disease of gray matter. However, convergent evidence has suggested that white matter abnormalities are also important components of Alzheimer's disease. We undertook a review of the recent findings of Alzheimer's disease related white matter aberrations identified in patients with Alzheimer's disease and using in-vitro and in-vivo models, and discuss the potential causes of white matter damage in Alzheimer's disease. In doing so, we aim to provide a renewed insight into white matter changes in Alzheimer's disease and related dementias. RECENT FINDINGS Neuroimaging studies have found that patients with preclinical Alzheimer's disease have widespread white matter abnormalities at a stage similar to those reported in Alzheimer's disease, whereas gray matter structures were relatively intact. In addition, demyelination of the white matter is reported to occur prior to the presence of amyloid-β plaques and neurofibrillary tangles in the presymptomatic stages of Alzheimer's disease. Furthermore, in a mouse model of Alzheimer's disease, axonal disease due to impaired axonal transport was shown to precede and drive downstream production and aggregation of amyloid β peptides. SUMMARY White matter abnormalities not only represent an early neuropathological event in Alzheimer's disease but may also play an important role in the pathogenesis and diagnosis of Alzheimer's disease.
Collapse
|
38
|
Li J, Wu RG, Meng FY, Wang Z, Wang CM, Wang YY, Zhang ZJ. Synergism and rules from combination of Baicalin, Jasminoidin and Desoxycholic acid in refined Qing Kai Ling for treat ischemic stroke mice model. PLoS One 2012; 7:e45811. [PMID: 23049867 PMCID: PMC3458908 DOI: 10.1371/journal.pone.0045811] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 08/22/2012] [Indexed: 12/31/2022] Open
Abstract
Refined Qing-Kai-Ling (QKL), a modified Chinese medicine, consists of three main ingredients (Baicalin, Jasminoidin and Desoxycholic acid), plays a synergistic effect on the treatment of the acute stage of ischemic stroke. However, the rules of the combination and synergism are still unknown. Based on the ischemic stroke mice model, all different kinds of combination of Baicalin, Jasminoidin, and Desoxycholic acid were investigated by the methods of neurological examination, microarray, and genomics analysis. As a result, it confirmed that the combination of three drugs offered a better therapeutical effect on ischemic stroke than monotherapy of each drug. Additionally, we used Ingenuity pathway Analysis (IPA) and principal component analysis (PCA) to extract the dominant information of expression changes in 373 ischemia-related genes. The results suggested that 5 principal components (PC1-5) could account for more than 95% energy in the gene data. Moreover, 3 clusters (PC1, PC2+PC5, and PC3+PC4) were addressed with cluster analysis. Furthermore, we matched PCs on the drug-target networks, the findings demonstrated that Baicalin related with PC1 that played the leading role in the combination; Jasminoidin related with PC2+PC5 that played a compensatory role; while Desoxycholic acid had the least performance alone which could relate with PC3+PC4 that played a compatible role. These manifestations were accorded with the principle of herbal formulae of Traditional Chinese Medicine (TCM), emperor-minister-adjuvant-courier. In conclusion, we firstly provided scientific evidence to the classic theory of TCM formulae, an initiating holistic viewpoint of combination therapy of TCM. This study also illustrated that PCA might be an applicable method to analyze the complicated data of drug combination.
Collapse
Affiliation(s)
- Jian Li
- School of Basic Medical Science, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Run-guo Wu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Fan-yun Meng
- School of Resources Science & Technology, Beijing Normal University, Beijing, China
| | - Zhong Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Traditional Chinese Medicine, Beijing, China
| | - Chang-ming Wang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Yong-yan Wang
- Institute of Basic Research in Clinical Medicine, China Academy of Traditional Chinese Medicine, Beijing, China
| | - Zhan-jun Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
- * E-mail:
| |
Collapse
|
39
|
Kalaria RN. Cerebrovascular disease and mechanisms of cognitive impairment: evidence from clinicopathological studies in humans. Stroke 2012; 43:2526-34. [PMID: 22879100 DOI: 10.1161/strokeaha.112.655803] [Citation(s) in RCA: 151] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Raj N Kalaria
- Institute for Ageing and Health, Campus for Ageing & Vitality, Newcastle upon Tyne, NE4 5PL, UK.
