1
|
Kang YJ, Clement EM, Park IH, Greenfield LJ, Smith BN, Lee SH. Vulnerability of cholecystokinin-expressing GABAergic interneurons in the unilateral intrahippocampal kainate mouse model of temporal lobe epilepsy. Exp Neurol 2021; 342:113724. [PMID: 33915166 DOI: 10.1016/j.expneurol.2021.113724] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/26/2021] [Accepted: 04/22/2021] [Indexed: 10/21/2022]
Abstract
Temporal lobe epilepsy (TLE) is characterized by recurrent spontaneous seizures and behavioral comorbidities. Reduced hippocampal theta oscillations and hyperexcitability that contribute to cognitive deficits and spontaneous seizures are present beyond the sclerotic hippocampus in TLE. However, the mechanisms underlying compromised network oscillations and hyperexcitability observed in circuits remote from the sclerotic hippocampus are largely unknown. Cholecystokinin (CCK)-expressing basket cells (CCKBCs) critically participate in hippocampal theta rhythmogenesis, and regulate neuronal excitability. Thus, we examined whether CCKBCs were vulnerable in nonsclerotic regions of the ventral hippocampus remote from dorsal sclerotic hippocampus using the intrahippocampal kainate (IHK) mouse model of TLE, targeting unilateral dorsal hippocampus. We found a decrease in the number of CCK+ interneurons in ipsilateral ventral CA1 regions from epileptic mice compared to those from sham controls. We also found that the number of boutons from CCK+ interneurons was reduced in the stratum pyramidale, but not in other CA1 layers, of ipsilateral hippocampus in epileptic mice, suggesting that CCKBCs are vulnerable. Electrical recordings showed that synaptic connectivity and strength from surviving CCKBCs to CA1 pyramidal cells (PCs) were similar between epileptic mice and sham controls. In agreement with reduced CCKBC number in TLE, electrical recordings revealed a significant reduction in amplitude and frequency of IPSCs in CA1 PCs evoked by carbachol (commonly used to excite CCK+ interneurons) in ventral CA1 regions from epileptic mice versus sham controls. These findings suggest that loss of CCKBCs beyond the hippocampal lesion may contribute to hyperexcitability and compromised network oscillations in TLE.
Collapse
Affiliation(s)
- Young-Jin Kang
- Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA; Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Ethan M Clement
- Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - In-Hyun Park
- Department of Genetics, Yale Stem Cell Center, Yale Child Study Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Lazar John Greenfield
- Department of Neurology, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Bret N Smith
- Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA
| | - Sang-Hun Lee
- Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA; Department of Neurology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| |
Collapse
|
2
|
Sharma G, Shin EJ, Sharma N, Nah SY, Mai HN, Nguyen BT, Jeong JH, Lei XG, Kim HC. Glutathione peroxidase-1 and neuromodulation: Novel potentials of an old enzyme. Food Chem Toxicol 2021; 148:111945. [PMID: 33359022 DOI: 10.1016/j.fct.2020.111945] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 12/15/2020] [Accepted: 12/18/2020] [Indexed: 12/14/2022]
Abstract
Glutathione peroxidase (GPx) acts in co-ordination with other signaling molecules to exert its own antioxidant role. We have demonstrated the protective effects of GPx,/GPx-1, a selenium-dependent enzyme, on various neurodegenerative disorders (i.e., Parkinson's disease, Alzheimer's disease, cerebral ischemia, and convulsive disorders). In addition, we summarized the recent findings indicating that GPx-1 might play a role as a neuromodulator in neuropsychiatric conditions, such as, stress, bipolar disorder, schizophrenia, and drug intoxication. In this review, we attempted to highlight the mechanistic scenarios mediated by the GPx/GPx-1 gene in impacting these neurodegenerative and neuropsychiatric disorders, and hope to provide new insights on the therapeutic interventions against these disorders.
Collapse
Affiliation(s)
- Garima Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Naveen Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, Seoul, 05029, Republic of Korea
| | - Huynh Nhu Mai
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea; Pharmacy Faculty, Can Tho University of Medicine and Pharmacy, Can Tho City, 900000, Viet Nam
| | - Bao Trong Nguyen
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea
| | - Ji Hoon Jeong
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Xin Gen Lei
- Department of Animal Science, Cornell University, Ithaca, NY, 14853, USA
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 24341, Republic of Korea.
| |
Collapse
|
3
|
Kagitani-Shimono K, Kato H, Kuwayama R, Tominaga K, Nabatame S, Kishima H, Hatazawa J, Taniike M. Clinical evaluation of neuroinflammation in child-onset focal epilepsy: a translocator protein PET study. J Neuroinflammation 2021; 18:8. [PMID: 33407581 PMCID: PMC7789379 DOI: 10.1186/s12974-020-02055-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 12/09/2020] [Indexed: 01/17/2023] Open
Abstract
Background Neuroinflammation is associated with various chronic neurological diseases, including epilepsy; however, neuroimaging approaches for visualizing neuroinflammation have not been used in the clinical routine yet. In this study, we used the translocator protein positron emission tomography (PET) with [11C] DPA713 to investigate neuroinflammation in the epileptogenic zone in patients with child-onset focal epilepsy. Methods Patients with intractable focal epilepsy were recruited at the Epilepsy Center of Osaka University; those who were taking any immunosuppressants or steroids were excluded. PET images were acquired for 60 min after intravenous administration of [11C] DPA713. The PET image of [11C] DPA713 was co-registered to individual’s magnetic resonance imaging (MRI), and the standardized uptake value ratio (SUVr) in regions of interest, which were created in non-lesions and lesions, was calculated using the cerebellum as a pseudo-reference region. In the case of epilepsy surgery, the correlation between SUVr in lesions and pathological findings was analyzed. Results Twenty-seven patients (mean age: 11.3 ± 6.2 years, male/female: 17/10) were included in this study. Of these, 85.1% showed increased uptake of [11C] DPA713 in the focal epileptic lesion. Three patients showed epileptic spasms, suggesting partial seizure onset, and all 18 patients with abnormal lesions on MRI were similarly highlighted by significant uptake of [11C] DPA713. DPA713-positive patients had a broad range of etiologies, including focal cortical dysplasia, tumors, infarction, and hippocampal sclerosis. Five out of nine MRI-negative patients showed abnormal [11C] DPA713 uptake. The SUVr of [11C] DPA713 in lesions was significantly higher than that in non-lesions. In seven patients who underwent epilepsy surgery, increased [11C] DPA713 uptake was associated with microglial activation. Conclusions This study indicates that [11C] DPA713 uptake has valuable sensitivity in the identification of epileptic foci in child-onset focal epilepsy, and inflammation is implicated in the pathophysiology in the epileptic foci caused by various etiologies. Further research is required to establish diagnostic tools for identifying focal epileptogenic zones. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-020-02055-1.
