1
|
Hervella P, Sampedro-Viana A, Fernández-Rodicio S, Rodríguez-Yáñez M, López-Dequidt I, Pumar JM, Mosqueira AJ, Bazarra-Barreiros M, Abengoza-Bello MT, Ortega-Espina S, Ouro A, Pérez-Mato M, Campos F, Sobrino T, Castillo J, Alonso-Alonso ML, Iglesias-Rey R. Precision Medicine for Blood Glutamate Grabbing in Ischemic Stroke. Int J Mol Sci 2024; 25:6554. [PMID: 38928260 PMCID: PMC11204254 DOI: 10.3390/ijms25126554] [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: 05/13/2024] [Revised: 06/01/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
Glutamate grabbers, such as glutamate oxaloacetate transaminase (GOT), have been proposed to prevent excitotoxicity secondary to high glutamate levels in stroke patients. However, the efficacy of blood glutamate grabbing by GOT could be dependent on the extent and severity of the disruption of the blood-brain barrier (BBB). Our purpose was to analyze the relationship between GOT and glutamate concentration with the patient's functional status differentially according to BBB serum markers (soluble tumor necrosis factor-like weak inducer of apoptosis (sTWEAK) and leukoaraiosis based on neuroimaging). This retrospective observational study includes 906 ischemic stroke patients. We studied the presence of leukoaraiosis and the serum levels of glutamate, GOT, and sTWEAK in blood samples. Functional outcome was assessed using the modified Rankin Scale (mRS) at 3 months. A significant negative correlation between GOT and glutamate levels at admission was shown in those patients with sTWEAK levels > 2900 pg/mL (Pearson's correlation coefficient: -0.249; p < 0.0001). This correlation was also observed in patients with and without leukoaraiosis (Pearson's correlation coefficients: -0.299; p < 0.001 vs. -0.116; p = 0.024). The logistic regression model confirmed the association of higher levels of GOT with lower odds of poor outcome at 3 months when sTWEAK levels were >2900 pg/mL (OR: 0.41; CI 95%: 0.28-0.68; p < 0.0001) or with leukoaraiosis (OR: 0.75; CI 95%: 0.69-0.82; p < 0.0001). GOT levels are associated with glutamate levels and functional outcomes at 3 months, but only in those patients with leukoaraiosis and elevated sTWEAK levels. Consequently, therapies targeting glutamate grabbing might be more effective in patients with BBB dysfunction.
Collapse
Grants
- SAF2017-84267-R, PDC2021-121455-I00 Spanish Ministry of Science and Innovation
- IN607A2022-03, IN607A2022/07 Xunta de Galicia
- PI17/01103, PI22/00938, PI21/01256/, DTS23/00103, RD16/0019/0001, RD21/0006/0003, CB22/05/00067, CPII17/00027, CPII19/00020, CP22/00061, FI22/00200 Instituto de Salud Carlos III
- EAPA_791/2018_ NEUROATLANTIC, 0624_2IQBIONEURO_6_E INTERREG
Collapse
Affiliation(s)
- Pablo Hervella
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.H.); (A.S.-V.); (S.F.-R.); (J.M.P.); (A.J.M.); (M.B.-B.); (M.T.A.-B.); (S.O.-E.); (J.C.)
| | - Ana Sampedro-Viana
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.H.); (A.S.-V.); (S.F.-R.); (J.M.P.); (A.J.M.); (M.B.-B.); (M.T.A.-B.); (S.O.-E.); (J.C.)
| | - Sabela Fernández-Rodicio
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.H.); (A.S.-V.); (S.F.-R.); (J.M.P.); (A.J.M.); (M.B.-B.); (M.T.A.-B.); (S.O.-E.); (J.C.)
| | - Manuel Rodríguez-Yáñez
- Stroke Unit, Department of Neurology, Hospital Clínico Universitario, 15706 Santiago de Compostela, Spain;
| | - Iria López-Dequidt
- Department of Neurology, Hospital Clínico Universitario de Ferrol, 15405 Ferrol, Spain;
| | - José M. Pumar
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.H.); (A.S.-V.); (S.F.-R.); (J.M.P.); (A.J.M.); (M.B.-B.); (M.T.A.-B.); (S.O.-E.); (J.C.)
- Department of Neuroradiology, Hospital Clínico Universitario, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Antonio J. Mosqueira
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.H.); (A.S.-V.); (S.F.-R.); (J.M.P.); (A.J.M.); (M.B.-B.); (M.T.A.-B.); (S.O.-E.); (J.C.)
- Department of Neuroradiology, Hospital Clínico Universitario, Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Marcos Bazarra-Barreiros
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.H.); (A.S.-V.); (S.F.-R.); (J.M.P.); (A.J.M.); (M.B.-B.); (M.T.A.-B.); (S.O.-E.); (J.C.)
| | - María Teresa Abengoza-Bello
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.H.); (A.S.-V.); (S.F.-R.); (J.M.P.); (A.J.M.); (M.B.-B.); (M.T.A.-B.); (S.O.-E.); (J.C.)
| | - Sara Ortega-Espina
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.H.); (A.S.-V.); (S.F.-R.); (J.M.P.); (A.J.M.); (M.B.-B.); (M.T.A.-B.); (S.O.-E.); (J.C.)
| | - Alberto Ouro
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (A.O.); (T.S.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María Pérez-Mato
- Translational Stroke Laboratory (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.P.-M.); (F.C.)
| | - Francisco Campos
- Translational Stroke Laboratory (TREAT), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (M.P.-M.); (F.C.)
| | - Tomás Sobrino
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (A.O.); (T.S.)
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José Castillo
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.H.); (A.S.-V.); (S.F.-R.); (J.M.P.); (A.J.M.); (M.B.-B.); (M.T.A.-B.); (S.O.-E.); (J.C.)
| | - Maria Luz Alonso-Alonso
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.H.); (A.S.-V.); (S.F.-R.); (J.M.P.); (A.J.M.); (M.B.-B.); (M.T.A.-B.); (S.O.-E.); (J.C.)
| | - Ramón Iglesias-Rey
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain; (P.H.); (A.S.-V.); (S.F.-R.); (J.M.P.); (A.J.M.); (M.B.-B.); (M.T.A.-B.); (S.O.-E.); (J.C.)
| |
Collapse
|
2
|
Yang A, Yang YT, Zhao XM. An augmented Mendelian randomization approach provides causality of brain imaging features on complex traits in a single biobank-scale dataset. PLoS Genet 2023; 19:e1011112. [PMID: 38150468 PMCID: PMC10775988 DOI: 10.1371/journal.pgen.1011112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 01/09/2024] [Accepted: 12/12/2023] [Indexed: 12/29/2023] Open
Abstract
Mendelian randomization (MR) is an effective approach for revealing causal risk factors that underpin complex traits and diseases. While MR has been more widely applied under two-sample settings, it is more promising to be used in one single large cohort given the rise of biobank-scale datasets that simultaneously contain genotype data, brain imaging data, and matched complex traits from the same individual. However, most existing multivariable MR methods have been developed for two-sample setting or a small number of exposures. In this study, we introduce a one-sample multivariable MR method based on partial least squares and Lasso regression (MR-PL). MR-PL is capable of considering the correlation among exposures (e.g., brain imaging features) when the number of exposures is extremely upscaled, while also correcting for winner's curse bias. We performed extensive and systematic simulations, and demonstrated the robustness and reliability of our method. Comprehensive simulations confirmed that MR-PL can generate more precise causal estimates with lower false positive rates than alternative approaches. Finally, we applied MR-PL to the datasets from UK Biobank to reveal the causal effects of 36 white matter tracts on 180 complex traits, and showed putative white matter tracts that are implicated in smoking, blood vascular function-related traits, and eating behaviors.
Collapse
Affiliation(s)
- Anyi Yang
- Department of Neurology, Zhongshan Hospital and Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, People’s Republic of China
- MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, People’s Republic of China
| | - Yucheng T. Yang
- Department of Neurology, Zhongshan Hospital and Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, People’s Republic of China
- MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, People’s Republic of China
| | - Xing-Ming Zhao
- Department of Neurology, Zhongshan Hospital and Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, People’s Republic of China
- MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, People’s Republic of China
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, People’s Republic of China
- International Human Phenome Institutes (Shanghai), Shanghai, People’s Republic of China
| |
Collapse
|
3
|
Guo B, Zhang J, Zhang W, Chen F, Liu B. Gut microbiota-derived short chain fatty acids act as mediators of the gut-brain axis targeting age-related neurodegenerative disorders: a narrative review. Crit Rev Food Sci Nutr 2023; 65:265-286. [PMID: 37897083 DOI: 10.1080/10408398.2023.2272769] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2023]
Abstract
Neurodegenerative diseases associated with aging are often accompanied by cognitive decline and gut microbiota disorder. But the impact of gut microbiota on these cognitive disturbances remains incompletely understood. Short chain fatty acids (SCFAs) are major metabolites produced by gut microbiota during the digestion of dietary fiber, serving as an energy source for gut epithelial cells and/or circulating to other organs, such as the liver and brain, through the bloodstream. SCFAs have been shown to cross the blood-brain barrier and played crucial roles in brain metabolism, with potential implications in mediating Alzheimer's disease (AD) and Parkinson's disease (PD). However, the underlying mechanisms that SCFAs might influence psychological functioning, including affective and cognitive processes and their neural basis, have not been fully elucidated. Furthermore, the dietary sources which determine these SCFAs production was not thoroughly evaluated yet. This comprehensive review explores the production of SCFAs by gut microbiota, their transportation through the gut-brain axis, and the potential mechanisms by which they influence age-related neurodegenerative disorders. Also, the review discusses the importance of dietary fiber sources and the challenges associated with harnessing dietary-derived SCFAs as promoters of neurological health in elderly individuals. Overall, this study suggests that gut microbiota-derived SCFAs and/or dietary fibers hold promise as potential targets and strategies for addressing age-related neurodegenerative disorders.
Collapse
Affiliation(s)
- Bingbing Guo
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China
| | - Jingyi Zhang
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China
| | - Weihao Zhang
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China
| | - Feng Chen
- Shenzhen Key Laboratory of Food Nutrition and Health, Institute for Innovative Development of Food Industry, Department of Food Science and Engineering, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, China
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Shenzhen University, Shenzhen, China
| | - Bin Liu
- Shenzhen Key Laboratory of Food Nutrition and Health, Institute for Innovative Development of Food Industry, Department of Food Science and Engineering, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, China
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Shenzhen University, Shenzhen, China
| |
Collapse
|
4
|
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: 0.5] [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
|
5
|
Wang Y, Du W, Sun Y, Zhang J, Ma C, Jin X. CRTC1 is a potential target to delay aging-induced cognitive deficit by protecting the integrity of the blood-brain barrier via inhibiting inflammation. J Cereb Blood Flow Metab 2023; 43:1042-1059. [PMID: 37086081 PMCID: PMC10291461 DOI: 10.1177/0271678x231169133] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 03/09/2023] [Accepted: 03/24/2023] [Indexed: 04/23/2023]
Abstract
Aging can cause attenuation in the functioning of multiple organs, and blood-brain barrier (BBB) breakdown could promote the occurrence of disorders of the central nervous system during aging. Since inflammation is considered to be an important factor underlying BBB injury during aging, vascular endothelial cell senescence serves as a critical pathological basis for the destruction of BBB integrity. In the current review, we have first introduced the concepts related to aging-induced cognitive deficit and BBB integrity damage. Thereafter, we reviewed the potential relationship between disruption of BBB integrity and cognition deficit and the role of inflammation, vascular endothelial cell senescence, and BBB injury. We have also briefly introduced the function of CREB-regulated transcription co-activator 1 (CRTC1) in cognition and aging-induced CRTC1 changes as well as the critical roles of CRTC1/cyclooxygenase-2 (COX-2) in regulating inflammation, endothelial cell senescence, and BBB injury. Finally, the underlying mechanisms have been summarized and we propose that CRTC1 could be a promising target to delay aging-induced cognitive deficit by protecting the integrity of BBB through promoting inhibition of inflammation-mediated endothelial cell senescence.
