1
|
Lin L, Hu X, Hong W, Pan T, Wang Z, Wang E, Wu G. A novel animal model of spontaneous epilepsy: Cdk5 knockout in pericyte-specific mice. Front Cell Neurosci 2024; 18:1474231. [PMID: 39479522 PMCID: PMC11521856 DOI: 10.3389/fncel.2024.1474231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/02/2024] [Indexed: 11/02/2024] Open
Abstract
Changes in neurovascular unit components and their interactions play a crucial role in epileptogenesis and the pathological process of epilepsy. Currently, there is a lack of animal models that can accurately reflect the etiological impact of cerebrovascular lesions on epilepsy. In this study, we constructed cyclin-dependent kinase 5 conditional knockout mice in Cspg4 (pericyte marker)-positive cells using the Cre-LoxP system. The results revealed that this strain of mice exhibited significant seizure behaviors and epileptiform brain waves, loss of hippocampal and amygdala neurons, astrogliosis, decreased pericyte coverage, and reduced AQP4 polar distribution. Herein, we have developed a novel mouse model of spontaneous epilepsy, providing a critical animal model for studying the involvement of neurovascular unit factors in the development and progression of epilepsy.
Collapse
Affiliation(s)
- Lin Lin
- Department of Pharmacy, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
- School of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaofei Hu
- Department of Neurology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Weijun Hong
- Department of Neurology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Tengwei Pan
- Department of Neurology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Zhiren Wang
- Department of Pharmacy, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - En Wang
- Department of Neurology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
| | - Gang Wu
- Department of Pharmacy, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, China
- Taizhou Key Laboratory of Pharmaceuticals Therapy and Translation Research, Linhai, Zhejiang, China
| |
Collapse
|
2
|
Liu J, Binding L, Puntambekar I, Patodia S, Lim YM, Mryzyglod A, Xiao F, Pan S, Mito R, de Tisi J, Duncan JS, Baxendale S, Koepp M, Thom M. Microangiopathy in temporal lobe epilepsy with diffusion MRI alterations and cognitive decline. Acta Neuropathol 2024; 148:49. [PMID: 39377933 PMCID: PMC11461556 DOI: 10.1007/s00401-024-02809-8] [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: 08/01/2024] [Revised: 09/23/2024] [Accepted: 09/28/2024] [Indexed: 10/09/2024]
Abstract
White matter microvascular alterations in temporal lobe epilepsy (TLE) may be relevant to acquired neurodegenerative processes and cognitive impairments associated with this condition. We quantified microvascular changes, myelin, axonal, glial and extracellular-matrix labelling in the gyral core and deep temporal lobe white matter regions in surgical resections from 44 TLE patients with or without hippocampal sclerosis. We compared this pathology data with in vivo pre-operative MRI diffusion measurements in co-registered regions and neuropsychological measures of cognitive impairment and decline. In resections, increased arteriolosclerosis was observed in TLE compared to non-epilepsy controls (greater sclerotic index, p < 0.001), independent of age. Microvascular changes included increased vascular densities in some regions but uniformly reduced mean vascular size (quantified with collagen-4, p < 0.05-0.0001), and increased pericyte coverage of small vessels and capillaries particularly in deep white matter (quantified with platelet-derived growth factor receptorβ and smooth muscle actin, p < 0.01) which was more marked the longer the duration of epilepsy (p < 0.05). We noted increased glial numbers (Olig2, Iba1) but reduced myelin (MAG, PLP) in TLE compared to controls, particularly prominent in deep white matter. Gene expression analysis showed a greater reduction of myelination genes in HS than non-HS cases and with age and correlation with diffusion MRI alterations. Glial densities and vascular size were increased with increased MRI diffusivity and vascular density with white matter abnormality quantified using fixel-based analysis. Increased perivascular space was associated with reduced fractional anisotropy as well as age-accelerated cognitive decline prior to surgery (p < 0.05). In summary, likely acquired microangiopathic changes in TLE, including vascular sclerosis, increased pericyte coverage and reduced small vessel size, may indicate a functional alteration in contractility of small vessels and haemodynamics that could impact on tissue perfusion. These morphological features correlate with white matter diffusion MRI alterations and might explain cognitive decline in TLE.
Collapse
Affiliation(s)
- Joan Liu
- Department of Clinical and Experimental Epilepsy, Department of Neuropathology, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
- Department of Neuroscience, University of Westminster, London, UK
| | - Lawrence Binding
- Department of Clinical and Experimental Epilepsy, Department of Neuropathology, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
- Centre for Medical Image Computing, Department of Computer Science, UCL, London, UK
| | - Isha Puntambekar
- Department of Clinical and Experimental Epilepsy, Department of Neuropathology, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Smriti Patodia
- Department of Clinical and Experimental Epilepsy, Department of Neuropathology, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Yau Mun Lim
- Department of Clinical and Experimental Epilepsy, Department of Neuropathology, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Alicja Mryzyglod
- Department of Clinical and Experimental Epilepsy, Department of Neuropathology, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Fenglai Xiao
- Department of Clinical and Experimental Epilepsy, Department of Neuropathology, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Shengning Pan
- Department of Statistical Science, University College London, Gower St., London, UK
| | - Remika Mito
- Department of Neuroscience and Mental Health, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Victoria, Australia
| | - Jane de Tisi
- Department of Clinical and Experimental Epilepsy, Department of Neuropathology, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - John S Duncan
- Department of Clinical and Experimental Epilepsy, Department of Neuropathology, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Sallie Baxendale
- Department of Clinical and Experimental Epilepsy, Department of Neuropathology, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Matthias Koepp
- Department of Clinical and Experimental Epilepsy, Department of Neuropathology, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK
| | - Maria Thom
- Department of Clinical and Experimental Epilepsy, Department of Neuropathology, UCL Queen Square Institute of Neurology, London, WC1N 3BG, UK.
| |
Collapse
|
3
|
Bastedo WE, Scott RW, Arostegui M, Underhill TM. Single-cell analysis of mesenchymal cells in permeable neural vasculature reveals novel diverse subpopulations of fibroblasts. Fluids Barriers CNS 2024; 21:31. [PMID: 38575991 PMCID: PMC10996213 DOI: 10.1186/s12987-024-00535-7] [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: 07/31/2023] [Accepted: 03/25/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND In the choroid plexus and pituitary gland, vasculature is known to have a permeable, fenestrated phenotype which allows for the free passage of molecules in contrast to the blood brain barrier observed in the rest of the CNS. The endothelium of these compartments, along with secretory, neural-lineage cells (choroid epithelium and pituitary endocrine cells) have been studied in detail, but less attention has been given to the perivascular mesenchymal cells of these compartments. METHODS The Hic1CreERT2 Rosa26LSL-TdTomato mouse model was used in conjunction with a PdgfraH2B-EGFP mouse model to examine mesenchymal cells, which can be subdivided into Pdgfra+ fibroblasts and Pdgfra- pericytes within the choroid plexus (CP) and pituitary gland (PG), by histological, immunofluorescence staining and single-cell RNA-sequencing analyses. RESULTS We found that both CP and PG possess substantial populations of distinct Hic1+ mesenchymal cells, including an abundance of Pdgfra+ fibroblasts. Within the pituitary, we identified distinct subpopulations of Hic1+ fibroblasts in the glandular anterior pituitary and the neurosecretory posterior pituitary. We also identified multiple distinct markers of CP, PG, and the meningeal mesenchymal compartment, including alkaline phosphatase, indole-n-methyltransferase and CD34. CONCLUSIONS Novel, distinct subpopulations of mesenchymal cells can be found in permeable vascular interfaces, including the CP, PG, and meninges, and make distinct contributions to both organs through the production of structural proteins, enzymes, transporters, and trophic molecules.
Collapse
Affiliation(s)
- William E Bastedo
- Department of Cellular and Physiological Sciences, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - R Wilder Scott
- Department of Cellular and Physiological Sciences, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- School of Biomedical Engineering and the Biomedical Research Centre, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Martin Arostegui
- Department of Cellular and Physiological Sciences, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - T Michael Underhill
- Department of Cellular and Physiological Sciences, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
- School of Biomedical Engineering and the Biomedical Research Centre, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
4
|
Kyriatzis G, Bernard A, Bôle A, Khrestchatisky M, Ferhat L. In the Rat Hippocampus, Pilocarpine-Induced Status Epilepticus Is Associated with Reactive Glia and Concomitant Increased Expression of CD31, PDGFRβ, and Collagen IV in Endothelial Cells and Pericytes of the Blood-Brain Barrier. Int J Mol Sci 2024; 25:1693. [PMID: 38338969 PMCID: PMC10855308 DOI: 10.3390/ijms25031693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
In humans and animal models, temporal lobe epilepsy (TLE) is associated with reorganization of hippocampal neuronal networks, gliosis, neuroinflammation, and loss of integrity of the blood-brain barrier (BBB). More than 30% of epilepsies remain intractable, and characterization of the molecular mechanisms involved in BBB dysfunction is essential to the identification of new therapeutic strategies. In this work, we induced status epilepticus in rats through injection of the proconvulsant drug pilocarpine, which leads to TLE. Using RT-qPCR, double immunohistochemistry, and confocal imaging, we studied the regulation of reactive glia and vascular markers at different time points of epileptogenesis (latent phase-3, 7, and 14 days; chronic phase-1 and 3 months). In the hippocampus, increased expression of mRNA encoding the glial proteins GFAP and Iba1 confirmed neuroinflammatory status. We report for the first time the concomitant induction of the specific proteins CD31, PDGFRβ, and ColIV-which peak at the same time points as inflammation-in the endothelial cells, pericytes, and basement membrane of the BBB. The altered expression of these proteins occurs early in TLE, during the latent phase, suggesting that they could be associated with the early rupture and pathogenicity of the BBB that will contribute to the chronic phase of epilepsy.