Collapse
Affiliation(s)
- Kuriko Kagitani-Shimono
- Department of Child Development, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan. .,Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Japan. .,Epilepsy Center, Osaka University Hospital, Suita, Japan.
| | - Hiroki Kato
- Department of Nuclear Medicine and Tracer Kinetics, Osaka University Graduate School of Medicine, Suita, Japan
| | - Ryoko Kuwayama
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Japan.,Epilepsy Center, Osaka University Hospital, Suita, Japan
| | - Koji Tominaga
- Department of Child Development, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan.,Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Japan.,Epilepsy Center, Osaka University Hospital, Suita, Japan
| | - Shin Nabatame
- Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Japan.,Epilepsy Center, Osaka University Hospital, Suita, Japan
| | - Haruhiko Kishima
- Epilepsy Center, Osaka University Hospital, Suita, Japan.,Department of Neurosurgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Jun Hatazawa
- Department of Nuclear Medicine and Tracer Kinetics, Osaka University Graduate School of Medicine, Suita, Japan.,Department of Quantum Cancer Therapy Research Center for Nuclear Physics, Osaka University, Suita, Japan
| | - Masako Taniike
- Department of Child Development, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan.,Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
4
|
Dong W, Ma Y, Guan F, Zhang X, Chen W, Zhang L, Zhang L. Ablation of C9orf72 together with excitotoxicity induces ALS in rats. FEBS J 2020; 288:1712-1723. [PMID: 32745320 DOI: 10.1111/febs.15501] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 05/19/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022]
Abstract
Pathogenesis of familial amyotrophic lateral sclerosis (ALS) linked to expansion of the chromosome 9 open reading frame 72 (C9orf72) hexanucleotide repeat that impairs C9orf72 expression. Loss of function of the C9orf72 protein is one of the three main proposed C9orf72-related ALS mechanisms. However, C9orf72 loss of function, by itself, is insufficient to cause severe phenotypes in mice. Excitotoxicity is another major disease mechanism of ALS. We speculate that loss of C9orf72 protein might cause ALS in combination with excitotoxicity. To date, the effect of C9orf72 deficiency in the background of SD rat has not been examined. To test our hypothesis, we generated a line of rat with a deletion of part of the C9orf72 gene ablating the encoded protein. These animals did not develop any ALS phenotypes; however, when they were treated with kainic acid, an excitotoxicity inducer, the rats developed motor deficits and showed loss of motor neurons (MNs), Golgi complex fragmentation, and abnormal vesicle trafficking. RNA sequencing revealed profound changes in the gene profiles that were primarily associated with neural activity. Our results demonstrated that C9orf72 ablation alone was not enough to cause ALS pathogenesis in rat; but the ablation sensitized MNs to other risk factors that synergistically caused the ALS. These results support a loss of function of C9orf72 mechanism of ALS.
Collapse
Affiliation(s)
- Wei Dong
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Peking Union Medicine College, Chinese Academy of Medical Sciences, Beijing, China.,Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuanwu Ma
- Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Diseases, Institute of Laboratory Animal Science, Peking Union Medicine College, Chinese Academy of Medical Sciences, Beijing, China
| | - Feifei Guan
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Peking Union Medicine College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xu Zhang
- Beijing Engineering Research Center for Experimental Animal Models of Human Diseases, Institute of Laboratory Animal Science, Peking Union Medicine College, Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Chen
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Peking Union Medicine College, Chinese Academy of Medical Sciences, Beijing, China
| | - Li Zhang
- Beijing Engineering Research Center for Experimental Animal Models of Human Diseases, Institute of Laboratory Animal Science, Peking Union Medicine College, Chinese Academy of Medical Sciences, Beijing, China
| | - Lianfeng Zhang
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Peking Union Medicine College, Chinese Academy of Medical Sciences, Beijing, China.,Neuroscience Center, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
5
|
Chali F, Milior G, Marty S, Morin-Brureau M, Le Duigou C, Savary E, Blugeon C, Jourdren L, Miles R. Lipid markers and related transcripts during excitotoxic neurodegeneration in kainate-treated mice. Eur J Neurosci 2019; 50:1759-1778. [PMID: 30767299 DOI: 10.1111/ejn.14375] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/30/2019] [Accepted: 01/31/2019] [Indexed: 12/16/2022]
Abstract
Lipid homeostasis is dysregulated in some neurodegenerative diseases and after brain injuries due to excess glutamate or lack of oxygen. However the kinetics and cell specificity of dysregulation in different groups of lipids during excitotoxic neuronal death are not clear. Here we examined the changes during excitotoxic neuronal death induced by injecting kainic acid (KA) into the CA1 region of mouse hippocampus. We compared neuronal loss and glial cell proliferation with changes in lipid-related transcripts and markers for different lipid groups, over 12 days after KA-treatment. As neurons showed initial signs of damage, transcripts and proteins linked to fatty acid oxidation were up-regulated. Cholesterol biosynthesis induced by transcripts controlled by the transcription factor Srebp2 seems to be responsible for a transient increase in neuronal free cholesterol at 1 to 2 days. In microglia, but not in neurons, Perilipin-2 associated lipid droplets were induced and properties of Nile red emissions suggest lipid contents change over time. After microglial expression of phagocytotic markers at 2 days, some neutral lipid deposits co-localized with lysosome markers of microglia and were detected within putative phagocytotic cups. These data delineate distinct lipid signals in neurons and glial cells during excitotoxic processes from initial neuronal damage to engagement of the lysosome-phagosome system.