| |
Collapse
|
40
|
Abstract
The augmentation index (AIx) is a measure of pulse wave reflection from peripheral muscular conduit arteries, and is assumed to increase with stiffened arteries. A white matter lesion (WML) is generally regarded as a chronic ischemic lesion, which is associated with cerebral small-vessel arteriosclerosis. The aim of this study is to elucidate the effect of pulse wave reflection on the brain. Carotid AIx was measured in a total of 205 neurologically normal adults. The correlations between AIx and WML were investigated. Two categories of WML were evaluated, periventricular hyperintensity (PVH) and deep and subcortical white matter hyperintensity (DSWMH). On univariate analysis, AIx was significantly correlated with the grades of PVH and DSWMH (P=0.0001, respectively). On multivariate analysis, AIx was correlated with PVH and DSWMH, independent of other risk factors such as age or blood pressure. Pulse wave reflection from small vessels may influence the genesis of WML.
Collapse
|
41
|
Moran C, Phan TG, Srikanth VK. Cerebral small vessel disease: a review of clinical, radiological, and histopathological phenotypes. Int J Stroke 2011; 7:36-46. [PMID: 22111922 DOI: 10.1111/j.1747-4949.2011.00725.x] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cerebral small vessel disease is difficult to directly visualize in vivo. Therefore, we rely on radiological phenotypes as surrogate markers of disease. The principal phenotypes of clinical interest are small, deep brain infarcts, cerebral white matter lesions, deep brain haemorrhages, and cerebral microbleeds. The causes or mechanisms underlying these phenotypes are understood in varying degrees of detail. This review aims to summarize recent knowledge regarding these phenotypes and place it in context with classical clinicopathological observations to provide mechanistic, clinical, and therapeutic insights into small vessel disease.
Collapse
Affiliation(s)
- Chris Moran
- Department of Medicine, Southern Clinical School, Monash University, Melbourne, Vic., Australia
| | | | | |
Collapse
|
42
|
Back SA, Kroenke CD, Sherman LS, Lawrence G, Gong X, Taber EN, Sonnen JA, Larson EB, Montine TJ. White matter lesions defined by diffusion tensor imaging in older adults. Ann Neurol 2011; 70:465-76. [PMID: 21905080 DOI: 10.1002/ana.22484] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECTIVE The cellular and molecular mechanisms underlying magnetic resonance imaging-defined white matter (WM) changes associated with age-related cognitive decline remain poorly defined. We tested the hypothesis that WM lesions in older adults, defined by diffusion tensor imaging (DTI), arise in the setting of vascular brain injury (VBI) and are characterized by increased free radical injury and aberrant oligodendrocyte lineage (OL) cell response to injury. METHODS We undertook a multimodal analysis of prefrontal cortex (PFC) WM from 25 autopsies derived from a population-based cohort where VBI and Alzheimer disease (AD) frequently coincide. Ex vivo high field strength DTI measurements of fractional anisotropy (FA), apparent diffusion coefficient, and axial and radial (D(⊥) ) diffusivity were measured at high magnetic field strength (11.7T) and analyzed relative to quantitative in vivo biomarkers of free radical injury, an OL-specific marker Olig2, and histologic evaluation of hyaluronan (HA), an inhibitor of OL maturation. RESULTS Coincident AD and VBI showed significant association with lower FA and a robust relationship between decreasing FA and increasing D(⊥) . Free radical injury to docosahexaenoate and adrenate in PFC WM was significantly elevated in cases with VBI independent of AD, and was inversely correlated with FA. Similarly, increased density of Olig2-immunoreactive cells in PFC WM was significantly associated with VBI independent of AD and colocalized with regions enriched in HA. INTERPRETATION DTI-defined PFC WM lesions in older individuals are characterized by free radical injury to myelin and neuroaxonal elements that coincides with pronounced expansion of the pool of OL cells in HA-rich regions.