Collapse
Affiliation(s)
| | | | | | - Michel Khrestchatisky
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France, Institut de Neurophysiopathologie, Faculté de Médecine, 27 Bd Jean Moulin, 13005 Marseille, France; (G.K.); (A.B.); (A.B.)
| | - Lotfi Ferhat
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France, Institut de Neurophysiopathologie, Faculté de Médecine, 27 Bd Jean Moulin, 13005 Marseille, France; (G.K.); (A.B.); (A.B.)
| |
Collapse
|
5
|
van Lanen RHGJ, Haeren RHL, Staals J, Dings JTA, Schijns OEMG, Hoogland G, van Kuijk SMJ, Kapsokalyvas D, van Zandvoort MAMJ, Vink H, Rijkers K. Cerebrovascular glycocalyx damage and microcirculation impairment in patients with temporal lobe epilepsy. J Cereb Blood Flow Metab 2023; 43:1737-1751. [PMID: 37231664 PMCID: PMC10581235 DOI: 10.1177/0271678x231179413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 04/02/2023] [Accepted: 05/02/2023] [Indexed: 05/27/2023]
Abstract
Temporal lobe epilepsy (TLE) is increasingly associated with blood-brain barrier dysfunction and microvascular alterations, yet the pathophysiological link is missing. An important barrier function is exerted by the glycocalyx, a gel-like layer coating the endothelium. To explore such associations, we used intraoperative videomicroscopy to quantify glycocalyx and microcirculation properties of the neocortex and hippocampus of 15 patients undergoing resective brain surgery as treatment for drug-resistant TLE, and 15 non-epileptic controls. Fluorescent lectin staining of neocortex and hippocampal tissue was used for blood vessel surface area quantification. Neocortical perfused boundary region, the thickness of the glycocalyx' impaired layer, was higher in patients (2.64 ± 0.52 µm) compared to controls (1.31 ± 0.29 µm), P < 0.01, indicative of reduced glycocalyx integrity in patients. Moreover, erythrocyte flow velocity analysis revealed an impaired ability of TLE patients to (de-)recruit capillaries in response to changing metabolic demands (R2 = 0.75, P < 0.01), indicating failure of neurovascular coupling mechanisms. Blood vessel quantification comparison between intraoperative measurements and resected tissue showed strong correlation (R2 = 0.94, P < 0.01). This is the first report on in vivo assessment of glycocalyx and microcirculation properties in TLE patients, confirming the pivotal role of cerebrovascular changes. Further assessment of the cerebral microcirculation in relation to epileptogenesis might open avenues for new therapeutic targets for drug-resistant epilepsy.
Collapse
Affiliation(s)
- Rick HGJ van Lanen
- Department of Neurosurgery, Maastricht University Medical Centre, Maastricht, The Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
| | - Roel HL Haeren
- Department of Neurosurgery, Maastricht University Medical Centre, Maastricht, The Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
| | - Julie Staals
- Department of Neurology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Jim TA Dings
- Department of Neurosurgery, Maastricht University Medical Centre, Maastricht, The Netherlands
- Academic Centre for Epileptology, Maastricht University Medical Centre and Kempenhaeghe, Maastricht/Heeze, The Netherlands
| | - Olaf EMG Schijns
- Department of Neurosurgery, Maastricht University Medical Centre, Maastricht, The Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- Academic Centre for Epileptology, Maastricht University Medical Centre and Kempenhaeghe, Maastricht/Heeze, The Netherlands
| | - Govert Hoogland
- Department of Neurosurgery, Maastricht University Medical Centre, Maastricht, The Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
| | - Sander MJ van Kuijk
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Dimitris Kapsokalyvas
- Department of Genetics & Cell Biology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
- Interdisciplinary Center for Clinical Research (IZKF), University Hospital RWTH Aachen, Aachen, Germany
| | - Marc AMJ van Zandvoort
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- Department of Genetics & Cell Biology, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
- Institute for Molecular Cardiovascular Research IMCAR, Universitätsklinikum, Aachen University, Aachen, Germany
| | - Hans Vink
- Department of Physiology, Maastricht University, Maastricht, The Netherlands
| | - Kim Rijkers
- Department of Neurosurgery, Maastricht University Medical Centre, Maastricht, The Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- Academic Centre for Epileptology, Maastricht University Medical Centre and Kempenhaeghe, Maastricht/Heeze, The Netherlands
| |
Collapse
|
6
|
Watanabe Y, Yamanaka G, Morichi S, Hayashi K, Suzuki S, Takeshita M, Morishita N, Ishida Y, Oana S, Takata F, Kawashima H. Altered serum levels of platelet-derived growth factor receptor β and cluster of differentiation 13 suggest a role for pericytes in West syndrome. Brain Dev 2023; 45:479-486. [PMID: 37263884 DOI: 10.1016/j.braindev.2023.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 05/12/2023] [Accepted: 05/17/2023] [Indexed: 06/03/2023]
Abstract
BACKGROUND Pericytes play a role in the maintenance of the blood-brain barrier and neuroinflammation, attracting attention as to whether they are also involved in the pathogenesis of epilepsy.This study aimed to explore the relationship between West syndrome and pericytes. METHODS Eighteen Japanese pediatric West syndrome patients and nine controls aged 2 years or younger were retrospectively enrolled in this study. We assessed theserumlevels of pericyte markers, serum PDGFRβ (platelet-derived growth factor receptorβ),CD13 (aminopeptidase N), and 27 cytokines in 17 pediatric patients with West syndrome and the control group. RESULTS Patients with West syndrome exhibited significantly increased CD13 and decreased PDGFRβ levels, compared with controls but not serum cytokine levels. These values did not differ significantly between symptomatic and idiopathic West syndrome. CONCLUSION Pericytes might be implicated in the pathogenesis of West syndrome.
Collapse
Affiliation(s)
- Yusuke Watanabe
- Department of Pediatrics and Adolescent Medicine, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku-ku, Tokyo 160-0023, Japan.
| | - Gaku Yamanaka
- Department of Pediatrics and Adolescent Medicine, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Shinichiro Morichi
- Department of Pediatrics and Adolescent Medicine, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Kanako Hayashi
- Department of Pediatrics and Adolescent Medicine, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Shinji Suzuki
- Department of Pediatrics and Adolescent Medicine, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Mika Takeshita
- Department of Pediatrics and Adolescent Medicine, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Natsumi Morishita
- Department of Pediatrics and Adolescent Medicine, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Yu Ishida
- Department of Pediatrics and Adolescent Medicine, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Shingo Oana
- Department of Pediatrics and Adolescent Medicine, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Fuyuko Takata
- Department of Pharmaceutical Care and Health Sciences, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan
| | - Hisashi Kawashima
- Department of Pediatrics and Adolescent Medicine, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| |
Collapse
|
7
|
Lv X, Zhang M, Cheng Z, Wang Q, Wang P, Xie Q, Ni M, Shen Y, Tang Q, Gao F. Changes in CSF sPDGFRβ level and their association with blood-brain barrier breakdown in Alzheimer's disease with or without small cerebrovascular lesions. Alzheimers Res Ther 2023; 15:51. [PMID: 36915135 PMCID: PMC10012584 DOI: 10.1186/s13195-023-01199-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 02/28/2023] [Indexed: 03/16/2023]
Abstract
BACKGROUND CSF-soluble platelet-derived growth factor receptor beta (sPDGFRβ) is closely associated with pericyte damage. However, the changes in CSF sPDGFRβ levels and their role in blood-brain barrier (BBB) leakage at different stages of Alzheimer's disease (AD), with or without cerebral small vessel disease (CSVD) burden, remain unclear. METHODS A total of 158 individuals from the China Aging and Neurodegenerative Disorder Initiative cohort were selected, including 27, 48, and 83 individuals with a clinical dementia rating (CDR) score of 0, 0.5, and 1-2, respectively. CSF total tau, phosphorylated tau181 (p-tau181), Aβ40, and Aβ42 were measured using the Simoa assay. Albumin and CSF sPDGFRβ were measured by commercial assay kits. CSVD burden was assessed by magnetic resonance imaging. RESULTS CSF sPDGFRβ was the highest level in the CDR 0.5 group. CSF sPDGFRβ was significantly correlated with the CSF/serum albumin ratio (Q-alb) in the CDR 0-0.5 group (β = 0.314, p = 0.008) but not in the CDR 1-2 group (β = - 0.117, p = 0.317). In the CDR 0-0.5 group, CSF sPDGFRβ exhibited a significant mediating effect between Aβ42/Aβ40 levels and Q-alb (p = 0.038). Q-alb, rather than CSF sPDGFRβ, showed a significant difference between individuals with or without CSVD burden. Furthermore, in the CDR 0.5 group, CSF sPDGFRβ was higher in subjects with progressive mild cognitive impairment than in those with stable mild cognitive impairment subjects (p < 0.001). Meanwhile, CSF sPDGFRβ was significantly associated with yearly changes in MMSE scores in the CDR 0.5 group (β = - 0.400, p = 0.020) and CDR 0.5 (A+) subgroup (β = - 0.542, p = 0.019). CONCLUSIONS We provide evidence that increased CSF sPDGFRβ is associated with BBB leakage in the early cognitive impairment stage of AD, which may contribute to cognitive impairment in AD progression.
Collapse
Affiliation(s)
- Xinyi Lv
- Department of Neurology, Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Mengguo Zhang
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhaozhao Cheng
- Department of Neurology, Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qiong Wang
- Department of Neurology, Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Peng Wang
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qiang Xie
- Department of Nuclear Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ming Ni
- Department of Nuclear Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yong Shen
- Department of Neurology, Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Anhui Province Key Laboratory of Biomedical Aging Research, Hefei, China
| | - Qiqiang Tang
- Department of Neurology, Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Feng Gao
- Department of Neurology, Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China. .,Anhui Province Key Laboratory of Biomedical Aging Research, Hefei, China.
| | | |
Collapse
|
8
|
Salehi A, Salari S, Jullienne A, Daglian J, Chen K, Baram TZ, Obenaus A. Vascular topology and blood flow are acutely impacted by experimental febrile status epilepticus. J Cereb Blood Flow Metab 2023; 43:84-98. [PMID: 35912523 PMCID: PMC9875348 DOI: 10.1177/0271678x221117625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Febrile status epilepticus (FSE) is an important risk factor for temporal lobe epilepsy and early identification of those at high risk for epilepsy is vital. In a rat model of FSE, we identified an acute (2 hrs) novel MRI signal where reduced T2 relaxation values in the basolateral amygdala (BLA) predicted epilepsy in adulthood; this T2 signal remains incompletely understood and we hypothesized that it may be influenced by vascular topology. Experimental FSE induced in rat pups reduced blood vessel density of the cortical vasculature in a lateralized manner at 2 hrs post FSE. Middle cerebral artery (MCA) exhibited abnormal topology in FSE pups but not in controls. In the BLA, significant vessel junction reductions and decreased vessel diameter were observed, together with a strong trend for reduced vessel length. Perfusion weighted MRI (PWI) was acutely increased cerebral blood flow (CBF) in cortex, amygdala and hippocampus of FSE pups that correlated to decreased T2 relaxation values compared to controls. This is consistent with increased levels of deoxyhemoglobin associated with increased metabolic demand. In summary, FSE acutely modifies vascular topological and CBF in cortex and BLA that may underlie acute MRI signal changes that predict progression to future epilepsy.