Collapse
Affiliation(s)
- Farah Chali
- Inserm U1127, CNRS UMR7225, Sorbonne Université, UPMC Université Paris 6 UMR S1127, Institut du Cerveau et de la Moelle Epinière, CHU Pitié-Salpêtrière, Paris, France
| | - Giampaolo Milior
- Inserm U1127, CNRS UMR7225, Sorbonne Université, UPMC Université Paris 6 UMR S1127, Institut du Cerveau et de la Moelle Epinière, CHU Pitié-Salpêtrière, Paris, France
| | - Serge Marty
- Inserm U1127, CNRS UMR7225, Sorbonne Université, UPMC Université Paris 6 UMR S1127, Institut du Cerveau et de la Moelle Epinière, CHU Pitié-Salpêtrière, Paris, France
| | - Mélanie Morin-Brureau
- Inserm U1127, CNRS UMR7225, Sorbonne Université, UPMC Université Paris 6 UMR S1127, Institut du Cerveau et de la Moelle Epinière, CHU Pitié-Salpêtrière, Paris, France
| | - Caroline Le Duigou
- Inserm U1127, CNRS UMR7225, Sorbonne Université, UPMC Université Paris 6 UMR S1127, Institut du Cerveau et de la Moelle Epinière, CHU Pitié-Salpêtrière, Paris, France
| | - Etienne Savary
- Inserm U1127, CNRS UMR7225, Sorbonne Université, UPMC Université Paris 6 UMR S1127, Institut du Cerveau et de la Moelle Epinière, CHU Pitié-Salpêtrière, Paris, France
| | - Corinne Blugeon
- Institut de Biologie de l'École normale supérieure (IBENS), École Normale Supérieure, CNRS, INSERM PSL Université Paris, Paris, France
| | - Laurent Jourdren
- Institut de Biologie de l'École normale supérieure (IBENS), École Normale Supérieure, CNRS, INSERM PSL Université Paris, Paris, France
| | - Richard Miles
- Inserm U1127, CNRS UMR7225, Sorbonne Université, UPMC Université Paris 6 UMR S1127, Institut du Cerveau et de la Moelle Epinière, CHU Pitié-Salpêtrière, Paris, France
| |
Collapse
|
6
|
Targeting the Mouse Ventral Hippocampus in the Intrahippocampal Kainic Acid Model of Temporal Lobe Epilepsy. eNeuro 2018; 5:eN-NWR-0158-18. [PMID: 30131968 PMCID: PMC6102375 DOI: 10.1523/eneuro.0158-18.2018] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 06/08/2018] [Accepted: 06/29/2018] [Indexed: 11/21/2022] Open
Abstract
Here we describe a novel mouse model of temporal lobe epilepsy (TLE) that moves the site of kainate injection from the rodent dorsal hippocampus (corresponding to the human posterior hippocampus) to the ventral hippocampus (corresponding to the human anterior hippocampus). We compare the phenotypes of this new model—with respect to seizures, cognitive impairment, affective deficits, and histopathology—to the standard dorsal intrahippocampal kainate model. Our results demonstrate that histopathological measures of granule cell dispersion and mossy fiber sprouting maximize near the site of kainate injection. Somewhat surprisingly, both the dorsal and ventral models exhibit similar spatial memory impairments in addition to similar electrographic and behavioral seizure burdens. In contrast, we find a more pronounced affective (anhedonic) phenotype specifically in the ventral model. These results demonstrate that the ventral intrahippocampal kainic acid model recapitulates critical pathologies of the dorsal model while providing a means to further study affective phenotypes such as depression in TLE.
Collapse
|
7
|
Sharma S, Carlson S, Puttachary S, Sarkar S, Showman L, Putra M, Kanthasamy AG, Thippeswamy T. Role of the Fyn-PKCδ signaling in SE-induced neuroinflammation and epileptogenesis in experimental models of temporal lobe epilepsy. Neurobiol Dis 2018; 110:102-121. [PMID: 29197620 PMCID: PMC5753797 DOI: 10.1016/j.nbd.2017.11.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 11/08/2017] [Accepted: 11/27/2017] [Indexed: 02/07/2023] Open
Abstract
Status epilepticus (SE) induces neuroinflammation and epileptogenesis, but the mechanisms are not yet fully delineated. The Fyn, a non-receptor Src family tyrosine kinase (SFK), and its immediate downstream target, PKCδ are emerging as potential mediators of neuroinflammation. In order to first determine the role of Fyn kinase signaling in SE, we tested the efficacy of a SFK inhibitor, saracatinib (25mg/kg, oral) in C57BL/6J mouse kainate model of acute seizures. Saracatinib pretreatment dampened SE severity and completely prevented mortality. We further utilized fyn-/- and fyn+/+ mice (wildtype control for the fyn-/- mice on same genetic background), and the rat kainate model, treated with saracatinib post-SE, to validate the role of Fyn/SFK in SE and epileptogenesis. We observed significant reduction in SE severity, epileptiform spikes, and electrographic non-convulsive seizures in fyn-/- mice when compared to fyn+/+ mice. Interestingly, significant reductions in phosphorylated pSrc-416 and PKCδ (pPKCδ-507) and naive PKCδ were observed in fyn-/- mice as compared to fyn+/+ mice suggesting that PKCδ signaling is a downstream mediator of Fyn in SE and epileptogenesis. Notably, fyn-/- mice also showed a reduction in key proinflammatory mediators TNF-α, IL-1β, and iNOS mRNA expression; serum IL-6 and IL-12 levels; and nitro-oxidative stress markers such as 4-HNE, gp91phox, and 3-NT in the hippocampus. Immunohistochemistry revealed a significant increase in reactive microgliosis and neurodegeneration in the hippocampus and hilus of dentate gyrus in fyn+/+ mice in contrast to fyn-/- mice. Interestingly, we did not observe upregulation of Fyn in pyramidal neurons of the hippocampus during post-SE in fyn+/+ mice, but it was upregulated in hilar neurons of the dentate gyrus when compared to naïve control. In reactive microglia, both Fyn and PKCδ were persistently upregulated during post-SE suggesting that Fyn-PKCδ may drive neuroinflammation during epileptogenesis. Since disabling the Fyn kinase prior to SE, either by treating with saracatinib or fyn gene knockout, suppressed seizures and the subsequent epileptogenic events, we further tested whether Fyn/SFK inhibition during post-SE modifies epileptogenesis. Telemetry-implanted, SE-induced, rats were treated with saracatinib and continuously monitored for a month. At 2h post-diazepam, the saracatinib (25mg/kg) or the vehicle was administered orally and repeated twice daily for first three days followed by a single dose/day for the next four days. The saracatinib post-treatment prevented epileptogenesis in >50% of the rats and significantly reduced spontaneous seizures and epileptiform spikes in the rest (one animal did not respond) when compared to the vehicle treated group, which had >24 seizures in a month. Collectively, the findings suggest that Fyn/SFK is a potential mediator of epileptogenesis and a therapeutic target to prevent/treat seizures and epileptogenesis.
