1
|
Serdar M, Walther KA, Gallert M, Kempe K, Obst S, Labusek N, Herrmann R, Herz J, Felderhoff-Müser U, Bendix I. Prenatal inflammation exacerbates hyperoxia-induced neonatal brain injury. J Neuroinflammation 2025; 22:57. [PMID: 40022130 PMCID: PMC11871844 DOI: 10.1186/s12974-025-03389-4] [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] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 02/20/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Premature born infants are at high risk to develop white matter injury (WMI). Hyperoxia and perinatal inflammation are main risk factors for preterm birth and associated brain injury. To date the majority of experimental studies have focused on isolated insults. However, clinically, WMI injury is a multifactorial disorder caused by a variety of triggers. To establish a clinically relevant rodent model of WMI, we combined prenatal inflammation with postnatal hyperoxia to investigate individual, and additive or synergistic effects on inflammatory processes, myelination and grey matter development. METHODS At embryonic day 20, pregnant Wistar rat dams received either a single intraperitoneal injection of 100 µg/ kg lipopolysaccharide (LPS) or sodium chloride. Offspring were either exposed to hyperoxia (80% O2) or normoxia (21% O2) from postnatal day 3 to 5. Animals were sacrificed immediately after hyperoxia or 6 days later, corresponding to term-equivalent age. White and grey matter development and neuroinflammatory responses were investigated at cellular and molecular levels applying immunohistochemistry, western blotting, real time PCR in brain tissues and multiplex protein expression analysis on serum samples. RESULTS Prenatal inflammation combined with postnatal hyperoxia resulted in reduced body weight and length in the offspring, accompanied by increased serum leptin levels at term equivalent age. The altered body parameters, like body weight, were associated with decreased brain volume, thinning of deep cortical layers and hypomyelination. As potential underlying mechanisms, we identified severe myelination deficits and an increased microglia activation associated with elevated inflammatory cytokine expression in brain tissues, while peripheral cytokine levels were reduced. Interestingly, effects on body size were mainly mediated by prenatal LPS, independent of hyperoxia, while oligodendrocyte degeneration was mainly induced by postnatal hyperoxia, independent of prenatal inflammation. However, for the majority of pathological changes, including brain size, myelination deficits, microglia activation and inflammatory cytokine expression, additive or synergistic effects were detected. CONCLUSION Prenatal inflammation combined with postnatal hyperoxia results in aggravated myelination deficits and inflammatory responses compared to single insults, making it an ideal model to improve our understanding of the complex pathophysiology underlying WMI and to evaluate urgently needed therapies.
Collapse
Affiliation(s)
- Meray Serdar
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Kay-Anja Walther
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Markus Gallert
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Karina Kempe
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Stefanie Obst
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Nicole Labusek
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Ralf Herrmann
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Josephine Herz
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Ursula Felderhoff-Müser
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany.
| | - Ivo Bendix
- Department of Paediatrics I, Neonatology and Experimental perinatal Neurosciences, Centre for Translational Neuro- and Behavioural Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany.
| |
Collapse
|
2
|
Cheng Y, Huang P, Lin L, Zhang J, Cheng Y, Zheng J, Wang Y, Pan X. Abnormal brain-heart electrophysiology in mild and severe orthostatic hypotension. J Hypertens 2024; 42:2094-2106. [PMID: 39207017 DOI: 10.1097/hjh.0000000000003838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION This study investigated the changes in cardiocerebral electrophysiology in patients with mild orthostatic hypotension (MOH) and severe orthostatic hypotension (SOH) and their relationship with the severity of orthostatic hypotension, psychiatric symptoms, and cognitive dysfunction. METHODS This study included 72 nonorthostatic hypotension (NOH), 17 with MOH, and 11 with SOH. Seated resting-state heart rate variability (HRV) and quantitative electroencephalogram parameters were synchronized and recorded. HRV measures in the time and frequency domains were analyzed, along with the peak frequency and power of the brain waves. RESULTS Abnormal neuronal activity was found in FP1 in patients with MOH, whereas it was more widespread in FP1, FP2, and O2 in patients with SOH ( P < 0.05). Cardiac and cerebral electrophysiological abnormalities were significantly associated with orthostatic hypotension severity, psychiatric symptoms, and cognitive dysfunction. CONCLUSION Abnormal EEG activity in patients are mainly manifested in the prefrontal and occipital lobes, especially in patients with SOH. These results may help patients to better understand the mechanisms underlying orthostatic hypotension severity and psychiatric and cognitive impairment in orthostatic hypotension.
Collapse
Affiliation(s)
- Yingzhe Cheng
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital
- Institute of Clinical Neurology
- Four Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou City
| | - Peilin Huang
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital
- Institute of Clinical Neurology
- Four Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou City
| | - Lin Lin
- Department of Neurology, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Jiejun Zhang
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital
- Institute of Clinical Neurology
- Four Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou City
- Center for Geriatrics, Hainan General Hospital, Hainan Province
| | - Yahui Cheng
- Shandong Second Medical University, Weifang City
| | - Jiahao Zheng
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital
- Institute of Clinical Neurology
- Four Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou City
| | - Yanping Wang
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou
| | - Xiaodong Pan
- Department of Neurology, Center for Cognitive Neurology, Fujian Medical University Union Hospital
- Fujian Institute of Geriatrics, Fujian Medical University Union Hospital
- Institute of Clinical Neurology
- Four Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou City
| |
Collapse
|
3
|
Korte N, Barkaway A, Wells J, Freitas F, Sethi H, Andrews SP, Skidmore J, Stevens B, Attwell D. Inhibiting Ca 2+ channels in Alzheimer's disease model mice relaxes pericytes, improves cerebral blood flow and reduces immune cell stalling and hypoxia. Nat Neurosci 2024; 27:2086-2100. [PMID: 39294491 PMCID: PMC11537984 DOI: 10.1038/s41593-024-01753-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 08/06/2024] [Indexed: 09/20/2024]
Abstract
Early in Alzheimer's disease (AD), pericytes constrict capillaries, increasing their hydraulic resistance and trapping of immune cells and, thus, decreasing cerebral blood flow (CBF). Therapeutic approaches to attenuate pericyte-mediated constriction in AD are lacking. Here, using in vivo two-photon imaging with laser Doppler and speckle flowmetry and magnetic resonance imaging, we show that Ca2+ entry via L-type voltage-gated calcium channels (CaVs) controls the contractile tone of pericytes. In AD model mice, we identifed pericytes throughout the capillary bed as key drivers of an immune reactive oxygen species (ROS)-evoked and pericyte intracellular calcium concentration ([Ca2+]i)-mediated decrease in microvascular flow. Blocking CaVs with nimodipine early in disease progression improved CBF, reduced leukocyte stalling at pericyte somata and attenuated brain hypoxia. Amyloid β (Aβ)-evoked pericyte contraction in human cortical tissue was also greatly reduced by CaV block. Lowering pericyte [Ca2+]i early in AD may, thus, offer a therapeutic strategy to enhance brain energy supply and possibly cognitive function in AD.
Collapse
Affiliation(s)
- Nils Korte
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, UK.
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Anna Barkaway
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, UK
| | - Jack Wells
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, UK
| | - Felipe Freitas
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, UK
| | - Huma Sethi
- Department of Neurosurgery, National Hospital for Neurology and Neurosurgery, London, UK
| | - Stephen P Andrews
- ALBORADA Drug Discovery Institute, University of Cambridge, Cambridge, UK
| | - John Skidmore
- ALBORADA Drug Discovery Institute, University of Cambridge, Cambridge, UK
| | - Beth Stevens
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Stanley Center, Broad Institute, Cambridge, MA, USA
| | - David Attwell
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, UK.
| |
Collapse
|
4
|
Askew KE, Beverley J, Sigfridsson E, Szymkowiak S, Emelianova K, Dando O, Hardingham GE, Duncombe J, Hennessy E, Koudelka J, Samarasekera N, Salman RA, Smith C, Tavares AAS, Gomez‐Nicola D, Kalaria RN, McColl BW, Horsburgh K. Inhibiting CSF1R alleviates cerebrovascular white matter disease and cognitive impairment. Glia 2024; 72:375-395. [PMID: 37909242 PMCID: PMC10952452 DOI: 10.1002/glia.24481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 09/27/2023] [Accepted: 10/04/2023] [Indexed: 11/02/2023]
Abstract
White matter abnormalities, related to poor cerebral perfusion, are a core feature of small vessel cerebrovascular disease, and critical determinants of vascular cognitive impairment and dementia. Despite this importance there is a lack of treatment options. Proliferation of microglia producing an expanded, reactive population and associated neuroinflammatory alterations have been implicated in the onset and progression of cerebrovascular white matter disease, in patients and in animal models, suggesting that targeting microglial proliferation may exert protection. Colony-stimulating factor-1 receptor (CSF1R) is a key regulator of microglial proliferation. We found that the expression of CSF1R/Csf1r and other markers indicative of increased microglial abundance are significantly elevated in damaged white matter in human cerebrovascular disease and in a clinically relevant mouse model of chronic cerebral hypoperfusion and vascular cognitive impairment. Using the mouse model, we investigated long-term pharmacological CSF1R inhibition, via GW2580, and demonstrated that the expansion of microglial numbers in chronic hypoperfused white matter is prevented. Transcriptomic analysis of hypoperfused white matter tissue showed enrichment of microglial and inflammatory gene sets, including phagocytic genes that were the predominant expression modules modified by CSF1R inhibition. Further, CSF1R inhibition attenuated hypoperfusion-induced white matter pathology and rescued spatial learning impairments and to a lesser extent cognitive flexibility. Overall, this work suggests that inhibition of CSF1R and microglial proliferation mediates protection against chronic cerebrovascular white matter pathology and cognitive deficits. Our study nominates CSF1R as a target for the treatment of vascular cognitive disorders with broader implications for treatment of other chronic white matter diseases.
Collapse
Affiliation(s)
| | - Joshua Beverley
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | - Emma Sigfridsson
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | - Stefan Szymkowiak
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
- UK Dementia Research InstituteUniversity of EdinburghEdinburghUK
| | - Katherine Emelianova
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
- UK Dementia Research InstituteUniversity of EdinburghEdinburghUK
| | - Owen Dando
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
- UK Dementia Research InstituteUniversity of EdinburghEdinburghUK
| | - Giles E. Hardingham
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
- UK Dementia Research InstituteUniversity of EdinburghEdinburghUK
| | - Jessica Duncombe
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | - Edel Hennessy
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| | - Juraj Koudelka
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
- UK Dementia Research InstituteUniversity of EdinburghEdinburghUK
| | - Neshika Samarasekera
- Centre for Clinical Brain Sciences and Sudden Death Brain BankUniversity of EdinburghEdinburghUK
| | - Rustam Al‐Shahi Salman
- Centre for Clinical Brain Sciences and Sudden Death Brain BankUniversity of EdinburghEdinburghUK
| | - Colin Smith
- Centre for Clinical Brain Sciences and Sudden Death Brain BankUniversity of EdinburghEdinburghUK
| | - Adriana A. S. Tavares
- British Heart Foundation Centre for Cardiovascular ScienceUniversity of EdinburghEdinburghUK
| | | | - Raj N. Kalaria
- Clinical and Translational Research InstituteNewcastle UniversityNewcastleUK
| | - Barry W. McColl
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
- UK Dementia Research InstituteUniversity of EdinburghEdinburghUK
| | - Karen Horsburgh
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUK
| |
Collapse
|
5
|
Cheng Y, Lin L, Jiang S, Huang P, Zhang J, Xin J, Xu H, Wang Y, Pan X. Aberrant microstructural integrity of white matter in mild and severe orthostatic hypotension: A NODDI study. CNS Neurosci Ther 2024; 30:e14586. [PMID: 38421091 PMCID: PMC10851318 DOI: 10.1111/cns.14586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 12/11/2023] [Accepted: 12/20/2023] [Indexed: 03/02/2024] Open
Abstract
OBJECTIVE Scarce evidence is available to elucidate the association between the abnormal microstructure of white matter (WM) and cognitive performance in patients with orthostatic hypotension (OH). This study investigated the microstructural integrity of WM in patients with mild OH (MOH) and severe OH (SOH) and evaluated the association of abnormal WM microstructure with the broad cognitive domains and cognition-related plasma biomarkers. METHODS Our study included 72 non-OH (NOH), 17 MOH, and 11 SOH participants. Across the groups, the WM integrity was analyzed by neurite orientation dispersion and density imaging (NODDI), and differences in WM microstructure were evaluated by nonparametric tests and post hoc models. The correlations between WM microstructure and broad cognitive domains and cognition-related plasma biomarkers were assessed by Spearman's correlation analysis. RESULTS The abnormal WM microstructure was localized to the WM fiber bundles in MOH patients but distributed widely in SOH cohorts (p < 0.05). Further analysis showed that the neurite density index of the left cingulate gyrus was negatively associated with amyloid β-40, glial fibrillary acidic protein, neurofilament light chain, phospho-tau181 (p < 0.05) but positively with global cognitive function (MOCA, MMSE, AER-III), memory, attention, language, language fluency, visuospatial function and amyloid β-40 / amyloid β-42 (p < 0.05). Additionally, other abnormal WM microstructures of OH were associated with broad cognitive domains and cognition-related plasma biomarkers to varying degrees. CONCLUSION The findings evidence that abnormal WM microstructures may present themselves as early as in the MOH phase and that these structural abnormalities are associated with cognitive functions and cognition-related plasma biomarkers.