Collapse
Affiliation(s)
- Arjang Salehi
- Department of Anatomy and Neurobiology, School of Medicine, University of California Irvine, Irvine CA, USA
| | - Sirus Salari
- Department of Pediatrics, School of Medicine, University of California Irvine, Irvine CA, USA
| | - Amandine Jullienne
- Department of Pediatrics, School of Medicine, University of California Irvine, Irvine CA, USA
| | - Jennifer Daglian
- Department of Anatomy and Neurobiology, School of Medicine, University of California Irvine, Irvine CA, USA
| | - Kevin Chen
- Department of Anatomy and Neurobiology, School of Medicine, University of California Irvine, Irvine CA, USA
| | - Tallie Z Baram
- Department of Anatomy and Neurobiology, School of Medicine, University of California Irvine, Irvine CA, USA.,Department of Pediatrics, School of Medicine, University of California Irvine, Irvine CA, USA
| | - Andre Obenaus
- Department of Anatomy and Neurobiology, School of Medicine, University of California Irvine, Irvine CA, USA.,Department of Pediatrics, School of Medicine, University of California Irvine, Irvine CA, USA
| |
Collapse
|
9
|
Hattori Y. The Multiple Roles of Pericytes in Vascular Formation and Microglial Functions in the Brain. Life (Basel) 2022; 12:1835. [PMID: 36362989 PMCID: PMC9699346 DOI: 10.3390/life12111835] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 10/15/2023] Open
Abstract
In the capillary walls, vascular endothelial cells are covered with mural cells, such as smooth muscle cells and pericytes. Although pericytes had been thought to play simply a structural role, emerging evidence has highlighted their multiple functions in the embryonic, postnatal, and adult brain. As the central nervous system (CNS) develops, the brain's vascular structure gradually matures into a hierarchical network, which is crucial for the proper development of neural lineage cells by providing oxygen and nutrients. Pericytes play an essential role in vascular formation and regulate blood‒brain barrier (BBB) integrity as a component of the neurovascular unit (NVU), in collaboration with other cells, such as vascular endothelial cells, astrocytes, neurons, and microglia. Microglia, the resident immune cells of the CNS, colonize the brain at embryonic day (E) 9.5 in mice. These cells not only support the development and maturation of neural lineage cells but also help in vascular formation through their extensive migration. Recent studies have demonstrated that pericytes directly contact microglia in the CNS, and their interactions have a profound effect on physiological and pathological aspects. This review summarizes the function of pericytes, focusing on the interplay between pericytes and microglia.
Collapse
Affiliation(s)
- Yuki Hattori
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
10
|
Ippolitov D, Arreza L, Munir MN, Hombach-Klonisch S. Brain Microvascular Pericytes—More than Bystanders in Breast Cancer Brain Metastasis. Cells 2022; 11:cells11081263. [PMID: 35455945 PMCID: PMC9028330 DOI: 10.3390/cells11081263] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/06/2022] [Accepted: 04/06/2022] [Indexed: 01/27/2023] Open
Abstract
Brain tissue contains the highest number of perivascular pericytes compared to other organs. Pericytes are known to regulate brain perfusion and to play an important role within the neurovascular unit (NVU). The high phenotypic and functional plasticity of pericytes make this cell type a prime candidate to aid physiological adaptations but also propose pericytes as important modulators in diverse pathologies in the brain. This review highlights known phenotypes of pericytes in the brain, discusses the diverse markers for brain pericytes, and reviews current in vitro and in vivo experimental models to study pericyte function. Our current knowledge of pericyte phenotypes as it relates to metastatic growth patterns in breast cancer brain metastasis is presented as an example for the crosstalk between pericytes, endothelial cells, and metastatic cells. Future challenges lie in establishing methods for real-time monitoring of pericyte crosstalk to understand causal events in the brain metastatic process.
Collapse
Affiliation(s)
- Danyyl Ippolitov
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (D.I.); (L.A.); (M.N.M.)
| | - Leanne Arreza
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (D.I.); (L.A.); (M.N.M.)
| | - Maliha Nuzhat Munir
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (D.I.); (L.A.); (M.N.M.)
| | - Sabine Hombach-Klonisch
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (D.I.); (L.A.); (M.N.M.)
- Department of Pathology, University of Manitoba, Winnipeg, MB R3E 0Z2, Canada
- Correspondence:
| |
Collapse
|
11
|
van Vliet EA, Marchi N. Neurovascular unit dysfunction as a mechanism of seizures and epilepsy during aging. Epilepsia 2022; 63:1297-1313. [PMID: 35218208 PMCID: PMC9321014 DOI: 10.1111/epi.17210] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/23/2022] [Accepted: 02/23/2022] [Indexed: 11/30/2022]
Abstract
The term neurovascular unit (NVU) describes the structural and functional liaison between specialized brain endothelium, glial and mural cells, and neurons. Within the NVU, the blood‐brain barrier (BBB) is the microvascular structure regulating neuronal physiology and immune cross‐talk, and its properties adapt to brain aging. Here, we analyze a research framework where NVU dysfunction, caused by acute insults or disease progression in the aging brain, represents a converging mechanism underlying late‐onset seizures or epilepsy and neurological or neurodegenerative sequelae. Furthermore, seizure activity may accelerate brain aging by sustaining regional NVU dysfunction, and a cerebrovascular pathology may link seizures to comorbidities. Next, we focus on NVU diagnostic approaches that could be tailored to seizure conditions in the elderly. We also examine the impending disease‐modifying strategies based on the restoration of the NVU and, more in general, the homeostatic control of anti‐ and pro‐inflammatory players. We conclude with an outlook on current pre‐clinical knowledge gaps and clinical challenges pertinent to seizure onset and conditions in an aging population.
Collapse
Affiliation(s)
- Erwin A van Vliet
- Amsterdam UMC, University of Amsterdam, dept. of (Neuro)pathology, Amsterdam, the Netherlands.,University of Amsterdam, Swammerdam Institute for Life Sciences, Center for Neuroscience, Amsterdam, the Netherlands
| | - Nicola Marchi
- Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
12
|
van Lanen RH, Melchers S, Hoogland G, Schijns OE, Zandvoort MAV, Haeren RH, Rijkers K. Microvascular changes associated with epilepsy: A narrative review. J Cereb Blood Flow Metab 2021; 41:2492-2509. [PMID: 33866850 PMCID: PMC8504411 DOI: 10.1177/0271678x211010388] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The blood-brain barrier (BBB) is dysfunctional in temporal lobe epilepsy (TLE). In this regard, microvascular changes are likely present. The aim of this review is to provide an overview of the current knowledge on microvascular changes in epilepsy, and includes clinical and preclinical evidence of seizure induced angiogenesis, barriergenesis and microcirculatory alterations. Anatomical studies show increased microvascular density in the hippocampus, amygdala, and neocortex accompanied by BBB leakage in various rodent epilepsy models. In human TLE, a decrease in afferent vessels, morphologically abnormal vessels, and an increase in endothelial basement membranes have been observed. Both clinical and experimental evidence suggests that basement membrane changes, such as string vessels and protrusions, indicate and visualize a misbalance between endothelial cell proliferation and barriergenesis. Vascular endothelial growth factor (VEGF) appears to play a crucial role. Following an altered vascular anatomy, its physiological functioning is affected as expressed by neurovascular decoupling that subsequently leads to hypoperfusion, disrupted parenchymal homeostasis and potentially to seizures". Thus, epilepsy might be a condition characterized by disturbed cerebral microvasculature in which VEGF plays a pivotal role. Additional physiological data from patients is however required to validate findings from models and histological studies on patient biopsies.
Collapse
Affiliation(s)
- Rick Hgj van Lanen
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, the Netherlands.,School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Stan Melchers
- Faculty of Health Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands
| | - Govert Hoogland
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, the Netherlands.,School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.,Academic Center for Epileptology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Olaf Emg Schijns
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, the Netherlands.,School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.,Academic Center for Epileptology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Marc Amj van Zandvoort
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.,Department of Molecular Cell Biology, School for Mental Health and Neuroscience, Maastricht University Medical Center, Maastricht, the Netherlands.,School for Cardiovascular Diseases, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Roel Hl Haeren
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, the Netherlands.,Department of Neurosurgery, Helsinki University Central Hospital, Helsinki, Finland
| | - Kim Rijkers
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, the Netherlands.,School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.,Academic Center for Epileptology, Maastricht University Medical Center, Maastricht, the Netherlands
| |
Collapse
|
13
|
The Neuroinflammatory Role of Pericytes in Epilepsy. Biomedicines 2021; 9:biomedicines9070759. [PMID: 34209145 PMCID: PMC8301485 DOI: 10.3390/biomedicines9070759] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/25/2021] [Accepted: 06/26/2021] [Indexed: 02/07/2023] Open
Abstract
Pericytes are a component of the blood-brain barrier (BBB) neurovascular unit, in which they play a crucial role in BBB integrity and are also implicated in neuroinflammation. The association between pericytes, BBB dysfunction, and the pathophysiology of epilepsy has been investigated, and links between epilepsy and pericytes have been identified. Here, we review current knowledge about the role of pericytes in epilepsy. Clinical evidence has shown an accumulation of pericytes with altered morphology in the cerebral vascular territories of patients with intractable epilepsy. In vitro, proinflammatory cytokines, including IL-1β, TNFα, and IL-6, cause morphological changes in human-derived pericytes, where IL-6 leads to cell damage. Experimental studies using epileptic animal models have shown that cerebrovascular pericytes undergo redistribution and remodeling, potentially contributing to BBB permeability. These series of pericyte-related modifications are promoted by proinflammatory cytokines, of which the most pronounced alterations are caused by IL-1β, a cytokine involved in the pathogenesis of epilepsy. Furthermore, the pericyte-glial scarring process in leaky capillaries was detected in the hippocampus during seizure progression. In addition, pericytes respond more sensitively to proinflammatory cytokines than microglia and can also activate microglia. Thus, pericytes may function as sensors of the inflammatory response. Finally, both in vitro and in vivo studies have highlighted the potential of pericytes as a therapeutic target for seizure disorders.
Collapse
|
14
|
Links between Immune Cells from the Periphery and the Brain in the Pathogenesis of Epilepsy: A Narrative Review. Int J Mol Sci 2021; 22:ijms22094395. [PMID: 33922369 PMCID: PMC8122797 DOI: 10.3390/ijms22094395] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/19/2021] [Accepted: 04/21/2021] [Indexed: 12/13/2022] Open
Abstract
Accumulating evidence has demonstrated that the pathogenesis of epilepsy is linked to neuroinflammation and cerebrovascular dysfunction. Peripheral immune cell invasion into the brain, along with these responses, is implicitly involved in epilepsy. This review explored the current literature on the association between the peripheral and central nervous systems in the pathogenesis of epilepsy, and highlights novel research directions for therapeutic interventions targeting these reactions. Previous experimental and human studies have demonstrated the activation of the innate and adaptive immune responses in the brain. The time required for monocytes (responsible for innate immunity) and T cells (involved in acquired immunity) to invade the central nervous system after a seizure varies. Moreover, the time between the leakage associated with blood–brain barrier (BBB) failure and the infiltration of these cells varies. This suggests that cell infiltration is not merely a secondary disruptive event associated with BBB failure, but also a non-disruptive event facilitated by various mediators produced by the neurovascular unit consisting of neurons, perivascular astrocytes, microglia, pericytes, and endothelial cells. Moreover, genetic manipulation has enabled the differentiation between peripheral monocytes and resident microglia, which was previously considered difficult. Thus, the evidence suggests that peripheral monocytes may contribute to the pathogenesis of seizures.