Collapse
Affiliation(s)
- Yanping Wang
- Department of Neurology, the Second Hospital of Jiaxing City, Jiaxing, China
| | - Weihong Du
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Yanyun Sun
- Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Junfang Zhang
- Department of Physiology, School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, China
| | - Chaolin Ma
- School of Life Science and Institute of Life Science, Nanchang University, Nanchang, China
| | - Xinchun Jin
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Histology and Embryology, School of Basic Medical Sciences, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
- Institute of Neuroscience, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
6
|
Vielee ST, Wise JP. Among Gerontogens, Heavy Metals Are a Class of Their Own: A Review of the Evidence for Cellular Senescence. Brain Sci 2023; 13:500. [PMID: 36979310 PMCID: PMC10046019 DOI: 10.3390/brainsci13030500] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023] Open
Abstract
Advancements in modern medicine have improved the quality of life across the globe and increased the average lifespan of our population by multiple decades. Current estimates predict by 2030, 12% of the global population will reach a geriatric age and live another 3-4 decades. This swelling geriatric population will place critical stress on healthcare infrastructures due to accompanying increases in age-related diseases and comorbidities. While much research focused on long-lived individuals seeks to answer questions regarding how to age healthier, there is a deficit in research investigating what aspects of our lives accelerate or exacerbate aging. In particular, heavy metals are recognized as a significant threat to human health with links to a plethora of age-related diseases, and have widespread human exposures from occupational, medical, or environmental settings. We believe heavy metals ought to be classified as a class of gerontogens (i.e., chemicals that accelerate biological aging in cells and tissues). Gerontogens may be best studied through their effects on the "Hallmarks of Aging", nine physiological hallmarks demonstrated to occur in aged cells, tissues, and bodies. Evidence suggests that cellular senescence-a permanent growth arrest in cells-is one of the most pertinent hallmarks of aging and is a useful indicator of aging in tissues. Here, we discuss the roles of heavy metals in brain aging. We briefly discuss brain aging in general, then expand upon observations for heavy metals contributing to age-related neurodegenerative disorders. We particularly emphasize the roles and observations of cellular senescence in neurodegenerative diseases. Finally, we discuss the observations for heavy metals inducing cellular senescence. The glaring lack of knowledge about gerontogens and gerontogenic mechanisms necessitates greater research in the field, especially in the context of the global aging crisis.
Collapse
Affiliation(s)
- Samuel T. Vielee
- Pediatrics Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
| | - John P. Wise
- Pediatrics Research Institute, Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
7
|
Tang X, Wei C, Zhang R, You J, Chen X. CCL21/CCR7 axis regulates demyelination and vascular cognitive impairment in a mouse model for chronic cerebral hypoperfusion. Neurol Res 2023; 45:248-259. [PMID: 36215431 DOI: 10.1080/01616412.2022.2132456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
OBJECTIVES White matter lesions (WML) are usually accompanied by cognitive decline, which consist of axonal loss and demyelination. CC chemokine ligand 21 (CCL21) and its receptor C-C chemokine receptor 7 (CCR7) belong to the chemokine family, which are involved in many diseases. However, their function in the central nervous system (CNS) is still unexplored. This study aimed to explore the role of CCL21/CCR7 axis in the pathological process of chronic ischemia-induced WML. METHODS Bilateral common carotid artery stenosis (BCAS) was employed in C57BL/6 mice as the in vivo WML model. Microarray analysis was performed to detect the overall molecular changes induced in the endothelial cells by BCAS. Q-PCR, Western blotting, and immunofluorescence staining were performed to evaluate expression levels of the related molecules. The mice were injected with LV-CCL21-GFP virus in the corpus callosum to overexpress CCL21. WML degree was determined via MRI, and cognitive ability was assessed by Y-maze and novel object recognition tests. Myelin sheath integrity was evaluated via immunofluorescence staining. RESULTS CCL21 was significantly downregulated in endothelial cells after BCAS and CCL21 overexpression alleviated BCAS-induced cognitive deficits and demyelination. Furthermore, CCR7 was found to be mainly expressed in oligodendrocytes (OLs) after exposed to hypoxia and CCR7 silencing blocked the protective effects induced by CCL21 overexpression. Conclusions CCL21/CCR7 axis may play a key role in demyelination induced by BCAS. This might provide a novel therapeutic target for WML.
Collapse
Affiliation(s)
- Xuelian Tang
- These authors have contributed equally to this work and share the first authorship
| | - Cunsheng Wei
- These authors have contributed equally to this work and share the first authorship
| | - Rui Zhang
- Department of Neurology, the Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, China
| | - Jie You
- Department of Neurology, the Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, China
| | - Xuemei Chen
- Department of Neurology, the Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, China
| |
Collapse
|
8
|
Abstract
Cerebral small vessel disease (CSVD) has emerged as a common factor driving age-dependent diseases, including stroke and dementia. CSVD-related dementia will affect a growing fraction of the aging population, requiring improved recognition, understanding, and treatments. This review describes evolving criteria and imaging biomarkers for the diagnosis of CSVD-related dementia. We describe diagnostic challenges, particularly in the context of mixed pathologies and the absence of highly effective biomarkers for CSVD-related dementia. We review evidence regarding CSVD as a risk factor for developing neurodegenerative disease and potential mechanisms by which CSVD leads to progressive brain injury. Finally, we summarize recent studies on the effects of major classes of cardiovascular medicines relevant to CSVD-related cognitive impairment. Although many key questions remain, the increased attention to CSVD has resulted in a sharper vision for what will be needed to meet the upcoming challenges imposed by this disease.
Collapse
Affiliation(s)
- Fanny M. Elahi
- Departments of Neurology and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY
- Neurology Service, VA Bronx Healthcare System, Bronx, NY
| | - Michael M. Wang
- Departments of Neurology and Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI
- Neurology Service, VA Ann Arbor Healthcare System, Ann Arbor, MI
| | | |
Collapse
|
9
|
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
|
10
|
Wang H, Liu D. Retrospective case-control study on screening risk factors of antibiotic-associated encephalopathy in patients with chronic kidney disease. BMJ Open 2022; 12:e064995. [PMID: 36526324 PMCID: PMC9764618 DOI: 10.1136/bmjopen-2022-064995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE The renal excretion function of patients with chronic kidney disease (CKD) is reduced, and the nervous system toxic reactions of antibiotics are prone to occur. The purpose of this study is to screen out some risk factors for patients with CKD to suffer from antibiotic-associated encephalopathy (AAE). DESIGN A case-control study. SETTING A tertiary hospital in China. PARTICIPANTS The medical records of patients who were hospitalised for CKD and infectious diseases in our hospital from January 2010 to December 2019. All patients used antibiotics to treat infectious diseases during hospitalisation. All patients were divided into two groups according to whether they developed AAE during hospitalisation. The patients with CKD without AAE were selected as the control group (n=120), and the patients with CKD with AAE were regarded as the AAE group (n=102). INTERVENTIONS This study systematically analysed its clinical manifestations, laboratory examinations, prognosis, etc, and summarised the risk factors related to AAE in patients with CKD. PRIMARY OUTCOME Screening risk factors of AAE in patients with CKD. RESULTS Logistic regression analysis showed that coronary heart disease, as well as abnormal indicators of haemoglobin, albumin, uric acid and blood phosphorus were independent risk factors for patients with CKD with AAE (OR values were 4.137, 0.963, 0.849, 0.996 0.161, respectively, all p<0.05). The case fatality rate (Pearson χ2=7.524, p=0.006), rehospitalisation rate (Pearson χ2=6.187, p=0.013) and treatment costs (t=-8.44, p<0.001) in encephalopathy group are significantly higher than the control group. CONCLUSIONS Patients with CKD with AAE will increase the case fatality rate and cause poor prognosis. Coronary heart disease, as well as decreased levels of haemoglobin, albumin, uric acid, and blood phosphorus are independent risk factors for patients with CKD with AAE. Timely intervention of these risk factors may reduce the incidence of AAE and improve the prognosis.
Collapse
Affiliation(s)
- Hongling Wang
- Department of Nephrology and Rheumatology, Tianjin Third Central Hospital, Tianjin, China
| | - Daquan Liu
- Department of Anatomy and Histology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
11
|
Zhong T, Qi Y, Li R, Zhou H, Ran B, Wang J, Cai Z. Contribution of intracranial artery stenosis to white matter hyperintensities progression in elderly Chinese patients: A 3-year retrospective longitudinal study. Front Neurol 2022; 13:922320. [PMID: 36212654 PMCID: PMC9539973 DOI: 10.3389/fneur.2022.922320] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Background and purposeThere have been controversial results in previous studies for the association between intracranial artery stenosis (ICAS) and white matter hyperintensities (WMHs), and the correlation of ICAS with the progression of WMHs is uncertain. The aim of this study was to investigate the association between ICAS and the progression of WMHs.MethodsIn this retrospective longitudinal study, we enrolled 302 patients aged 60 years and older who had received two brain MRI scans with a 3-year interval and was examined by CTA in the first MRI scan. We measured the stenosis of major intracranial arteries by CTA and assessed the progression of WMHs using the modified Rotterdam Progression scale (mRPS). We performed binary logistic regression analyses and established linear regression model to determine the relationship between the degree of ICAS and the progression of WMHs.ResultsA total of 302 patients were enrolled, of which 48.3% experienced WMHs progression. After adjustment for confounding factors, the patients with Grade 2 ICAS had an OR of 2.8 (95% CI 1.4–5.5), and those with Grade 3 ICAS had an OR of 3.0 (95% CI 1.2–7.3) for the progression of WMHs. The ICAS degree remained associated with PVWMHs but had an attenuated relation to SCWMHs. ICAS severity was significantly associated with WMHs progression scores, higher for Grade 3 ICAS [β (SE) = 0.18 (0.18)] followed by Grade 2 ICAS [β (SE) = 0.10 (0.15)] compared with Grade 1 ICAS.ConclusionsPatients with more severe ICAS are more likely to have WMHs progression and have distinct relevancy to PVWMHs and SCWMHs, which may provide clues for understanding mechanisms of WMHs progression.
Collapse
Affiliation(s)
- Tingting Zhong
- Chongqing Medical University, Chongqing, China
- Department of Cardiology, Chongqing General Hospital, Chongqing, China
- Department of Neurology, Chongqing General Hospital, Chongqing, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, China
| | - Yunwen Qi
- Chongqing Medical University, Chongqing, China
- Department of Neurology, Chongqing General Hospital, Chongqing, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, China
| | - Rui Li
- Stroke Center and Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Huadong Zhou
- Department of Neurology and Centre for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Boli Ran
- Department of Cardiology, Chongqing General Hospital, Chongqing, China
| | - Jiao Wang
- Department of Cardiology, Chongqing General Hospital, Chongqing, China
| | - ZhiYou Cai
- Chongqing Medical University, Chongqing, China
- Department of Neurology, Chongqing General Hospital, Chongqing, China
- Chongqing Key Laboratory of Neurodegenerative Diseases, Chongqing, China
- *Correspondence: ZhiYou Cai
| |
Collapse
|
12
|
Abstract
Understanding normal brain aging physiology is essential to improving healthy human longevity, differentiation, and early detection of diseases, such as neurodegenerative diseases, which are an enormous social and economic burden. Functional decline, such as reduced physical activity and cognitive abilities, is typically associated with brain aging. The authors summarize the aging brain mechanism and effects of aging on the brain observed by brain structural MR imaging and advanced neuroimaging techniques, such as diffusion tensor imaging and functional MR imaging.