Collapse
Affiliation(s)
- Shaunik Sharma
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames 50011, USA
| | - Steven Carlson
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames 50011, USA
| | - Sreekanth Puttachary
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames 50011, USA
| | - Souvarish Sarkar
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames 50011, USA
| | - Lucas Showman
- W.M. Keck Metabolomics Research Laboratory, Iowa State University, Ames 50011, USA
| | - Marson Putra
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames 50011, USA
| | - Anumantha G Kanthasamy
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames 50011, USA
| | - Thimmasettappa Thippeswamy
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames 50011, USA.
| |
Collapse
|
8
|
Fahrenhold M, Rakic S, Classey J, Brayne C, Ince PG, Nicoll JAR, Boche D. TREM2 expression in the human brain: a marker of monocyte recruitment? Brain Pathol 2017; 28:595-602. [PMID: 28987033 PMCID: PMC6221091 DOI: 10.1111/bpa.12564] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 10/02/2017] [Indexed: 01/04/2023] Open
Abstract
Mutation in the triggering receptor expressed on myeloid cells (TREM) 2 gene has been identified as a risk factor for several neurodegenerative diseases including Alzheimer's disease (AD). Experimental studies using animal models of AD have highlighted a number of functions associated with TREM2 and its expression by microglial cells. It has therefore been assumed that this is also the case in humans. However, there is very limited information concerning the cellular expression of TREM2 in the human brain. As part of investigations of microglia using post‐mortem resources provided by the Medical Research Council Cognitive Function and Ageing Studies (MRC‐CFAS), we immunostained the cerebral cortex of 299 participants for TREM2 using the Sigma antibody HPA010917 and compared with the macrophage/microglial markers Iba1 and CD68. As expected, Iba1 and CD68 labeled microglia and perivascular macrophages. However, in most cases (284/299), the TREM2 antibody labelled monocytes within vascular lumens, but not microglia or perivascular macrophages. In contrast, in 5 out of 6 cases with acute infarcts, TREM2 immunoreaction identified cells within the brain parenchyma interpreted as recruited monocytes. Six cases with old infarcts contained phagocytic foamy macrophages which were CD68‐positive but TREM2 negative. Our observations, using the HPA010917 anti‐TREM2 antibody, suggest that TREM2 is not expressed by microglia but instead seems to be a marker of recruited monocytes in the human brain. This finding has implications with regards to the role of TREM2 as a risk factor, emphasizing the importance of systemic immune responses in the development and progression of Alzheimer's disease.
Collapse
Affiliation(s)
- Marie Fahrenhold
- Clinical Neurosciences, Clinical and Experimental Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - Sonja Rakic
- Clinical Neurosciences, Clinical and Experimental Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - John Classey
- Clinical Neurosciences, Clinical and Experimental Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - Carol Brayne
- Cambridge Institute of Public Health, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Paul G Ince
- Sheffield Institute for Translational Neuroscience, Sheffield University, Sheffield, S10 2HQ, UK
| | - James A R Nicoll
- Clinical Neurosciences, Clinical and Experimental Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK.,Department of Cellular Pathology, University Hospital Southampton NHS Foundation Trust, Southampton, Southampton SO16 6YD, UK
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | | |
Collapse
|
9
|
Schidlitzki A, Twele F, Klee R, Waltl I, Römermann K, Bröer S, Meller S, Gerhauser I, Rankovic V, Li D, Brandt C, Bankstahl M, Töllner K, Löscher W. A combination of NMDA and AMPA receptor antagonists retards granule cell dispersion and epileptogenesis in a model of acquired epilepsy. Sci Rep 2017; 7:12191. [PMID: 28939854 PMCID: PMC5610327 DOI: 10.1038/s41598-017-12368-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 09/08/2017] [Indexed: 01/01/2023] Open
Abstract
Epilepsy may arise following acute brain insults, but no treatments exist that prevent epilepsy in patients at risk. Here we examined whether a combination of two glutamate receptor antagonists, NBQX and ifenprodil, acting at different receptor subtypes, exerts antiepileptogenic effects in the intrahippocampal kainate mouse model of epilepsy. These drugs were administered over 5 days following kainate. Spontaneous seizures were recorded by video/EEG at different intervals up to 3 months. Initial trials showed that drug treatment during the latent period led to higher mortality than treatment after onset of epilepsy, and further, that combined therapy with both drugs caused higher mortality at doses that appear safe when used singly. We therefore refined the combined-drug protocol, using lower doses. Two weeks after kainate, significantly less mice of the NBQX/ifenprodil group exhibited electroclinical seizures compared to vehicle controls, but this effect was lost at subsequent weeks. The disease modifying effect of the treatment was associated with a transient prevention of granule cell dispersion and less neuronal degeneration in the dentate hilus. These data substantiate the involvement of altered glutamatergic transmission in the early phase of epileptogenesis. Longer treatment with NBQX and ifenprodil may shed further light on the apparent temporal relationship between dentate gyrus reorganization and development of spontaneous seizures.
Collapse
Affiliation(s)
- Alina Schidlitzki
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
- Center for Systems Neuroscience, 30559, Hannover, Germany
| | - Friederike Twele
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
| | - Rebecca Klee
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
| | - Inken Waltl
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
- Center for Systems Neuroscience, 30559, Hannover, Germany
| | - Kerstin Römermann
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
| | - Sonja Bröer
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
| | - Sebastian Meller
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
- Center for Systems Neuroscience, 30559, Hannover, Germany
| | - Ingo Gerhauser
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Vladan Rankovic
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
- Institute for Auditory Neuroscience at University Medical Center Göttingen & German Primate Center, Göttingen, Germany
| | - Dandan Li
- Center for Systems Neuroscience, 30559, Hannover, Germany
- Department of Pathology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Claudia Brandt
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
| | - Marion Bankstahl
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
- Center for Systems Neuroscience, 30559, Hannover, Germany
| | - Kathrin Töllner
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559, Hannover, Germany.