Collapse
|
15
|
Riew TR, Jin X, Kim S, Kim HL, Lee MY. Temporal dynamics of cells expressing NG2 and platelet-derived growth factor receptor-β in the fibrotic scar formation after 3-nitropropionic acid-induced acute brain injury. Cell Tissue Res 2021; 385:539-555. [PMID: 33864501 DOI: 10.1007/s00441-021-03438-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/18/2021] [Indexed: 12/22/2022]
Abstract
Neuron-glia antigen 2 (NG2) proteoglycan and platelet-derived growth factor receptor beta (PDGFR-β) are widely used markers of pericytes, which are considered cells that form fibrotic scars in response to central nervous system insults. However, the exact phenotypes of NG2- and PDGFR-β-expressing cells, as well as the origin of the fibrotic scar after central nervous system insults, are still elusive. In the present study, we directly examined the identities and distributions of NG2- and PDGFR-β-positive cells in the control and lesioned striatum injured by the mitochondrial toxin 3-nitropropionic acid. Immunoelectron microscopy and correlative light and electron microscopy clearly distinguished NG2 and PDGFR-β expression in the vasculature during the post-injury period. Vascular smooth muscle cells and pericytes expressed NG2, which was prominently increased after the injury. NG2 expression was restricted to these vascular mural cells until 14 days post-lesion. By contrast, PDGFR-β-positive cells were perivascular fibroblasts located abluminal to smooth muscle cells or pericytes. These PDGFR-β-expressing cells formed extravascular networks associated with collagen fibrils at 14 days post-lesion. We also found that in the injured striatal parenchyma, PDGFR-β could be used as a complementary marker of resting and reactive NG2 glia because activated microglia/macrophages shared only the NG2 expression with NG2 glia in the lesioned striatum. These data indicate that NG2 and PDGFR-β label different vascular mural and parenchymal cells in the healthy and injured brain, suggesting that fibrotic scar-forming cells most likely originate in PDGFR-β-positive perivascular fibroblasts rather than in NG2-positive pericytes.
Collapse
Affiliation(s)
- Tae-Ryong Riew
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Xuyan Jin
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.,Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Soojin Kim
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Hong Lim Kim
- Integrative Research Support Center, Laboratory of Electron Microscope, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Mun-Yong Lee
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea. .,Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| |
Collapse
|
16
|
Liu JYW, Reeves C, van der Pijl R, Thom M. Glial regenerative cell types in the superficial cortex in cortical dysplasia subtypes. Epilepsy Res 2020; 169:106529. [PMID: 33370704 PMCID: PMC7829594 DOI: 10.1016/j.eplepsyres.2020.106529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 11/23/2020] [Accepted: 12/04/2020] [Indexed: 10/29/2022]
Abstract
PURPOSE Focal Cortical Dysplasias (FCD) are localized malformative brain lesions in epilepsy. FCD3a associated with hippocampal sclerosis, affects the superficial cortex and is presumed to have an 'acquired' rather than developmental origin. Precursor cells may arise outside neurogenic zones including cortical layer I. Our aim was to characterise subsets of glial progenitor cells in the superficial cortical layers, known to be involved in gliosis and gliogenesis and that could distinguish FCD3a from other subtypes. METHODS Using immunohistochemistry we quantified the density of glial progenitor subsets in superficial cortex layers using markers against PAX6, GFAP, Olig2 and PDGFRβ and proliferation marker MCM2 in ten FCD3a cases compared to 18 other FCD types and 11 non-FCD controls. KEY FINDINGS Glial progenitor cells types were present in the cortical layer I and II in all FCD groups. GFAP cells frequently expressed PAX6 and significantly higher GFAP/PAX6 than GFAP/MCM2 cell densities were identified in the FCD3a group (p < 0.05). Olig2 cell densities were significantly higher in FCD3b than FCD3a (p = 0.002) and significantly higher GFAP/MCM2 compared to PDGFRβ/MCM2 cell densities were identified in both FCD3b and FCD2 groups. There was no correlation between cell densities and the age of patients at surgery and between cortical regions. SIGNIFICANCE Immature and proliferative glial populations across FCD variants reflect reactive cell types and differences may provide insight into underlying pathomechanisms. Higher PAX6 expression in astroglial cells in FCD3a may indicate a switch to astrocytic maturation and enhanced superficial gliosis. Higher Olig2 and GFAP/MCM2 densities in FCD3b may reflect margins of the tumour infiltration zone rather than true cortical dysplasia.
Collapse
Affiliation(s)
- Joan Y W Liu
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK; School of Life Sciences, University of Westminster, London, W1W 6UW, UK
| | - Cheryl Reeves
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK
| | - Rianne van der Pijl
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK
| | - Maria Thom
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK; Division of Neuropathology, National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG, UK.
| |
Collapse
|
17
|
Kyyriäinen J, Tapiala J, Lipponen A, Ekolle Ndode-Ekane X, Pitkänen A. Plau/Plaur double-deficiency did not worsen lesion severity or vascular integrity after traumatic brain injury. Neurosci Lett 2020; 729:134935. [PMID: 32360936 DOI: 10.1016/j.neulet.2020.134935] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/06/2020] [Accepted: 03/25/2020] [Indexed: 12/27/2022]
Abstract
Binding of urokinase-type plasminogen activator receptor (uPAR) to its ligand uPA or to its plasma membrane partner, platelet-derived growth factor receptor β (PDGFRβ), promotes neuroprotection, cell proliferation, and angiogenesis. Following injury, single deficiency in uPA or uPAR leads in increased tissue loss and compromised vascular remodeling. We hypothesized that double-deficiency of uPAR (Plaur) and uPA (Plau) would result in increased lesion area and poor vascular integrity after traumatic brain injury (TBI). TBI was induced by lateral fluid-percussion injury in Plau/Plaur double-knockout (dKO) and wild-type (Wt) mice. The cortical lesion area was quantified in unfolded cortical maps prepared from thionin-stained sections at 4 d or 30 d post-TBI. The density of PDGFRβ+ pericytes and blood vessels was calculated from immunostained sections. Blood-brain barrier leakage was analyzed using ImageJ® from IgG-immunostained sections. Genotype had no effect on the total area of the cortical lesion at 4 d or 30 d post-TBI (p > 0.05) or its progression as the overall lesion area was comparable at 4 d and 30 d post-TBI in both genotypes (p > 0.05). Subfield analysis, however, indicated that damage to the visual cortex at 4 d post-TBI in dKO-TBI mice was 53 % of that in Wt-TBI mice (p < 0.05). Both genotypes had a higher density of PDGFRβ-positive pericytes at 4 d than at 30 d post-TBI (p < 0.05), but no genotype effect was detected between these time-points (p > 0.05). TBI-induced increase in the density of PDGFRβ+ blood vessels at the region adjacent to the lesion core was comparable in both genotypes (p > 0.05). Genotype had no effect on TBI-induced IgG leakage into the perilesional cortical parenchyma (p > 0.05). Contrary to our expectations, Plau/Plaur double-deficiency did not aggravate TBI-related structural outcome.
Collapse
Affiliation(s)
- Jenni Kyyriäinen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FI-70211, Kuopio, Finland
| | - Jesse Tapiala
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FI-70211, Kuopio, Finland
| | - Anssi Lipponen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FI-70211, Kuopio, Finland
| | - Xavier Ekolle Ndode-Ekane
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FI-70211, Kuopio, Finland
| | - Asla Pitkänen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FI-70211, Kuopio, Finland.
| |
Collapse
|
18
|
Riew TR, Jin X, Kim HL, Kim S, Lee MY. Ultrastructural and Molecular Characterization of Platelet-derived growth factor Beta-Positive Leptomeningeal Cells in the Adult Rat Brain. Mol Neurobiol 2019; 57:1484-1501. [PMID: 31773411 DOI: 10.1007/s12035-019-01793-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 09/22/2019] [Indexed: 02/01/2023]
Abstract
The leptomeninges, referring to the arachnoid and pia mater and their projections into the perivascular compartments in the central nervous system, actively participate in diverse biological processes including fluid homeostasis, immune cell infiltrations, and neurogenesis, yet their detailed cellular and molecular identities remain elusive. This study aimed to characterize platelet-derived growth factor beta (PDGFR-β)-expressing cells in the leptomeninges in the adult rat brain using light and electron microscopy. PDGFR-β+ cells were observed in the inner arachnoid, arachnoid trabeculae, pia mater, and leptomeningeal sheath of the subarachnoid vessels, thereby forming a cellular network throughout the leptomeninges. Leptomeningeal PDGFR-β+ cells were commonly characterized by large euchromatic nuclei, thin branching processes forming web-like network, and the expression of the intermediate filaments nestin and vimentin. These cells were typical of active fibroblasts with a well-developed rough endoplasmic reticulum and close spatial correlation with collagen fibrils. Leptomeningeal PDGFR-β+ cells ensheathing the vasculature in the subarachnoid space joined with pial PDGFR-β+ cells upon entering the cortical parenchyma, yet perivascular PDGFR-β+ cells in these penetrating vessels underwent abrupt changes in their morphological and molecular characteristics: they became more flattened with loss of immunoreactivity for nestin and vimentin and deficient collagen deposition, which was indicative of inactive fibroblasts termed fibrocytes. In the cortical parenchyma, PDGFR-β immunoreactivity was almost exclusively localized to larger caliber vessels, and significantly decreased in capillary-like microvessels. Collectively, our data identify PDGFR-β as a novel cellular marker for leptomeningeal fibroblasts comprising the leptomeninges and perivascular adventitial cells of the subarachnoid and penetrating large-sized cortical vasculatures.
Collapse
Affiliation(s)
- Tae-Ryong Riew
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Korea
| | - Xuyan Jin
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Korea.,Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Hong Lim Kim
- Integrative Research Support Center, Laboratory of Electron Microscope, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Soojin Kim
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Korea
| | - Mun-Yong Lee
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Korea. .,Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
| |
Collapse
|
19
|
Arango-Lievano M, Boussadia B, De Terdonck LDT, Gault C, Fontanaud P, Lafont C, Mollard P, Marchi N, Jeanneteau F. Topographic Reorganization of Cerebrovascular Mural Cells under Seizure Conditions. Cell Rep 2019; 23:1045-1059. [PMID: 29694884 DOI: 10.1016/j.celrep.2018.03.110] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 01/04/2018] [Accepted: 03/22/2018] [Indexed: 12/27/2022] Open
Abstract
Reorganization of the neurovascular unit has been suggested in the epileptic brain, although the dynamics and functional significance remain unclear. Here, we tracked the in vivo dynamics of perivascular mural cells as a function of electroencephalogram (EEG) activity following status epilepticus. We segmented the cortical vascular bed to provide a size- and type-specific analysis of mural cell plasticity topologically. We find that mural cells are added and removed from veins, arterioles, and capillaries after seizure induction. Loss of mural cells is proportional to seizure severity and vascular pathology (e.g., rigidity, perfusion, and permeability). Treatment with platelet-derived growth factor subunits BB (PDGF-BB) reduced mural cell loss, vascular pathology, and epileptiform EEG activity. We propose that perivascular mural cells play a pivotal role in seizures and are potential targets for reducing pathophysiology.