Collapse
Affiliation(s)
- Yoshiaki Ota
- Division of Neuroradiology, Department of Radiology, University of Michigan, 1500 East Medical Center Drive, UH B2, Ann Arbor, MI 48109, USA
| | - Gaurang Shah
- Division of Neuroradiology, Department of Radiology, University of Michigan, 1500 East Medical Center Drive, UH B2, Ann Arbor, MI 48109, USA.
| |
Collapse
|
13
|
Uddin MA, Akhter MS, Kubra KT, Barabutis N. Hsp90 inhibition protects brain endothelial cells against LPS-induced injury. Biofactors 2022; 48:926-933. [PMID: 35266593 PMCID: PMC10131175 DOI: 10.1002/biof.1833] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 02/21/2022] [Indexed: 02/06/2023]
Abstract
Dysfunction of the blood-brain barrier (BBB) endothelium increases infiltration of lymphocytes and innate immune cells in the brain, leading to the development of neurological disorders. Heat shock protein 90 (Hsp90) inhibitors are anti-inflammatory agents and P53 inducers, which reduce the production of reactive oxygen species (ROS) in a diverse variety of human tissues. In this study, we investigate the effects of those compounds in LPS-induced brain endothelial inflammation, by utilizing human cerebral microvascular endothelial cells (hCMEC/D3). Our results suggest that Hsp90 inhibitors suppress inflammation by inhibiting the LPS-induced signal transducer and activator of transcription 3 (STAT3); and P38 activation. Moreover, those compounds reduce the P53 suppressors murine double minute 2 (MDM2) and murine double minute 4 (MDM4). Immunoglobulin heavy chain binding protein/glucose-regulated protein 78 (BiP/Grp78)-a key element of endothelial barrier integrity-was also increased by Hsp90 inhibition. Hence, we conclude that application of Hsp90 inhibitors in diseases related to BBB dysfunction may deliver a novel therapeutic possibility in the affected population.
Collapse
Affiliation(s)
- Mohammad A Uddin
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, USA
| | - Mohammad S Akhter
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, USA
| | - Khadeja-Tul Kubra
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, USA
| | - Nektarios Barabutis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, USA
| |
Collapse
|
14
|
Brown RB, Tozer DJ, Loubière L, Hong YT, Fryer TD, Williams GB, Graves MJ, Aigbirhio FI, O’Brien JT, Markus HS. MINocyclinE to Reduce inflammation and blood brain barrier leakage in small Vessel diseAse (MINERVA) trial study protocol. Eur Stroke J 2022; 7:323-330. [PMID: 36082255 PMCID: PMC9445404 DOI: 10.1177/23969873221100338] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 04/24/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Cerebral small vessel disease (SVD) is a common cause of stroke and cognitive impairment. Recent data has implicated neuroinflammation and increased blood-brain barrier (BBB) permeability in its pathogenesis, but whether such processes are causal and can be therapeutically modified is uncertain. In a rodent model of SVD, minocycline was associated with reduced white matter lesions, inflammation and BBB permeability. Aims: To determine whether blood-brain barrier permeability (measured using dynamic contrast-enhanced MRI) and microglial activation (measured by positron emission tomography using the radioligand 11C-PK11195) can be modified in SVD. Design: Phase II randomised double blind, placebo-controlled trial of minocycline 100 mg twice daily for 3 months in 44 participants with moderate to severe SVD defined as a clinical lacunar stroke and confluent white matter hyperintensities. Outcomes: Primary outcome measures are volume and intensity of focal increases of blood-brain barrier permeability and microglial activation determined using PET-MRI imaging. Secondary outcome measures include inflammatory biomarkers in serum, and change in conventional MRI markers and cognitive performance over 1 year follow up. Discussion: The MINERVA trial aims to test whether minocycline can influence novel pathological processes thought to be involved in SVD progression, and will provide insights into whether central nervous system inflammation in SVD can be therapeutically modulated.
Collapse
Affiliation(s)
- Robin B Brown
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Daniel J Tozer
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Laurence Loubière
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Young T Hong
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Tim D Fryer
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Guy B Williams
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - Martin J Graves
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - Franklin I Aigbirhio
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Wolfson Brain Imaging Centre, University of Cambridge, Cambridge, UK
| | - John T O’Brien
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - Hugh S Markus
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| |
Collapse
|
15
|
Wardlaw JM, Benveniste H, Williams A. Cerebral Vascular Dysfunctions Detected in Human Small Vessel Disease and Implications for Preclinical Studies. Annu Rev Physiol 2022; 84:409-434. [PMID: 34699267 DOI: 10.1146/annurev-physiol-060821-014521] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cerebral small vessel disease (SVD) is highly prevalent and a common cause of ischemic and hemorrhagic stroke and dementia, yet the pathophysiology is poorly understood. Its clinical expression is highly varied, and prognostic implications are frequently overlooked in clinics; thus, treatment is currently confined to vascular risk factor management. Traditionally, SVD is considered the small vessel equivalent of large artery stroke (occlusion, rupture), but data emerging from human neuroimaging and genetic studies refute this, instead showing microvessel endothelial dysfunction impacting on cell-cell interactions and leading to brain damage. These dysfunctions reflect defects that appear to be inherited and secondary to environmental exposures, including vascular risk factors. Interrogation in preclinical models shows consistent and converging molecular and cellular interactions across the endothelial-glial-neural unit that increasingly explain the human macroscopic observations and identify common patterns of pathology despite different triggers. Importantly, these insights may offer new targets for therapeutic intervention focused on restoring endothelial-glial physiology.
Collapse
Affiliation(s)
- Joanna M Wardlaw
- Division of Neuroimaging Sciences, Centre for Clinical Brain Sciences; UK Dementia Research Institute; and Edinburgh Imaging, University of Edinburgh, Edinburgh, United Kingdom;
| | - Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Anna Williams
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
16
|
Rudilosso S, Rodríguez-Vázquez A, Urra X, Arboix A. The Potential Impact of Neuroimaging and Translational Research on the Clinical Management of Lacunar Stroke. Int J Mol Sci 2022; 23:1497. [PMID: 35163423 PMCID: PMC8835925 DOI: 10.3390/ijms23031497] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/20/2022] [Accepted: 01/24/2022] [Indexed: 12/21/2022] Open
Abstract
Lacunar infarcts represent one of the most frequent subtypes of ischemic strokes and may represent the first recognizable manifestation of a progressive disease of the small perforating arteries, capillaries, and venules of the brain, defined as cerebral small vessel disease. The pathophysiological mechanisms leading to a perforating artery occlusion are multiple and still not completely defined, due to spatial resolution issues in neuroimaging, sparsity of pathological studies, and lack of valid experimental models. Recent advances in the endovascular treatment of large vessel occlusion may have diverted attention from the management of patients with small vessel occlusions, often excluded from clinical trials of acute therapy and secondary prevention. However, patients with a lacunar stroke benefit from early diagnosis, reperfusion therapy, and secondary prevention measures. In addition, there are new developments in the knowledge of this entity that suggest potential benefits of thrombolysis in an extended time window in selected patients, as well as novel therapeutic approaches targeting different pathophysiological mechanisms involved in small vessel disease. This review offers a comprehensive update in lacunar stroke pathophysiology and clinical perspective for managing lacunar strokes, in light of the latest insights from imaging and translational studies.
Collapse
Affiliation(s)
- Salvatore Rudilosso
- Comprehensive Stroke Center, Hospital Clínic of Barcelona, 08036 Barcelona, Spain; (S.R.); (A.R.-V.); (X.U.)
| | - Alejandro Rodríguez-Vázquez
- Comprehensive Stroke Center, Hospital Clínic of Barcelona, 08036 Barcelona, Spain; (S.R.); (A.R.-V.); (X.U.)
| | - Xabier Urra
- Comprehensive Stroke Center, Hospital Clínic of Barcelona, 08036 Barcelona, Spain; (S.R.); (A.R.-V.); (X.U.)
| | - Adrià Arboix
- Cerebrovascular Division, Department of Neurology, Hospital Universitari del Sagrat Cor, Universitat de Barcelona, 08034 Barcelona, Spain
| |
Collapse
|
17
|
Blinkouskaya Y, Caçoilo A, Gollamudi T, Jalalian S, Weickenmeier J. Brain aging mechanisms with mechanical manifestations. Mech Ageing Dev 2021; 200:111575. [PMID: 34600936 PMCID: PMC8627478 DOI: 10.1016/j.mad.2021.111575] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 09/09/2021] [Accepted: 09/22/2021] [Indexed: 12/14/2022]
Abstract
Brain aging is a complex process that affects everything from the subcellular to the organ level, begins early in life, and accelerates with age. Morphologically, brain aging is primarily characterized by brain volume loss, cortical thinning, white matter degradation, loss of gyrification, and ventricular enlargement. Pathophysiologically, brain aging is associated with neuron cell shrinking, dendritic degeneration, demyelination, small vessel disease, metabolic slowing, microglial activation, and the formation of white matter lesions. In recent years, the mechanics community has demonstrated increasing interest in modeling the brain's (bio)mechanical behavior and uses constitutive modeling to predict shape changes of anatomically accurate finite element brain models in health and disease. Here, we pursue two objectives. First, we review existing imaging-based data on white and gray matter atrophy rates and organ-level aging patterns. This data is required to calibrate and validate constitutive brain models. Second, we review the most critical cell- and tissue-level aging mechanisms that drive white and gray matter changes. We focuse on aging mechanisms that ultimately manifest as organ-level shape changes based on the idea that the integration of imaging and mechanical modeling may help identify the tipping point when normal aging ends and pathological neurodegeneration begins.
Collapse
Affiliation(s)
- Yana Blinkouskaya
- Department of Mechanical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, United States
| | - Andreia Caçoilo
- Department of Mechanical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, United States
| | - Trisha Gollamudi
- Department of Biomedical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, United States
| | - Shima Jalalian
- Department of Mechanical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, United States
| | - Johannes Weickenmeier
- Department of Mechanical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030, United States.
| |
Collapse
|
18
|
Liao FF, Lin G, Chen X, Chen L, Zheng W, Raghow R, Zhou FM, Shih AY, Tan XL. Endothelial Nitric Oxide Synthase-Deficient Mice: A Model of Spontaneous Cerebral Small-Vessel Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1932-1945. [PMID: 33711310 PMCID: PMC8647425 DOI: 10.1016/j.ajpath.2021.02.022] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 02/04/2021] [Accepted: 02/24/2021] [Indexed: 02/08/2023]
Abstract
Age-related cerebral small-vessel disease (CSVD) is a major cause of stroke and dementia. Despite a widespread acceptance of small-vessel arteriopathy, lacunar infarction, diffuse white matter injury, and cognitive impairment as four cardinal features of CSVD, a unifying pathologic mechanism of CSVD remains elusive. Herein, we introduce partial endothelial nitric oxide synthase (eNOS)-deficient mice as a model of age-dependent, spontaneous CSVD. These mice developed cerebral hypoperfusion and blood-brain barrier leakage at a young age, which progressively worsened with advanced age. Their brains exhibited elevated oxidative stress, astrogliosis, cerebral amyloid angiopathy, microbleeds, microinfarction, and white matter pathology. Partial eNOS-deficient mice developed gait disturbances at middle age, and hippocampus-dependent memory deficits at older ages. These mice also showed enhanced expression of bone morphogenetic protein 4 (BMP4) in brain pericytes before myelin loss and white matter pathology. Because BMP4 signaling not only promotes astrogliogenesis but also blocks oligodendrocyte differentiation, we posit that paracrine actions of BMP4, localized within the neurovascular unit, promote white matter disorganization and neurodegeneration. These observations point to BMP4 signaling pathway in the aging brain vasculature as a potential therapeutic target. Finally, because studies in partial eNOS-deficient mice corroborated recent clinical evidence that blood-brain barrier disruption is a primary cause of white matter pathology, the mechanism of impaired nitric oxide signaling-mediated CSVD warrants further investigation.