- Center for Systems Neuroscience, 30559, Hannover, Germany.
| |
Collapse
|
10
|
Bar-Klein G, Klee R, Brandt C, Bankstahl M, Bascuñana P, Töllner K, Dalipaj H, Bankstahl JP, Friedman A, Löscher W. Isoflurane prevents acquired epilepsy in rat models of temporal lobe epilepsy. Ann Neurol 2017; 80:896-908. [PMID: 27761920 DOI: 10.1002/ana.24804] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 09/16/2016] [Accepted: 10/10/2016] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Acquired epilepsy is a devastating long-term risk of various brain insults, including trauma, stroke, infections, and status epilepticus (SE). There is no preventive treatment for patients at risk. Attributable to the complex alterations involved in epileptogenesis, it is likely that multitargeted approaches are required for epilepsy prevention. We report novel preclinical findings with isoflurane, which exerts various nonanesthetic effects that may be relevant for antiepileptogenesis. METHODS The effects of isoflurane were investigated in two rat models of SE-induced epilepsy: intrahippocampal kainate and systemic administration of paraoxon. Isoflurane was either administered during (kainate) or after (paraoxon) induction of SE. Magnetic resonance imaging was used to assess blood-brain barrier (BBB) dysfunction. Positron emission tomography was used to visualize neuroinflammation. Long-term electrocorticographic recordings were used to monitor spontaneous recurrent seizures. Neuronal damage was assessed histologically. RESULTS In the absence of isoflurane, spontaneous recurrent seizures were common in the majority of rats in both models. When isoflurane was administered during kainate injection, duration and severity of SE were not affected, but only few rats developed spontaneous recurrent seizures. A similar antiepileptogenic effect was found when paraoxon-treated rats were exposed to isoflurane after SE. Moreover, in the latter model, isoflurane prevented BBB dysfunction and neurodegeneration, whereas isoflurane reduced neuroinflammation in the kainate model. INTERPRETATION Given that isoflurane is a widely used volatile anesthetic, and is used for inhalational long-term sedation in critically ill patients at risk to develop epilepsy, our findings hold a promising potential to be successfully translated into the clinic. Ann Neurol 2016;80:896-908.
Collapse
Affiliation(s)
- Guy Bar-Klein
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, the Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Rebecca Klee
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Claudia Brandt
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Marion Bankstahl
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| | - Pablo Bascuñana
- Department of Nuclear Medicine, Hannover Medical School, Germany
| | - Kathrin Töllner
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Hotjensa Dalipaj
- Department of Medical Neuroscience, Dalhousie University, Halifax, Canada
| | - Jens P Bankstahl
- Department of Nuclear Medicine, Hannover Medical School, Germany
| | - Alon Friedman
- Departments of Physiology and Cell Biology, Cognitive and Brain Sciences, the Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Department of Medical Neuroscience, Dalhousie University, Halifax, Canada
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Hannover, Germany.,Center for Systems Neuroscience, Hannover, Germany
| |
Collapse
|
11
|
Klee R, Brandt C, Töllner K, Löscher W. Various modifications of the intrahippocampal kainate model of mesial temporal lobe epilepsy in rats fail to resolve the marked rat-to-mouse differences in type and frequency of spontaneous seizures in this model. Epilepsy Behav 2017; 68:129-140. [PMID: 28167446 DOI: 10.1016/j.yebeh.2016.11.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 11/18/2016] [Accepted: 11/21/2016] [Indexed: 11/19/2022]
Abstract
Temporal lobe epilepsy (TLE) is the most common type of acquired epilepsy in adults. TLE can develop after diverse brain insults, including traumatic brain injury, infections, stroke, or prolonged status epilepticus (SE). Post-SE rodent models of TLE are widely used to understand mechanisms of epileptogenesis and develop treatments for epilepsy prevention. In this respect, the intrahippocampal kainate model of TLE in mice is of interest, because highly frequent spontaneous electrographic seizures develop in the kainate focus, allowing evaluation of both anti-seizure and anti-epileptogenic effects of novel drugs with only short EEG recording periods, which is not possible in any other model of TLE, including the intrahippocampal kainate model in rats. In the present study, we investigated whether the marked mouse-to-rat difference in occurrence and frequency of spontaneous seizures is due to a species difference or to technical variables, such as anesthesia during kainate injection, kainate dose, or location of kainate injection and EEG electrode in the hippocampus. When, as in the mouse model, anesthesia was used during kainate injection, only few rats developed epilepsy, although severity or duration of SE was not affected by isoflurane. In contrast, most rats developed epilepsy when kainate was injected without anesthesia. However, frequent electrographic seizures as observed in mice did not occur in rats, irrespective of location of kainate injection (CA1, CA3) or EEG recording electrode (CA1, CA3, dentate gyrus) or dose of kainate injected. These data indicate marked phenotypic differences between mice and rats in this model. Further studies should explore the mechanisms underlying this species difference.
Collapse
Affiliation(s)
- Rebecca Klee
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany, and Center for Systems Neuroscience, Hannover, Germany
| | - Claudia Brandt
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany, and Center for Systems Neuroscience, Hannover, Germany
| | - Kathrin Töllner
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany, and Center for Systems Neuroscience, Hannover, Germany
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany, and Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
12
|
Leaw B, Zhu D, Tan J, Muljadi R, Saad MI, Mockler JC, Wallace EM, Lim R, Tolcos M. Human amnion epithelial cells rescue cell death via immunomodulation of microglia in a mouse model of perinatal brain injury. Stem Cell Res Ther 2017; 8:46. [PMID: 28241859 PMCID: PMC5330154 DOI: 10.1186/s13287-017-0496-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 01/19/2017] [Accepted: 02/09/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Human amnion epithelial cells (hAECs) are clonogenic and have been proposed to reduce inflammatory-induced tissue injury. Perturbation of the immune response is implicated in the pathogenesis of perinatal brain injury; modulating this response could thus be a novel therapy for treating or preventing such injury. The immunomodulatory properties of hAECs have been shown in other animal models, but a detailed investigation of the effects on brain immune cells following injury has not been undertaken. Here, we investigate the effects of hAECs on microglia, the first immune responders to injury within the brain. METHODS We generated a mouse model combining neonatal inflammation and perinatal hyperoxia, both of which are risk factors associated with perinatal brain injury. On embryonic day 16 we administered lipopolysaccharide (LPS), or saline (control), intra-amniotically to C57Bl/6 J mouse pups. On postnatal day (P)0, LPS pups were placed in hyperoxia (65% oxygen) and control pups in normoxia for 14 days. Pups were given either hAECs or saline intravenously on P4. RESULTS At P14, relative to controls, LPS and hyperoxia pups had reduced body weight, increased density of apoptotic cells (TUNEL) in the cortex, striatum and white matter, astrocytes (GFAP) in the white matter and activated microglia (CD68) in the cortex and striatum, but no change in total microglia density (Iba1). hAEC administration rescued the decreased body weight and reduced apoptosis and astrocyte areal coverage in the white matter, but increased the density of total and activated microglia. We then stimulated primary microglia (CD45lowCD11b+) with LPS for 24 h, followed by co-culture with hAEC conditioned medium for 48 h. hAEC conditioned medium increased microglial phagocytic activity, decreased microglia apoptosis and decreased M1 activation markers (CD86). Stimulating hAECs for 24 h with LPS did not alter release of cytokines known to modulate microglia activity. CONCLUSIONS These data demonstrate that hAECs can directly immunomodulate brain microglia, probably via release of trophic factors. This observation offers promise that hAECs may afford therapeutic utility in the management of perinatal brain injury.