Collapse
Affiliation(s)
- Margarita Arango-Lievano
- Departments of Neuroscience & Physiology, Laboratory of Stress Hormones & Plasticity, Institut de Génomique Fonctionnelle, INSERM, CNRS, University of Montpellier, 34093 Montpellier, France.
| | - Badreddine Boussadia
- Department of Neuroscience, Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Institut de Génomique Fonctionnelle, INSERM, CNRS, University of Montpellier, 34093 Montpellier, France
| | - Lucile Du Trieu De Terdonck
- Departments of Neuroscience & Physiology, Laboratory of Stress Hormones & Plasticity, Institut de Génomique Fonctionnelle, INSERM, CNRS, University of Montpellier, 34093 Montpellier, France
| | - Camille Gault
- Departments of Neuroscience & Physiology, Laboratory of Stress Hormones & Plasticity, Institut de Génomique Fonctionnelle, INSERM, CNRS, University of Montpellier, 34093 Montpellier, France
| | - Pierre Fontanaud
- Department of Physiology, Laboratory of Networks and Rhythms in Endocrine Glands, Institut de Génomique Fonctionnelle, INSERM, CNRS, University of Montpellier, 34093 Montpellier, France
| | - Chrystel Lafont
- Department of Physiology, Laboratory of Networks and Rhythms in Endocrine Glands, Institut de Génomique Fonctionnelle, INSERM, CNRS, University of Montpellier, 34093 Montpellier, France
| | - Patrice Mollard
- Department of Physiology, Laboratory of Networks and Rhythms in Endocrine Glands, Institut de Génomique Fonctionnelle, INSERM, CNRS, University of Montpellier, 34093 Montpellier, France
| | - Nicola Marchi
- Department of Neuroscience, Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Institut de Génomique Fonctionnelle, INSERM, CNRS, University of Montpellier, 34093 Montpellier, France.
| | - Freddy Jeanneteau
- Departments of Neuroscience & Physiology, Laboratory of Stress Hormones & Plasticity, Institut de Génomique Fonctionnelle, INSERM, CNRS, University of Montpellier, 34093 Montpellier, France.
| |
Collapse
|
20
|
Klement W, Blaquiere M, Zub E, deBock F, Boux F, Barbier E, Audinat E, Lerner-Natoli M, Marchi N. A pericyte-glia scarring develops at the leaky capillaries in the hippocampus during seizure activity. Epilepsia 2019; 60:1399-1411. [PMID: 31135065 DOI: 10.1111/epi.16019] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 04/28/2019] [Accepted: 04/28/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Inflammatory cerebrovascular damage occurs in epilepsy. Here, we tested the hypothesis that a pericyte-glia scar forms around the outer wall of hippocampal capillaries in a model of temporal lobe epilepsy associated with hippocampal sclerosis. We studied the participation of stromal cells expressing platelet-derived growth factor receptor beta (PDGFRβ) and extracellular matrix modifications to the perivascular scar during epileptogenesis. METHODS We used NG2DsRed/C57BL6 mice and induced status epilepticus (SE) followed by epileptogenesis and spontaneous recurrent seizures (SRS) by means of unilateral intrahippocampal injection of kainic acid (KA). For pharmacological assessment, we used organotypic hippocampal cultures (OHCs) where ictal electrographic activity was elicited by KA or bicuculline. RESULTS NG2DsRed pericytes, GFAP astroglia, and IBA1 microglia are reactive and converge to form a pericapillary multicellular scar in the CA hippocampal regions during epileptogenesis and at SRS. The capillaries are leaky as indicated by fluorescein entering the parenchyma from the peripheral blood. Concomitantly, PDGFRβ transcript and protein levels were significantly increased. Within the regional scar, a fibrotic-like PDGFRβ mesh developed around the capillaries, peaking at 1 week post-SE and regressing, but not resolving, at SRS. Abnormal distribution or accumulation of extracellular matrix collagens III/IV occurred in the CA regions during seizure progression. PDGFRβ/DAPI cells were in direct contact with or adjacent to the damaged NG2DsRed pericytes at the capillary interface, consistent with the notion of stromal cell reactivity or fibroblast formation. Inducing electrographic activity in OHCs was sufficient to augment PDGFRβ reactivity around the capillaries. The latter effect was pharmacologically mimicked by treating OHCs with the PDGFRβ agonist PDGF-BB and it was diminished by the PDGFRβ inhibitor imatinib. SIGNIFICANCE The reported multicellular activation and scar are traits of perivascular inflammation and hippocampal sclerosis in experimental epilepsy, with an implication for neurovascular dysfunction. Modulation of PDGFRβ could be exploited to target inflammation in this chronic disease setting.
Collapse
Affiliation(s)
- Wendy Klement
- Laboratory of Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Marine Blaquiere
- Laboratory of Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Emma Zub
- Laboratory of Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Frederic deBock
- Laboratory of Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Fabien Boux
- Grenoble Neuroscience Institute, GIN, Inserm U 1216 - Grenoble University, La Tronche, France
| | - Emmanuel Barbier
- Grenoble Neuroscience Institute, GIN, Inserm U 1216 - Grenoble University, La Tronche, France
| | - Etienne Audinat
- Laboratory of Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Mireille Lerner-Natoli
- Laboratory of Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Nicola Marchi
- Laboratory of Cerebrovascular and Glia Research, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| |
Collapse
|
21
|
Reeves C, Pradim-Jardim A, Sisodiya SM, Thom M, Liu JYW. Spatiotemporal dynamics of PDGFRβ expression in pericytes and glial scar formation in penetrating brain injuries in adults. Neuropathol Appl Neurobiol 2019; 45:609-627. [PMID: 30636077 PMCID: PMC6767497 DOI: 10.1111/nan.12539] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 10/17/2018] [Indexed: 12/30/2022]
Abstract
Aims Understanding the spatiotemporal dynamics of reactive cell types following brain injury is important for future therapeutic interventions. We have previously used penetrating cortical injuries following intracranial recordings as a brain repair model to study scar‐forming nestin‐expressing cells. We now explore the relationship between nestin‐expressing cells, PDGFRβ+ pericytes and Olig2+ glia, including their proliferation and functional maturation. Methods In 32 cases, ranging from 3 to 461 days post injury (dpi), immunohistochemistry for PDGFRβ, nestin, GFAP, Olig2, MCM2, Aquaporin 4 (Aq4), Glutamine Synthetase (GS) and Connexin 43 (Cx43) was quantified for cell densities, labelling index (LI) and cellular co‐expression at the injury site compared to control regions. Results PDGFRβ labelling highlighted both pericytes and multipolar parenchymal cells. PDGFRβ LI and PDGFRβ+/MCM2+ cells significantly increased in injury Zones at 10–13 dpi with migration of pericytes away from vessels with increased co‐localization of PDGRFβ with nestin compared to control regions (P < 0.005). Olig2+/MCM2+ cell populations peaked at 13 dpi with significantly higher cell densities at injury sites than in control regions (P < 0.01) and decreasing with dpi (P < 0.05). Cx43 LI was reduced in acute injuries but increased with dpi (P < 0.05) showing significant cellular co‐localization with nestin and GFAP (P < 0.005 and P < 0.0001) but not PDGFRβ. Conclusions These findings indicate that PDGFRβ+ and Olig2+ cells contribute to the proliferative fraction following penetrating brain injuries, with evidence of pericyte migration. Dynamic changes in Cx43 in glial cell types with dpi suggest functional alterations during temporal stages of brain repair.
Collapse
Affiliation(s)
- C Reeves
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, Queen Square, London.,Department of Neuropathology, UCL Queen Square Institute of Neurology, Queen Square, London
| | - A Pradim-Jardim
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, Queen Square, London.,Department of Neurology and Neurosurgery, Universidade Federal de Sao Paulo, UNIFESP, Sao Paulo/SP, Brazil
| | - S M Sisodiya
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, Queen Square, London.,Chalfont Centre for Epilepsy, Chesham Lane, Chalfont St Peter, Bucks, SL9 0RJ, UK
| | - M Thom
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, Queen Square, London.,Department of Neuropathology, UCL Queen Square Institute of Neurology, Queen Square, London
| | - J Y W Liu
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, Queen Square, London.,Department of Neuropathology, UCL Queen Square Institute of Neurology, Queen Square, London.,School of life Sciences, University of Westminster, London, W1W 6UW, UK
| |
Collapse
|
22
|
Geranmayeh MH, Rahbarghazi R, Farhoudi M. Targeting pericytes for neurovascular regeneration. Cell Commun Signal 2019; 17:26. [PMID: 30894190 PMCID: PMC6425710 DOI: 10.1186/s12964-019-0340-8] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 03/13/2019] [Indexed: 02/06/2023] Open
Abstract
Pericytes, as a key cellular part of the blood-brain barrier, play an important role in the maintenance of brain neurovascular unit. These cells participate in brain homeostasis by regulating vascular development and integrity mainly through secreting various factors. Pericytes per se show different restorative properties after blood-brain barrier injury. Upon the occurrence of brain acute and chronic diseases, pericytes provoke immune cells to regulate neuro-inflammatory conditions. Loss of pericytes in distinct neurologic disorders intensifies blood-brain barrier permeability and leads to vascular dementia. The therapeutic potential of pericytes is originated from the unique morphological shape, location, and their ability in providing vast paracrine and juxtacrine interactions. A subset of pericytes possesses multipotentiality and exhibit trans-differentiation capacity in the context of damaged tissue. This review article aimed to highlight the critical role of pericytes in restoration of the blood-brain barrier after injury by focusing on the dynamics of pericytes and cross-talk with other cell types.
Collapse
Affiliation(s)
- Mohammad Hossein Geranmayeh
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Neurosciences Research Center (NSRC), Imam Reza Medical Center, Tabriz University of Medical Sciences, Golgasht St., Azadi Ave, Tabriz, 5166614756, Iran
| | - Reza Rahbarghazi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mehdi Farhoudi
- Neurosciences Research Center (NSRC), Imam Reza Medical Center, Tabriz University of Medical Sciences, Golgasht St., Azadi Ave, Tabriz, 5166614756, Iran.
| |
Collapse
|
23
|
Stebbins MJ, Gastfriend BD, Canfield SG, Lee MS, Richards D, Faubion MG, Li WJ, Daneman R, Palecek SP, Shusta EV. Human pluripotent stem cell-derived brain pericyte-like cells induce blood-brain barrier properties. SCIENCE ADVANCES 2019; 5:eaau7375. [PMID: 30891496 PMCID: PMC6415958 DOI: 10.1126/sciadv.aau7375] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 01/29/2019] [Indexed: 05/18/2023]
Abstract
Brain pericytes play important roles in the formation and maintenance of the neurovascular unit (NVU), and their dysfunction has been implicated in central nervous system disorders. While human pluripotent stem cells (hPSCs) have been used to model other NVU cell types, including brain microvascular endothelial cells (BMECs), astrocytes, and neurons, hPSC-derived brain pericyte-like cells have not been integrated into these models. In this study, we generated neural crest stem cells (NCSCs), the embryonic precursor to forebrain pericytes, from hPSCs and subsequently differentiated NCSCs to brain pericyte-like cells. These cells closely resembled primary human brain pericytes and self-assembled with endothelial cells. The brain pericyte-like cells induced blood-brain barrier properties in BMECs, including barrier enhancement and reduced transcytosis. Last, brain pericyte-like cells were incorporated with iPSC-derived BMECs, astrocytes, and neurons to form an isogenic human model that should prove useful for the study of the NVU.