Collapse
Affiliation(s)
- Francesca-Fang Liao
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee.
| | - Geng Lin
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee; Department of Histology and Embryology, Basic Medical University, China Medical University, Shenyang, China
| | - Xingyong Chen
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee; Department of Neurology, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou, China
| | - Ling Chen
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee; Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Wei Zheng
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee; Department of Histology and Embryology, Basic Medical University, China Medical University, Shenyang, China
| | - Rajendra Raghow
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee
| | - Fu-Ming Zhou
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee
| | - Andy Y Shih
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington
| | - Xing-Lin Tan
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee; Department of Neurology, Nanhai Hospital of Southern Medical University, Foshan, China
| |
Collapse
|
19
|
Frías-Anaya E, Gromnicova R, Kraev I, Rogachevsky V, Male DK, Crea F, Hawkes CA, Romero IA. Age-related ultrastructural neurovascular changes in the female mouse cortex and hippocampus. Neurobiol Aging 2021; 101:273-284. [PMID: 33579556 DOI: 10.1016/j.neurobiolaging.2020.12.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 11/29/2020] [Accepted: 12/05/2020] [Indexed: 10/22/2022]
Abstract
Blood-brain barrier (BBB) breakdown occurs in aging and neurodegenerative diseases. Although age-associated alterations have previously been described, most studies focused in male brains; hence, little is known about BBB breakdown in females. This study measured ultrastructural features in the aging female BBB using transmission electron microscopy and 3-dimensional reconstruction of cortical and hippocampal capillaries from 6- and 24-month-old female C57BL/6J mice. Aged cortical capillaries showed more changes than hippocampal capillaries. Specifically, the aged cortex showed thicker basement membrane, higher number and volume of endothelial pseudopods, decreased endothelial mitochondrial number, larger pericyte mitochondria, higher pericyte-endothelial cell contact, and increased tight junction tortuosity compared with young animals. Only increased basement membrane thickness and pericyte mitochondrial volume were observed in the aged hippocampus. Regional comparison revealed significant differences in endothelial pseudopods and tight junctions between the cortex and hippocampus of 24-month-old mice. Therefore, the aging female BBB shows region-specific ultrastructural alterations that may lead to oxidative stress and abnormal capillary blood flow and barrier stability, potentially contributing to cerebrovascular diseases, particularly in postmenopausal women.
Collapse
Affiliation(s)
- Eduardo Frías-Anaya
- School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Radka Gromnicova
- School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Igor Kraev
- School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Vadim Rogachevsky
- Institute of Cell Biophysics RAS, Pushchino Federal Research Centre for Biological Research, Pushchino, Russia
| | - David K Male
- School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Francesco Crea
- School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Cheryl A Hawkes
- School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, UK; Department of Biomedical and Life Sciences, Lancaster University, Lancaster, UK
| | - Ignacio A Romero
- School of Life, Health and Chemical Sciences, The Open University, Milton Keynes, UK.
| |
Collapse
|
20
|
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: 1.8] [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
|
21
|
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: 1.0] [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
|
22
|
Zhou YN, Gao HY, Zhao FF, Liang YC, Gao Y, Liu XH, Wang T, Wang ZG, Wu QJ. The study on analysis of risk factors for severity of white matter lesions and its correlation with cerebral microbleeds in the elderly with lacunar infarction. Medicine (Baltimore) 2020; 99:e18865. [PMID: 31977887 PMCID: PMC7004709 DOI: 10.1097/md.0000000000018865] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
This study aimed to explore the risk factors for severity of white matter lesions and its correlation with in the elderly with lacunar infarction.Patients (range, 70-85 years) with lacunar infarction treated in a hospital in China from 2016 to 2017were enrolled. Fazekas rating scale (0-6 points) was used to assess severity of white matter lesions. Risk factors for the severity of white matter lesions and correlation between cerebral microbleeds and white matter lesions in the elderly with lacunar infarction were studied.The elderly (81-85 years' old, odds ratio [OR]: 2.423, 95% confidence interval [CI]: 1.795-3.271, P = .018; 76∼80 years' old, OR: 3.113, 95% CI: 1.723-5.625, P = .043), carotid atherosclerosis (OR: 3.062, 95% CI:1.715-5.468, P < .001), history of hypertension (OR: 3.694, 95% CI: 2.031-6.717, P < .001) were risk factors for the severity of white matter lesions. The white matter lesions score increased corresponding to increase in the cerebral microbleeds grade (P < .001). The white matter lesions score was higher in the cerebral microbleeds combined with the white matter lesions group than in the white matter lesions group (P < .01). After correcting the effects of age, there was a correlation between white matter lesions and cerebral microbleeds (P < .001). Logistic analysis revealed that the patients' age (81-85 years' old, OR: 2.722, 95% CI: 1.985-3.734, P = .019; 76∼80 years' old, OR: 1.857, 95% CI: 1.075-3.207, P = .031), history of hypertension (OR: 2.931, 95% CI: 1.136-7.567, P = 0.0.036), systolic blood pressure (OR: 1.049, 95% CI: 1.015-1.084, P = .007), high-sensitivity C-reactive protein (OR: 1.504, 95% CI: 1.254-1.803, P < .001), homocysteine (OR: 1.076, 95% CI: 1.020-1.136, P = .009), and carotid atherosclerosis (OR: 1.389, 95% CI: 1.103-1.748, P = .010) were significant risk factors for combined cerebral microbleeds with white matter lesions in patients with lacunar infarction.The elderly, carotid atherosclerosis, history of hypertension were risk factors for the severity of white matter lesions. Cerebral microbleeds were positively correlated with the severity of white matter lesions.
Collapse
Affiliation(s)
- Yu-Ni Zhou
- Department of Neurology, Jining Psychiatric Hospital, Jining
| | - Hao-Yuan Gao
- Department of Cardiology, The Second Affiliated Hospital of Shandong First Medical University
| | - Fang-Fang Zhao
- Department of Neurology, Tai’an City Central Hospital, Tai’an
| | - Ying-Chun Liang
- Department of Neurology, Tai’an City Central Hospital, Tai’an
| | - Yuan Gao
- Department of Psychiatry, Jining Psychiatric Hospital, Jining, Shandong
| | - Xin-Hong Liu
- Department of Neurology, Tai’an City Central Hospital, Tai’an
| | - Tao Wang
- Department of Neurology, Tai’an City Central Hospital, Tai’an
| | - Zhi-Gao Wang
- Department of Statistics, East China Normal University, Shanghai
| | - Qing-Jian Wu
- Department of Emergency, Jining NO.1 People's Hospital, Jining, Shandong, China
| |
Collapse
|
23
|
Coelho S, Pozo JM, Costantini M, Highley JR, Mozumder M, Simpson JE, Ince PG, Frangi AF. Histological data of axons, astrocytes, and myelin in deep subcortical white matter populations. Data Brief 2019; 23:103762. [PMID: 31372422 PMCID: PMC6660516 DOI: 10.1016/j.dib.2019.103762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 02/07/2019] [Accepted: 02/08/2019] [Indexed: 11/15/2022] Open
Abstract
This immunohistochemistry dataset contains the main structures in deep subcortical white matter (axons, astrocytes, and myelinated axons) in a representative cohort of an ageing population. A set of samples from 90 subjects of the Cognitive Function and Ageing Study (CFAS) were analysed, stratified into three groups of 30 subjects each, in relation to the presence of age-associated deep subcortical lesions. High-resolution microscopy data enables the extraction of valuable information, such as volume fractions, for the construction and validation of diffusion MRI (dMRI) models. The dataset provided here was used in Coelho et al. [1].
Collapse
Affiliation(s)
- Santiago Coelho
- Centre for Computational Imaging & Simulation Technologies in Biomedicine (CISTIB) and Leeds Institute for Cardiac and Metabolic Medicine (LICAMM), School of Computing & School of Medicine, University of Leeds, Leeds, UK.,CISTIB, Electronic and Electrical Engineering Department, The University of Sheffield, Sheffield, UK
| | - Jose M Pozo
- Centre for Computational Imaging & Simulation Technologies in Biomedicine (CISTIB) and Leeds Institute for Cardiac and Metabolic Medicine (LICAMM), School of Computing & School of Medicine, University of Leeds, Leeds, UK.,CISTIB, Electronic and Electrical Engineering Department, The University of Sheffield, Sheffield, UK
| | - Marina Costantini
- CISTIB, Electronic and Electrical Engineering Department, The University of Sheffield, Sheffield, UK
| | - J Robin Highley
- Sheffield Institute for Translational Neuroscience (SITraN), The University of Sheffield, Sheffield, UK
| | - Meghdoot Mozumder
- CISTIB, Electronic and Electrical Engineering Department, The University of Sheffield, Sheffield, UK
| | - Julie E Simpson
- Sheffield Institute for Translational Neuroscience (SITraN), The University of Sheffield, Sheffield, UK
| | - Paul G Ince
- Sheffield Institute for Translational Neuroscience (SITraN), The University of Sheffield, Sheffield, UK
| | - Alejandro F Frangi
- Centre for Computational Imaging & Simulation Technologies in Biomedicine (CISTIB) and Leeds Institute for Cardiac and Metabolic Medicine (LICAMM), School of Computing & School of Medicine, University of Leeds, Leeds, UK.,CISTIB, Electronic and Electrical Engineering Department, The University of Sheffield, Sheffield, UK
| |
Collapse
|
24
|
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: 71] [Impact Index Per Article: 11.8] [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
|
25
|
Local volume fraction distributions of axons, astrocytes, and myelin in deep subcortical white matter. Neuroimage 2018; 179:275-287. [DOI: 10.1016/j.neuroimage.2018.06.040] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 05/31/2018] [Accepted: 06/11/2018] [Indexed: 01/28/2023] Open
|
26
|
Pan Y, Nicolazzo JA. Impact of aging, Alzheimer's disease and Parkinson's disease on the blood-brain barrier transport of therapeutics. Adv Drug Deliv Rev 2018; 135:62-74. [PMID: 29665383 DOI: 10.1016/j.addr.2018.04.009] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 01/17/2018] [Accepted: 04/07/2018] [Indexed: 01/01/2023]
Abstract
Older people are at a greater risk of medicine-induced toxicity resulting from either increased drug sensitivity or age-related pharmacokinetic changes. The scenario is further complicated with the two most prevalent age-related neurodegenerative diseases, Alzheimer's disease (AD) and Parkinson's disease (PD). With aging, AD and PD, there is growing evidence of altered structure and function of the blood-brain barrier (BBB), including modifications to tight junctions and efflux transporters, such as P-glycoprotein. The subsequent impact on CNS drug exposure and risk of neurotoxicity from systemically-acting medicines is less well characterized. The purpose of this review, therefore, is to provide an overview of the multiple changes that occur to the BBB as a result of aging, AD and PD, and the impact that such changes have on CNS exposure of drugs, based on studies conducted in aged rodents or rodent models of disease, and in elderly people with and without AD or PD.