Collapse
Affiliation(s)
- Yingzhe Cheng
- Department of Neurology, Center for Cognitive NeurologyFujian Medical University Union HospitalFuzhou CityChina
- Fujian Institute of GeriatricsFujian Medical University Union HospitalFuzhou CityChina
- Institute of Clinical NeurologyFujian Medical UniversityFuzhou CityChina
- Fujian Key Laboratory of Molecular NeurologyFujian Medical UniversityFuzhou CityChina
| | - Lin Lin
- Department of Neurology, Center for Cognitive NeurologyFujian Medical University Union HospitalFuzhou CityChina
- Fujian Institute of GeriatricsFujian Medical University Union HospitalFuzhou CityChina
- Institute of Clinical NeurologyFujian Medical UniversityFuzhou CityChina
- Fujian Key Laboratory of Molecular NeurologyFujian Medical UniversityFuzhou CityChina
| | - Shaofan Jiang
- Department of RadiologyFujian Medical University Union HospitalFuzhou CityChina
- Fujian Key Laboratory of Intelligent Imaging and Precision Radiotherapy for TumorsFujian Medical UniversityFuzhou CityChina
| | - Peilin Huang
- Department of Neurology, Center for Cognitive NeurologyFujian Medical University Union HospitalFuzhou CityChina
- Fujian Institute of GeriatricsFujian Medical University Union HospitalFuzhou CityChina
- Institute of Clinical NeurologyFujian Medical UniversityFuzhou CityChina
- Fujian Key Laboratory of Molecular NeurologyFujian Medical UniversityFuzhou CityChina
| | - Jiejun Zhang
- Department of Neurology, Center for Cognitive NeurologyFujian Medical University Union HospitalFuzhou CityChina
- Fujian Institute of GeriatricsFujian Medical University Union HospitalFuzhou CityChina
- Institute of Clinical NeurologyFujian Medical UniversityFuzhou CityChina
- Fujian Key Laboratory of Molecular NeurologyFujian Medical UniversityFuzhou CityChina
- Center for GeriatricsHainan General HospitalHainanChina
| | - Jiawei Xin
- Department of Neurology, Center for Cognitive NeurologyFujian Medical University Union HospitalFuzhou CityChina
- Fujian Institute of GeriatricsFujian Medical University Union HospitalFuzhou CityChina
- Institute of Clinical NeurologyFujian Medical UniversityFuzhou CityChina
- Fujian Key Laboratory of Molecular NeurologyFujian Medical UniversityFuzhou CityChina
| | - Haibin Xu
- Fujian Medical UniversityFuzhou CityChina
| | - Yanping Wang
- Department of EndocrinologyFujian Medical University Union HospitalFuzhou CityChina
| | - Xiaodong Pan
- Department of Neurology, Center for Cognitive NeurologyFujian Medical University Union HospitalFuzhou CityChina
- Fujian Institute of GeriatricsFujian Medical University Union HospitalFuzhou CityChina
- Institute of Clinical NeurologyFujian Medical UniversityFuzhou CityChina
- Fujian Key Laboratory of Molecular NeurologyFujian Medical UniversityFuzhou CityChina
| |
Collapse
|
6
|
Ishikawa H, Shindo A, Mizutani A, Tomimoto H, Lo EH, Arai K. A brief overview of a mouse model of cerebral hypoperfusion by bilateral carotid artery stenosis. J Cereb Blood Flow Metab 2023; 43:18-36. [PMID: 36883344 PMCID: PMC10638994 DOI: 10.1177/0271678x231154597] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/23/2022] [Accepted: 01/04/2023] [Indexed: 03/09/2023]
Abstract
Vascular cognitive impairment (VCI) refers to all forms of cognitive disorder related to cerebrovascular diseases, including vascular mild cognitive impairment, post-stroke dementia, multi-infarct dementia, subcortical ischemic vascular dementia (SIVD), and mixed dementia. Among the causes of VCI, more attention has been paid to SIVD because the causative cerebral small vessel pathologies are frequently observed in elderly people and because the gradual progression of cognitive decline often mimics Alzheimer's disease. In most cases, small vessel diseases are accompanied by cerebral hypoperfusion. In mice, prolonged cerebral hypoperfusion is induced by bilateral carotid artery stenosis (BCAS) with surgically implanted metal micro-coils. This cerebral hypoperfusion BCAS model was proposed as a SIVD mouse model in 2004, and the spreading use of this mouse SIVD model has provided novel data regarding cognitive dysfunction and histological/genetic changes by cerebral hypoperfusion. Oxidative stress, microvascular injury, excitotoxicity, blood-brain barrier dysfunction, and secondary inflammation may be the main mechanisms of brain damage due to prolonged cerebral hypoperfusion, and some potential therapeutic targets for SIVD have been proposed by using transgenic mice or clinically used drugs in BCAS studies. This review article overviews findings from the studies that used this hypoperfused-SIVD mouse model, which were published between 2004 and 2021.
Collapse
Affiliation(s)
- Hidehiro Ishikawa
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Akihiro Shindo
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Akane Mizutani
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hidekazu Tomimoto
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Eng H Lo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
7
|
Lana D, Magni G, Landucci E, Wenk GL, Pellegrini-Giampietro DE, Giovannini MG. Phenomic Microglia Diversity as a Druggable Target in the Hippocampus in Neurodegenerative Diseases. Int J Mol Sci 2023; 24:13668. [PMID: 37761971 PMCID: PMC10531074 DOI: 10.3390/ijms241813668] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/31/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
Phenomics, the complexity of microglia phenotypes and their related functions compels the continuous study of microglia in disease animal models to find druggable targets for neurodegenerative disorders. Activation of microglia was long considered detrimental for neuron survival, but more recently it has become apparent that the real scenario of microglia morphofunctional diversity is far more complex. In this review, we discuss the recent literature on the alterations in microglia phenomics in the hippocampus of animal models of normal brain aging, acute neuroinflammation, ischemia, and neurodegenerative disorders, such as AD. Microglia undergo phenomic changes consisting of transcriptional, functional, and morphological changes that transform them into cells with different properties and functions. The classical subdivision of microglia into M1 and M2, two different, all-or-nothing states is too simplistic, and does not correspond to the variety of phenotypes recently discovered in the brain. We will discuss the phenomic modifications of microglia focusing not only on the differences in microglia reactivity in the diverse models of neurodegenerative disorders, but also among different areas of the brain. For instance, in contiguous and highly interconnected regions of the rat hippocampus, microglia show a differential, finely regulated, and region-specific reactivity, demonstrating that microglia responses are not uniform, but vary significantly from area to area in response to insults. It is of great interest to verify whether the differences in microglia reactivity may explain the differential susceptibility of different brain areas to insults, and particularly the higher sensitivity of CA1 pyramidal neurons to inflammatory stimuli. Understanding the spatiotemporal heterogeneity of microglia phenomics in health and disease is of paramount importance to find new druggable targets for the development of novel microglia-targeted therapies in different CNS disorders. This will allow interventions in three different ways: (i) by suppressing the pro-inflammatory properties of microglia to limit the deleterious effect of their activation; (ii) by modulating microglia phenotypic change to favor anti-inflammatory properties; (iii) by influencing microglia priming early in the disease process.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| | - Giada Magni
- Institute of Applied Physics “Nello Carrara”, National Research Council (IFAC-CNR), Via Madonna del Piano 10, 50019 Florence, Italy;
| | - Elisa Landucci
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| | - Gary L. Wenk
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA;
| | - Domenico Edoardo Pellegrini-Giampietro
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| | - Maria Grazia Giovannini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| |
Collapse
|
8
|
Rajeev V, Chai YL, Poh L, Selvaraji S, Fann DY, Jo DG, De Silva TM, Drummond GR, Sobey CG, Arumugam TV, Chen CP, Lai MKP. Chronic cerebral hypoperfusion: a critical feature in unravelling the etiology of vascular cognitive impairment. Acta Neuropathol Commun 2023; 11:93. [PMID: 37309012 PMCID: PMC10259064 DOI: 10.1186/s40478-023-01590-1] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/14/2023] Open
Abstract
Vascular cognitive impairment (VCI) describes a wide spectrum of cognitive deficits related to cerebrovascular diseases. Although the loss of blood flow to cortical regions critically involved in cognitive processes must feature as the main driver of VCI, the underlying mechanisms and interactions with related disease processes remain to be fully elucidated. Recent clinical studies of cerebral blood flow measurements have supported the role of chronic cerebral hypoperfusion (CCH) as a major driver of the vascular pathology and clinical manifestations of VCI. Here we review the pathophysiological mechanisms as well as neuropathological changes of CCH. Potential interventional strategies for VCI are also reviewed. A deeper understanding of how CCH can lead to accumulation of VCI-associated pathology could potentially pave the way for early detection and development of disease-modifying therapies, thus allowing preventive interventions instead of symptomatic treatments.
Collapse
Affiliation(s)
- Vismitha Rajeev
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
| | - Yuek Ling Chai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
| | - Luting Poh
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
| | - Sharmelee Selvaraji
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore, Singapore, Singapore
| | - David Y Fann
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - T Michael De Silva
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Grant R Drummond
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Christopher G Sobey
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Thiruma V Arumugam
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Christopher P Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
- NUS Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore.
- NUS Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
9
|
Deng S, Shu S, Zhai L, Xia S, Cao X, Li H, Bao X, Liu P, Xu Y. Optogenetic Stimulation of mPFC Alleviates White Matter Injury-Related Cognitive Decline after Chronic Ischemia through Adaptive Myelination. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2202976. [PMID: 36529961 PMCID: PMC9929132 DOI: 10.1002/advs.202202976] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 09/08/2022] [Indexed: 06/07/2023]
Abstract
White matter injury (WMI), which reflects myelin loss, contributes to cognitive decline or dementia caused by cerebral vascular diseases. However, because pharmacological agents specifically for WMI are lacking, novel therapeutic strategies need to be explored. It is recently found that adaptive myelination is required for homeostatic control of brain functions. In this study, adaptive myelination-related strategies are applied to explore the treatment for ischemic WMI-related cognitive dysfunction. Here, bilateral carotid artery stenosis (BCAS) is used to model ischemic WMI-related cognitive impairment and uncover that optogenetic and chemogenetic activation of glutamatergic neurons in the medial prefrontal cortex (mPFC) promote the differentiation of oligodendrocyte precursor cells (OPCs) in the corpus callosum, leading to improvements in myelin repair and working memory. Mechanistically, these neuromodulatory techniques exert a therapeutic effect by inducing the secretion of Wnt2 from activated neuronal axons, which acts on oligodendrocyte precursor cells and drives oligodendrogenesis and myelination. Thus, this study suggests that neuromodulation is a promising strategy for directing myelin repair and cognitive recovery through adaptive myelination in the context of ischemic WMI.
Collapse
Affiliation(s)
- Shiji Deng
- Department of NeurologyDrum Tower HospitalMedical School and The State Key Laboratory of Pharmaceutical BiotechnologyInstitute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjing210008China
| | - Shu Shu
- Department of NeurologyDrum Tower HospitalMedical School and The State Key Laboratory of Pharmaceutical BiotechnologyInstitute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjing210008China
| | - Lili Zhai
- Department of NeurologyDrum Tower HospitalMedical School and The State Key Laboratory of Pharmaceutical BiotechnologyInstitute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjing210008China
| | - Shengnan Xia
- Department of NeurologyDrum Tower HospitalMedical School and The State Key Laboratory of Pharmaceutical BiotechnologyInstitute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjing210008China
| | - Xiang Cao
- Department of NeurologyDrum Tower HospitalMedical School and The State Key Laboratory of Pharmaceutical BiotechnologyInstitute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjing210008China
| | - Huiya Li
- Department of NeurologyDrum Tower HospitalMedical School and The State Key Laboratory of Pharmaceutical BiotechnologyInstitute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjing210008China
| | - Xinyu Bao
- Department of NeurologyDrum Tower HospitalMedical School and The State Key Laboratory of Pharmaceutical BiotechnologyInstitute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjing210008China
| | - Pinyi Liu
- Department of NeurologyDrum Tower HospitalMedical School and The State Key Laboratory of Pharmaceutical BiotechnologyInstitute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjing210008China
| | - Yun Xu
- Department of NeurologyDrum Tower HospitalMedical School and The State Key Laboratory of Pharmaceutical BiotechnologyInstitute of Translational Medicine for Brain Critical DiseasesNanjing UniversityNanjing210008China
- Jiangsu Key Laboratory for Molecular MedicineMedical School of Nanjing UniversityNanjing210008China
- Jiangsu Provincial Key Discipline of NeurologyNanjing210008China
- Nanjing Neurology Medical CenterNanjing210008China
| |
Collapse
|
10
|
Bian Z, Hu X, Liu X, Yu H, Bian Y, Sun H, Fukui Y, Morihara R, Ishiura H, Yamashita T. Protective Effects of Rivaroxaban on White Matter Integrity and Remyelination in a Mouse Model of Alzheimer's Disease Combined with Cerebral Hypoperfusion. J Alzheimers Dis 2023; 96:609-622. [PMID: 37840489 DOI: 10.3233/jad-230413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by cognitive dysfunction and memory loss that is accompanied by pathological changes to white matter. Some clinical and animal research revealed that AD combined with chronic cerebral hypoperfusion (CCH) exacerbates AD progression by inducing blood-brain barrier dysfunction and fibrinogen deposition. Rivaroxaban, an anticoagulant, has been shown to reduce the rates of dementia in atrial fibrillation patients, but its effects on white matter and the underlying mechanisms are unclear. OBJECTIVE The main purpose of this study was to explore the therapeutic effect of rivaroxaban on the white matter of AD+CCH mice. METHODS In this study, the therapeutic effects of rivaroxaban on white matter in a mouse AD+CCH model were investigated to explore the potential mechanisms involving fibrinogen deposition, inflammation, and oxidative stress on remyelination in white matter. RESULTS The results indicate that rivaroxaban significantly attenuated fibrinogen deposition, fibrinogen-related microglia activation, oxidative stress, and enhanced demyelination in AD+CCH mice, leading to improved white matter integrity, reduced axonal damage, and restored myelin loss. CONCLUSIONS These findings suggest that long-term administration of rivaroxaban might reduce the risk of dementia.