Collapse
Affiliation(s)
- Matthew J. Stebbins
- Department of Chemical and Biological Engineering, University of Wisconsin–Madison, Madison, WI, USA
| | - Benjamin D. Gastfriend
- Department of Chemical and Biological Engineering, University of Wisconsin–Madison, Madison, WI, USA
| | - Scott G. Canfield
- Department of Chemical and Biological Engineering, University of Wisconsin–Madison, Madison, WI, USA
| | - Ming-Song Lee
- Department of Orthopedics and Rehabilitation, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Drew Richards
- Department of Chemical and Biological Engineering, University of Wisconsin–Madison, Madison, WI, USA
| | - Madeline G. Faubion
- Department of Chemical and Biological Engineering, University of Wisconsin–Madison, Madison, WI, USA
| | - Wan-Ju Li
- Department of Orthopedics and Rehabilitation, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Richard Daneman
- Departments of Neuroscience and Pharmacology, University of California, San Diego, San Diego, CA, USA
| | - Sean P. Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin–Madison, Madison, WI, USA
| | - Eric V. Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin–Madison, Madison, WI, USA
| |
Collapse
|
24
|
Prager O, Kamintsky L, Hasam‐Henderson LA, Schoknecht K, Wuntke V, Papageorgiou I, Swolinsky J, Muoio V, Bar‐Klein G, Vazana U, Heinemann U, Friedman A, Kovács R. Seizure‐induced microvascular injury is associated with impaired neurovascular coupling and blood–brain barrier dysfunction. Epilepsia 2019; 60:322-336. [DOI: 10.1111/epi.14631] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 12/03/2018] [Accepted: 12/03/2018] [Indexed: 01/04/2023]
Affiliation(s)
- Ofer Prager
- Departments of Physiology and Cell Biology Cognitive and Brain Sciences, Biomedical Engineering, Zlotowski Center for Neuroscience, Ben‐Gurion University of the Negev Beer‐Sheva Israel
| | - Lyna Kamintsky
- Departments of Physiology and Cell Biology Cognitive and Brain Sciences, Biomedical Engineering, Zlotowski Center for Neuroscience, Ben‐Gurion University of the Negev Beer‐Sheva Israel
- Department of Medical Neuroscience Faculty of Medicine Dalhousie University Halifax Nova Scotia Canada
| | - Luisa A. Hasam‐Henderson
- Institute for Neurophysiology Charité– Medical University Berlin, corporate member of Free University of Berlin, Humboldt University of Berlin, and Berlin Institute of Health Berlin Germany
| | - Karl Schoknecht
- Neuroscience Research Center Charité—Medical University Berlin, corporate member of Free University of Berlin, Humboldt University of Berlin, and Berlin Institute of Health Berlin Germany
| | - Vera Wuntke
- Institute for Neurophysiology Charité– Medical University Berlin, corporate member of Free University of Berlin, Humboldt University of Berlin, and Berlin Institute of Health Berlin Germany
| | - Ismini Papageorgiou
- Institute for Neurophysiology Charité– Medical University Berlin, corporate member of Free University of Berlin, Humboldt University of Berlin, and Berlin Institute of Health Berlin Germany
| | - Jutta Swolinsky
- Institute for Neurophysiology Charité– Medical University Berlin, corporate member of Free University of Berlin, Humboldt University of Berlin, and Berlin Institute of Health Berlin Germany
| | - Valeria Muoio
- Institute for Neurophysiology Charité– Medical University Berlin, corporate member of Free University of Berlin, Humboldt University of Berlin, and Berlin Institute of Health Berlin Germany
| | - Guy Bar‐Klein
- McKusick‐Nathans Institute of Genetic Medicine Johns Hopkins University School of Medicine Baltimore Maryland
- Howard Hughes Medical Institute Chevy Chase Maryland
| | - Udi Vazana
- Departments of Physiology and Cell Biology Cognitive and Brain Sciences, Biomedical Engineering, Zlotowski Center for Neuroscience, Ben‐Gurion University of the Negev Beer‐Sheva Israel
| | | | - Alon Friedman
- Departments of Physiology and Cell Biology Cognitive and Brain Sciences, Biomedical Engineering, Zlotowski Center for Neuroscience, Ben‐Gurion University of the Negev Beer‐Sheva Israel
- Department of Medical Neuroscience Faculty of Medicine Dalhousie University Halifax Nova Scotia Canada
| | - Richard Kovács
- Department of Medical Neuroscience Faculty of Medicine Dalhousie University Halifax Nova Scotia Canada
| |
Collapse
|
25
|
Riew TR, Choi JH, Kim HL, Jin X, Lee MY. PDGFR-β-Positive Perivascular Adventitial Cells Expressing Nestin Contribute to Fibrotic Scar Formation in the Striatum of 3-NP Intoxicated Rats. Front Mol Neurosci 2018; 11:402. [PMID: 30455628 PMCID: PMC6230557 DOI: 10.3389/fnmol.2018.00402] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 10/15/2018] [Indexed: 11/13/2022] Open
Abstract
Perivascular cells expressing platelet-derived growth factor receptor beta (PDGFR-β) have recently been implicated in fibrotic scar formation after acute brain injury, but their precise identity and detailed morphological characteristics remain elusive. This study sought to characterize and define the cellular phenotype of vascular-associated cells expressing PDGFR-β in the striatum of rats treated with the mitochondrial toxin 3-nitropropionic acid (3-NP). In the control striatum, PDGFR-β-positive cells were invariably localized on the abluminal side of smooth muscle cells of larger caliber vessels, and demonstrated morphological features typical of perivascular fibroblasts. PDGFR-β expression increased and expanded to almost all vessels, including microvessels in the lesion core, at 7 days after 3-NP injection. The cells expressing PDGFR-β had ultrastructural features of fibroblasts undergoing active collagen synthesis: large euchromatic nuclei with a prominent nucleolus, well-developed rough endoplasmic reticulum (rER) with dilated cisterns and extracellular collagen fibrils. By 14 days, PDGFR-β-positive cells had somata located at a distance from the vasculature, and their highly ramified, slender processes overlapped with those from other cells, thus forming a plexus of processes in the extravascular space of the lesion core. In addition, their ultrastructural morphology and spatial correlation with activated microglia/macrophages were elaborated by three-dimensional reconstruction. Using a correlative light- and electron-microscopy technique, we found that the intermediate filament proteins nestin and vimentin were induced in PDGFRβ-positive fibroblasts in the lesion core. Collectively, our data suggest that perivascular PDGFR-β-positive fibroblasts are distinct from other vascular cell types, including pericytes and contribute to fibrotic scar formation in the lesion core after acute brain injury. Nestin and vimentin play critical roles in the structural dynamics of these reactive fibroblasts.
Collapse
Affiliation(s)
- Tae-Ryong Riew
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jeong-Heon Choi
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Hong Lim Kim
- Integrative Research Support Center, Laboratory of Electron Microscope, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Xuyan Jin
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Mun-Yong Lee
- Department of Anatomy, Catholic Neuroscience Institute, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
26
|
Liu JYW, Matarin M, Reeves C, McEvoy AW, Miserocchi A, Thompson P, Sisodiya SM, Thom M. Doublecortin-expressing cell types in temporal lobe epilepsy. Acta Neuropathol Commun 2018; 6:60. [PMID: 30005693 PMCID: PMC6045867 DOI: 10.1186/s40478-018-0566-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/03/2018] [Indexed: 12/14/2022] Open
Abstract
Doublecortin (DCX) is widely regarded as a marker of immature and migrating neurons during development. While DCX expression persists in adults, particularly in the temporal lobe and neurogenic regions, it is unknown how seizures influence its expression. The aim of the present study was to explore the distribution and characteristics of DCX-expressing cells in surgical and postmortem samples from 40 adult and paediatric patients, with epilepsy and with or without hippocampal sclerosis (HS), compared to post mortem controls. The hippocampus (pes and body), parahippocampal gyrus, amygdala, temporal pole and temporal cortex were examined with DCX immunohistochemistry using four commercially-available DCX antibodies, labelled cells were quantified in different regions of interest as well as their co-expression with cell type specific markers (CD68, Iba1, GFAP, GFAP∂, nestin, SOX2, CD34, OLIG2, PDGFRβ, NeuN) and cell cycle marker (MCM2). Histological findings were compared with clinical data, as well as gene expression data obtained from the temporal cortex of 83 temporal lobe epilepsy cases with HS. DCX immunohistochemistry identified immature (Nestin−/NeuN−) neurons in layer II of the temporal neocortex in patients with and without epilepsy. Their number declined significantly with age but was not associated with the presence of hippocampal sclerosis, seizure semiology or memory dysfunction. DCX+ cells were prominent in the paralaminar nuclei and periamygdalar cortex and these declined with age but were not significantly associated with epilepsy history. DCX expressing cells with ramified processes were prominent in all regions, particularly in the hippocampal subgranular zone, where significantly increased numbers were observed in epilepsy samples compared to controls. DCX ramified cells co-expressed Iba1, CD68 and PDGFRβ, and less frequently MCM2, OLIG2 and SOX2, but no co-localization was observed with CD34, nestin or GFAP/GFAP ∂. Gene expression data from neocortical samples in patients with TLE and HS supported ongoing DCX expression in adults. We conclude that DCX identifies a range of morphological cell types in temporal lobe epilepsy, including immature populations, glial and microglial cell types. Their clinical relevance and biological function requires further study but we show some evidence for alteration with age and in epilepsy.
Collapse
|
27
|
Smyth LCD, Rustenhoven J, Scotter EL, Schweder P, Faull RLM, Park TIH, Dragunow M. Markers for human brain pericytes and smooth muscle cells. J Chem Neuroanat 2018; 92:48-60. [PMID: 29885791 DOI: 10.1016/j.jchemneu.2018.06.001] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 04/20/2018] [Accepted: 06/06/2018] [Indexed: 01/24/2023]
Abstract
Brain pericytes and vascular smooth muscle cells (vSMCs) are a critical component of the neurovascular unit and are important in regulating cerebral blood flow and blood-brain barrier integrity. Identification of subtypes of mural cells in tissue and in vitro is important to any study of their function, therefore we identified distinct mural cell morphologies in neurologically normal post-mortem human brain. Further, the distribution of mural cell markers platelet-derived growth factor receptor-β (PDGFRβ), α-smooth muscle actin (αSMA), CD13, neural/glial antigen-2 (NG2), CD146 and desmin was examined. We determined that PDGFRβ, NG2, CD13, and CD146 were expressed in capillary-associated pericytes. NG2, and CD13 were also present on vSMCs in large vessels, however abundant CD146 and desmin staining was also detected in vSMCs on large vessels, co-labelling with αSMA. To determine whether cultures recapitulated observations from tissue, primary human brain pericytes derived from neurologically normal autopsies were analysed for the presence of pericyte markers by immunocytochemistry, western blotting and qPCR. The proteins observed in brain pericytes in tissue (PDGFRβ, αSMA, desmin, CD146, CD13, and NG2) were present in vitro, validating a panel of proteins that can be used to label brain pericytes and vSMCs in tissue and in vitro. Finally, we showed that the proteins CD146 and desmin that are expressed on large vessels in situ, are also selective markers of a smooth muscle cell phenotype in vitro.