Collapse
Affiliation(s)
- Yijun Pan
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia
| | - Joseph A Nicolazzo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 399 Royal Parade, Parkville, Victoria 3052, Australia.
| |
Collapse
|
27
|
Stamatovic SM, Martinez-Revollar G, Hu A, Choi J, Keep RF, Andjelkovic AV. Decline in Sirtuin-1 expression and activity plays a critical role in blood-brain barrier permeability in aging. Neurobiol Dis 2018; 126:105-116. [PMID: 30196051 DOI: 10.1016/j.nbd.2018.09.006] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 08/20/2018] [Accepted: 09/05/2018] [Indexed: 01/05/2023] Open
Abstract
Accumulating evidence suggest that cerebral microvascular disease increases with advancing age and is associated with lacunar stroke, leukoaraiosis, vascular dementia and Alzheimer disease. Increased blood brain barrier (BBB) permeability/leakage takes "center stage" in ongoing age-related vascular/brain parenchymal injury. Although significant effort has been made in defining the gene mutations and risk factors involved in microvascular alterations in vascular dementia and Alzheimer disease, the intra- and intercellular pathogenic mechanisms responsible for vascular hyperpermeability are still largely unknown. The present study aimed to reveal the ongoing senescence process in brain endothelial cells and its effect on BBB integrity in healthy/non-disease conditions. An analysis of BBB integrity during the life span of C56Bl6 mice (young, 2-6 months; middle-aged, 6-12, months; old, 16-22 months) showed increased BBB permeability for different molecular sized tracers (sodium fluorescein, inulin and 20 kDa dextran) in aged mice which was accompanied by modifications in tight junction (TJ) complex organization, manifested as altered TJ protein expression (particularly claudin-5). A gene screening analysis of aging associated markers in brain microvessels isolated from "aged" mice (C56Bl6, 18-20 months) and human brain samples showed a significant decline in sirtuin-1 expression (Sirt1; ~2.8-fold) confirmed at mRNA and protein levels and by activation assay. Experiments in Sirt1 transgenic mice and brain endothelial cell-specific Sirt1 knockout mice indicated that Sirt1 affects BBB integrity, with loss increasing permeability. Similarly, in vitro, overexpressing Sirt1 or increasing Sirt1 activity with an agonist (Sirt1720) protected against senescence-induced brain endothelial barrier hyperpermeability, stabilized claudin-5/ZO-1 interactions and rescued claudin-5 expression. These findings reveal a novel role of Sirt1 in modulating aging-associated BBB persistent leakage.
Collapse
Affiliation(s)
- Svetlana M Stamatovic
- Department of Pathology, Medical School, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Anna Hu
- Department of Pathology, Medical School, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jennifer Choi
- Department of Pathology, Medical School, University of Michigan, Ann Arbor, MI 48109, USA
| | - Richard F Keep
- Department of Neurosurgery, Medical School, University of Michigan, Ann Arbor, MI 48109, USA; Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Anuska V Andjelkovic
- Department of Pathology, Medical School, University of Michigan, Ann Arbor, MI 48109, USA; Department of Neurosurgery, Medical School, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
28
|
Brown R, Benveniste H, Black SE, Charpak S, Dichgans M, Joutel A, Nedergaard M, Smith KJ, Zlokovic BV, Wardlaw JM. Understanding the role of the perivascular space in cerebral small vessel disease. Cardiovasc Res 2018; 114:1462-1473. [PMID: 29726891 PMCID: PMC6455920 DOI: 10.1093/cvr/cvy113] [Citation(s) in RCA: 231] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 04/18/2018] [Accepted: 05/02/2018] [Indexed: 12/17/2022] Open
Abstract
Small vessel diseases (SVDs) are a group of disorders that result from pathological alteration of the small blood vessels in the brain, including the small arteries, capillaries and veins. Of the 35-36 million people that are estimated to suffer from dementia worldwide, up to 65% have an SVD component. Furthermore, SVD causes 20-25% of strokes, worsens outcome after stroke and is a leading cause of disability, cognitive impairment and poor mobility. Yet the underlying cause(s) of SVD are not fully understood. Magnetic resonance imaging has confirmed enlarged perivascular spaces (PVS) as a hallmark feature of SVD. In healthy tissue, these spaces are proposed to form part of a complex brain fluid drainage system which supports interstitial fluid exchange and may also facilitate clearance of waste products from the brain. The pathophysiological signature of PVS and what this infers about their function and interaction with cerebral microcirculation, plus subsequent downstream effects on lesion development in the brain has not been established. Here we discuss the potential of enlarged PVS to be a unique biomarker for SVD and related brain disorders with a vascular component. We propose that widening of PVS suggests presence of peri-vascular cell debris and other waste products that form part of a vicious cycle involving impaired cerebrovascular reactivity, blood-brain barrier dysfunction, perivascular inflammation and ultimately impaired clearance of waste proteins from the interstitial fluid space, leading to accumulation of toxins, hypoxia, and tissue damage. Here, we outline current knowledge, questions and hypotheses regarding understanding the brain fluid dynamics underpinning dementia and stroke through the common denominator of SVD.
Collapse
Affiliation(s)
- Rosalind Brown
- Centre for Clinical Brain Sciences, The University of Edinburgh, Chancellor's Building, Edinburgh, UK
| | - Helene Benveniste
- Department of Anesthesiology, Yale School of Medicine, New Haven, USA
| | - Sandra E Black
- LC Campbell Cognitive Neurology Research Unit, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
- Department of Medicine (Neurology), Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
- Hurvitz Brain Sciences Program, Sunnybrook Health Sciences Center, University of Toronto, Toronto, Canada
- Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - Serge Charpak
- INSERM U1128, Laboratory of Neurophysiology and New Microscopies, Université Paris Descartes, Paris, France
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-Universität LMU, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE, Munich), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Anne Joutel
- Genetics and Pathogenesis of Cerebrovascular Diseases, INSERM, Université Paris Diderot-Paris 7, Paris, France
- DHU NeuroVasc, Sorbonne Paris Cité, Paris, France
| | - Maiken Nedergaard
- Section for Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
- Division of Glia Disease and Therapeutics, Center for Translational Neuromedicine, University of Rochester Medical School, Rochester, USA
| | - Kenneth J Smith
- Department of Neuroinflammation, UCL Institute of Neurology, London, UK
| | - Berislav V Zlokovic
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Joanna M Wardlaw
- Centre for Clinical Brain Sciences, Chancellor's Building, Edinburgh, UK
- UK Dementia Research Institute at The University of Edinburgh, Chancellor's Building, Edinburgh, UK
- Row Fogo Centre for Research into Ageing and the Brain, The University of Edinburgh, Chancellor's Building, Edinburgh, UK
| |
Collapse
|
29
|
Hunter S, Smailagic N, Brayne C. Dementia Research: Populations, Progress, Problems, and Predictions. J Alzheimers Dis 2018; 64:S119-S143. [DOI: 10.3233/jad-179927] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Sally Hunter
- Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Nadja Smailagic
- Institute of Public Health, University of Cambridge, Cambridge, UK
| | - Carol Brayne
- Institute of Public Health, University of Cambridge, Cambridge, UK
| |
Collapse
|
30
|
Morgan SV, Garwood CJ, Jennings L, Simpson JE, Castelli LM, Heath PR, Mihaylov SR, Vaquéz-Villaseñor I, Minshull TC, Ince PG, Dickman MJ, Hautbergue GM, Wharton SB. Proteomic and cellular localisation studies suggest non-tight junction cytoplasmic and nuclear roles for occludin in astrocytes. Eur J Neurosci 2018; 47:1444-1456. [PMID: 29738614 PMCID: PMC6079634 DOI: 10.1111/ejn.13933] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 04/16/2018] [Indexed: 12/16/2022]
Abstract
Occludin is a component of tight junctions, which are essential structural components of the blood–brain barrier. However, occludin is expressed in cells without tight junctions, implying additional functions. We determined the expression and localisation of occludin in astrocytes in cell culture and in human brain tissue, and sought novel binding partners using a proteomic approach. Expression was investigated by immunocytochemistry and immunoblotting in the 1321N1 astrocytoma cell line and ScienCell human primary astrocytes, and by immunohistochemistry in human autopsy brain tissue. Recombinant N‐ and C‐terminal occludin was used to pull‐down proteins from 1321N1 cell lysates and protein‐binding partners identified by mass spectrometry analysis. Occludin was expressed in both the cytoplasm and nucleus of astrocytes in vitro and in vivo. Mass spectrometry identified binding to nuclear and cytoplasmic proteins, particularly those related to RNA metabolism and nuclear function. Occludin is expressed in several subcellular compartments of brain cell‐types that do not form tight junctions and the expression patterns in cell culture reflect those in human brain tissue, indicating they are suitable model systems. Proteomic analysis suggests that occludin has novel functions in neuroepithelial cells that are unrelated to tight junction formation. Further research will establish the roles of these functions in both cellular physiology and in disease states.
Collapse
Affiliation(s)
- Sarah V Morgan
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Claire J Garwood
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Luke Jennings
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Julie E Simpson
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Lydia M Castelli
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Paul R Heath
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Simeon R Mihaylov
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | | | - Thomas C Minshull
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK
| | - Paul G Ince
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Mark J Dickman
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield, UK
| | - Guillaume M Hautbergue
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Stephen B Wharton
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| |
Collapse
|
31
|
Cardiovascular disease and brain health: Focus on white matter hyperintensities. IJC HEART & VASCULATURE 2018; 19:63-69. [PMID: 29946567 PMCID: PMC6016077 DOI: 10.1016/j.ijcha.2018.04.006] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 04/25/2018] [Accepted: 04/26/2018] [Indexed: 12/28/2022]
Abstract
Diseases affecting the brain contribute to a substantial proportion of morbidity and mortality in the general population. Conditions such as stroke, dementia and cognitive impairment have a prominent impact on global public health. Despite the heterogeneous clinical manifestations of these conditions and their diverse prognostic implications, current evidence supports a role for cardiovascular disease as a common pathophysiological ground. Brain white matter hyperintensities (WMH) are patchy white matter signal hyperintensity on T2-weighted magnetic resonance imaging sequences commonly found in elderly individuals. WMH appear to have a vascular pathogenesis and have been shown to confer an increased risk of stroke and cognitive decline. Indeed, they were proposed as a marker for central nervous system frailty. Cardiovascular diseases seem to play a key role in the etiology of WMH. Carotid atherosclerosis and atrial fibrillation were shown to be associated with higher WMH burden, while adequate blood pressure control has been reported reducing WMH progression. Aim of the present work is to review the available evidence linking WMH to cardiovascular disease, highlighting the complex interplay between cerebral and cardiovascular health.