Collapse
Affiliation(s)
- Zhihong Bian
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Xinran Hu
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Xia Liu
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haibo Yu
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Yuting Bian
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Hongming Sun
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Yusuke Fukui
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Ryuta Morihara
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Hiroyuki Ishiura
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Toru Yamashita
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| |
Collapse
|
11
|
Chen X, Chen L, Lin G, Wang Z, Kodali MC, Li M, Chen H, Lebovitz SG, Ortyl TC, Li L, Ismael S, Singh P, Malik KU, Ishrat T, Zhou FM, Zheng W, Liao FF. White matter damage as a consequence of vascular dysfunction in a spontaneous mouse model of chronic mild chronic hypoperfusion with eNOS deficiency. Mol Psychiatry 2022; 27:4754-4769. [PMID: 35948662 PMCID: PMC9734049 DOI: 10.1038/s41380-022-01701-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 06/20/2022] [Accepted: 07/01/2022] [Indexed: 12/14/2022]
Abstract
Vascular cognitive impairment and dementia (VCID) is the second most common form of dementia after Alzheimer's disease (AD). Currently, the mechanistic insights into the evolution and progression of VCID remain elusive. White matter change represents an invariant feature. Compelling clinical neuroimaging and pathological evidence suggest a link between white matter changes and neurodegeneration. Our prior study detected hypoperfused lesions in mice with partial deficiency of endothelial nitric oxide (eNOS) at very young age, precisely matching to those hypoperfused areas identified in preclinical AD patients. White matter tracts are particularly susceptible to the vascular damage induced by chronic hypoperfusion. Using immunohistochemistry, we detected severe demyelination in the middle-aged eNOS-deficient mice. The demyelinated areas were confined to cortical and subcortical areas including the corpus callosum and hippocampus. The intensity of demyelination correlated with behavioral deficits of gait and associative recognition memory performances. By Evans blue angiography, we detected blood-brain barrier (BBB) leakage as another early pathological change affecting frontal and parietal cortex in eNOS-deficient mice. Sodium nitrate fortified drinking water provided to young and middle-aged eNOS-deficient mice completely prevented non-perfusion, BBB leakage, and white matter pathology, indicating that impaired endothelium-derived NO signaling may have caused these pathological events. Furthermore, genome-wide transcriptomic analysis revealed altered gene clusters most related to mitochondrial respiratory pathways selectively in the white matter of young eNOS-deficient mice. Using eNOS-deficient mice, we identified BBB breakdown and hypoperfusion as the two earliest pathological events, resulting from insufficient vascular NO signaling. We speculate that the compromised BBB and mild chronic hypoperfusion trigger vascular damage, along with oxidative stress and astrogliosis, accounting for the white matter pathological changes in the eNOS-deficient mouse model. We conclude that eNOS-deficient mice represent an ideal spontaneous evolving model for studying the earliest events leading to white matter changes, which will be instrumental to future therapeutic testing of drug candidates and for targeting novel/specific vascular mechanisms contributing to VCID and AD.
Collapse
Affiliation(s)
- Xingyong Chen
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA
- Department of Neurology, Fujian Provincial Hospital, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, PR China
| | - Ling Chen
- Department of Cell Biology and Genetics, The school of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350001, PR China
| | - Geng Lin
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA
- Teaching Center of Basic Medical Experiment, China Medical University, Shenyang, Liaoning, 110122, PR China
| | - Zhengjun Wang
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA
| | - Mahesh C Kodali
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA
| | - Mingqi Li
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA
| | - Huimin Chen
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA
| | - Sarah G Lebovitz
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA
| | - Tyler C Ortyl
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA
| | - Lexiao Li
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA
| | - Saifudeen Ismael
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA
| | - Purnima Singh
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA
| | - Kafait U Malik
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA
| | - Tauheed Ishrat
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA
| | - Fu-Ming Zhou
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA
| | - Wei Zheng
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA.
- Department of Histology and Embryology, Basic Medical University, China Medical University, Shenyang, Liaoning, 110122, PR China.
| | - Francesca-Fang Liao
- Department of Pharmacology, Addiction Science, Toxicology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, 38163, USA.
| |
Collapse
|
12
|
Ma J, Liu F, Wang Y, Ma L, Niu Y, Wang J, Ye Z, Zhang J. Frequency-dependent white-matter functional network changes associated with cognitive deficits in subcortical vascular cognitive impairment. Neuroimage Clin 2022; 36:103245. [PMID: 36451351 PMCID: PMC9668649 DOI: 10.1016/j.nicl.2022.103245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 10/07/2022] [Accepted: 10/21/2022] [Indexed: 11/11/2022]
Abstract
Vascular cognitive impairment (VCI) refers to all forms of cognitive decline associated with cerebrovascular diseases, in which white matter (WM) is highly vulnerable. Although previous studies have shown that blood oxygen level-dependent (BOLD) signals inside WM can effectively reflect neural activities, whether WM BOLD signal alterations are present and their roles underlying cognitive impairment in VCI remain largely unknown. In this study, 36 subcortical VCI (SVCI) patients and 36 healthy controls were enrolled to evaluate WM dysfunction. Specifically, fourteen distinct WM networks were identified from resting-state functional MRI using K-means clustering analysis. Subsequently, between-network functional connectivity (FC) and within-network BOLD signal amplitude of WM networks were calculated in three frequency bands (band A: 0.01-0.15 Hz, band B: 0.08-0.15 Hz, and band C: 0.01-0.08 Hz). Patients with SVCI manifested decreased FC mainly in bilateral parietal WM regions, forceps major, superior and inferior longitudinal fasciculi. These connections extensively linked with distinct WM networks and with gray-matter networks such as frontoparietal control, dorsal and ventral attention networks, which exhibited frequency-specific alterations in SVCI. Additionally, extensive amplitude reductions were found in SVCI, showing frequency-dependent properties in parietal, anterior corona radiate, pre/post central, superior and inferior longitudinal fasciculus networks. Furthermore, these decreased FC and amplitudes showed significant positive correlations with cognitive performances in SVCI, and high diagnostic performances for SVCI especially combining all bands. Our study indicated that VCI-related cognitive deficits were characterized by frequency-dependent WM functional abnormalities, which offered novel applicable neuromarkers for VCI.
Collapse
Affiliation(s)
- Juanwei Ma
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China; National Clinical Research Center for Cancer, Tianjin, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, China; Tianjin's Clinical Research Center for Cancer, Tianjin, China; Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Feng Liu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Yang Wang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Lin Ma
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Yali Niu
- Department of Rehabilitation, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jing Wang
- Department of Rehabilitation, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Zhaoxiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China; National Clinical Research Center for Cancer, Tianjin, China; Key Laboratory of Cancer Prevention and Therapy, Tianjin, China; Tianjin's Clinical Research Center for Cancer, Tianjin, China.
| | - Jing Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
13
|
Yu W, Li Y, Hu J, Wu J, Huang Y. A Study on the Pathogenesis of Vascular Cognitive Impairment and Dementia: The Chronic Cerebral Hypoperfusion Hypothesis. J Clin Med 2022; 11:jcm11164742. [PMID: 36012981 PMCID: PMC9409771 DOI: 10.3390/jcm11164742] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/27/2022] [Accepted: 08/02/2022] [Indexed: 11/16/2022] Open
Abstract
The pathogenic mechanisms underlying vascular cognitive impairment and dementia (VCID) remain controversial due to the heterogeneity of vascular causes and complexity of disease neuropathology. However, one common feature shared among all these vascular causes is cerebral blood flow (CBF) dysregulation, and chronic cerebral hypoperfusion (CCH) is the universal consequence of CBF dysregulation, which subsequently results in an insufficient blood supply to the brain, ultimately contributing to VCID. The purpose of this comprehensive review is to emphasize the important contributions of CCH to VCID and illustrate the current findings about the mechanisms involved in CCH-induced VCID pathological changes. Specifically, evidence is mainly provided to support the molecular mechanisms, including Aβ accumulation, inflammation, oxidative stress, blood-brain barrier (BBB) disruption, trophic uncoupling and white matter lesions (WMLs). Notably, there are close interactions among these multiple mechanisms, and further research is necessary to elucidate the hitherto unsolved questions regarding these interactions. An enhanced understanding of the pathological features in preclinical models could provide a theoretical basis, ultimately achieving the shift from treatment to prevention.
Collapse
Affiliation(s)
- Weiwei Yu
- Department of Neurology, Peking University Shenzhen Hospital, 1120 Lianhua Road, Futian District, Shenzhen 518036, China
| | - Yao Li
- Department of Neurology, Peking University Shenzhen Hospital, 1120 Lianhua Road, Futian District, Shenzhen 518036, China
| | - Jun Hu
- Department of Neurology, Peking University Shenzhen Hospital, 1120 Lianhua Road, Futian District, Shenzhen 518036, China
| | - Jun Wu
- Department of Neurology, Peking University Shenzhen Hospital, 1120 Lianhua Road, Futian District, Shenzhen 518036, China
- Correspondence: (J.W.); (Y.H.); Tel.: +86-0755-8392-2833 (J.W.); +86-010-83572857 (Y.H.)
| | - Yining Huang
- Department of Neurology, Peking University First Hospital, 8 Xishiku Street Xicheng District, Beijing 100034, China
- Correspondence: (J.W.); (Y.H.); Tel.: +86-0755-8392-2833 (J.W.); +86-010-83572857 (Y.H.)
| |
Collapse
|
14
|
Ishiyama H, Tanaka T, Saito S, Koyama T, Kitamura A, Inoue M, Fukushima N, Morita Y, Koga M, Toyoda K, Kuriyama N, Urushitani M, Ihara M. Plasma mid-regional pro-adrenomedullin: A biomarker of the ischemic penumbra in hyperacute stroke. Brain Pathol 2022; 33:e13110. [PMID: 35916272 PMCID: PMC10041162 DOI: 10.1111/bpa.13110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 07/15/2022] [Indexed: 12/01/2022] Open
Abstract
Reperfusion therapy has improved the outcomes of ischemic stroke but also emphasized the importance of ischemic penumbra. However, blood biomarkers are currently unavailable for this region. Adrenomedullin (ADM) is a neuroprotective peptide, secreted in a compensatory response to brain ischemia. We thus investigated whether an increase in mid-regional pro-ADM (MR-proADM), a stable peptide fragment of the ADM precursor, could act as a biomarker by predicting the ischemic penumbra in hyperacute ischemic stroke (HAIS). We prospectively enrolled consecutive HAIS patients (n = 119; median age, 77 years; male, 59.7%) admitted to our institutes from July 2017 to March 2019 and evaluated plasma MR-proADM levels within 4.5 h of onset. MR-proADM levels in HAIS were compared to healthy controls (n = 1298; median age, 58 years; male, 33.2%) in the Japan Multi-Institutional Collaborative Cohort Study from 2013 to 2017. Furthermore, we evaluated whether MR-proADM levels were associated with the penumbra estimated by clinical-diffusion mismatch (CDM) (National Institute of Health Stroke Scale [NIHSS] ≥8, diffusion ischemic core volume ≤25 ml), or magnetic resonance angiography-diffusion-weighted imaging mismatch (MDM) (NIHSS ≥5, a proximal vessel occlusion with core volume ≤25 ml, or a proximal vessel stenosis/distal vessel occlusion with core volume ≤15 ml). In a case-control study, multivariate logistic analysis showed a significant association between HAIS and MR-proADM ≥0.54 nmol/L (adjusted odds ratio, 7.92 [95% CI, 4.17-15.02], p < 0.001). Though MR-proADM levels in HAIS did not correlate with the ischemic core volume (rs = 0.09, p = 0.348), they were higher in HAIS with CDM (n = 34; 0.81 vs. 0.61 nmol/L, p < 0.001) or MDM (n = 26; 0.83 vs. 0.62 nmol/L, p = 0.002). These differences remained significant after adjusting baseline factors (adjusted odds ratio, 4.06 [95% CI, 1.31-12.55], p = 0.015 and 4.65 [1.35-16.11], p = 0.015, respectively). Plasma MR-proADM is elevated in HAIS, especially in those with a substantial penumbra, suggesting potential as a blood biomarker in this region.
Collapse
Affiliation(s)
- Hiroyuki Ishiyama
- Department of NeurologyNational Cerebral and Cardiovascular CenterSuitaJapan
| | - Tomotaka Tanaka
- Department of NeurologyNational Cerebral and Cardiovascular CenterSuitaJapan
| | - Satoshi Saito
- Department of NeurologyNational Cerebral and Cardiovascular CenterSuitaJapan
| | - Teruhide Koyama
- Department of Epidemiology for Community Health and MedicineKyoto Prefectural University of MedicineKyotoJapan
| | - Akihiro Kitamura
- Department of NeurologyShiga University of Medical ScienceOtsuJapan
| | - Manabu Inoue
- Department of Cerebrovascular MedicineNational Cerebral and Cardiovascular CenterSuitaJapan
| | - Naoya Fukushima
- Department of NeurologyNational Cerebral and Cardiovascular CenterSuitaJapan
| | - Yoshiaki Morita
- Department of RadiologyNational Cerebral and Cardiovascular CenterSuitaJapan
| | - Masatoshi Koga
- Department of Cerebrovascular MedicineNational Cerebral and Cardiovascular CenterSuitaJapan
| | - Kazunori Toyoda
- Department of Cerebrovascular MedicineNational Cerebral and Cardiovascular CenterSuitaJapan
| | - Nagato Kuriyama
- Department of Epidemiology for Community Health and MedicineKyoto Prefectural University of MedicineKyotoJapan
- Shizuoka Graduate University of Public HealthShizuokaJapan
| | | | - Masafumi Ihara
- Department of NeurologyNational Cerebral and Cardiovascular CenterSuitaJapan
| |
Collapse
|
15
|
Agrawal S, Schneider JA. Vascular pathology and pathogenesis of cognitive impairment and dementia in older adults. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2022; 3:100148. [PMID: 36324408 PMCID: PMC9616381 DOI: 10.1016/j.cccb.2022.100148] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 05/25/2022] [Accepted: 06/23/2022] [Indexed: 12/24/2022]
Abstract
It is well recognized that brains of older people often harbor cerebrovascular disease pathology including vessel disease and vascular-related tissue injuries and that this is associated with vascular cognitive impairment and contributes to dementia. Here we review vascular pathologies, cognitive impairment, and dementia. We highlight the importance of mixed co-morbid AD/non-AD neurodegenerative and vascular pathology that has been collected in multiple clinical pathologic studies, especially in community-based studies. We also provide an update of vascular pathologies from the Rush Memory and Aging Project and Religious Orders Study cohorts with special emphasis on the differences across age in persons with and without dementia. Finally, we discuss neuropathological perspectives on the interpretation of clinical-pathological studies and emerging data in community-based studies.