Collapse
Affiliation(s)
- Stephen A Back
- Department of Pediatrics, Oregon Health and Science University, Portland, OR, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Brain Iron Dysregulation and the Risk of Ageing White Matter Lesions. Neuromolecular Med 2011; 13:289-99. [DOI: 10.1007/s12017-011-8161-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2011] [Accepted: 09/23/2011] [Indexed: 11/26/2022]
|
44
|
Gorelick PB, Scuteri A, Black SE, Decarli C, Greenberg SM, Iadecola C, Launer LJ, Laurent S, Lopez OL, Nyenhuis D, Petersen RC, Schneider JA, Tzourio C, Arnett DK, Bennett DA, Chui HC, Higashida RT, Lindquist R, Nilsson PM, Roman GC, Sellke FW, Seshadri S. Vascular contributions to cognitive impairment and dementia: a statement for healthcare professionals from the american heart association/american stroke association. Stroke 2011; 42:2672-713. [PMID: 21778438 PMCID: PMC3778669 DOI: 10.1161/str.0b013e3182299496] [Citation(s) in RCA: 2575] [Impact Index Per Article: 198.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND PURPOSE This scientific statement provides an overview of the evidence on vascular contributions to cognitive impairment and dementia. Vascular contributions to cognitive impairment and dementia of later life are common. Definitions of vascular cognitive impairment (VCI), neuropathology, basic science and pathophysiological aspects, role of neuroimaging and vascular and other associated risk factors, and potential opportunities for prevention and treatment are reviewed. This statement serves as an overall guide for practitioners to gain a better understanding of VCI and dementia, prevention, and treatment. METHODS Writing group members were nominated by the writing group co-chairs on the basis of their previous work in relevant topic areas and were approved by the American Heart Association Stroke Council Scientific Statement Oversight Committee, the Council on Epidemiology and Prevention, and the Manuscript Oversight Committee. The writing group used systematic literature reviews (primarily covering publications from 1990 to May 1, 2010), previously published guidelines, personal files, and expert opinion to summarize existing evidence, indicate gaps in current knowledge, and, when appropriate, formulate recommendations using standard American Heart Association criteria. All members of the writing group had the opportunity to comment on the recommendations and approved the final version of this document. After peer review by the American Heart Association, as well as review by the Stroke Council leadership, Council on Epidemiology and Prevention Council, and Scientific Statements Oversight Committee, the statement was approved by the American Heart Association Science Advisory and Coordinating Committee. RESULTS The construct of VCI has been introduced to capture the entire spectrum of cognitive disorders associated with all forms of cerebral vascular brain injury-not solely stroke-ranging from mild cognitive impairment through fully developed dementia. Dysfunction of the neurovascular unit and mechanisms regulating cerebral blood flow are likely to be important components of the pathophysiological processes underlying VCI. Cerebral amyloid angiopathy is emerging as an important marker of risk for Alzheimer disease, microinfarction, microhemorrhage and macrohemorrhage of the brain, and VCI. The neuropathology of cognitive impairment in later life is often a mixture of Alzheimer disease and microvascular brain damage, which may overlap and synergize to heighten the risk of cognitive impairment. In this regard, magnetic resonance imaging and other neuroimaging techniques play an important role in the definition and detection of VCI and provide evidence that subcortical forms of VCI with white matter hyperintensities and small deep infarcts are common. In many cases, risk markers for VCI are the same as traditional risk factors for stroke. These risks may include but are not limited to atrial fibrillation, hypertension, diabetes mellitus, and hypercholesterolemia. Furthermore, these same vascular risk factors may be risk markers for Alzheimer disease. Carotid intimal-medial thickness and arterial stiffness are emerging as markers of arterial aging and may serve as risk markers for VCI. Currently, no specific treatments for VCI have been approved by the US Food and Drug Administration. However, detection and control of the traditional risk factors for stroke and cardiovascular disease may be effective in the prevention of VCI, even in older people. CONCLUSIONS Vascular contributions to cognitive impairment and dementia are important. Understanding of VCI has evolved substantially in recent years, based on preclinical, neuropathologic, neuroimaging, physiological, and epidemiological studies. Transdisciplinary, translational, and transactional approaches are recommended to further our understanding of this entity and to better characterize its neuropsychological profile. There is a need for prospective, quantitative, clinical-pathological-neuroimaging studies to improve knowledge of the pathological basis of neuroimaging change and the complex interplay between vascular and Alzheimer disease pathologies in the evolution of clinical VCI and Alzheimer disease. Long-term vascular risk marker interventional studies beginning as early as midlife may be required to prevent or postpone the onset of VCI and Alzheimer disease. Studies of intensive reduction of vascular risk factors in high-risk groups are another important avenue of research.