Collapse
|
32
|
Blood-Brain Barrier Damage as the Starting Point of Leukoaraiosis Caused by Cerebral Chronic Hypoperfusion and Its Involved Mechanisms: Effect of Agrin and Aquaporin-4. BIOMED RESEARCH INTERNATIONAL 2018; 2018:2321797. [PMID: 29682525 PMCID: PMC5846350 DOI: 10.1155/2018/2321797] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 08/07/2017] [Accepted: 08/29/2017] [Indexed: 11/17/2022]
Abstract
White matter lesion (WML) is popular in the patients aged over 65. Brain edema and blood-brain barrier (BBB) dysfunction due to cerebral chronic hypoperfusion (CCH) contributed to WML. Preserving astrocyte polarity is vital for BBB integrity. In our experiment, CCH model is established by bilateral carotid arteries occlusion (2VO). Leukoaraiosis was verified by fiber density stain, and brain edema was evaluated using brain water content measuring. The expressions of agrin and aquaporin-4 (AQP4) were evaluated, as well as the integrity of BBB. Astrocyte polarity was assessed by visualizing the distribution of AQP4 on astrocyte end-feet membranes. The results showed that expression of AQP4 firstly increased and then decreased, as agrin expression decreased gradually. At 3 days after 2VO, AQP4 and agrin displayed the most opposite expression with the former increasing and the latter decreasing; at the same time, brain edema reached high point as well as BBB permeability, and astrocyte polarity was degeneration. In the later phase, brain edema and BBB permeability were getting recovered, but WML was getting more evident. In accordance with that, agrin and AQP4 expression decreased significantly with astrocyte polarity reducing. We speculated that agrin and AQP4 played key roles in development of WML by mediating BBB damage in CCH, and BBB dysfunction due to reduced astrocyte polarity is the starting point of WMH.
Collapse
|
33
|
Giannoni P, Badaut J, Dargazanli C, Fayd'Herbe De Maudave A, Klement W, Costalat V, Marchi N. The pericyte-glia interface at the blood-brain barrier. Clin Sci (Lond) 2018; 132:361-374. [PMID: 29439117 DOI: 10.1042/cs20171634] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/04/2018] [Accepted: 01/04/2018] [Indexed: 12/30/2022]
Abstract
The cerebrovasculature is a multicellular structure with varying rheological and permeability properties. The outer wall of the brain capillary endothelium is enclosed by pericytes and astrocyte end feet, anatomically assembled to guarantee barrier functions. We, here, focus on the pericyte modifications occurring in disease conditions, reviewing evidence supporting the interplay amongst pericytes, the endothelium, and glial cells in health and pathology. Deconstruction and reactivity of pericytes and glial cells around the capillary endothelium occur in response to traumatic brain injury, epilepsy, and neurodegenerative disorders, impacting vascular permeability and participating in neuroinflammation. As this represents a growing field of research, addressing the multicellular reorganization occurring at the outer wall of the blood-brain barrier (BBB) in response to an acute insult or a chronic disease could disclose novel disease mechanisms and therapeutic targets.
Collapse
Affiliation(s)
| | - Jerome Badaut
- Laboratory of Brain Molecular Imaging, CNRS UMR5287, University of Bordeaux, France
- Basic Science Departments, Loma Linda University School of Medicine, CA, U.S.A
| | - Cyril Dargazanli
- Neuroradiology, University Hospital, Montpellier, France
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Alexis Fayd'Herbe De Maudave
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Wendy Klement
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Vincent Costalat
- Neuroradiology, University Hospital, Montpellier, France
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Nicola Marchi
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| |
Collapse
|
34
|
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: 16.9] [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
|
35
|
Hainsworth AH, Minett T, Andoh J, Forster G, Bhide I, Barrick TR, Elderfield K, Jeevahan J, Markus HS, Bridges LR. Neuropathology of White Matter Lesions, Blood-Brain Barrier Dysfunction, and Dementia. Stroke 2017; 48:2799-2804. [PMID: 28855392 DOI: 10.1161/strokeaha.117.018101] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 07/28/2017] [Accepted: 08/01/2017] [Indexed: 01/17/2023]
Abstract
BACKGROUND AND PURPOSE We tested whether blood-brain barrier dysfunction in subcortical white matter is associated with white matter abnormalities or risk of clinical dementia in older people (n=126; mean age 86.4, SD: 7.7 years) in the MRC CFAS (Medical Research Council Cognitive Function and Ageing Study). METHODS Using digital pathology, we quantified blood-brain barrier dysfunction (defined by immunohistochemical labeling for the plasma marker fibrinogen). This was assessed within subcortical white matter tissue samples harvested from postmortem T2 magnetic resonance imaging (MRI)-detected white matter hyperintensities, from normal-appearing white matter (distant from coexistent MRI-defined hyperintensities), and from equivalent areas in MRI normal brains. Histopathologic lesions were defined using a marker for phagocytic microglia (CD68, clone PGM1). RESULTS Extent of fibrinogen labeling was not significantly associated with white matter abnormalities defined either by MRI (odds ratio, 0.90; 95% confidence interval, 0.79-1.03; P=0.130) or by histopathology (odds ratio, 0.93; 95% confidence interval, 0.77-1.12; P=0.452). Among participants with normal MRI (no detectable white matter hyperintensities), increased fibrinogen was significantly related to decreased risk of clinical dementia (odds ratio, 0.74; 95% confidence interval, 0.58-0.94; P=0.013). Among participants with histological lesions, increased fibrinogen was related to increased risk of dementia (odds ratio, 2.26; 95% confidence interval, 1.25-4.08; P=0.007). CONCLUSIONS Our data suggest that some degree of blood-brain barrier dysfunction is common in older people and that this may be related to clinical dementia risk, additional to standard MRI biomarkers.
Collapse
Affiliation(s)
- Atticus H Hainsworth
- From the Cell Biology and Genetics Research Centre (A.H.H., J.A., I.B., L.R.B.) and Neuroscience Research Centre (A.H.H., J.A., I.B., T.R.B., L.R.B.), Molecular and Clinical Sciences Research Institute, St George's University of London, United Kingdom; Department of Neurology (A.H.H.) and Department of Cellular Pathology (K.E., J.J., L.R.B.), St George's University Hospitals NHS Foundation Trust, London, United Kingdom; Department of Public Health and Primary Care (T.M.), Department of Radiology (T.M.), and Stroke Research Group, Department of Clinical Neurosciences (H.S.M.), University of Cambridge, United Kingdom; and The Sheffield Institute for Translational Neuroscience, University of Sheffield, United Kingdom (G.F.).
| | - Thais Minett
- From the Cell Biology and Genetics Research Centre (A.H.H., J.A., I.B., L.R.B.) and Neuroscience Research Centre (A.H.H., J.A., I.B., T.R.B., L.R.B.), Molecular and Clinical Sciences Research Institute, St George's University of London, United Kingdom; Department of Neurology (A.H.H.) and Department of Cellular Pathology (K.E., J.J., L.R.B.), St George's University Hospitals NHS Foundation Trust, London, United Kingdom; Department of Public Health and Primary Care (T.M.), Department of Radiology (T.M.), and Stroke Research Group, Department of Clinical Neurosciences (H.S.M.), University of Cambridge, United Kingdom; and The Sheffield Institute for Translational Neuroscience, University of Sheffield, United Kingdom (G.F.)
| | - Joycelyn Andoh
- From the Cell Biology and Genetics Research Centre (A.H.H., J.A., I.B., L.R.B.) and Neuroscience Research Centre (A.H.H., J.A., I.B., T.R.B., L.R.B.), Molecular and Clinical Sciences Research Institute, St George's University of London, United Kingdom; Department of Neurology (A.H.H.) and Department of Cellular Pathology (K.E., J.J., L.R.B.), St George's University Hospitals NHS Foundation Trust, London, United Kingdom; Department of Public Health and Primary Care (T.M.), Department of Radiology (T.M.), and Stroke Research Group, Department of Clinical Neurosciences (H.S.M.), University of Cambridge, United Kingdom; and The Sheffield Institute for Translational Neuroscience, University of Sheffield, United Kingdom (G.F.)
| | - Gillian Forster
- From the Cell Biology and Genetics Research Centre (A.H.H., J.A., I.B., L.R.B.) and Neuroscience Research Centre (A.H.H., J.A., I.B., T.R.B., L.R.B.), Molecular and Clinical Sciences Research Institute, St George's University of London, United Kingdom; Department of Neurology (A.H.H.) and Department of Cellular Pathology (K.E., J.J., L.R.B.), St George's University Hospitals NHS Foundation Trust, London, United Kingdom; Department of Public Health and Primary Care (T.M.), Department of Radiology (T.M.), and Stroke Research Group, Department of Clinical Neurosciences (H.S.M.), University of Cambridge, United Kingdom; and The Sheffield Institute for Translational Neuroscience, University of Sheffield, United Kingdom (G.F.)
| | - Ishaan Bhide
- From the Cell Biology and Genetics Research Centre (A.H.H., J.A., I.B., L.R.B.) and Neuroscience Research Centre (A.H.H., J.A., I.B., T.R.B., L.R.B.), Molecular and Clinical Sciences Research Institute, St George's University of London, United Kingdom; Department of Neurology (A.H.H.) and Department of Cellular Pathology (K.E., J.J., L.R.B.), St George's University Hospitals NHS Foundation Trust, London, United Kingdom; Department of Public Health and Primary Care (T.M.), Department of Radiology (T.M.), and Stroke Research Group, Department of Clinical Neurosciences (H.S.M.), University of Cambridge, United Kingdom; and The Sheffield Institute for Translational Neuroscience, University of Sheffield, United Kingdom (G.F.)
| | - Thomas R Barrick
- From the Cell Biology and Genetics Research Centre (A.H.H., J.A., I.B., L.R.B.) and Neuroscience Research Centre (A.H.H., J.A., I.B., T.R.B., L.R.B.), Molecular and Clinical Sciences Research Institute, St George's University of London, United Kingdom; Department of Neurology (A.H.H.) and Department of Cellular Pathology (K.E., J.J., L.R.B.), St George's University Hospitals NHS Foundation Trust, London, United Kingdom; Department of Public Health and Primary Care (T.M.), Department of Radiology (T.M.), and Stroke Research Group, Department of Clinical Neurosciences (H.S.M.), University of Cambridge, United Kingdom; and The Sheffield Institute for Translational Neuroscience, University of Sheffield, United Kingdom (G.F.)
| | - Kay Elderfield
- From the Cell Biology and Genetics Research Centre (A.H.H., J.A., I.B., L.R.B.) and Neuroscience Research Centre (A.H.H., J.A., I.B., T.R.B., L.R.B.), Molecular and Clinical Sciences Research Institute, St George's University of London, United Kingdom; Department of Neurology (A.H.H.) and Department of Cellular Pathology (K.E., J.J., L.R.B.), St George's University Hospitals NHS Foundation Trust, London, United Kingdom; Department of Public Health and Primary Care (T.M.), Department of Radiology (T.M.), and Stroke Research Group, Department of Clinical Neurosciences (H.S.M.), University of Cambridge, United Kingdom; and The Sheffield Institute for Translational Neuroscience, University of Sheffield, United Kingdom (G.F.)
| | - Jamuna Jeevahan
- From the Cell Biology and Genetics Research Centre (A.H.H., J.A., I.B., L.R.B.) and Neuroscience Research Centre (A.H.H., J.A., I.B., T.R.B., L.R.B.), Molecular and Clinical Sciences Research Institute, St George's University of London, United Kingdom; Department of Neurology (A.H.H.) and Department of Cellular Pathology (K.E., J.J., L.R.B.), St George's University Hospitals NHS Foundation Trust, London, United Kingdom; Department of Public Health and Primary Care (T.M.), Department of Radiology (T.M.), and Stroke Research Group, Department of Clinical Neurosciences (H.S.M.), University of Cambridge, United Kingdom; and The Sheffield Institute for Translational Neuroscience, University of Sheffield, United Kingdom (G.F.)