Collapse
Affiliation(s)
- Leon C D Smyth
- Department of Pharmacology and Clinical Pharmacology, Auckland, New Zealand; Centre for Brain Research, Auckland, New Zealand
| | - Justin Rustenhoven
- Department of Pharmacology and Clinical Pharmacology, Auckland, New Zealand; Centre for Brain Research, Auckland, New Zealand
| | - Emma L Scotter
- Department of Pharmacology and Clinical Pharmacology, Auckland, New Zealand; Centre for Brain Research, Auckland, New Zealand
| | - Patrick Schweder
- Centre for Brain Research, Auckland, New Zealand; Auckland City Hospital, Auckland, New Zealand
| | - Richard L M Faull
- Centre for Brain Research, Auckland, New Zealand; Department of Anatomy and Medical Imaging, Auckland, New Zealand
| | - Thomas I H Park
- Department of Pharmacology and Clinical Pharmacology, Auckland, New Zealand; Centre for Brain Research, Auckland, New Zealand; Department of Anatomy and Medical Imaging, Auckland, New Zealand
| | - Mike Dragunow
- Department of Pharmacology and Clinical Pharmacology, Auckland, New Zealand; Centre for Brain Research, Auckland, New Zealand.
| |
Collapse
|
28
|
Giannoni P, Badaut J, Dargazanli C, Fayd'Herbe De Maudave A, Klement W, Costalat V, Marchi N. The pericyte-glia interface at the blood-brain barrier. Clin Sci (Lond) 2018; 132:361-374. [PMID: 29439117 DOI: 10.1042/cs20171634] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/04/2018] [Accepted: 01/04/2018] [Indexed: 12/30/2022]
Abstract
The cerebrovasculature is a multicellular structure with varying rheological and permeability properties. The outer wall of the brain capillary endothelium is enclosed by pericytes and astrocyte end feet, anatomically assembled to guarantee barrier functions. We, here, focus on the pericyte modifications occurring in disease conditions, reviewing evidence supporting the interplay amongst pericytes, the endothelium, and glial cells in health and pathology. Deconstruction and reactivity of pericytes and glial cells around the capillary endothelium occur in response to traumatic brain injury, epilepsy, and neurodegenerative disorders, impacting vascular permeability and participating in neuroinflammation. As this represents a growing field of research, addressing the multicellular reorganization occurring at the outer wall of the blood-brain barrier (BBB) in response to an acute insult or a chronic disease could disclose novel disease mechanisms and therapeutic targets.
Collapse
Affiliation(s)
| | - Jerome Badaut
- Laboratory of Brain Molecular Imaging, CNRS UMR5287, University of Bordeaux, France
- Basic Science Departments, Loma Linda University School of Medicine, CA, U.S.A
| | - Cyril Dargazanli
- Neuroradiology, University Hospital, Montpellier, France
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Alexis Fayd'Herbe De Maudave
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Wendy Klement
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Vincent Costalat
- Neuroradiology, University Hospital, Montpellier, France
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Nicola Marchi
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| |
Collapse
|
29
|
Klement W, Garbelli R, Zub E, Rossini L, Tassi L, Girard B, Blaquiere M, Bertaso F, Perroy J, de Bock F, Marchi N. Seizure progression and inflammatory mediators promote pericytosis and pericyte-microglia clustering at the cerebrovasculature. Neurobiol Dis 2018; 113:70-81. [PMID: 29432809 DOI: 10.1016/j.nbd.2018.02.002] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 02/05/2018] [Accepted: 02/07/2018] [Indexed: 10/18/2022] Open
Abstract
BACKGROUND Cerebrovascular dysfunction and inflammation occur in epilepsy. Here we asked whether pericytes, a pivotal cellular component of brain capillaries, undergo pathological modifications during experimental epileptogenesis and in human epilepsy. We evaluated whether pro-inflammatory cytokines, present in the brain during seizures, contribute to pericyte morphological modifications. METHODS In vivo, unilateral intra-hippocampal kainic acid (KA) injections were performed in NG2DsRed/C57BL6 mice to induce status epilepticus (SE), epileptogenesis, and spontaneous recurrent seizures (SRS). NG2DsRed mice were used to visualize pericytes during seizure progression. The effect triggered by recombinant IL-1β, TNFα, or IL-6 on pericytes was evaluated in NG2DsRed hippocampal slices and in human-derived cell culture. Human brain specimens obtained from temporal lobe epilepsy (TLE) with or without sclerosis (HS) and focal cortical dysplasia (FCD-IIb) were evaluated for pericyte-microglial cerebrovascular assembly. RESULTS A disarray of NG2DsRed+ pericyte soma and ramifications was found 72 h post-SE and 1 week post-SE (epileptogenesis) in the hippocampus. Pericyte modifications topographically overlapped with IBA1+ microglia clustering around the capillaries with cases of pericytes lodged within the microglial cells. Microglial clustering around the NG2DsRed pericytes lingered at SRS. Pericyte proliferation (Ki67+) occurred 72 h post-SE and during epileptogenesis and returned towards control levels at SRS. Human epileptic brain tissues showed pericyte-microglia assemblies with IBA1/HLA microglial cells outlining the capillary wall in TLE-HS and FCD-IIb specimens. Inflammatory mediators contributed to pericyte modifications, in particular IL-1β elicited pericyte morphological changes and pericyte-microglia clustering in NG2DsRed hippocampal slices. Modifications also occurred when pro-inflammatory cytokines were added to an in vitro culture of pericytes. CONCLUSIONS These results indicate the occurrence of pericytosis during seizures and introduce a pericyte-microglial mediated mechanism of blood-brain barrier dysfunction in epilepsy.
Collapse
Affiliation(s)
- Wendy Klement
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Rita Garbelli
- Clinical Epileptology and Experimental Neurophysiology Unit, Fondazione IRCCS, Istituto Neurologico C. Besta, Milano, Italy
| | - Emma Zub
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Laura Rossini
- Clinical Epileptology and Experimental Neurophysiology Unit, Fondazione IRCCS, Istituto Neurologico C. Besta, Milano, Italy
| | - Laura Tassi
- C. Munari Epilepsy Surgery Centre, Ospedale Niguarda, Milano, Italy
| | - Benoit Girard
- Laboratory of Pathophysiology of Synaptic Transmission, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Marine Blaquiere
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Federica Bertaso
- Laboratory of Pathophysiology of Synaptic Transmission, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Julie Perroy
- Laboratory of Pathophysiology of Synaptic Transmission, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Frederic de Bock
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Nicola Marchi
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France.
| |
Collapse
|
30
|
Cerebrovascular heterogeneity and neuronal excitability. Neurosci Lett 2018; 667:75-83. [DOI: 10.1016/j.neulet.2017.01.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 01/04/2017] [Accepted: 01/06/2017] [Indexed: 01/01/2023]
|
31
|
Liu J, Reeves C, Jacques T, McEvoy A, Miserocchi A, Thompson P, Sisodiya S, Thom M. Nestin-expressing cell types in the temporal lobe and hippocampus: Morphology, differentiation, and proliferative capacity. Glia 2018; 66:62-77. [PMID: 28925561 PMCID: PMC5724502 DOI: 10.1002/glia.23211] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/30/2017] [Accepted: 08/09/2017] [Indexed: 12/13/2022]
Abstract
Nestin is expressed in immature neuroepithelial and progenitor cell types and transiently upregulated in proliferative neuroglial cells responding to acute brain injury, including following seizures. In 36 temporal lobe (TLobe) specimens from patients with TLobe epilepsy (age range 8-60 years) we studied the number, distribution and morphology of nestin-expressing cells (NEC) in the pes, hippocampus body, parahippocampal gyrus, amygdala, temporal cortex and pole compared with post mortem control tissues from 26 cases (age range 12 gestational weeks to 76 years). The proliferative fraction of NEC was evaluated in selected regions, including recognized niches, using MCM2. Their differentiation was explored with neuronal (DCX, mushashi, βIII tubulin, NeuN) and glial (GFAP, GFAPdelta, glutamine synthetase, aquaporin4, EAAT1) markers, both in sections or following culture. Findings were correlated with clinical parameters. A stereotypical pattern in the distribution and morphologies of NEC was observed, reminiscent of patterns in the developing brain, with increased densities in epilepsy than adult controls (p < .001). Findings included MCM2-positive radial glial-like cells in the periventricular white matter and rows of NEC in the hippocampal fimbria and sulcus. Nestin cells represented 29% of the hippocampal proliferative fraction in epilepsy cases; 20% co-expressed βIII tubulin in culture compared with 28% with GFAP. Significant correlations were noted between age at surgery, memory deficits and nestin populations. TLobe NEC with ongoing proliferative capacity likely represent vestiges of developmental migratory streams and resident reactive cell populations of potential relevance to hippocampal epileptogenesis, TLobe pathology, and co-morbidities, including memory decline.