Collapse
Affiliation(s)
- Sonal Agrawal
- Rush Alzheimer's Disease Center, Rush University Medical Center, Jelke Building, 1750 W. Harrison Street, Chicago 60612, IL, USA
- Department of Pathology, Rush University Medical Center, Chicago, IL, USA
| | - Julie A. Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, Jelke Building, 1750 W. Harrison Street, Chicago 60612, IL, USA
- Department of Pathology, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
16
|
Yu M, Zheng X, Cheng F, Shao B, Zhuge Q, Jin K. Metformin, Rapamycin, or Nicotinamide Mononucleotide Pretreatment Attenuate Cognitive Impairment After Cerebral Hypoperfusion by Inhibiting Microglial Phagocytosis. Front Neurol 2022; 13:903565. [PMID: 35769369 PMCID: PMC9234123 DOI: 10.3389/fneur.2022.903565] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/10/2022] [Indexed: 12/02/2022] Open
Abstract
Vascular cognitive impairment (VCI) is the second leading form of dementia after Alzheimer's disease (AD) plaguing the elder population. Despite the enormous prevalence of VCI, the biological basis of this disease has been much less well-studied than that of AD, with no specific therapy currently existing to prevent or treat VCI. As VCI mainly occurs in the elderly, the role of anti-aging drugs including metformin, rapamycin, and nicotinamide mono nucleotide (NMN), and the underlying mechanism remain uncertain. Here, we examined the role of metformin, rapamycin, and NMN in cognitive function, white matter integrity, microglial response, and phagocytosis in a rat model of VCI by bilateral common carotid artery occlusion (BCCAO). BCCAO-induced chronic cerebral hypoperfusion could cause spatial working memory deficits and white matter lesions (WMLs), along with increasing microglial activation and phagocytosis compared to sham-operated rats. We found the cognitive impairment was significantly improved in BCCAO rats pretreated with these three drugs for 14 days before BCCAO compared with the vehicle group by the analysis of the Morris water maze and new object recognition tests. Pretreatment of metformin, rapamycin, or NMN also increased myelin basic protein (MBP, a marker for myelin) expression and reduced SMI32 (a marker for demyelinated axons) intensity and SMI32/MBP ratio compared with the vehicle group, suggesting that these drugs could ameliorate BCCAO-induced WMLs. The findings were confirmed by Luxol fast blue (LFB) stain, which is designed for staining myelin/myelinated axons. We further found that pretreatment of metformin, rapamycin, or NMN reduced microglial activation and the number of M1 microglia, but increased the number of M2 microglia compared to the vehicle group. Importantly, the number of MBP+/Iba1+/CD68+ microglia was significantly reduced in the BCCAO rats pretreated with these three drugs compared with the vehicle group, suggesting that these drugs suppress microglial phagocytosis. No significant difference was found between the groups pretreated with metformin, rapamycin, or NMN. Our data suggest that metformin, rapamycin, or NMN could protect or attenuate cognitive impairment and WMLs by modifying microglial polarization and inhibiting phagocytosis. The findings may open a new avenue for VCI treatment.
Collapse
Affiliation(s)
- Mengdi Yu
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Xiaoying Zheng
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Fangyu Cheng
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Bei Shao
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
| | - Qichuan Zhuge
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang, China
- *Correspondence: Qichuan Zhuge
| | - Kunlin Jin
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX, United States
- Kunlin Jin
| |
Collapse
|
17
|
Tian Z, Ji X, Liu J. Neuroinflammation in Vascular Cognitive Impairment and Dementia: Current Evidence, Advances, and Prospects. Int J Mol Sci 2022; 23:ijms23116224. [PMID: 35682903 PMCID: PMC9181710 DOI: 10.3390/ijms23116224] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/26/2022] [Accepted: 05/31/2022] [Indexed: 02/04/2023] Open
Abstract
Vascular cognitive impairment and dementia (VCID) is a major heterogeneous brain disease caused by multiple factors, and it is the second most common type of dementia in the world. It is caused by long-term chronic low perfusion in the whole brain or local brain area, and it eventually develops into severe cognitive dysfunction syndrome. Because of the disease’s ambiguous classification and diagnostic criteria, there is no clear treatment strategy for VCID, and the association between cerebrovascular pathology and cognitive impairment is controversial. Neuroinflammation is an immunological cascade reaction mediated by glial cells in the central nervous system where innate immunity resides. Inflammatory reactions could be triggered by various damaging events, including hypoxia, ischemia, and infection. Long-term chronic hypoperfusion-induced ischemia and hypoxia can overactivate neuroinflammation, causing apoptosis, blood–brain barrier damage and other pathological changes, triggering or aggravating the occurrence and development of VCID. In this review, we will explore the mechanisms of neuroinflammation induced by ischemia and hypoxia caused by chronic hypoperfusion and emphasize the important role of neuroinflammation in the development of VCID from the perspective of immune cells, immune mediators and immune signaling pathways, so as to provide valuable ideas for the prevention and treatment of the disease.
Collapse
Affiliation(s)
- Zhengming Tian
- Laboratory of Brain Disorders, Beijing Institute of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing 100069, China;
| | - Xunming Ji
- Laboratory of Brain Disorders, Beijing Institute of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing 100069, China;
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100069, China
- Correspondence: (X.J.); (J.L.); Tel.: +86-13520729063 (J.L.)
| | - Jia Liu
- Laboratory of Brain Disorders, Beijing Institute of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Capital Medical University, Beijing 100069, China;
- Correspondence: (X.J.); (J.L.); Tel.: +86-13520729063 (J.L.)
| |
Collapse
|
18
|
Porcu M, Cocco L, Cau R, Suri JS, Mannelli L, Puig J, Qi Y, Paraskevas KI, Saba L. Mid-term effects of carotid endarterectomy on cognition and white matter status evaluated by whole brain diffusion tensor imaging metrics: A preliminary analysis. Eur J Radiol 2022; 151:110314. [PMID: 35452954 DOI: 10.1016/j.ejrad.2022.110314] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/03/2022] [Accepted: 04/08/2022] [Indexed: 12/24/2022]
Abstract
PURPOSE To analyze the mid-term (12 months) effects of carotid endarterectomy (CEA) on cognition and on the microstructural properties of the whole brain white matter in terms of derived diffusion Tensor imaging (DTI) metrics. METHODS We analyzed a population of 19 asymptomatic patients with extra-cranial internal carotid artery stenosis (eICA) eligible for CEA. All patients underwent cognitive evaluation with the Italian version of the Mini-Mental State Examination corrected for age and schooling, and with a Magnetic Resonance Imaging (MRI) investigation on a 1.5 Tesla MRI scanner, that included a 34-directions Diffusion Weighted Imaging (DWI) sequence for DTI metrics analysis. The global fractional anisotropy (gFA), global mean diffusivity (gMD), global radial diffusivity (gRD) and global axial diffusivity (gAD) were calculated for each patient. Both the cognitive and the imaging evaluation were performed at baseline (PRE-CEA) and 12 months after CEA (POST-CEA). Two-tailed Paerson's correlation test and paired samples t-test were used for evaluating the correlation between PRE-CEA and POST-CEA values, adopting a p-value of <0.05 as statistically significant. RESULTS A statistically significant increase of the MMSE scores (p < 0.0001), as well as for gFA (p < 0.0001), and a statistically significant reduction of gMD (p = 0.027) and gRD (p = 0.0005) was observed 12 months following uncomplicated CEA. CONCLUSIONS These findings suggest that CEA is associated with a general improvement of the WM microstructure of the whole brain.
Collapse
Affiliation(s)
- Michele Porcu
- Department of Radiology, AOU Cagliari, University of Cagliari, Italy.
| | - Luigi Cocco
- Department of Radiology, AOU Cagliari, University of Cagliari, Italy
| | - Riccardo Cau
- Department of Radiology, AOU Cagliari, University of Cagliari, Italy
| | - Jasjit S Suri
- Stroke Monitoring and Diagnostic Division, AtheroPoint™, Roseville, CA, USA
| | | | - Josep Puig
- Department of Radiology (IDI) and Girona Biomedical Research Institute (IDIBGI), Hospital Universitari de Girona Dr Josep Trueta, Girona, Spain
| | - Yang Qi
- Xuanwu Hospital, Capital Medical University, No.45 Changchun Street, Xicheng District, Beijing, China
| | | | - Luca Saba
- Department of Radiology, AOU Cagliari, University of Cagliari, Italy
| |
Collapse
|
19
|
Teng X, Hu P, Chen Y, Zang Y, Ye X, Ou J, Chen G, Shi YS. A novel
Lgi1
mutation causes white matter abnormalities and impairs motor coordination in mice. FASEB J 2022; 36:e22212. [DOI: 10.1096/fj.202101652r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/18/2022] [Accepted: 02/03/2022] [Indexed: 12/22/2022]
Affiliation(s)
- Xiao‐Yu Teng
- Minister of Education Key Laboratory of Model Animal for Disease Study Model Animal Research Center, Medical School Nanjing University Nanjing China
| | - Ping Hu
- Department of Prenatal Diagnosis State Key Laboratory of Reproductive Medicine Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital Nanjing China
| | - Yangyang Chen
- Minister of Education Key Laboratory of Model Animal for Disease Study Model Animal Research Center, Medical School Nanjing University Nanjing China
| | - Yanyu Zang
- Minister of Education Key Laboratory of Model Animal for Disease Study Model Animal Research Center, Medical School Nanjing University Nanjing China
| | - Xiaolian Ye
- Minister of Education Key Laboratory of Model Animal for Disease Study Model Animal Research Center, Medical School Nanjing University Nanjing China
| | - Jingmin Ou
- Department of General Surgery Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine Shanghai China
| | - Guiquan Chen
- Minister of Education Key Laboratory of Model Animal for Disease Study Model Animal Research Center, Medical School Nanjing University Nanjing China
| | - Yun Stone Shi
- Minister of Education Key Laboratory of Model Animal for Disease Study Model Animal Research Center, Medical School Nanjing University Nanjing China
- State Key Laboratory of Pharmaceutical Biotechnology Department of Neurology Affiliated Drum Tower Hospital of Nanjing University Medical School Nanjing University Nanjing China
- Institute for Brain Sciences Nanjing University Nanjing China
- Chemistry and Biomedicine Innovation Center Nanjing University Nanjing China
| |
Collapse
|
20
|
Li M, Kitamura A, Beverley J, Koudelka J, Duncombe J, Lennen R, Jansen MA, Marshall I, Platt B, Wiegand UK, Carare RO, Kalaria RN, Iliff JJ, Horsburgh K. Impaired Glymphatic Function and Pulsation Alterations in a Mouse Model of Vascular Cognitive Impairment. Front Aging Neurosci 2022; 13:788519. [PMID: 35095472 PMCID: PMC8793139 DOI: 10.3389/fnagi.2021.788519] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 12/07/2021] [Indexed: 12/24/2022] Open
Abstract
Large vessel disease and carotid stenosis are key mechanisms contributing to vascular cognitive impairment (VCI) and dementia. Our previous work, and that of others, using rodent models, demonstrated that bilateral common carotid stenosis (BCAS) leads to cognitive impairment via gradual deterioration of the neuro-glial-vascular unit and accumulation of amyloid-β (Aβ) protein. Since brain-wide drainage pathways (glymphatic) for waste clearance, including Aβ removal, have been implicated in the pathophysiology of VCI via glial mechanisms, we hypothesized that glymphatic function would be impaired in a BCAS model and exacerbated in the presence of Aβ. Male wild-type and Tg-SwDI (model of microvascular amyloid) mice were subjected to BCAS or sham surgery which led to a reduction in cerebral perfusion and impaired spatial learning acquisition and cognitive flexibility. After 3 months survival, glymphatic function was evaluated by cerebrospinal fluid (CSF) fluorescent tracer influx. We demonstrated that BCAS caused a marked regional reduction of CSF tracer influx in the dorsolateral cortex and CA1-DG molecular layer. In parallel to these changes increased reactive astrogliosis was observed post-BCAS. To further investigate the mechanisms that may lead to these changes, we measured the pulsation of cortical vessels. BCAS impaired vascular pulsation in pial arteries in WT and Tg-SwDI mice. Our findings show that BCAS influences VCI and that this is paralleled by impaired glymphatic drainage and reduced vascular pulsation. We propose that these additional targets need to be considered when treating VCI.
Collapse
Affiliation(s)
- Mosi Li
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Medical School, UK Dementia Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Akihiro Kitamura
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Department of Neurology, Shiga University of Medical Science, Otsu, Japan
| | - Joshua Beverley
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Juraj Koudelka
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Jessica Duncombe
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Ross Lennen
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Maurits A Jansen
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Ian Marshall
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Bettina Platt
- School of Medicine, Medical Sciences and Nutrition, College of Life Sciences and Medicine, University of Aberdeen, Aberdeen, United Kingdom
| | - Ulrich K Wiegand
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Roxana O Carare
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Rajesh N Kalaria
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Jeffrey J Iliff
- VISN 20 Mental Illness Research, Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, United States
- Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, United States
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, United States
| | - Karen Horsburgh
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
21
|
Poh L, Sim WL, Jo DG, Dinh QN, Drummond GR, Sobey CG, Chen CLH, Lai MKP, Fann DY, Arumugam TV. The role of inflammasomes in vascular cognitive impairment. Mol Neurodegener 2022; 17:4. [PMID: 35000611 PMCID: PMC8744307 DOI: 10.1186/s13024-021-00506-8] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 12/02/2021] [Indexed: 12/11/2022] Open
Abstract
There is an increasing prevalence of Vascular Cognitive Impairment (VCI) worldwide, and several studies have suggested that Chronic Cerebral Hypoperfusion (CCH) plays a critical role in disease onset and progression. However, there is a limited understanding of the underlying pathophysiology of VCI, especially in relation to CCH. Neuroinflammation is a significant contributor in the progression of VCI as increased systemic levels of the proinflammatory cytokine interleukin-1β (IL-1β) has been extensively reported in VCI patients. Recently it has been established that CCH can activate the inflammasome signaling pathways, involving NLRP3 and AIM2 inflammasomes that critically regulate IL-1β production. Given that neuroinflammation is an early event in VCI, it is important that we understand its molecular and cellular mechanisms to enable development of disease-modifying treatments to reduce the structural brain damage and cognitive deficits that are observed clinically in the elderly. Hence, this review aims to provide a comprehensive insight into the molecular and cellular mechanisms involved in the pathogenesis of CCH-induced inflammasome signaling in VCI.