Collapse
Affiliation(s)
- Bryan Leaw
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168 Australia
| | - Dandan Zhu
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168 Australia
| | - Jean Tan
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168 Australia
| | - Ruth Muljadi
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168 Australia
| | - Mohamed I. Saad
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168 Australia
| | - Joanne C. Mockler
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168 Australia
| | - Euan M. Wallace
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168 Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168 Australia
| | - Rebecca Lim
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168 Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168 Australia
| | - Mary Tolcos
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright Street, Clayton, VIC 3168 Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC 3168 Australia
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083 Australia
| |
Collapse
|
13
|
Sawano T, Tsuchihashi R, Morii E, Watanabe F, Nakane K, Inagaki S. Homology analysis detects topological changes of Iba1 localization accompanied by microglial activation. Neuroscience 2017; 346:43-51. [PMID: 28077279 DOI: 10.1016/j.neuroscience.2016.12.052] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 12/21/2016] [Accepted: 12/29/2016] [Indexed: 01/01/2023]
Abstract
The state of microglial activation provides important information about the central nervous system. However, a reliable index of microglial activation in histological samples has yet to be established. Here, we show that microglial activation induces topological changes of Iba1 localization that can be detected by analysis based on homology theory. Analysis of homology was applied to images of Iba1-stained tissue sections, and the 0-dimentional Betti number (b0: the number of solid components) and the 1-dimentional Betti number (b1: the number of windows surrounded by solid components) were obtained. We defined b1/b0 as the Homology Value (HV), and investigated its validity as an index of microglial activation using cerebral ischemia model mice. Microglial activation was accompanied by changes to Iba1 localization and morphology of microglial processes. In single microglial cells, the change of Iba1 localization increased b1. Conversely, thickening or retraction of microglial processes decreased b0. Consequently, microglial activation increased the HV. The HV of a tissue area increased with proximity to the ischemic core and showed a high degree of concordance with the number of microglia expressing activation makers. Furthermore, the HV of human metastatic brain tumor tissue also increased with proximity to the tumor. These results suggest that our index, based on homology theory, can be used to correctly evaluate microglial activation in various tissue images.
Collapse
Affiliation(s)
- Toshinori Sawano
- Group of Neurobiology, Division of Health Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Ryo Tsuchihashi
- Group of Neurobiology, Division of Health Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Eiichi Morii
- Department of Pathology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Fumiya Watanabe
- Group of Neurobiology, Division of Health Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Kazuaki Nakane
- Division of Health Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan.
| | - Shinobu Inagaki
- Group of Neurobiology, Division of Health Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan.
| |
Collapse
|
14
|
Abstract
Lidocaine, the most commonly used local anesthetic, inhibits glutamate release from nerve terminals. Given the involvement of glutamate neurotoxicity in the pathogenesis of various neurological disorders, this study investigated the role of lidocaine in hippocampal neuronal death and inflammatory events induced by an i.p. injection of kainic acid (KA) (15 mg/kg), a glutamate analog. The results showed that KA significantly led to neuronal death in the CA3 pyramidal layers of the hippocampus and this effect was attenuated by the systemic administration of lidocaine (0.8 or 4 mg/kg, i.p.) 30 min before KA injection. Moreover, KA-induced microglia activation and gene expression of proinflammatory cytokines, namely, interleukin-1β, interleukin-6, and tumor necrosis factor-α, in the hippocampus were reduced by the lidocaine pretreatment. Altogether, the results suggest that lidocaine can effectively treat glutamate excitotoxicity-related brain disorders.
Collapse
|
15
|
Spranger M, Fontana A. REVIEW ■ : Activation of Microglia: A Dangerous Interlude in Immune Function in the Brain. Neuroscientist 2016. [DOI: 10.1177/107385849600200515] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Microglial cells are representatives of the immune system in the CNS parenchyma. Their most characteristic property is their ability to modify their behavior in response to diverse signals from other cells in a variety of experimental conditions and human diseases, both acute and chronic. The transformation from a quiescent state into phagocytic brain macrophages is under strict control and accompanied by the production of several secretory products. These include cytokines, excitatory amino acids, and reactive oxygen metabolites by which the activated microglial cells correspond with other cells of the brain and immune system. Thus, they represent an essential host defense and repair system, and may be responsible for tissue destruction and neuronal death, depending on the balance of activating and inhibitory signals. NEUROSCIENTIST 2:293-299, 1996
Collapse
|
16
|
Eyo UB, Murugan M, Wu LJ. Microglia-Neuron Communication in Epilepsy. Glia 2016; 65:5-18. [PMID: 27189853 DOI: 10.1002/glia.23006] [Citation(s) in RCA: 187] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 04/20/2016] [Accepted: 04/28/2016] [Indexed: 12/22/2022]
Abstract
Epilepsy has remained a significant social concern and financial burden globally. Current therapeutic strategies are based primarily on neurocentric mechanisms that have not proven successful in at least a third of patients, raising the need for novel alternative and complementary approaches. Recent evidence implicates glial cells and neuroinflammation in the pathogenesis of epilepsy with the promise of targeting these cells to complement existing strategies. Specifically, microglial involvement, as a major inflammatory cell in the epileptic brain, has been poorly studied. In this review, we highlight microglial reaction to experimental seizures, discuss microglial control of neuronal activities, and propose the functions of microglia during acute epileptic phenotypes, delayed neurodegeneration, and aberrant neurogenesis. Future research that would help fill in the current gaps in our knowledge includes epilepsy-induced alterations in basic microglial functions, neuro-microglial interactions during chronic epilepsy, and microglial contribution to developmental seizures. Studying the role of microglia in epilepsy could inform therapies to better alleviate the disease. GLIA 2016;65:5-18.