| | - Hugh S Markus
- From the Cell Biology and Genetics Research Centre (A.H.H., J.A., I.B., L.R.B.) and Neuroscience Research Centre (A.H.H., J.A., I.B., T.R.B., L.R.B.), Molecular and Clinical Sciences Research Institute, St George's University of London, United Kingdom; Department of Neurology (A.H.H.) and Department of Cellular Pathology (K.E., J.J., L.R.B.), St George's University Hospitals NHS Foundation Trust, London, United Kingdom; Department of Public Health and Primary Care (T.M.), Department of Radiology (T.M.), and Stroke Research Group, Department of Clinical Neurosciences (H.S.M.), University of Cambridge, United Kingdom; and The Sheffield Institute for Translational Neuroscience, University of Sheffield, United Kingdom (G.F.)
| | - Leslie R Bridges
- From the Cell Biology and Genetics Research Centre (A.H.H., J.A., I.B., L.R.B.) and Neuroscience Research Centre (A.H.H., J.A., I.B., T.R.B., L.R.B.), Molecular and Clinical Sciences Research Institute, St George's University of London, United Kingdom; Department of Neurology (A.H.H.) and Department of Cellular Pathology (K.E., J.J., L.R.B.), St George's University Hospitals NHS Foundation Trust, London, United Kingdom; Department of Public Health and Primary Care (T.M.), Department of Radiology (T.M.), and Stroke Research Group, Department of Clinical Neurosciences (H.S.M.), University of Cambridge, United Kingdom; and The Sheffield Institute for Translational Neuroscience, University of Sheffield, United Kingdom (G.F.)
| |
Collapse
|
36
|
Valdés Hernández MDC, González-Castro V, Chappell FM, Sakka E, Makin S, Armitage PA, Nailon WH, Wardlaw JM. Application of Texture Analysis to Study Small Vessel Disease and Blood-Brain Barrier Integrity. Front Neurol 2017; 8:327. [PMID: 28769863 PMCID: PMC5515862 DOI: 10.3389/fneur.2017.00327] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 06/22/2017] [Indexed: 11/13/2022] Open
Abstract
Objectives We evaluate the alternative use of texture analysis for evaluating the role of blood–brain barrier (BBB) in small vessel disease (SVD). Methods We used brain magnetic resonance imaging from 204 stroke patients, acquired before and 20 min after intravenous gadolinium administration. We segmented tissues, white matter hyperintensities (WMH) and applied validated visual scores. We measured textural features in all tissues pre- and post-contrast and used ANCOVA to evaluate the effect of SVD indicators on the pre-/post-contrast change, Kruskal–Wallis for significance between patient groups and linear mixed models for pre-/post-contrast variations in cerebrospinal fluid (CSF) with Fazekas scores. Results Textural “homogeneity” increase in normal tissues with higher presence of SVD indicators was consistently more overt than in abnormal tissues. Textural “homogeneity” increased with age, basal ganglia perivascular spaces scores (p < 0.01) and SVD scores (p < 0.05) and was significantly higher in hypertensive patients (p < 0.002) and lacunar stroke (p = 0.04). Hypertension (74% patients), WMH load (median = 1.5 ± 1.6% of intracranial volume), and age (mean = 65.6 years, SD = 11.3) predicted the pre/post-contrast change in normal white matter, WMH, and index stroke lesion. CSF signal increased with increasing SVD post-contrast. Conclusion A consistent general pattern of increasing textural “homogeneity” with increasing SVD and post-contrast change in CSF with increasing WMH suggest that texture analysis may be useful for the study of BBB integrity.
Collapse
Affiliation(s)
- Maria Del C Valdés Hernández
- Department of Neuroimaging Sciences, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Victor González-Castro
- Department of Neuroimaging Sciences, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Francesca M Chappell
- Department of Neuroimaging Sciences, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Eleni Sakka
- Department of Neuroimaging Sciences, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Stephen Makin
- Department of Neuroimaging Sciences, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Paul A Armitage
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - William H Nailon
- Department of Oncology Physics, University of Edinburgh, Edinburgh, United Kingdom
| | - Joanna M Wardlaw
- Department of Neuroimaging Sciences, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
37
|
Goodall EF, Wang C, Simpson JE, Baker DJ, Drew DR, Heath PR, Saffrey MJ, Romero IA, Wharton SB. Age-associated changes in the blood-brain barrier: comparative studies in human and mouse. Neuropathol Appl Neurobiol 2017; 44:328-340. [PMID: 28453876 PMCID: PMC5900918 DOI: 10.1111/nan.12408] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 04/24/2017] [Accepted: 04/28/2017] [Indexed: 12/22/2022]
Abstract
Aims While vascular pathology is a common feature of a range of neurodegenerative diseases, we hypothesized that vascular changes occur in association with normal ageing. Therefore, we aimed to characterize age‐associated changes in the blood–brain barrier (BBB) in human and mouse cohorts. Methods Immunohistochemistry and Evans blue assays were used to characterize BBB dysfunction (tight junction protein expression and serum plasma protein accumulation), vascular pathology (pericyte loss and vascular density) and glial pathology (astrocyte and microglial density) in ageing neurological control human prefrontal cortex (a total of 23 cases from 5 age groups representing the spectrum of young adult to old age: 20–30 years, 31–45 years, 46–60 years, 61–75 years and 75+) and C57BL/6 mice (3 months, 12 months, 18 months and 24 months, n = 5/6 per group). Results Quantification of the tight junction protein ZO‐1 within the cortex and cerebellum of the mouse cohort showed a significant trend to both increased number (cortex P < 0.001, cerebellum P < 0.001) and length (cortex P < 0.001, cerebellum P < 0.001) of junctional breaks associated with increasing age. GFAP expression significantly correlated with ageing in the mice (P = 0.037). In the human cohort, assessment of human protein accumulation (albumin, fibrinogen and human IgG) demonstrated cells morphologically resembling clasmatodendritic astrocytes, indicative of BBB dysfunction. Semiquantitative assessment of astrogliosis in the cortex expression revealed an association with age (P = 0.003), while no age‐associated changes in microglial pathology, microvascular density or pericyte coverage were detected. Conclusions This study demonstrates BBB dysfunction in normal brain ageing, both in human and mouse cohorts.
Collapse
Affiliation(s)
- E F Goodall
- Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield, UK
| | - C Wang
- School of Life Science, Health and Chemical Sciences, Faculty of Science, The Open University, Milton Keynes, UK
| | - J E Simpson
- Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield, UK
| | - D J Baker
- Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield, UK
| | - D R Drew
- Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield, UK
| | - P R Heath
- Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield, UK
| | - M J Saffrey
- School of Life Science, Health and Chemical Sciences, Faculty of Science, The Open University, Milton Keynes, UK
| | - I A Romero
- School of Life Science, Health and Chemical Sciences, Faculty of Science, The Open University, Milton Keynes, UK
| | - S B Wharton
- Sheffield Institute for Translational Neuroscience, The University of Sheffield, Sheffield, UK
| |
Collapse
|
38
|
Srinivasan V, Braidy N, Chan EKW, Xu YH, Chan DKY. Genetic and environmental factors in vascular dementia: an update of blood brain barrier dysfunction. Clin Exp Pharmacol Physiol 2016; 43:515-21. [PMID: 26859837 DOI: 10.1111/1440-1681.12558] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 02/01/2016] [Accepted: 02/01/2016] [Indexed: 11/29/2022]
Abstract
Vascular dementia (VaD) describes a combination of both cognitive and behavioural manifestations associated with variable brain lesions of vascular origin. While vascular risk factors have been implicated in VaD, the relationship is most evident when the factors are considered together and not individually. This review will examine the significance of the integrity of blood brain barrier (BBB) tight junction (TJ) proteins - occludin and claudins in the pathophysiology of VaD. Specifically, some of the genetic contributors to VaD, namely those responsible for the integrity of the BBB, will be reviewed in detail. Moreover, environmental factors will be considered in conjunction with these genes to examine how the interaction of environmental and genetic factors contributes to one's susceptibility to VaD.
Collapse
Affiliation(s)
- Vivek Srinivasan
- Department of Aged Care and Rehabilitation, Bankstown-Lidcombe Hospital, Bankstown, Australia.,Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Nady Braidy
- Department of Aged Care and Rehabilitation, Bankstown-Lidcombe Hospital, Bankstown, Australia.,Ingham Institute, Liverpool, Australia.,School of Psychiatry, Centre for Healthy Brain Ageing, Sydney, Australia
| | - Eunice K W Chan
- Faculty of Medicine, Western Sydney University, Macarthur, Australia
| | - Ying-Hua Xu
- Department of Aged Care and Rehabilitation, Bankstown-Lidcombe Hospital, Bankstown, Australia.,Faculty of Medicine, University of New South Wales, Sydney, Australia.,Ingham Institute, Liverpool, Australia
| | - Daniel K Y Chan
- Department of Aged Care and Rehabilitation, Bankstown-Lidcombe Hospital, Bankstown, Australia.,Faculty of Medicine, University of New South Wales, Sydney, Australia.,Ingham Institute, Liverpool, Australia
| |
Collapse
|
39
|
Vascular Contributions to Cognitive Impairment and Treatments with Traditional Chinese Medicine. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:9627258. [PMID: 28042305 PMCID: PMC5141557 DOI: 10.1155/2016/9627258] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 10/12/2016] [Accepted: 10/18/2016] [Indexed: 12/21/2022]
Abstract
The prevalence of cognitive impairment and dementia caused by cerebrovascular disease is likely to increase with the global aging population. Vascular contributions to cognitive impairment and dementia (VCID) is a wide spectrum term used to include a diverse heterogeneous group of cognitive syndromes with vascular factors regardless of the cause of pathogenesis. VCID ranges from mild cognitive impairment to full-blown dementia with vascular dementia (VaD) as the most severe stage. It is further complexed by the coexistence of other forms of dementia such as Alzheimer's disease (AD). Recent researches in the functions of the neurovascular unit (NVU) suggest that dysfunction of the NVU might be the cause of primary vascular events in the brain that leads to further neurodegeneration. In this review, we have briefly summarized various forms of VCID. There is currently no standard therapy for VCID or dementia. Given the fact that Traditional Chinese Medicine (TCM) has gained popularity worldwide, we also reviewed recent scientific and clinical findings on various antidementia TCM for the treatment of VCID, including Salvia miltiorrhiza, Huperzia serrata, Ligusticum chuanxiong, Ginkgo biloba, Panax ginseng, and also TCM formula Sailuotong capsule (SLT) and Fufangdanshen tablets (FFDS).