Collapse
Affiliation(s)
- Luting Poh
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wei Liang Sim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Quynh Nhu Dinh
- Centre for Cardiovascular Biology and Disease Research, Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC Australia
| | - Grant R. Drummond
- Centre for Cardiovascular Biology and Disease Research, Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC Australia
| | - Christopher G. Sobey
- Centre for Cardiovascular Biology and Disease Research, Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC Australia
| | - Christopher Li-Hsian Chen
- Memory Aging and Cognition Centre, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mitchell K. P. Lai
- Memory Aging and Cognition Centre, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - David Y. Fann
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Centre for Healthy Longevity, National University Health System (NUHS), Singapore, Singapore
| | - Thiruma V. Arumugam
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
- Centre for Cardiovascular Biology and Disease Research, Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC Australia
| |
Collapse
|
22
|
Chai X, Li X, Zhang W, Tan X, Wang H, Yang Z. Legumain knockout improved cognitive impairment via reducing neuroinflammation in right unilateral common carotid artery occlusion mice. Life Sci 2021; 285:119944. [PMID: 34509465 DOI: 10.1016/j.lfs.2021.119944] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/28/2021] [Accepted: 09/03/2021] [Indexed: 12/26/2022]
Abstract
AIMS Chronic cerebral hypoperfusion (CCH) is a state of chronic cerebral blood flow reduction, and it is the main cause of cognitive impairment and neurodegenerative diseases. The abnormal upregulation of legumain, a lysosomal cysteine protease, trigger synaptic plasticity impairment and neuroinflammation, which are involved in the underlying pathophysiology of CCH. At present, few studies have reported the role of legumain in cognitive impairment caused by CCH. In our study, we aimed to investigate the involvement of legumain knockout in cognitive function and neuroinflammation in a CCH mouse model. MAIN METHODS In this study, right unilateral common carotid artery occlusion (rUCCAO) was used to simulate the pathological state of cerebral ischemic injury. Various behavioural tests were executed to assess cognitive performance. In vivo electrophysiological recordings were used to measure synaptic functions. Western blotting, Golgi staining, haematoxylin/eosin staining, and immunofluorescence assays were conducted to examine pathological changes and molecular mechanisms. KEY FINDINGS The data showed that the level of legumain was significantly increased in the hippocampus of mice subjected to rUCCAO. Legumain knockout significantly improved cognitive function and synaptic plasticity induced by rUCCAO, suggesting that legumain knockout-regulation effectively protected against CCH-induced behavioural dysfunctions. Moreover, legumain knockout suppressed rUCCAO-induced microglial activation, reduced the abnormal expression of inflammatory cytokines and the inflammasome complex, and impeded the activation of P65 and pyroptosis. SIGNIFICANCE These findings suggest that legumain is an effective regulator of CCH, and may be an ideal target for the development of cerebral ischemia treatments in the future.
Collapse
Affiliation(s)
- Xueqing Chai
- Medical School, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, Nankai University, Tianjin 300071, China
| | - Xiaolin Li
- Medical School, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, Nankai University, Tianjin 300071, China
| | - Wenxin Zhang
- Medical School, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, Nankai University, Tianjin 300071, China
| | - Xiaoyue Tan
- Medical School, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, Nankai University, Tianjin 300071, China
| | - Haiyun Wang
- Department of Anesthesiology, The Third Central Clinical College of Tianjin Medical University, Tianjin, China
| | - Zhuo Yang
- Medical School, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials for Ministry of Education, Nankai University, Tianjin 300071, China.
| |
Collapse
|
23
|
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
|
24
|
Li Z, Dolui S, Habes M, Bassett DS, Wolk D, Detre JA. Predicted disconnectome associated with progressive periventricular white matter ischemia. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2021; 2:100022. [PMID: 36324715 PMCID: PMC9616229 DOI: 10.1016/j.cccb.2021.100022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 11/21/2022]
Abstract
We used a virtual lesion DTI fiber tracking approach with healthy subject DTI data and simulated periventricular white matter (PVWM) lesion masks to predict the sequence of connectivity changes associated with progressive PVWM ischemia. We found that the optic radiations, inferior fronto-occipital fasciculus, inferior longitudinal fasciculus, corpus callosum, temporopontine tract and fornix were affected in early simulated ischemic injury, and that the connectivity of subcortical, cerebellar, and visual regions were significantly disrupted with increasing simulated lesion severity. The results of this study provide insights into the spatial-temporal changes of the brain structural connectome under progressive PVWM ischemia. The virtual lesion approach provides a meaningful proxy to the spatial-temporal changes of the brain's structural connectome and can be used to further characterize the cognitive sequelae of progressive PVWM ischemia in both normal aging and dementia.
Collapse
Affiliation(s)
- Zhengjun Li
- Departments of Neurology, University of Pennsylvania, 3W Gates Pavilion, 3400 Spruce Street, Philadelphia, PA 19104, USA
- Bioengineering, USA
- Physics & Astronomy, USA
- Electrical and Systems Engineering, University of Pennsylvania Perelman School of Medicine, USA
| | - Sudipto Dolui
- Radiology, USA
- Bioengineering, USA
- Physics & Astronomy, USA
- Electrical and Systems Engineering, University of Pennsylvania Perelman School of Medicine, USA
| | - Mohamad Habes
- Departments of Neurology, University of Pennsylvania, 3W Gates Pavilion, 3400 Spruce Street, Philadelphia, PA 19104, USA
- Radiology, USA
- Bioengineering, USA
- Physics & Astronomy, USA
- Electrical and Systems Engineering, University of Pennsylvania Perelman School of Medicine, USA
- Biggs institute neuroimaging core (BINC), Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Sciences Center, USA
| | - Danielle S. Bassett
- Departments of Neurology, University of Pennsylvania, 3W Gates Pavilion, 3400 Spruce Street, Philadelphia, PA 19104, USA
- Psychiatry, USA
- Bioengineering, USA
- Physics & Astronomy, USA
- Electrical and Systems Engineering, University of Pennsylvania Perelman School of Medicine, USA
- The Santa Fe Institute, USA
| | - David Wolk
- Departments of Neurology, University of Pennsylvania, 3W Gates Pavilion, 3400 Spruce Street, Philadelphia, PA 19104, USA
- Bioengineering, USA
- Physics & Astronomy, USA
- Electrical and Systems Engineering, University of Pennsylvania Perelman School of Medicine, USA
| | - John A. Detre
- Departments of Neurology, University of Pennsylvania, 3W Gates Pavilion, 3400 Spruce Street, Philadelphia, PA 19104, USA
- Radiology, USA
- Bioengineering, USA
- Physics & Astronomy, USA
- Electrical and Systems Engineering, University of Pennsylvania Perelman School of Medicine, USA
| |
Collapse
|
25
|
Takase H, Hamanaka G, Ohtomo R, Ishikawa H, Chung KK, Mandeville ET, Lok J, Fornage M, Herrup K, Tse KH, Lo EH, Arai K. Transcriptome Profiling of Mouse Corpus Callosum After Cerebral Hypoperfusion. Front Cell Dev Biol 2021; 9:685261. [PMID: 34222254 PMCID: PMC8248229 DOI: 10.3389/fcell.2021.685261] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/12/2021] [Indexed: 02/01/2023] Open
Abstract
White matter damage caused by cerebral hypoperfusion is a major hallmark of subcortical ischemic vascular dementia (SIVD), which is the most common subtype of vascular cognitive impairment and dementia (VCID) syndrome. In an aging society, the number of SIVD patients is expected to increase; however, effective therapies have yet to be developed. To understand the pathological mechanisms, we analyzed the profiles of the cells of the corpus callosum after cerebral hypoperfusion in a preclinical SIVD model. We prepared cerebral hypoperfused mice by subjecting 2-month old male C57BL/6J mice to bilateral carotid artery stenosis (BCAS) operation. BCAS-hypoperfusion mice exhibited cognitive deficits at 4 weeks after cerebral hypoperfusion, assessed by novel object recognition test. RNA samples from the corpus callosum region of sham- or BCAS-operated mice were then processed using RNA sequencing. A gene set enrichment analysis using differentially expressed genes between sham and BCAS-operated mice showed activation of oligodendrogenesis pathways along with angiogenic responses. This database of transcriptomic profiles of BCAS-hypoperfusion mice will be useful for future studies to find a therapeutic target for SIVD.
Collapse
Affiliation(s)
- Hajime Takase
- Neuroprotection Research Laboratory, Department of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Gen Hamanaka
- Neuroprotection Research Laboratory, Department of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Ryo Ohtomo
- Neuroprotection Research Laboratory, Department of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Hidehiro Ishikawa
- Neuroprotection Research Laboratory, Department of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Kelly K Chung
- Neuroprotection Research Laboratory, Department of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Emiri T Mandeville
- Neuroprotection Research Laboratory, Department of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Josephine Lok
- Neuroprotection Research Laboratory, Department of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Myriam Fornage
- Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States.,Human Genetics Center, Division of Epidemiology, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Karl Herrup
- Department of Neurobiology and ADRC, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kai-Hei Tse
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Eng H Lo
- Neuroprotection Research Laboratory, Department of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| | - Ken Arai
- Neuroprotection Research Laboratory, Department of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States
| |
Collapse
|
26
|
Feng T, Yamashita T, Sasaki R, Tadokoro K, Matsumoto N, Hishikawa N, Abe K. Protective effects of edaravone on white matter pathology in a novel mouse model of Alzheimer's disease with chronic cerebral hypoperfusion. J Cereb Blood Flow Metab 2021; 41:1437-1448. [PMID: 33106078 PMCID: PMC8142121 DOI: 10.1177/0271678x20968927] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
White matter lesions (WMLs) caused by cerebral chronic hypoperfusion (CCH) may contribute to the pathophysiology of Alzheimer's disease (AD). However, the underlying mechanisms and therapeutic approaches have yet to be totally identified. In the present study, we investigated a potential therapeutic effect of the free radical scavenger edaravone (EDA) on WMLs in our previously reported novel mouse model of AD (APP23) plus CCH with motor and cognitive deficits. Relative to AD with CCH mice at 12 months (M) of age, EDA strongly improved CCH-induced WMLs in the corpus callosum of APP23 mice at 12 M by improving the disruption of white matter integrity, enhancing the proliferation of oligodendrocyte progenitor cells, attenuating endothelium/astrocyte unit dysfunction, and reducing neuroinflammation and oxidative stress. The present study demonstrates that the long-term administration of EDA may provide a promising therapeutic approach for WMLs in AD plus CCH disease with cognitive deficits.
Collapse
Affiliation(s)
- Tian Feng
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Toru Yamashita
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Ryo Sasaki
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Koh Tadokoro
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Namiko Matsumoto
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Nozomi Hishikawa
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Koji Abe
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
27
|
Jiang T, Luo J, Pan X, Zheng H, Yang H, Zhang L, Hu X. Physical exercise modulates the astrocytes polarization, promotes myelin debris clearance and remyelination in chronic cerebral hypoperfusion rats. Life Sci 2021; 278:119526. [PMID: 33894268 DOI: 10.1016/j.lfs.2021.119526] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 04/01/2021] [Accepted: 04/13/2021] [Indexed: 10/21/2022]
Abstract
AIMS White matter damage is the main pathological feature of chronic cerebral hypoperfusion (CCH) and glial activation is crucial in this process. Physical exercise has protective effects on CCH, but the mechanism is unclear. Therefore, this study focuses on investigating the influence of physical exercise on activated astrocytes polarization and its role in CCH. MAIN METHODS Rats were given wheel running 48 h after 2VO (2 vessel occlusion) surgery. The cognitive function was evaluated by Morris water maze and novel object recognition test. Inflammatory cytokines expressions were detected by ELISA. Astrocytes polarization was analyzed by immunofluorescence. Myelin debris clearance and remyelination were detected by immunofluorescence and transmission electron microscopy. KEY FINDINGS Astrocytes were activated and mainly switched to A1 phenotype in rats 2 and 3 months after 2VO. Myelin debris deposition and limited remyelination can be observed at the corresponding time. Whereas physical exercise can improve the cognitive function of 2VO rats, downregulate the expression of inflammatory factors IL-1α, C1q and TNF, upregulate the release of TGFβ, and promote activated astrocytes transformation from A1 to A2 phenotype. In addition, it can also enhance myelin debris removal and remyelination. SIGNIFICANCE These findings suggest that the benefits of physical exercise on white matter repair and cognition improvement may be related to its regulation of astrocytes polarization, which contributes to myelin debris clearance and effective remyelination in CCH.
Collapse
Affiliation(s)
- Ting Jiang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China; Department of Neurorehabilitation, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Jing Luo
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Xiaona Pan
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Haiqing Zheng
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Huaichun Yang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Liying Zhang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China.
| | - Xiquan Hu
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China.
| |
Collapse
|
28
|
Cui Y, Jin X, Choi JY, Kim BG. Modeling subcortical ischemic white matter injury in rodents: unmet need for a breakthrough in translational research. Neural Regen Res 2021; 16:638-642. [PMID: 33063714 PMCID: PMC8067929 DOI: 10.4103/1673-5374.295313] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Subcortical ischemic white matter injury (SIWMI), pathological correlate of white matter hyperintensities or leukoaraiosis on magnetic resonance imaging, is a common cause of cognitive decline in elderly. Despite its high prevalence, it remains unknown how various components of the white matter degenerate in response to chronic ischemia.This incomplete knowledge is in part due to a lack of adequate animal model. The current review introduces various SIWMI animal models and aims to scrutinize their advantages and disadvantages primarily in regard to the pathological manifestations of white matter components. The SIWMI animal models are categorized into 1) chemically induced SIWMI models, 2) vascular occlusive SIWMI models, and 3) SIWMI models with comorbid vascular risk factors. Chemically induced models display consistent lesions in predetermined areas of the white matter, but the abrupt evolution of lesions does not appropriately reflect the progressive pathological processes in human white matter hyperintensities. Vascular occlusive SIWMI models often do not exhibit white matter lesions that are sufficiently unequivocal to be quantified. When combined with comorbid vascular risk factors (specifically hypertension), however, they can produce progressive and definitive white matter lesions including diffuse rarefaction, demyelination, loss of oligodendrocytes, and glial activation, which are by far the closest to those found in human white matter hyperintensities lesions. However, considerable surgical mortality and unpredictable natural deaths during a follow-up period would necessitate further refinements in these models. In the meantime, in vitro SIWMI models that recapitulate myelinated white matter track may be utilized to study molecular mechanisms of the ischemic white matter injury. Appropriate in vivo and in vitro SIWMI models will contribute in a complementary manner to making a breakthrough in developing effective treatment to prevent progression of white matter hyperintensities.