Collapse
|
45
|
Xiong YY, Mok V. Age-related white matter changes. J Aging Res 2011; 2011:617927. [PMID: 21876810 PMCID: PMC3163144 DOI: 10.4061/2011/617927] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Revised: 06/28/2011] [Accepted: 06/28/2011] [Indexed: 11/20/2022] Open
Abstract
Age-related white matter changes (WMC) are considered manifestation of arteriolosclerotic small vessel disease and are related to age and vascular risk factors. Most recent studies have shown that WMC are associated with a host of poor outcomes, including cognitive impairment, dementia, urinary incontinence, gait disturbances, depression, and increased risk of stroke and death. Although the clinical relevance of WMC has been extensively studied, to date, only very few clinical trials have evaluated potential symptomatic or preventive treatments for WMC. In this paper, we reviewed the current understanding in the pathophysiology, epidemiology, clinical importance, chemical biomarkers, and treatments of age-related WMC.
Collapse
Affiliation(s)
- Yun Yun Xiong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Shatin 999077, Hong Kong
| | | |
Collapse
|
46
|
Heterogeneity in age-related white matter changes. Acta Neuropathol 2011; 122:171-85. [PMID: 21706175 DOI: 10.1007/s00401-011-0851-x] [Citation(s) in RCA: 225] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Revised: 06/15/2011] [Accepted: 06/16/2011] [Indexed: 10/18/2022]
Abstract
White matter changes occur endemically in routine magnetic resonance imaging (MRI) scans of elderly persons. MRI appearance and histopathological correlates of white matter changes are heterogeneous. Smooth periventricular hyperintensities, including caps around the ventricular horns, periventricular lining and halos are likely to be of non-vascular origin. They relate to a disruption of the ependymal lining with subependymal widening of the extracellular space and have to be differentiated from subcortical and deep white matter abnormalities. For the latter a distinction needs to be made between punctate, early confluent and confluent types. Although punctate white matter lesions often represent widened perivascular spaces without substantial ischemic tissue damage, early confluent and confluent lesions correspond to incomplete ischemic destruction. Punctate abnormalities on MRI show a low tendency for progression, while early confluent and confluent changes progress rapidly. The causative and modifying pathways involved in the occurrence of sporadic age-related white matter changes are still incompletely understood, but recent microarray and genome-wide association approaches increased the notion of pathways that might be considered as targets for therapeutic intervention. The majority of differentially regulated transcripts in white matter lesions encode genes associated with immune function, cell cycle, proteolysis, and ion transport. Genome-wide association studies identified six SNPs mapping to a locus on chromosome 17q25 to be related to white matter lesion load in the general population. We also report first and preliminary data that demonstrate apolipoprotein E (ApoE) immunoreactivity in white matter lesions and support epidemiological findings indicating that ApoE is another factor possibly related to white matter lesion occurrence. Further insights come from modern MRI techniques, such as diffusion tensor and magnetization transfer imaging, as they provide tools for the characterization of normal-appearing brain tissue beyond what can be expected from standard MRI scans. There is a need for additional pre- and postmortem studies in humans, including these new imaging techniques.