Collapse
Affiliation(s)
- Ukpong B Eyo
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey
| | - Madhuvika Murugan
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey
| | - Long-Jun Wu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey
| |
Collapse
|
17
|
Choi JH, Jang M, Kim EJ, Kim H, Ye SK, Cho IH. Oriental Medicine Woohwangchungsimwon Attenuates Kainic Acid-Induced Seizures and Neuronal Cell Death in the Hippocampus. Rejuvenation Res 2016; 19:394-405. [PMID: 26981959 DOI: 10.1089/rej.2015.1779] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Woohwangchungsimwon (WCW) is an oriental medicine that has been extensively prescribed in Asia to patients with apoplexy, high blood pressure, acute/chronic convulsion, and so on. However, the potential therapeutic value of WCW in treating the pathologic brain has not yet been fully investigated. In the present study, we evaluated whether WCW has beneficial effects on kainic acid (KA)-induced excitotoxicity. An intraperitoneal injection of KA (40 mg/kg) and an intracerebroventricular injection of KA (0.2 μg) produced typical seizure behavior and neuronal cell death in the CA1 and CA3 pyramidal layers of the hippocampus, respectively. However, the systemic administration of WCW significantly attenuated the seizure behavior and neuronal cell death. WCW was found to exert the best protective effect when it was administrated 2 hours before a KA injection. Moreover, this WCW-induced neuroprotection was accompanied by a reduction in microglia activation and tumor necrosis factor-alpha, interleukin (IL)-1β, IL-6, inducible nitric oxide synthase, and cyclooxyganase-2 in the hippocampus. These results suggest that WCW has therapeutic potential to suppress KA-induced pathogenesis in the brain by inhibiting inflammation.
Collapse
Affiliation(s)
- Jong Hee Choi
- 1 Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University , Seoul, Republic of Korea.,2 Brain Korea 21 Plus Program, College of Korean Medicine, Kyung Hee University , Seoul, Republic of Korea
| | - Minhee Jang
- 1 Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University , Seoul, Republic of Korea
| | - Eun-Jeong Kim
- 1 Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University , Seoul, Republic of Korea.,2 Brain Korea 21 Plus Program, College of Korean Medicine, Kyung Hee University , Seoul, Republic of Korea
| | - Hocheol Kim
- 2 Brain Korea 21 Plus Program, College of Korean Medicine, Kyung Hee University , Seoul, Republic of Korea.,3 Department of Herbology, College of Korean Medicine, Kyung Hee University , Seoul, Republic of Korea.,4 Institute of Korean Medicine, College of Korean Medicine, Kyung Hee University , Seoul, Republic of Korea
| | - Sang-Kyu Ye
- 5 Department of Pharmacology, Seoul National University College of Medicine , Seoul, Republic of Korea
| | - Ik-Hyun Cho
- 1 Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University , Seoul, Republic of Korea.,2 Brain Korea 21 Plus Program, College of Korean Medicine, Kyung Hee University , Seoul, Republic of Korea.,4 Institute of Korean Medicine, College of Korean Medicine, Kyung Hee University , Seoul, Republic of Korea
| |
Collapse
|
18
|
Twele F, Töllner K, Brandt C, Löscher W. Significant effects of sex, strain, and anesthesia in the intrahippocampal kainate mouse model of mesial temporal lobe epilepsy. Epilepsy Behav 2016; 55:47-56. [PMID: 26736063 DOI: 10.1016/j.yebeh.2015.11.027] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 11/24/2015] [Accepted: 11/24/2015] [Indexed: 01/12/2023]
Abstract
The intrahippocampal kainate mouse model of mesial temporal lobe epilepsy is increasingly being used for studies on epileptogenesis and antiepileptogenesis. Almost all previous studies used male mice for this purpose, and no study is available in this or other models of acquired epilepsy that directly compared epileptogenesis in female and male rodents. Epidemiological studies suggest that gender may affect susceptibility to epilepsy and its prognosis; therefore, one goal of this study was to investigate whether sex has an influence on latent period and epileptogenesis in the intrahippocampal kainate model in mice. Another aspect that was examined in the present study was whether mouse strain differences in epileptogenesis exist. Finally, we examined the effects of different types of anesthesia (chloral hydrate, isoflurane) on kainate-induced status epilepticus (SE) and epileptogenesis. Continuous (24/7) video-EEG monitoring was used during SE and the 2 weeks following SE as well as 4-6 weeks after SE. In male NMRI mice with chloral hydrate anesthesia during kainate injection, SE was followed by a seizure-free latent period of 10-14 days if hippocampal paroxysmal discharges (HPDs) recorded from the kainate focus were considered the onset of epilepsy. Anesthesia with isoflurane led to a more rapid onset and higher severity of SE, and not all male NMRI mice exhibited a seizure-free latent period. Female NMRI mice differed from male animals in the lack of any clear latent period, independently of anesthesia type. Furthermore, HPDs were only rarely observed. These problems were not resolved by decreasing the dose of kainate or using other strains (C57BL/6, FVB/N) of female mice. The present data are the first to demonstrate marked sex-related differences in the latent period following brain injury in a rodent model of acquired epilepsy. Furthermore, our data demonstrate that the choice of anesthestic agent during kainate administration affects SE severity and as a consequence, the latent period, which may explain some of the differences reported for this model in the literature.
Collapse
Affiliation(s)
- Friederike Twele
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Kathrin Töllner
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Claudia Brandt
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|
19
|
Shin EJ, Jeong JH, Chung CK, Kim DJ, Wie MB, Park ES, Chung YH, Nam Y, Tran TV, Lee SY, Kim HJ, Ong WY, Kim HC. Ceruloplasmin is an endogenous protectant against kainate neurotoxicity. Free Radic Biol Med 2015; 84:355-372. [PMID: 25843655 DOI: 10.1016/j.freeradbiomed.2015.03.031] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Revised: 03/10/2015] [Accepted: 03/14/2015] [Indexed: 01/08/2023]
Abstract
To determine the role of ceruloplasmin (Cp) in epileptic seizures, we used a kainate (KA) seizure animal model and examined hippocampal samples from epileptic patients. Treatment with KA resulted in a time-dependent decrease in Cp protein expression in the hippocampus of rats. Cp-positive cells were colocalized with neurons or reactive astrocytes in KA-treated rats and epileptic patient samples. KA-induced seizures, initial oxidative stress (i.e., hydroxyl radical formation, lipid peroxidation, protein oxidation, and synaptosomal reactive oxygen species), altered iron status (increasing Fe(2+) accumulation and L-ferritin-positive reactive microglial cells and decreasing H-ferritin-positive neurons), and impaired glutathione homeostasis and neurodegeneration (i.e., Fluoro-Nissl and Fluoro-Jade B staining analyses) were more pronounced in Cp antisense oligonucleotide (ASO)- than in Cp sense oligonucleotide-treated rats. Consistently, Cp ASO facilitated KA-induced lactate dehydrogenase (LDH) release, Fe(2+) accumulation, and glutathione loss in neuron-rich and mixed cultures. However, Cp ASO did not alter KA-induced LDH release or Fe(2+) accumulation in the astroglial culture, but did facilitate impairment in glutathione homeostasis in the same culture. Importantly, treatment with human Cp protein resulted in a significant attenuation against these neurotoxicities induced by Cp ASO. Our results suggest that Cp-mediated neuroprotection occurs via the inhibition of seizure-associated oxidative damage (including impairment in glutathione homeostasis), Fe(2+) accumulation, and alterations in ferritin immunoreactivity. Moreover, interactive modulation between neurons and glia was found to be important for Cp upregulation in the attenuation of epileptic damage in both animals and humans.