Collapse
Affiliation(s)
- Yuexian Cui
- Department of Brain Science, Ajou University School of Medicine; Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Korea; Department of Neurology, Yanbian University Hospital, Yanji, Jilin Province, China
| | - Xuelian Jin
- Department of Brain Science, Ajou University School of Medicine; Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Korea; Department of Nephrology, Suqian First Hospital, Suqian, Jiangsu Province, China
| | - Jun Young Choi
- Department of Brain Science, Ajou University School of Medicine; Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine; Department of Neurology, Ajou University School of Medicine, Suwon, Korea
| | - Byung Gon Kim
- Department of Brain Science, Ajou University School of Medicine; Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine; Department of Neurology, Ajou University School of Medicine, Suwon, Korea
| |
Collapse
|
29
|
Cullen CL, Pepper RE, Clutterbuck MT, Pitman KA, Oorschot V, Auderset L, Tang AD, Ramm G, Emery B, Rodger J, Jolivet RB, Young KM. Periaxonal and nodal plasticities modulate action potential conduction in the adult mouse brain. Cell Rep 2021; 34:108641. [PMID: 33472075 DOI: 10.1016/j.celrep.2020.108641] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 11/18/2020] [Accepted: 12/21/2020] [Indexed: 12/25/2022] Open
Abstract
Central nervous system myelination increases action potential conduction velocity. However, it is unclear how myelination is coordinated to ensure the temporally precise arrival of action potentials and facilitate information processing within cortical and associative circuits. Here, we show that myelin sheaths, supported by mature oligodendrocytes, remain plastic in the adult mouse brain and undergo subtle structural modifications to influence action potential conduction velocity. Repetitive transcranial magnetic stimulation and spatial learning, two stimuli that modify neuronal activity, alter the length of the nodes of Ranvier and the size of the periaxonal space within active brain regions. This change in the axon-glial configuration is independent of oligodendrogenesis and robustly alters action potential conduction velocity. Because aptitude in the spatial learning task was found to correlate with action potential conduction velocity in the fimbria-fornix pathway, modifying the axon-glial configuration may be a mechanism that facilitates learning in the adult mouse brain.
Collapse
Affiliation(s)
- Carlie L Cullen
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Renee E Pepper
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | | | - Kimberley A Pitman
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Viola Oorschot
- Ramaciotti Centre for Cryo-Electron Microscopy, Monash University, Melbourne, VIC 3800, Australia
| | - Loic Auderset
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia
| | - Alexander D Tang
- Experimental and Regenerative Neuroscience, School of Biological Sciences, University of Western Australia, Perth, WA 6009, Australia
| | - Georg Ramm
- Ramaciotti Centre for Cryo-Electron Microscopy, Monash University, Melbourne, VIC 3800, Australia
| | - Ben Emery
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health and Science University, Portland, OR 97239-3098, USA
| | - Jennifer Rodger
- Experimental and Regenerative Neuroscience, School of Biological Sciences, University of Western Australia, Perth, WA 6009, Australia; Perron Institute for Neurological and Translational Research, Perth, WA 6009, Australia
| | - Renaud B Jolivet
- Département de Physique Nucléaire et Corpusculaire, University of Geneva, 1211 Geneva 4, Switzerland
| | - Kaylene M Young
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7000, Australia.
| |
Collapse
|
30
|
Quick S, Moss J, Rajani RM, Williams A. A Vessel for Change: Endothelial Dysfunction in Cerebral Small Vessel Disease. Trends Neurosci 2020; 44:289-305. [PMID: 33308877 DOI: 10.1016/j.tins.2020.11.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/24/2020] [Accepted: 11/11/2020] [Indexed: 01/08/2023]
Abstract
The blood vessels of the brain are lined with endothelial cells and it has been long known that these help to regulate blood flow to the brain. However, there is increasing evidence that these cells also interact with the surrounding brain tissue. These interactions change when the endothelial cells become dysfunctional and have an impact in diseases such as cerebral small vessel disease, the leading cause of vascular dementia. In this review, we focus on what endothelial dysfunction is, what causes it, how it leads to surrounding brain pathology, how researchers can investigate it with current models, and where this might lead in the future for dementia therapies.
Collapse
Affiliation(s)
- Sophie Quick
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Jonathan Moss
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Rikesh M Rajani
- UK Dementia Research Institute at UCL, University College London, London, UK
| | - Anna Williams
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK.
| |
Collapse
|
31
|
Sigfridsson E, Marangoni M, Hardingham GE, Horsburgh K, Fowler JH. Deficiency of Nrf2 exacerbates white matter damage and microglia/macrophage levels in a mouse model of vascular cognitive impairment. J Neuroinflammation 2020; 17:367. [PMID: 33261626 PMCID: PMC7709339 DOI: 10.1186/s12974-020-02038-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 11/17/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Chronic cerebral hypoperfusion causes damage to the brain's white matter underpinning vascular cognitive impairment. Inflammation and oxidative stress have been proposed as key pathophysiological mechanisms of which the transcription factor Nrf2 is a master regulator. We hypothesised that white matter pathology, microgliosis, blood-brain barrier breakdown and behavioural deficits induced by chronic hypoperfusion would be exacerbated in mice deficient in the transcription factor Nrf2. METHODS Mice deficient in Nrf2 (male heterozygote or homozygous for Nrf2 knockout) or wild-type littermates on a C57Bl6/J background underwent bilateral carotid artery stenosis (BCAS) to induce chronic cerebral hypoperfusion or sham surgery and survived for a further 6 weeks. White matter pathology was assessed with MAG immunohistochemistry as a marker of altered axon-glial integrity; alterations to astrocytes and microglia/macrophages were assessed with GFAP and Iba1 immunohistochemistry, and blood-brain barrier breakdown was assessed with IgG immunohistochemistry. Behavioural alterations were assessed using 8-arm radial arm maze, and alterations to Nrf2-related and inflammatory-related genes were assessed with qRT-PCR. RESULTS Chronic cerebral hypoperfusion induced white matter pathology, elevated microglial/macrophage levels and blood-brain barrier breakdown in white matter tracts that were increased in Nrf2+/- mice and further exacerbated by the complete absence of Nrf2. Chronic hypoperfusion induced white matter astrogliosis and induced an impairment in behaviour assessed with radial arm maze; however, these measures were not affected by Nrf2 deficiency. Although Nrf2-related antioxidant gene expression was not altered by chronic cerebral hypoperfusion, there was evidence for elevated pro-inflammatory related gene expression following chronic hypoperfusion that was not affected by Nrf2 deficiency. CONCLUSIONS The results demonstrate that the absence of Nrf2 exacerbates white matter pathology and microgliosis following cerebral hypoperfusion but does not affect behavioural impairment.
Collapse
Affiliation(s)
- Emma Sigfridsson
- Centre for Discovery Brain Sciences, University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK
| | - Martina Marangoni
- Centre for Discovery Brain Sciences, University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK
- Present address: Department of Health Sciences, University of Florence, Florence, Italy
| | - Giles E. Hardingham
- Centre for Discovery Brain Sciences, University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK
- The UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Karen Horsburgh
- Centre for Discovery Brain Sciences, University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK
| | - Jill H. Fowler
- Centre for Discovery Brain Sciences, University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK
| |
Collapse
|
32
|
Lana D, Ugolini F, Giovannini MG. An Overview on the Differential Interplay Among Neurons-Astrocytes-Microglia in CA1 and CA3 Hippocampus in Hypoxia/Ischemia. Front Cell Neurosci 2020; 14:585833. [PMID: 33262692 PMCID: PMC7686560 DOI: 10.3389/fncel.2020.585833] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 10/09/2020] [Indexed: 12/13/2022] Open
Abstract
Neurons have been long regarded as the basic functional cells of the brain, whereas astrocytes and microglia have been regarded only as elements of support. However, proper intercommunication among neurons-astrocytes-microglia is of fundamental importance for the functional organization of the brain. Perturbation in the regulation of brain energy metabolism not only in neurons but also in astrocytes and microglia may be one of the pathophysiological mechanisms of neurodegeneration, especially in hypoxia/ischemia. Glial activation has long been considered detrimental for survival of neurons, but recently it appears that glial responses to an insult are not equal but vary in different brain areas. In this review, we first take into consideration the modifications of the vascular unit of the glymphatic system and glial metabolism in hypoxic conditions. Using the method of triple-labeling fluorescent immunohistochemistry coupled with confocal microscopy (TIC), we recently studied the interplay among neurons, astrocytes, and microglia in chronic brain hypoperfusion. We evaluated the quantitative and morpho-functional alterations of the neuron-astrocyte-microglia triads comparing the hippocampal CA1 area, more vulnerable to ischemia, to the CA3 area, less vulnerable. In these contiguous and interconnected areas, in the same experimental hypoxic conditions, astrocytes and microglia show differential, finely regulated, region-specific reactivities. In both areas, astrocytes and microglia form triad clusters with apoptotic, degenerating neurons. In the neuron-astrocyte-microglia triads, the cell body of a damaged neuron is infiltrated and bisected by branches of astrocyte that create a microscar around it while a microglial cell phagocytoses the damaged neuron. These coordinated actions are consistent with the scavenging and protective activities of microglia. In hypoxia, the neuron-astrocyte-microglia triads are more numerous in CA3 than in CA1, further indicating their protective effects. These data, taken from contiguous and interconnected hippocampal areas, demonstrate that glial response to the same hypoxic insult is not equal but varies significantly. Understanding the differences of glial reactivity is of great interest to explain the differential susceptibility of hippocampal areas to hypoxia/ischemia. Further studies may evidence the differential reactivity of glia in different brain areas, explaining the higher or lower sensitivity of these areas to different insults and whether glia may represent a target for future therapeutic interventions.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Filippo Ugolini
- Department of Health Sciences, Section of Anatomopathology, University of Florence, Florence, Italy
| | - Maria G Giovannini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| |
Collapse
|
33
|
Chen M, Yang LL, Hu ZW, Qin C, Zhou LQ, Duan YL, Bosco DB, Wu LJ, Zhan KB, Xu SB, Tian DS. Deficiency of microglial Hv1 channel is associated with activation of autophagic pathway and ROS production in LPC-induced demyelination mouse model. J Neuroinflammation 2020; 17:333. [PMID: 33158440 PMCID: PMC7646080 DOI: 10.1186/s12974-020-02020-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 10/30/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is an immune-mediated demyelinated disease of the central nervous system. Activation of microglia is involved in the pathogenesis of myelin loss. OBJECTIVE This study is focused on the role of Hv1 in regulating demyelination and microglial activation through reactive oxygen species (ROS) production after lysophosphatidylcholine (LPC)-mediated demyelination. We also explored autophagy in this process. METHODS A model of demyelination using two-point LPC injection into the corpus callosum was established. LFB staining, immunofluorescence, Western blot, and electron microscopy were used to study the severity of demyelination. Microglial phenotype and autophagy were detected by immunofluorescence and Western blot. Morris water maze was used to test spatial learning and memory ability. RESULTS We have identified that LPC-mediated myelin damage was reduced by Hv1 deficiency. Furthermore, we found that ROS and autophagy of microglia increased in the demyelination region, which was also inhibited by Hv1 knockout. CONCLUSION These results suggested that microglial Hv1 deficiency ameliorates demyelination through inhibition of ROS-mediated autophagy and microglial phenotypic transformation.
Collapse
Affiliation(s)
- Man Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
| | - Lin-Lin Yang
- Department of Neurology, Second Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Zi-Wei Hu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
| | - Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
| | - Luo-Qi Zhou
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China
| | - Ya-Ling Duan
- Department of Neurology, Second Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, People's Republic of China
| | - Dale B Bosco
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Ke-Bin Zhan
- Department of Neurology, Second Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, People's Republic of China.
| | - Sha-Bei Xu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China.
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, People's Republic of China.
| |
Collapse
|
34
|
Yang T, Zhang F. Targeting Transcription Factor Nrf2 (Nuclear Factor Erythroid 2-Related Factor 2) for the Intervention of Vascular Cognitive Impairment and Dementia. Arterioscler Thromb Vasc Biol 2020; 41:97-116. [PMID: 33054394 DOI: 10.1161/atvbaha.120.314804] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Vascular cognitive impairment and dementia (VCID) is an age-related, mild to severe mental disability due to a broad panel of cerebrovascular disorders. Its pathobiology involves neurovascular dysfunction, blood-brain barrier disruption, white matter damage, microRNAs, oxidative stress, neuroinflammation, and gut microbiota alterations, etc. Nrf2 (Nuclear factor erythroid 2-related factor 2) is the master regulator of redox status and controls the transcription of a panel of antioxidative and anti-inflammatory genes. By interacting with NF-κB (nuclear factor-κB), Nrf2 also fine-tunes the cellular oxidative and inflammatory balance. Aging is associated with Nrf2 dysfunction, and increasing evidence has proved the role of Nrf2 in mitigating the VCID process. Based on VCID pathobiologies and Nrf2 studies from VCID and other brain diseases, we point out several hypothetical Nrf2 targets for VCID management, including restoration of endothelial function and neurovascular coupling, preservation of blood-brain barrier integrity, reduction of amyloidopathy, promoting white matter integrity, and mitigating oxidative stress and neuroinflammation. Collectively, the Nrf2 pathway could be a promising direction for future VCID research. Targeting Nrf2 would shed light on VCID managing strategies.