Collapse
|
47
|
Fornage M, Debette S, Bis JC, Schmidt H, Ikram MA, Dufouil C, Sigurdsson S, Lumley T, DeStefano AL, Fazekas F, Vrooman HA, Shibata DK, Maillard P, Zijdenbos A, Smith AV, Gudnason H, de Boer R, Cushman M, Mazoyer B, Heiss G, Vernooij MW, Enzinger C, Glazer NL, Beiser A, Knopman DS, Cavalieri M, Niessen WJ, Harris TB, Petrovic K, Lopez OL, Au R, Lambert JC, Hofman A, Gottesman RF, Garcia M, Heckbert SR, Atwood LD, Catellier DJ, Uitterlinden AG, Yang Q, Smith NL, Aspelund T, Romero JR, Rice K, Taylor KD, Nalls MA, Rotter JI, Sharrett R, van Duijn CM, Amouyel P, Wolf PA, Gudnason V, van der Lugt A, Boerwinkle E, Psaty BM, Seshadri S, Tzourio C, Breteler MMB, Mosley TH, Schmidt R, Longstreth WT, DeCarli C, Launer LJ. Genome-wide association studies of cerebral white matter lesion burden: the CHARGE consortium. Ann Neurol 2011; 69:928-39. [PMID: 21681796 PMCID: PMC3122147 DOI: 10.1002/ana.22403] [Citation(s) in RCA: 151] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE White matter hyperintensities (WMHs) detectable by magnetic resonance imaging are part of the spectrum of vascular injury associated with aging of the brain and are thought to reflect ischemic damage to the small deep cerebral vessels. WMHs are associated with an increased risk of cognitive and motor dysfunction, dementia, depression, and stroke. Despite a significant heritability, few genetic loci influencing WMH burden have been identified. METHODS We performed a meta-analysis of genome-wide association studies (GWASs) for WMH burden in 9,361 stroke-free individuals of European descent from 7 community-based cohorts. Significant findings were tested for replication in 3,024 individuals from 2 additional cohorts. RESULTS We identified 6 novel risk-associated single nucleotide polymorphisms (SNPs) in 1 locus on chromosome 17q25 encompassing 6 known genes including WBP2, TRIM65, TRIM47, MRPL38, FBF1, and ACOX1. The most significant association was for rs3744028 (p(discovery) = 4.0 × 10(-9) ; p(replication) = 1.3 × 10(-7) ; p(combined) = 4.0 × 10(-15) ). Other SNPs in this region also reaching genome-wide significance were rs9894383 (p = 5.3 × 10(-9) ), rs11869977 (p = 5.7 × 10(-9) ), rs936393 (p = 6.8 × 10(-9) ), rs3744017 (p = 7.3 × 10(-9) ), and rs1055129 (p = 4.1 × 10(-8) ). Variant alleles at these loci conferred a small increase in WMH burden (4-8% of the overall mean WMH burden in the sample). INTERPRETATION This large GWAS of WMH burden in community-based cohorts of individuals of European descent identifies a novel locus on chromosome 17. Further characterization of this locus may provide novel insights into the pathogenesis of cerebral WMH.