Collapse
Affiliation(s)
- Joan Liu
- Department of Clinical and Experimental EpilepsyUCL Institute of Neurology, Queen SquareLondonWC1N 3BGUnited Kingdom
- Divisions of NeuropathologyNational Hospital for Neurology and Neurosurgery, Queen SquareLondonWCN1BGUK
- Department of Biomedical SciencesUniversity of WestminsterLondonW1W 6UWUnited Kingdom
| | - Cheryl Reeves
- Department of Clinical and Experimental EpilepsyUCL Institute of Neurology, Queen SquareLondonWC1N 3BGUnited Kingdom
- Divisions of NeuropathologyNational Hospital for Neurology and Neurosurgery, Queen SquareLondonWCN1BGUK
| | - Thomas Jacques
- Department of NeuropathologyUCL Institute of Child Health and Great Ormond Street Hospital for ChildrenLondonUnited Kingdom
| | - Andrew McEvoy
- Department of Clinical and Experimental EpilepsyUCL Institute of Neurology, Queen SquareLondonWC1N 3BGUnited Kingdom
- Neurosurgery at the National Hospital for Neurology and Neurosurgery, Queen SquareLondonWCN1BGUnited Kingdom
| | - Anna Miserocchi
- Neurosurgery at the National Hospital for Neurology and Neurosurgery, Queen SquareLondonWCN1BGUnited Kingdom
| | - Pamela Thompson
- Department of Clinical and Experimental EpilepsyUCL Institute of Neurology, Queen SquareLondonWC1N 3BGUnited Kingdom
- The Chalfont Centre for Epilepsy, Chesham Lane, Chalfont St PeterBuckinghamshireSL9 0RJUnited Kingdom
- Department of NeuropsychologyNational Hospital for Neurology and Neurosurgery, Queen SquareLondonWCN1BGUK
| | - Sanjay Sisodiya
- Department of Clinical and Experimental EpilepsyUCL Institute of Neurology, Queen SquareLondonWC1N 3BGUnited Kingdom
- The Chalfont Centre for Epilepsy, Chesham Lane, Chalfont St PeterBuckinghamshireSL9 0RJUnited Kingdom
- Department of NeurologyNational Hospital for Neurology and Neurosurgery, Queen SquareLondonWCN1BGUK
| | - Maria Thom
- Department of Clinical and Experimental EpilepsyUCL Institute of Neurology, Queen SquareLondonWC1N 3BGUnited Kingdom
- Divisions of NeuropathologyNational Hospital for Neurology and Neurosurgery, Queen SquareLondonWCN1BGUK
| |
Collapse
|
32
|
Thom M, Liu J, Bongaarts A, Reinten RJ, Paradiso B, Jäger HR, Reeves C, Somani A, An S, Marsdon D, McEvoy A, Miserocchi A, Thorne L, Newman F, Bucur S, Honavar M, Jacques T, Aronica E. Multinodular and vacuolating neuronal tumors in epilepsy: dysplasia or neoplasia? Brain Pathol 2017; 28:155-171. [PMID: 28833756 PMCID: PMC5887881 DOI: 10.1111/bpa.12555] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 06/27/2017] [Accepted: 08/01/2017] [Indexed: 12/28/2022] Open
Abstract
Multinodular and vacuolating neuronal tumor (MVNT) is a new pattern of neuronal tumour included in the recently revised WHO 2016 classification of tumors of the CNS. There are 15 reports in the literature to date. They are typically associated with late onset epilepsy and a neoplastic vs. malformative biology has been questioned. We present a series of ten cases and compare their pathological and genetic features to better characterized epilepsy‐associated malformations including focal cortical dysplasia type II (FCDII) and low‐grade epilepsy‐associated tumors (LEAT). Clinical and neuroradiology data were reviewed and a broad immunohistochemistry panel was applied to explore neuronal and glial differentiation, interneuronal populations, mTOR pathway activation and neurodegenerative changes. Next generation sequencing was performed for targeted multi‐gene analysis to identify mutations common to epilepsy lesions including FCDII and LEAT. All of the surgical cases in this series presented with seizures, and were located in the temporal lobe. There was a lack of any progressive changes on serial pre‐operative MRI and a mean age at surgery of 45 years. The vacuolated cells of the lesion expressed mature neuronal markers (neurofilament/SMI32, MAP2, synaptophysin). Prominent labelling of the lesional cells for developmentally regulated proteins (OTX1, TBR1, SOX2, MAP1b, CD34, GFAPδ) and oligodendroglial lineage markers (OLIG2, SMI94) was observed. No mutations were detected in the mTOR pathway genes, BRAF, FGFR1 or MYB. Clinical, pathological and genetic data could indicate that MVNT aligns more with a malformative lesion than a true neoplasm with origin from a progenitor neuro‐glial cell type showing aberrant maturation.
Collapse
Affiliation(s)
- Maria Thom
- Departments of Clinical and Experimental Epilepsy and Neuropathology, UCL Institute of Neurology and the National Hospital for Neurology and Neurosurgery, Queen Square, London WCN1BG, UK
| | - Joan Liu
- Departments of Clinical and Experimental Epilepsy and Neuropathology, UCL Institute of Neurology and the National Hospital for Neurology and Neurosurgery, Queen Square, London WCN1BG, UK
| | - Anika Bongaarts
- Department of (Neuro)Pathology, Academic Medical Center, Amsterdam, The Netherlands
| | - Roy J Reinten
- Department of (Neuro)Pathology, Academic Medical Center, Amsterdam, The Netherlands
| | - Beatrice Paradiso
- Departments of Clinical and Experimental Epilepsy and Neuropathology, UCL Institute of Neurology and the National Hospital for Neurology and Neurosurgery, Queen Square, London WCN1BG, UK.,Cardiovascular Pathology Unit, Department of Cardiac, Thoracic and Vascular Sciences University of Padua Medical School, Padova, Italy
| | - Hans Rolf Jäger
- Neuroradiological Academic Unit, Department of Brain Repair and Rehabilitation, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
| | - Cheryl Reeves
- Departments of Clinical and Experimental Epilepsy and Neuropathology, UCL Institute of Neurology and the National Hospital for Neurology and Neurosurgery, Queen Square, London WCN1BG, UK
| | - Alyma Somani
- Departments of Clinical and Experimental Epilepsy and Neuropathology, UCL Institute of Neurology and the National Hospital for Neurology and Neurosurgery, Queen Square, London WCN1BG, UK
| | - Shu An
- Departments of Clinical and Experimental Epilepsy and Neuropathology, UCL Institute of Neurology and the National Hospital for Neurology and Neurosurgery, Queen Square, London WCN1BG, UK
| | - Derek Marsdon
- Departments of Clinical and Experimental Epilepsy and Neuropathology, UCL Institute of Neurology and the National Hospital for Neurology and Neurosurgery, Queen Square, London WCN1BG, UK
| | - Andrew McEvoy
- Victor Horsley Department of Neurosurgery, National Hospital for Neurology and Neurosurgery, Queen Square, London WC1N 3BG, UK
| | - Anna Miserocchi
- Victor Horsley Department of Neurosurgery, National Hospital for Neurology and Neurosurgery, Queen Square, London WC1N 3BG, UK
| | - Lewis Thorne
- Victor Horsley Department of Neurosurgery, National Hospital for Neurology and Neurosurgery, Queen Square, London WC1N 3BG, UK
| | - Fay Newman
- Neurosurgery Department, Brighton and Sussex University Hospitals, Brighton, UK
| | - Sorin Bucur
- Neurosurgery Department, Brighton and Sussex University Hospitals, Brighton, UK
| | - Mrinalini Honavar
- Department of Anatomic Pathology, Hospital Pedro Hispano, Matosinhos, Portugal
| | - Tom Jacques
- Neuropathology Department, Great Ormond Street Hospital, London, UK
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Academic Medical Center, Amsterdam, The Netherlands.,Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, The Netherlands
| |
Collapse
|
33
|
Kyyriäinen J, Ekolle Ndode-Ekane X, Pitkänen A. Dynamics of PDGFRβ expression in different cell types after brain injury. Glia 2016; 65:322-341. [PMID: 27778377 DOI: 10.1002/glia.23094] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 10/11/2016] [Accepted: 10/13/2016] [Indexed: 01/26/2023]
Abstract
Platelet-derived growth factor receptor β (PDGFRβ) is upregulated after brain injury and its depletion results in the blood-brain barrier (BBB) damage. We investigated the time-window and localization of PDGFRβ expression in mice with intrahippocampal kainic acid-induced status epilepticus (SE) and in rats with lateral fluid-percussion-induced traumatic brain injury (TBI). Tissue immunohistochemistry was evaluated at several time-points after SE and TBI. The distribution of PDGFRβ was analyzed, and its cell type-specific expression was verified with double/triple-labeling of astrocytes (GFAP), NG2 cells, and endothelial cells (RECA-1). In normal mouse hippocampus, we found evenly distributed PDGFRβ+ parenchymal cells. In double-labeling, all NG2+ and 40%-60% GFAP+ cells were PDGFRβ+. After SE, PDGFRβ+ cells clustered in the ipsilateral hilus (178% of that in controls at fourth day, 225% at seventh day, P < 0.05) and in CA3 (201% at seventh day, P < 0.05), but the total number of PDGFRβ+ cells was not altered. As in controls, PDGFRβ-immunoreactivity was detected in parenchymal NG2+ and GFAP+ cells. We also observed PDGFRβ+ structural pericytes, detached reactive pericytes, and endothelial cells. After TBI, PDGFRβ+ cells clustered in the perilesional cortex and thalamus, particularly during the first post-injury week. PDGFRβ immunopositivity was observed in NG2+ and GFAP+ cells, structural pericytes, detached reactive pericytes, and endothelial cells. In some animals, PDGFRβ vascular staining was observed around the cortical glial scar for up to 3 months. Our data revealed an acute accumulation of PDGFRβ+ BBB-related cells in degenerating brain areas, which can be long lasting, suggesting an active role for PDGFRβ-signaling in blood vessel and post-injury tissue recovery. GLIA 2017;65:322-341.
Collapse
Affiliation(s)
- Jenni Kyyriäinen
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, FI-70211, Finland
| | - Xavier Ekolle Ndode-Ekane
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, FI-70211, Finland
| | - Asla Pitkänen
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, FI-70211, Finland
| |
Collapse
|
34
|
Sagare AP, Sweeney MD, Makshanoff J, Zlokovic BV. Shedding of soluble platelet-derived growth factor receptor-β from human brain pericytes. Neurosci Lett 2015; 607:97-101. [PMID: 26407747 DOI: 10.1016/j.neulet.2015.09.025] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 09/19/2015] [Accepted: 09/21/2015] [Indexed: 10/23/2022]
Abstract
Platelet-derived growth factor receptor-β (PDGFRβ) is expressed in the brain by vascular mural cells-brain capillary pericytes and arterial vascular smooth muscle cells (VSMCs). Recent evidence shows that blood-brain barrier (BBB) disruption and increased permeability, especially in the hippocampus, positively correlates with elevated levels of soluble PDGFRβ (sPDGFRβ) in cerebrospinal fluid (CSF) in patients with mild dementia. To determine which vascular cell type(s) contributes to increased sPDGFRβ in CSF, we compared PDGFRβ expression and sPDGFRβ shedding in response to injury in early passage primary cultures of human brain pericytes, brain arterial VSMCs, and brain endothelial cells. PDGFRβ protein was undetectable in endothelial cells, but was found both in pericytes and VSMCs. PDGFRβ relative protein abundance was by 4.2-fold (p<0.05) higher in pericytes compared to VSMCs. Hypoxia (1% O2) or amyloid-β peptide (25 μM) compared to normoxia (21% O2) both increased over 48 h shedding of sPDGFRβ and its levels in the culture medium from pericytes cultures, but not from VSMCs cultures, by 4.3-fold and 4.6-fold, respectively, compared to the basal sPDGFRβ levels in the medium (1.43±0.15 ng/ml). This was associated with the corresponding loss of cell-associated PDGFRβ from pericytes and no change in cellular levels of PDGFRβ in VSMCs. Thus, sPDGFRβ is a biomarker of pericyte injury, and elevated sPDGFRβ levels in biofluids in patients with dementia and/or other neurodegenerative disorders likely reflects pericyte injury, which supports the potential for sPDGFRβ to be developed and validated as a biomarker of brain pericyte injury and BBB dysfunction.
Collapse
Affiliation(s)
- Abhay P Sagare
- Department of Physiology and Biophysics, Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Melanie D Sweeney
- Department of Physiology and Biophysics, Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jacob Makshanoff
- Department of Physiology and Biophysics, Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Berislav V Zlokovic
- Department of Physiology and Biophysics, Center for Neurodegeneration and Regeneration, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|