Collapse
Affiliation(s)
- Tuo Yang
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, PA
| | - Feng Zhang
- Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, PA
| |
Collapse
|
35
|
Lubetzki C, Sol-Foulon N, Desmazières A. Nodes of Ranvier during development and repair in the CNS. Nat Rev Neurol 2020; 16:426-439. [DOI: 10.1038/s41582-020-0375-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2020] [Indexed: 01/01/2023]
|
36
|
Regenhardt RW, Takase H, Lo EH, Lin DJ. Translating concepts of neural repair after stroke: Structural and functional targets for recovery. Restor Neurol Neurosci 2020; 38:67-92. [PMID: 31929129 PMCID: PMC7442117 DOI: 10.3233/rnn-190978] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Stroke is among the most common causes of adult disability worldwide, and its disease burden is shifting towards that of a long-term condition. Therefore, the development of approaches to enhance recovery and augment neural repair after stroke will be critical. Recovery after stroke involves complex interrelated systems of neural repair. There are changes in both structure (at the molecular, cellular, and tissue levels) and function (in terms of excitability, cortical maps, and networks) that occur spontaneously within the brain. Several approaches to augment neural repair through enhancing these changes are under study. These include identifying novel drug targets, implementing rehabilitation strategies, and developing new neurotechnologies. Each of these approaches has its own array of different proposed mechanisms. Current investigation has emphasized both cellular and circuit-based targets in both gray and white matter, including axon sprouting, dendritic branching, neurogenesis, axon preservation, remyelination, blood brain barrier integrity, blockade of extracellular inhibitory signals, alteration of excitability, and promotion of new brain cortical maps and networks. Herein, we review for clinicians recovery after stroke, basic elements of spontaneous neural repair, and ongoing work to augment neural repair. Future study requires alignment of basic, translational, and clinical research. The field continues to grow while becoming more clearly defined. As thrombolysis changed stroke care in the 1990 s and thrombectomy in the 2010 s, the augmentation of neural repair and recovery after stroke may revolutionize care for these patients in the coming decade.
Collapse
Affiliation(s)
- Robert W Regenhardt
- Department of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
| | - Hajime Takase
- Department of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
| | - Eng H Lo
- Department of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
- Department of Radiology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
| | - David J Lin
- Department of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
- Center for Neurotechnology and Neurorecovery, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114
| |
Collapse
|
37
|
Yu Y, Luo X, Li C, Ding F, Wang M, Xie M, Yu Z, Ransom BR, Wang W. Microglial Hv1 proton channels promote white matter injuries after chronic hypoperfusion in mice. J Neurochem 2019; 152:350-367. [PMID: 31769505 DOI: 10.1111/jnc.14925] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 11/16/2019] [Accepted: 11/19/2019] [Indexed: 12/30/2022]
Abstract
Microglia are critical in damage/repair processes during ischemic white matter injury (WMI). Voltage-gated proton channel (Hv1) is expressed in microglia and contributes to nicotinamide adenine dinucleotide phosphate oxidase complex-dependent production of reactive oxygen species (ROS). Recent findings have shown that Hv1 is involved in regulating luminal pH of M1-polarized microglial phagosomes and inhibits endocytosis in microglia. We previously reported that Hv1 facilitated production of ROS and pro-inflammatory cytokines in microglia and enhanced damage to oligodendrocyte progenitor cells from oxygen and glucose deprivation. To investigate the role of Hv1 in hypoperfusion-induced WMI, we employed mice that were genetically devoid of Hv1 (Hv1-/- ), as well as a model of subcortical vascular dementia via bilateral common carotid artery stenosis. Integrity of myelin was assessed using immunofluorescent staining and transmission electron microscopy, while cognitive impairment was assessed using an eight-arm radial maze test. Hv1 deficiency was found to attenuate bilateral common carotid artery stenosis-induced disruption of white matter integrity and impairment of working memory. Immunofluorescent staining and western blotting were used to assay changes in oligodendrocytes, OPCs, and microglial polarization. Compared with that in wild-type (WT) mice, Hv1-/- mice exhibited reduced ROS generation, decreased pro-inflammatory cytokines production, and an M2-dominant rather than M1-dominant microglial polarization. Furthermore, Hv1-/- mice exhibited enhanced OPC proliferation and differentiation into oligodendrocytes. Results of mouse-derived microglia-OPC co-cultures suggested that PI3K/Akt signaling was involved in Hv1-deficiency-induced M2-type microglial polarization and concomitant OPC differentiation. These results suggest that microglial Hv1 is a promising therapeutic target for reducing ischemic WMI and cognitive impairment.
Collapse
Affiliation(s)
- Ying Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Luo
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunyu Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengfei Ding
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Minghuan Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Minjie Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiyuan Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bruce R Ransom
- Department of Neurology, University of Washington School of Medicine HMC, Seattle, WA, USA
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
38
|
Chapman TW, Hill RA. Myelin plasticity in adulthood and aging. Neurosci Lett 2019; 715:134645. [PMID: 31765728 DOI: 10.1016/j.neulet.2019.134645] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/06/2019] [Accepted: 11/21/2019] [Indexed: 12/31/2022]
Abstract
The central nervous system maintains the potential for molecular and cellular plasticity throughout life. This flexibility underlies fundamental features of neural circuitry including the brain's ability to sense, store, and properly adapt to everchanging external stimuli on time scales from seconds to years. Evidence for most forms of plasticity are centered around changes in neuronal structure and synaptic strength, however recent data suggests that myelinating oligodendrocytes exhibit certain forms of plasticity in the adult. This plasticity ranges from the generation of entirely new myelinating cells to more subtle changes in myelin sheath length, thickness, and distribution along axons. The extent to which these changes dynamically modify axonal function and neural circuitry and whether they are directly related to mechanisms of learning and memory remains an open question. Here we describe different forms of myelin plasticity, highlight some recent evidence for changes in myelination throughout life, and discuss how defects in these forms of plasticity could be associated with cognitive decline in aging.
Collapse
Affiliation(s)
- Timothy W Chapman
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - Robert A Hill
- Department of Biological Sciences, Dartmouth College, Hanover, NH, USA.
| |
Collapse
|
39
|
Zhang SQ, Ding FF, Liu Q, Tian YY, Wang W, Qin C. Autophagy inhibition exerts neuroprotection on white matter ischemic damage after chronic cerebral hypoperfusion in mice. Brain Res 2019; 1721:146337. [PMID: 31319064 DOI: 10.1016/j.brainres.2019.146337] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 07/12/2019] [Accepted: 07/13/2019] [Indexed: 12/13/2022]
Abstract
Autophagy plays vital roles in the pathophysiology of many central nervous system diseases. Emerging evidence indicates that autophagy has both detrimental and protective effects in ischemic cerebral injury. This study aimed to investigate the temporal pattern of autophagy activation in the white matter of bilateral common carotid artery stenosis (BCAS) mouse model by immunofluorescence and western blotting. The effect of wortmannin, an autophagy inhibitor, against hypoperfusion induced white matter injury (WMI) was studied by immunofluorescence and eight-arm radial maze test. We found that autophagy was initially activated in the white matter 3 days after BCAS, and then suppressed by day 10, and was activated again at day 30. Administration of wortmannin during the first three days after BCAS revealed protective effects on axon-glia integrity and against the cognitive injury induced by the chronic hypoperfusion. The results indicated the possible link between autophagy and white matter ischemic damage after chronic cerebral hypoperfusion. Modulation of autophagy in a time course dependent manner may broaden the insight on the treatment of WMI.
Collapse
Affiliation(s)
- Shuo-Qi Zhang
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Feng-Fei Ding
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Qian Liu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Ye-Ye Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China
| | - Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, PR China.
| |
Collapse
|
40
|
Wang M, Qin C, Luo X, Wang J, Wang X, Xie M, Hu J, Cao J, Hu T, Goldman SA, Nedergaard M, Wang W. Astrocytic connexin 43 potentiates myelin injury in ischemic white matter disease. Am J Cancer Res 2019; 9:4474-4493. [PMID: 31285774 PMCID: PMC6599652 DOI: 10.7150/thno.31942] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 04/19/2019] [Indexed: 01/05/2023] Open
Abstract
Rational: Myelin loss is a characteristic feature of both ischemic white matter disease and its associated vascular dementia, and is a hallmark of chronic cerebral hypoperfusion due to carotid artery stenosis. Yet the cellular mechanisms involved in ischemic dysmyelination are not well-understood, and no effective treatment has emerged to prevent or slow hypoperfusion-related demyelination. In a study employing the bilateral common carotid artery stenosis (BCAS) mouse model, we found reduced cerebral blood flow velocity and arteriolar pulsatility, and confirmed that prolonged BCAS provoked myelin disruption. These pathological features were associated with marked cognitive decline, in the absence of evident damage to axons. Methods: To assess the role of astroglial communication in BCAS-associated demyelination, we investigated the effect of deleting or inhibiting connexin 43 (Cx43), a constituent of astroglial gap junctions and hemichannels. Results: Genetic deletion and pharmacological inhibition of gap junctions both protected myelin integrity and rescued cognitive decline in the BCAS-treated mice. Gap junction inhibition also suppressed the transient increase in extracellular glutamate observed in the callosal white matter of wild-type mice exposed to BCAS. Conclusion: These findings suggest that astrocytic Cx43 may be a viable target for attenuating the demyelination and cognitive decline associated with chronic cerebral hypoperfusion.
Collapse
|
41
|
Wang F, Cao Y, Ma L, Pei H, Rausch WD, Li H. Dysfunction of Cerebrovascular Endothelial Cells: Prelude to Vascular Dementia. Front Aging Neurosci 2018; 10:376. [PMID: 30505270 PMCID: PMC6250852 DOI: 10.3389/fnagi.2018.00376] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 10/29/2018] [Indexed: 12/19/2022] Open
Abstract
Vascular dementia (VaD) is the second most common type of dementia after Alzheimer's disease (AD), characterized by progressive cognitive impairment, memory loss, and thinking or speech problems. VaD is usually caused by cerebrovascular disease, during which, cerebrovascular endothelial cells (CECs) are vulnerable. CEC dysfunction occurs before the onset of VaD and can eventually lead to dysregulation of cerebral blood flow and blood-brain barrier damage, followed by the activation of glia and inflammatory environment in the brain. White matter, neuronal axons, and synapses are compromised in this process, leading to cognitive impairment. The present review summarizes the mechanisms underlying CEC impairment during hypoperfusion and pathological role of CECs in VaD. Through the comprehensive examination and summarization, endothelial nitric oxide synthase (eNOS)/nitric oxide (NO) signaling pathway, Ras homolog gene family member A (RhoA) signaling pathway, and CEC-derived caveolin-1 (CAV-1) are proposed to serve as targets of new drugs for the treatment of VaD.
Collapse
Affiliation(s)
- Feixue Wang
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Yu Cao
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Lina Ma
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Hui Pei
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Wolf Dieter Rausch
- Department for Biomedical Sciences, Institute of Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Hao Li
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
42
|
Arena F, Bardini P, Blasi F, Gianolio E, Marini GM, La Cava F, Valbusa G, Aime S. Gadolinium presence, MRI hyperintensities, and glucose uptake in the hypoperfused rat brain after repeated administrations of gadodiamide. Neuroradiology 2018; 61:163-173. [DOI: 10.1007/s00234-018-2120-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 10/19/2018] [Indexed: 12/30/2022]
|
43
|
Qin C, Liu Q, Hu ZW, Zhou LQ, Shang K, Bosco DB, Wu LJ, Tian DS, Wang W. Microglial TLR4-dependent autophagy induces ischemic white matter damage via STAT1/6 pathway. Theranostics 2018; 8:5434-5451. [PMID: 30555556 PMCID: PMC6276098 DOI: 10.7150/thno.27882] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 10/03/2018] [Indexed: 12/28/2022] Open
Abstract
Rationale: Ischemic white matter damage frequently results in myelin loss, accompanied with microglial activation. We previously found that directing microglia towards an anti-inflammatory phenotype provided a beneficial microenvironment and helped maintain white matter integrity during chronic cerebral hypoperfusion. However, the molecular mechanisms underlying microglial polarization remain elusive. Methods: Hypoperfusion induced white matter damage mice model and lipopolysaccharide (LPS) induced primary cultured microglia were established. Autophagy activation in microglia was detected both in vivo and in vitro by immunofluorescence, Western blot and electron microscopy. Autophagy inhibitors/agonist were administrated to investigate the role of autophagic process in modulating microglial phenotypes. Quantitative real time-polymerase chain reaction and Western blot were carried out to investigate the possible pathway. Results: We identified rapid accumulation of autophagosomes in primary cultured microglia exposed to LPS and within activated microglia during white matter ischemic damage. Autophagy inhibitors switched microglial function from pro-inflammatory to anti-inflammatory phenotype. Furthermore, we found TLR4, one of the major receptors binding LPS, was most highly expressed on microglia in corpus callosum during white matter ischemic damage, and TLR4 deficiency could mimic the phenomenon in microglial functional transformation, and exhibit a protective activity in chronic cerebral hypoperfusion. Whereas, the anti-inflammatory phenotype of microglia in TLR4 deficiency group was largely abolished by the activation of autophagic process. Finally, our transcriptional analysis confirmed that the up-regulation of STAT1 and down-regulation of STAT6 in microglia exposure to LPS could be reversed by autophagy inhibition. Conclusion: These results indicated that TLR4-dependent autophagy regulates microglial polarization and induces ischemic white matter damage via STAT1/6 pathway.
Collapse
|
44
|
Comparison of ion channel inhibitor combinations for limiting secondary degeneration following partial optic nerve transection. Exp Brain Res 2018; 237:161-171. [DOI: 10.1007/s00221-018-5414-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 10/21/2018] [Indexed: 11/25/2022]
|
45
|
Sigfridsson E, Marangoni M, Johnson JA, Hardingham GE, Fowler JH, Horsburgh K. Astrocyte-specific overexpression of Nrf2 protects against optic tract damage and behavioural alterations in a mouse model of cerebral hypoperfusion. Sci Rep 2018; 8:12552. [PMID: 30135571 PMCID: PMC6105641 DOI: 10.1038/s41598-018-30675-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 07/30/2018] [Indexed: 12/19/2022] Open
Abstract
Mouse models have shown that cerebral hypoperfusion causes white matter disruption and memory impairment relevant to the study of vascular cognitive impairment and dementia. The associated mechanisms include inflammation and oxidative stress are proposed to drive disruption of myelinated axons within hypoperfused white matter. The aim of this study was to determine if increased endogenous anti-oxidant and anti-inflammatory signalling in astrocytes was protective in a model of mild cerebral hypoperfusion. Transgenically altered mice overexpressing the transcription factor Nrf2 (GFAP-Nrf2) and wild type littermates were subjected to bilateral carotid artery stenosis or sham surgery. Behavioural alterations were assessed using the radial arm maze and tissue was collected for pathology and transcriptome analysis six weeks post-surgery. GFAP-Nrf2 mice showed less pronounced behavioural impairments compared to wild types following hypoperfusion, paralleled by reduced optic tract white matter disruption and astrogliosis. There was no effect of hypoperfusion on anti-oxidant gene alterations albeit the levels were increased in GFAP-Nrf2 mice. Instead, pro-inflammatory gene expression was determined to be significantly upregulated in the optic tract of hypoperfused wild type mice but differentially affected in GFAP-Nrf2 mice. In particular, complement components (C4 and C1q) were increased in wild type hypoperfused mice but expressed at levels similar to controls in hypoperfused GFAP-Nrf2 mice. This study provides evidence that overexpression of Nrf2 in astrocytes exerts beneficial effects through repression of inflammation and supports the potential use of Nrf2-activators in the amelioration of cerebrovascular-related inflammation and white matter degeneration.