Collapse
|
40
|
Mollenhauer B, Parnetti L, Rektorova I, Kramberger MG, Pikkarainen M, Schulz-Schaeffer WJ, Aarsland D, Svenningsson P, Farotti L, Verbeek MM, Schlossmacher MG. Biological confounders for the values of cerebrospinal fluid proteins in Parkinson's disease and related disorders. J Neurochem 2016; 139 Suppl 1:290-317. [DOI: 10.1111/jnc.13390] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 09/11/2015] [Accepted: 09/21/2015] [Indexed: 12/26/2022]
Affiliation(s)
- Brit Mollenhauer
- Paracelsus-Elena-Klinik; Kassel Germany
- University Medical Center (Department of Neuropathology); Georg-August University Goettingen; Goettingen Germany
| | - Lucilla Parnetti
- Centro Disturbi della Memoria- Unità Valutativa Alzheimer; Clinica Neurologica; Università di Perugia; Perugia Italy
| | - Irena Rektorova
- Applied Neuroscience Group; CEITEC MU; Masaryk University; Brno Czech Republic
| | - Milica G. Kramberger
- Department of Neurology; University Medical Center Ljubljana; Ljubljana Slovenia
- Division for Neurogeriatrics; Department of NVS; Karolinska Institutet; Center for Alzheimer Research; Stockholm Sweden
- Centre for Age-Related Medicine; Stavanger University Hospital; Stavanger Norway
| | - Maria Pikkarainen
- Institute of Clinical Medicine / Neurology; University of Eastern Finland; Kuopio Finland
| | - Walter J. Schulz-Schaeffer
- University Medical Center (Department of Neuropathology); Georg-August University Goettingen; Goettingen Germany
| | - Dag Aarsland
- Division for Neurogeriatrics; Department of NVS; Karolinska Institutet; Center for Alzheimer Research; Stockholm Sweden
- Centre for Age-Related Medicine; Stavanger University Hospital; Stavanger Norway
| | - Per Svenningsson
- Department for Clinical Neuroscience; Karolinska Institute; Stockholm Sweden
| | - Lucia Farotti
- Centro Disturbi della Memoria- Unità Valutativa Alzheimer; Clinica Neurologica; Università di Perugia; Perugia Italy
| | - Marcel M. Verbeek
- Department of Neurology; Department of Laboratory Medicine; Donders Institute for Brain, Cognition and Behaviour; Radboud University Medical Centre; Nijmegen The Netherlands
| | - Michael G. Schlossmacher
- Program in Neuroscience and Division of Neurology; The Ottawa Hospital; University of Ottawa Brain & Mind Research Institute; Ottawa Ontario Canada
| |
Collapse
|
41
|
Tight junction disruption of blood–brain barrier in white matter lesions in chronic hypertensive rats. Neuroreport 2015; 26:1039-43. [DOI: 10.1097/wnr.0000000000000464] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
42
|
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: 76] [Impact Index Per Article: 7.6] [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
|
43
|
Wardlaw JM, Valdés Hernández MC, Muñoz-Maniega S. What are white matter hyperintensities made of? Relevance to vascular cognitive impairment. J Am Heart Assoc 2015; 4:001140. [PMID: 26104658 PMCID: PMC4599520 DOI: 10.1161/jaha.114.001140] [Citation(s) in RCA: 603] [Impact Index Per Article: 60.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Joanna M Wardlaw
- Division of Neuroimaging Sciences and Brain Research Imaging Centre, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom (J.M.W., M.C.V.H., S.M.M.)
| | - Maria C Valdés Hernández
- Division of Neuroimaging Sciences and Brain Research Imaging Centre, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom (J.M.W., M.C.V.H., S.M.M.)
| | - Susana Muñoz-Maniega
- Division of Neuroimaging Sciences and Brain Research Imaging Centre, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom (J.M.W., M.C.V.H., S.M.M.)
| |
Collapse
|
44
|
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: 62] [Impact Index Per Article: 5.6] [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
|
45
|
Kim Y, Kim YK, Kim NK, Kim SH, Kim OJ, Oh SH. Circulating matrix metalloproteinase-9 level is associated with cerebral white matter hyperintensities in non-stroke individuals. Eur Neurol 2014; 72:234-40. [PMID: 25248031 DOI: 10.1159/000362876] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 04/04/2014] [Indexed: 11/19/2022]
Abstract
BACKGROUNDS The pathogenesis of cerebral white matter hyperintensities (WMH) has been poorly understood. Our aim was to investigate the association of circulating proteins, the biomarkers of inflammation, blood-brain barrier (BBB) dysfunction, and thrombosis with WMH in non-stroke individuals. METHODS Demographic, laboratory, and brain magnetic resonance imaging parameters were prospectively analyzed in 137 subjects. The relationship between plasma interleukin-6, tumor necrosis factor-α, matrx-metalloproteinase-9 (MMP-9), plasminogen activator inhibitor-1 and overt WMH (Fazekas grading score ≥2) was analyzed. RESULTS In univariate analysis, old age, high blood pressure, history of hypertension, and elevated plasma MMP-9 level were associated with overt WMH. In multivariate analysis, plasma MMP-9 still maintained a significant association with WMH. Plasma MMP-9 level was weakly but significantly associated with WMH volume (r = 0.232, p = 0.006). All the other circulating proteins examined failed to demonstrate a significant relationship with WMH. CONCLUSIONS Plasma MMP-9 is associated with pathophysiology of WMH development.
Collapse
Affiliation(s)
- Yoon Kim
- Department of Neurology, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea
| | | | | | | | | | | |
Collapse
|
46
|
Oakley R, Tharakan B. Vascular hyperpermeability and aging. Aging Dis 2014; 5:114-25. [PMID: 24729937 DOI: 10.14336/ad.2014.0500114] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 01/31/2014] [Accepted: 02/09/2014] [Indexed: 12/25/2022] Open
Abstract
Vascular hyperpermeability, the excessive leakage of fluid and proteins from blood vessels to the interstitial space, commonly occurs in traumatic and ischemic injuries. This hyperpermeability causes tissue vasogenic edema, which often leads to multiple organ failure resulting in patient death. Vascular hyperpermeability occurs most readily in small blood vessels as their more delicate physical constitution makes them an easy target for barrier dysfunction. A single layer of endothelial cells, linked to one another by cell adhesion molecules, covers the interior surface of each blood vessel. The cell adhesion molecules play a key role in maintaining barrier functions like the regulation of permeability. Aging is a major risk factor for microvascular dysfunction and hyperpermeability. Apart from age-related remodeling of the vascular wall, endothelial barrier integrity and function declines with the advancement of age. Studies that address the physiological and molecular basis of vascular permeability regulation in aging are currently very limited. There have been many cellular and molecular mechanisms proposed to explain aging-related endothelial dysfunction but their true relationship to barrier dysfunction and hyperpermeability is not clearly known. Among the several mechanisms that promote vascular dysfunction and hyperpermeability, the following are considered major contributors: oxidative stress, inflammation, and the activation of apoptotic signaling pathways. In this review we highlighted (a) the physiological, cellular and molecular changes that occur in the vascular system as a product of aging; (b) the potential mechanisms by which aging leads to barrier dysfunction and vascular hyperpermeability in the peripheral and the blood-brain barrier; (c) the mechanisms by which the age-related increases in oxidative stress, inflammatory markers and apoptotic signaling etc. cause endothelial dysfunction and their relationship to hyperpermeability; and (d) the relationship between aging, vascular permeability and traumatic injuries.
Collapse
Affiliation(s)
| | - Binu Tharakan
- Department of Surgery, Texas A&M University Health Science Center College of Medicine & Baylor Scott & White Healthcare, Temple, Texas, USA
| |
Collapse
|
47
|
Chung SJ, Kim JH, Cho JH, Kim GS, Choi SA, Lee PH, Lee JH. Subcortical vascular dementia (SVaD) without hypertension (HTN) may be a unique subtype of vascular dementia (VaD). Arch Gerontol Geriatr 2014; 58:231-5. [DOI: 10.1016/j.archger.2013.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 10/15/2013] [Accepted: 10/21/2013] [Indexed: 10/26/2022]
|
48
|
Abstract
The term cerebral small vessel disease (SVD) describes a range of neuroimaging, pathological, and associated clinical features. Clinical features range from none, to discrete focal neurological symptoms (eg, stroke), to insidious global neurological dysfunction and dementia. The burden on public health is substantial. The pathogenesis of SVD is largely unknown. Although the pathological processes leading to the arteriolar disease are associated with vascular risk factors and are believed to result from an intrinsic cerebral arteriolar occlusive disease, little is known about how these processes result in brain disease, how SVD lesions contribute to neurological or cognitive symptoms, and the association with risk factors. Pathology often shows end-stage disease, which makes identification of the earliest stages difficult. Neuroimaging provides considerable insights; although the small vessels are not easily seen themselves, the effects of their malfunction on the brain can be tracked with detailed brain imaging. We discuss potential mechanisms, detectable with neuroimaging, that might better fit the available evidence and provide testable hypotheses for future study.
Collapse
|
49
|
Species-Dependent Blood-Brain Barrier Disruption of Lipopolysaccharide: Amelioration by Colistin In Vitro and In Vivo. Antimicrob Agents Chemother 2013; 57:4336-4342. [PMID: 23796941 DOI: 10.1128/aac.00765-13] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 06/20/2013] [Indexed: 11/20/2022] Open
Abstract
The aim of this study was to use in vitro and in vivo models to assess the impact of lipopolysaccharide (LPS) from two different bacterial species on blood-brain barrier (BBB) integrity and brain uptake of colistin. Following repeated administration of LPS from Pseudomonas aeruginosa, the brain-to-plasma ratio of [14C]sucrose in Swiss outbred mice was not significantly increased. Furthermore, while the brain uptake of colistin in mice increased 3-fold following administration of LPS from Salmonella enterica, LPS from P. aeruginosa had no significant effect on colistin brain uptake. This apparent species-dependent effect did not appear to correlate with differences in plasma cytokine levels, as the concentrations of tumor necrosis factor alpha and interleukin-6 following administration of each LPS were not different (P > 0.05). To clarify whether this species-specific effect of LPS was due to direct effects on the BBB, human brain capillary endothelial (hCMEC/D3) cells were treated with LPS from P. aeruginosa or S. enterica and claudin-5 expression was measured by Western blotting. S. enterica LPS significantly (P < 0.05) reduced claudin-5 expression at a concentration of 7.5 μg/ml. In contrast, P. aeruginosa LPS decreased (P < 0.05) claudin-5 expression only at the highest concentration tested (i.e., 30 μg/ml). Coadministration of therapeutic concentrations of colistin ameliorated the S. enterica LPS-induced reduction in claudin-5 expression in hCMEC/D3 cells and the perturbation in BBB function in mice. This study demonstrates that BBB disruption induced by LPS is species dependent, at least between P. aeruginosa and S. enterica, and can be ameliorated by colistin.
Collapse
|
50
|
Triggers and effectors of oxidative stress at blood-brain barrier level: relevance for brain ageing and neurodegeneration. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:297512. [PMID: 23533687 PMCID: PMC3606793 DOI: 10.1155/2013/297512] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 01/27/2013] [Accepted: 01/31/2013] [Indexed: 01/23/2023]
Abstract
As fundamental research advances, it is becoming increasingly clear that a clinically expressed disease implies a mixture of intertwining molecular disturbances. Oxidative stress is one of such pathogenic pathways involved in virtually all central nervous system pathologies, infectious, inflammatory, or degenerative in nature. Since brain homeostasis largely depends on integrity of blood-brain barrier (BBB), many studies focused lately on BBB alteration in a wide spectrum of brain diseases. The proper two-way molecular transfer through BBB depends on several factors, including the functional status of its tight junction (TJ) complexes of proteins sealing neighbour endothelial cells. Although there is abundant experimental work showing that oxidative stress associates BBB permeability alteration, less is known about its implications, at molecular level, in TJ protein expression or TJ-related cell signalling. In this paper, oxidative stress is presented as a common pathway for different brain pathogenic mechanisms which lead to BBB dysregulation. We revise here oxidative-induced molecular mechanisms of BBB disruption and TJ protein expression alteration, in relation to ageing and neurodegeneration.
Collapse
|