Collapse
Affiliation(s)
- Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, Division of Epidemiology, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Simpson JE, Wharton SB, Cooper J, Gelsthorpe C, Baxter L, Forster G, Shaw PJ, Savva G, Matthews FE, Brayne C, Ince PG. Alterations of the blood–brain barrier in cerebral white matter lesions in the ageing brain. Neurosci Lett 2010; 486:246-51. [PMID: 20887772 DOI: 10.1016/j.neulet.2010.09.063] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Revised: 09/07/2010] [Accepted: 09/22/2010] [Indexed: 11/30/2022]
Affiliation(s)
- Julie E Simpson
- Academic Unit of Pathology, Department of Neuroscience, University of Sheffield, Sheffield, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Polvikoski TM, van Straaten ECW, Barkhof F, Sulkava R, Aronen HJ, Niinistö L, Oinas M, Scheltens P, Erkinjuntti T, Kalaria RN. Frontal lobe white matter hyperintensities and neurofibrillary pathology in the oldest old. Neurology 2010; 75:2071-8. [PMID: 21048201 DOI: 10.1212/wnl.0b013e318200d6f9] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Current studies suggest an interaction between vascular mechanisms and neurodegenerative processes that leads to late-onset Alzheimer disease (AD). We tested whether AD pathology was associated with white matter hyperintensities (WMH) or cerebral infarcts in the oldest old individuals. METHODS Brains from 132 subjects over 85 years old, who came to autopsy from the Vantaa 85+ population-based cohort, were scanned by postmortem MRI and examined for neuropathologic changes. Coronal images were analyzed to determine the degree of frontal and parietal periventricular WMH (PVWMH) and deep WMH (DWMH) and cerebral infarcts. Neuropathologic variables included Consortium to Establish a Registry for Alzheimer's Disease scores for neuritic plaques and Braak staging among subjects in 5 groups: normal aging (NA), borderline with insufficient AD pathology, AD, AD plus other pathology, and other primary degenerative diseases. RESULTS Frontal DWMH were detected in >50% of the sample. Both frontal PVWMH and DWMH were significantly more extensive in the AD group compared to the NA group or the NA and borderline groups combined. Frontal PVWMH and DWMH were also associated with increased Braak staging (p = 0.03) and the neuritic plaque load (p = 0.01). Further analysis revealed there were a greater number of cerebral infarcts associated with frontal DWMH (p = 0.03) but not with frontal PVWMH. CONCLUSIONS Our study showed an association between neurofibrillary pathology and frontal PVWMH and DWMH (rather than parietal), as a surrogate of small vessel disease, particularly in very old community-dwelling individuals.
Collapse
Affiliation(s)
- T M Polvikoski
- Institute for Ageing and Health, Newcastle University, Newcastle upon Tyne, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Xu H, Stamova B, Jickling G, Tian Y, Zhan X, Ander BP, Liu D, Turner R, Rosand J, Goldstein LB, Furie KL, Verro P, Johnston SC, Sharp FR, Decarli CS. Distinctive RNA expression profiles in blood associated with white matter hyperintensities in brain. Stroke 2010; 41:2744-9. [PMID: 20966416 DOI: 10.1161/strokeaha.110.591875] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND AND PURPOSE White matter hyperintensities (WMH) are areas of high signal detected by T2 and fluid-attenuated inversion recovery sequences on brain MRI. Although associated with aging, cerebrovascular risk factors, and cognitive impairment, the pathogenesis of WMH remains unclear. Thus, RNA expression was assessed in the blood of individuals with and without extensive WMH to search for evidence of oxidative stress, inflammation, and other abnormalities described in WMH lesions in brain. METHODS Subjects included 20 with extensive WMH (WMH+), 45% of whom had Alzheimer disease, and 18 with minimal WMH (WMH-), 44% of whom had Alzheimer disease. All subjects were clinically evaluated and underwent quantitative MRI. Total RNA from whole blood was processed on human whole genome Affymetrix HU133 Plus 2.0 microarrays. RNA expression was analyzed using an analysis of covariance. RESULTS Two hundred forty-one genes were differentially regulated at ± 1.2-fold difference (P < 0.005) in subjects with WMH+ as compared to WMH-, regardless of cognitive status and 50 genes were differentially regulated with ± 1.5-fold difference (P < 0.005). Cluster and principal components analyses showed that the expression profiles for these genes distinguished WMH+ from WMH- subjects. Function analyses suggested that WMH-specific genes were associated with oxidative stress, inflammation, detoxification, and hormone signaling, and included genes associated with oligodendrocyte proliferation, axon repair, long-term potentiation, and neurotransmission. CONCLUSIONS The unique RNA expression profile in blood associated with WMH is consistent with roles of systemic oxidative stress and inflammation, as well as other potential processes in the pathogenesis or consequences of WMH.
Collapse
Affiliation(s)
- Huichun Xu
- Department of Neurology and M.I.N.D. Institute, University of California at Davis, Sacramento, CA 95817, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|