Collapse
Affiliation(s)
- Emma Sigfridsson
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Martina Marangoni
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Jeffrey A Johnson
- Division of Pharmaceutical Sciences, University of Wisconsin, Madison, USA
- Molecular and Environmental Toxicology Center, University of Wisconsin, Madison, USA
- Center for Neuroscience, University of Wisconsin, Madison, USA
- Waisman Center, University of Wisconsin, Madison, USA
| | - Giles E Hardingham
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
- The UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Jill H Fowler
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK.
| | - Karen Horsburgh
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
46
|
Fowler JH, McQueen J, Holland PR, Manso Y, Marangoni M, Scott F, Chisholm E, Scannevin RH, Hardingham GE, Horsburgh K. Dimethyl fumarate improves white matter function following severe hypoperfusion: Involvement of microglia/macrophages and inflammatory mediators. J Cereb Blood Flow Metab 2018; 38:1354-1370. [PMID: 28606007 PMCID: PMC6077928 DOI: 10.1177/0271678x17713105] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The brain's white matter is highly vulnerable to reductions in cerebral blood flow via mechanisms that may involve elevated microgliosis and pro-inflammatory pathways. In the present study, the effects of severe cerebral hypoperfusion were investigated on white matter function and inflammation. Male C57Bl/6J mice underwent bilateral common carotid artery stenosis and white matter function was assessed at seven days with electrophysiology in response to evoked compound action potentials (CAPs) in the corpus callosum. The peak latency of CAPs and axonal refractoriness was increased following hypoperfusion, indicating a marked functional impairment in white matter, which was paralleled by axonal and myelin pathology and increased density and numbers of microglia/macrophages. The functional impairment in peak latency was significantly correlated with increased microglia/macrophages. Dimethyl fumarate (DMF; 100 mg/kg), a drug with anti-inflammatory properties, was found to reduce peak latency but not axonal refractoriness. DMF had no effect on hypoperfusion-induced axonal and myelin pathology. The density of microglia/macrophages was significantly increased in vehicle-treated hypoperfused mice, whereas DMF-treated hypoperfused mice had similar levels to that of sham-treated mice. The study suggests that increased microglia/macrophages following cerebral hypoperfusion contributes to the functional impairment in white matter that may be amenable to modulation by DMF.
Collapse
Affiliation(s)
- Jill H Fowler
- 1 Centre for Neuroregeneration, University of Edinburgh, Edinburgh, UK
| | - Jamie McQueen
- 1 Centre for Neuroregeneration, University of Edinburgh, Edinburgh, UK.,2 Centre for Integrative Physiology, University of Edinburgh, Edinburgh, UK
| | - Philip R Holland
- 1 Centre for Neuroregeneration, University of Edinburgh, Edinburgh, UK.,3 Current Address: Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Yasmina Manso
- 1 Centre for Neuroregeneration, University of Edinburgh, Edinburgh, UK.,4 Current Address: Developmental Neurobiology and Regeneration Lab, Parc Científic de Barcelona, Spain
| | - Martina Marangoni
- 1 Centre for Neuroregeneration, University of Edinburgh, Edinburgh, UK.,5 Current Address: Department of Health Sciences, University of Florence, Florence, Italy
| | - Fiona Scott
- 1 Centre for Neuroregeneration, University of Edinburgh, Edinburgh, UK
| | - Emma Chisholm
- 1 Centre for Neuroregeneration, University of Edinburgh, Edinburgh, UK
| | | | - Giles E Hardingham
- 2 Centre for Integrative Physiology, University of Edinburgh, Edinburgh, UK.,7 The UK Dementia Research Institute at The University of Edinburgh
| | - Karen Horsburgh
- 1 Centre for Neuroregeneration, University of Edinburgh, Edinburgh, UK.,8 Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
47
|
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
|
48
|
Bordeleau M, ElAli A, Rivest S. Severe chronic cerebral hypoperfusion induces microglial dysfunction leading to memory loss in APPswe/PS1 mice. Oncotarget 2017; 7:11864-80. [PMID: 26918610 PMCID: PMC4914254 DOI: 10.18632/oncotarget.7689] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 02/05/2016] [Indexed: 12/12/2022] Open
Abstract
Cerebral vasculature plays a key role in controlling brain homeostasis. Cerebral vasculature dysfunction, associated to irregularities in cerebral blood perfusion, has been proposed to directly contribute to Alzheimer's disease (AD) pathogenesis. More precisely, chronic cerebral hypoperfusion, which impairs brain homeostasis, was demonstrated to take place even before cognitive decline. However, the mechanisms underlying the implication of chronic cerebral hypoperfusion in AD pathogenesis remain elusive. Therefore, this study aims at investigating the role of severe chronic cerebral hypoperfusion (SCCH) in AD pathogenesis. For this purpose, SCCH was induced in young APPswe/PS1 in order to evaluate the progression of AD-like pathology in these mice. We observed that SCCH accelerated the cognitive decline of young APPswe/PS1 mice, which was associated with an increased amyloid plaque number in brain parenchyma. In addition, SCCH reduced the activity of extracellular signal-regulated kinases 1/2 (ERK1/2), which has been shown to play an important role in the adaptive responses of neurons. Importantly, SCCH impaired the function of microglial cells, which are implicated in amyloid-β (Aβ) elimination. In vitro approaches underlined the ability of a low-glucose microenvironment to decrease the general activity and phagocytic capacity of microglia. By using a new model of SCCH, our study unravels new insights into the implication of severe chronic cerebral hypoperfusion in AD pathogenesis, mainly by altering microglial cell activity and consequently Aβ clearance.
Collapse
Affiliation(s)
- Maude Bordeleau
- Neuroscience Laboratory, CHU de Québec Research Center (CHUL), Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, Canada
| | - Ayman ElAli
- Neuroscience Laboratory, CHU de Québec Research Center (CHUL), Department of Psychiatry and Neuroscience, Faculty of Medicine, Laval University, Québec, Canada
| | - Serge Rivest
- Neuroscience Laboratory, CHU de Québec Research Center (CHUL), Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, Canada
| |
Collapse
|
49
|
Qin C, Fan WH, Liu Q, Shang K, Murugan M, Wu LJ, Wang W, Tian DS. Fingolimod Protects Against Ischemic White Matter Damage by Modulating Microglia Toward M2 Polarization via STAT3 Pathway. Stroke 2017; 48:3336-3346. [PMID: 29114096 DOI: 10.1161/strokeaha.117.018505] [Citation(s) in RCA: 268] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 09/26/2017] [Accepted: 10/02/2017] [Indexed: 12/25/2022]
Abstract
BACKGROUND AND PURPOSE White matter (WM) ischemic injury, a major neuropathological feature of cerebral small vessel diseases, is an important cause of vascular cognitive impairment in later life. The pathogenesis of demyelination after WM ischemic damage are often accompanied by microglial activation. Fingolimod (FTY720) was approved for the treatment of multiple sclerosis for its immunosuppression property. In this study, we evaluated the neuroprotective potential of FTY720 in a WM ischemia model. METHODS Chronic WM ischemic injury model was induced by bilateral carotid artery stenosis. Cognitive function, WM integrity, microglial activation, and potential pathway involved in microglial polarization were assessed after bilateral carotid artery stenosis. RESULTS Disruption of WM integrity was characterized by demyelination in the corpus callosum and disorganization of Ranvier nodes using Luxol fast blue staining, immunofluorescence staining, and electron microscopy. In addition, radial maze test demonstrated that working memory performance was decreased at 1-month post-bilateral carotid artery stenosis-induced injury. Interestingly, FTY720 could reduce cognitive decline and ameliorate the disruption of WM integrity. Mechanistically, cerebral hypoperfusion induced microglial activation, production of associated proinflammatory cytokines, and priming of microglial polarization toward the M1 phenotype, whereas FTY720 attenuated microglia-mediated neuroinflammation after WM ischemia and promoted oligodendrocytogenesis by shifting microglia toward M2 polarization. FTY720's effect on microglial M2 polarization was largely suppressed by selective signal transducer and activator of transcription 3 (STAT3) blockade in vitro, revealing that FTY720-enabled shift of microglia from M1 to M2 polarization state was possibly mediated by STAT3 signaling. CONCLUSIONS Our study suggested that FTY720 might be a potential therapeutic drug targeting brain inflammation by skewing microglia toward M2 polarization after chronic cerebral hypoperfusion.
Collapse
Affiliation(s)
- Chuan Qin
- From the Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (C.Q., W.-H.F., Q.L., K.S., W.W., D.-S.T.); Department of Neurology, General Hospital of the Yangtze River Shipping, Wuhan, China (W.-H.F.); Department of Neurology, Mayo Clinic, Rochester, MN (M.M., L.-J.W.); and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ (M.M., L.-J.W.)
| | - Wen-Hui Fan
- From the Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (C.Q., W.-H.F., Q.L., K.S., W.W., D.-S.T.); Department of Neurology, General Hospital of the Yangtze River Shipping, Wuhan, China (W.-H.F.); Department of Neurology, Mayo Clinic, Rochester, MN (M.M., L.-J.W.); and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ (M.M., L.-J.W.)
| | - Qian Liu
- From the Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (C.Q., W.-H.F., Q.L., K.S., W.W., D.-S.T.); Department of Neurology, General Hospital of the Yangtze River Shipping, Wuhan, China (W.-H.F.); Department of Neurology, Mayo Clinic, Rochester, MN (M.M., L.-J.W.); and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ (M.M., L.-J.W.)
| | - Ke Shang
- From the Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (C.Q., W.-H.F., Q.L., K.S., W.W., D.-S.T.); Department of Neurology, General Hospital of the Yangtze River Shipping, Wuhan, China (W.-H.F.); Department of Neurology, Mayo Clinic, Rochester, MN (M.M., L.-J.W.); and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ (M.M., L.-J.W.)
| | - Madhuvika Murugan
- From the Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (C.Q., W.-H.F., Q.L., K.S., W.W., D.-S.T.); Department of Neurology, General Hospital of the Yangtze River Shipping, Wuhan, China (W.-H.F.); Department of Neurology, Mayo Clinic, Rochester, MN (M.M., L.-J.W.); and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ (M.M., L.-J.W.)
| | - Long-Jun Wu
- From the Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (C.Q., W.-H.F., Q.L., K.S., W.W., D.-S.T.); Department of Neurology, General Hospital of the Yangtze River Shipping, Wuhan, China (W.-H.F.); Department of Neurology, Mayo Clinic, Rochester, MN (M.M., L.-J.W.); and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ (M.M., L.-J.W.)
| | - Wei Wang
- From the Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (C.Q., W.-H.F., Q.L., K.S., W.W., D.-S.T.); Department of Neurology, General Hospital of the Yangtze River Shipping, Wuhan, China (W.-H.F.); Department of Neurology, Mayo Clinic, Rochester, MN (M.M., L.-J.W.); and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ (M.M., L.-J.W.)
| | - Dai-Shi Tian
- From the Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China (C.Q., W.-H.F., Q.L., K.S., W.W., D.-S.T.); Department of Neurology, General Hospital of the Yangtze River Shipping, Wuhan, China (W.-H.F.); Department of Neurology, Mayo Clinic, Rochester, MN (M.M., L.-J.W.); and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ (M.M., L.-J.W.).
| |
Collapse
|
50
|
Ohtomo R, Bannai T, Ohtomo G, Shindo A, Tomimoto H, Tsuji S, Iwata A. Cilostazol alleviates white matter degeneration caused by chronic cerebral hypoperfusion in mice: Implication of its mechanism from gene expression analysis. Neurosci Lett 2017; 662:247-252. [PMID: 29080698 DOI: 10.1016/j.neulet.2017.10.045] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 10/23/2017] [Accepted: 10/24/2017] [Indexed: 11/18/2022]
Abstract
Cilostazol is known to alleviate white matter demyelination due to chronic cerebral hypoperfusion in rodent models, although their pharmacological mechanisms remain unclear. In this study, we investigated the protective effect of cilostazol in relation to gene expression profile. Bilateral common carotid artery stenosis (BCAS) mice were treated with oral administration of cilostazol or placebo starting from a week after the surgery. Demyelination of the cingulum was compared between the 2 groups 2, 6, and 10 weeks after initial drug administration. Also, to examine temporal gene expression change during demyelination, DNA microarray analysis was conducted using samples from the corpus callosum of 2nd and 6th week BCAS mice. For genes that showed more than 2-fold up-regulation, their increase was validated by qPCR. Finally, to determine the effect of cilostazol towards those genes, their expression in the corpus callosum of 6-week placebo-treated and cilostazol-treated BCAS mice was compared by qPCR. Amelioration of myelin loss was observed in cilostazol-treated group, showing significant difference with those observed in placebo group after 10-week treatment. Gene ontology analysis of the 17 up-regulated (FDR<0.01) genes showed that majority of the genes were related to cell development processes. Among the validated genes, expression of Btg2 was significantly promoted in the corpus callosum of BCAS mice by administration of cilostazol. Results of this study suggest that activation of Btg2 may be one of the key pharmacological effects of cilostazol towards the white matter during chronic ischemia.
Collapse
Affiliation(s)
- Ryo Ohtomo
- Department of Neurology, The University of Tokyo Graduate School of Medicine, Japan.
| | - Taro Bannai
- Department of Neurology, The University of Tokyo Graduate School of Medicine, Japan
| | - Gaku Ohtomo
- Department of Neurology, The University of Tokyo Graduate School of Medicine, Japan
| | - Akihiro Shindo
- Department of Neurology, Mie University Graduate School of Medicine, Japan
| | - Hidekazu Tomimoto
- Department of Neurology, Mie University Graduate School of Medicine, Japan
| | - Shoji Tsuji
- Department of Neurology, The University of Tokyo Graduate School of Medicine, Japan
| | - Atsushi Iwata
- Department of Neurology, The University of Tokyo Graduate School of Medicine, Japan
| |
Collapse
|