Collapse
Affiliation(s)
- Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 200-701, South Korea
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul 156-756, South Korea
| | - Chun Kee Chung
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul National University Hospital, Seoul 110-799, South Korea
| | - Dae-Joong Kim
- Department of Anatomy and Cell Biology, Medical School, Kangwon National University, Chunchon 200-701, South Korea
| | - Myung-Bok Wie
- Department of Veterinary Medicine, Kangwon National University, Chunchon 200-701, South Korea
| | - Eon Sub Park
- Department of Pathology, College of Medicine, Chung-Ang University, Seoul 156-756, South Korea
| | - Yoon Hee Chung
- Department of Anatomy, College of Medicine, Chung-Ang University, Seoul 156-756, South Korea
| | - Yunsung Nam
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 200-701, South Korea
| | - The-Vinh Tran
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 200-701, South Korea
| | - Sung Youl Lee
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 200-701, South Korea
| | - Hwa-Jung Kim
- College of Pharmacy, Ewha Women׳s University, Seoul 120-750, South Korea
| | - Wei-Yi Ong
- Department of Anatomy, National University of Singapore, Singapore 119260, Singapore
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 200-701, South Korea.
| |
Collapse
|
20
|
Naringin Attenuates Autophagic Stress and Neuroinflammation in Kainic Acid-Treated Hippocampus In Vivo. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:354326. [PMID: 26124853 PMCID: PMC4466392 DOI: 10.1155/2015/354326] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 05/27/2015] [Indexed: 01/21/2023]
Abstract
Kainic acid (KA) is well known as a chemical compound to study epileptic seizures and neuronal excitotoxicity. KA-induced excitotoxicity causes neuronal death by induction of autophagic stress and microglia-derived neuroinflammation, suggesting that the control of KA-induced effects may be important to inhibit epileptic seizures with neuroprotection. Naringin, a flavonoid in grapefruit and citrus fruits, has anti-inflammatory and antioxidative activities, resulting in neuroprotection in animal models from neurodegenerative diseases such as Parkinson's disease and Alzheimer's disease. In the present study, we examined its beneficial effects involved in antiautophagic stress and antineuroinflammation in the KA-treated hippocampus. Our results showed that naringin treatment delayed the onset of KA-induced seizures and decreased the occurrence of chronic spontaneous recurrent seizures (SRS) in KA-treated mice. Moreover, naringin treatment protected hippocampal CA1 neurons in the KA-treated hippocampus, ameliorated KA-induced autophagic stress, confirmed by the expression of microtubule-associated protein light chain 3 (LC3), and attenuated an increase in tumor necrosis factor-α (TNFα) in activated microglia. These results suggest that naringin may have beneficial effects of preventing epileptic events and neuronal death through antiautophagic stress and antineuroinflammation in the hippocampus in vivo.
Collapse
|
21
|
Cipriani R, Chara JC, Rodríguez-Antigüedad A, Matute C. FTY720 attenuates excitotoxicity and neuroinflammation. J Neuroinflammation 2015; 12:86. [PMID: 25953296 PMCID: PMC4429813 DOI: 10.1186/s12974-015-0308-6] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 04/27/2015] [Indexed: 11/18/2022] Open
Abstract
Background FTY720 (fingolimod, Gilenya™), a structural analog of sphingosine-1-phosphate (S1P), is the first oral drug approved for treatment the relapsing-remitting form of multiple sclerosis (MS), and its efficacy has been related to induced lymphopenia and consequent immunosuppression via modulation of S1P1 receptors (S1P1R). However, due to its lipophilic nature, FTY720 crosses the blood brain barrier (BBB) and could act directly on neural cells. In this study, we investigated the effectiveness of FTY720 as a neuroprotective agent using in vitro and in vivo models of excitotoxic neuronal death and examined if FTY720 exerts a direct action on neurons, or/and an indirect modulation of inflammation-mediated neurodegeneration as a possible mechanism of neuroprotection. Methods Primary neuronal and organotypic cortical cultures were treated with N-methyl-D-aspartic acid (NMDA) to induce excitotoxic cell death (measured by lactate dehydrogenase (LDH) assay or propidium iodide uptake, respectively). The effects of FTY720 treatment (10, 100 and 1,000 nM) on neuronal survival were examined. As an in vivo model of neuronal death and inflammation, we used intracerebroventricular (icv) administration of kainic acid (KA; 0.5 μg/2 μl) in Sprague–Dawley rats. FTY720 was applied icv (1 μg/2 μl), together with KA, plus intraperitoneally (ip; 1 mg/kg) 24 h before, and daily, until sacrifice 3 days after icv. Rats were evaluated for neurological score, neuronal loss in CA3 hippocampal region and activation of microglia at the lesion site. In addition, we tested FTY720 as a modulator of microglia responses using microglial cell cultures activated with lipopolysaccharide (LPS) and its effects in stress signalling pathways using western blotting for p38 and JNK1/2 mitogen-activated protein kinases (MAPKs). Results FTY720 was able to reduce excitotoxic neuronal death in vitro. Moreover, in vivo repeated FTY720 administration attenuated KA-induced neurodegeneration and microgliosis at the CA3 lesion site. Furthermore, FTY720 negatively modulates p38 MAPK in LPS-activated microglia, whereas it had no effect on JNK1/2 activation. Conclusions These data support a role for FTY720 as a neuroprotective agent against excitotoxin-induced neuronal death and as a negative modulator of neuroinflammation by targeting the p38 MAPK stress signalling pathway in microglia. Electronic supplementary material The online version of this article (doi:10.1186/s12974-015-0308-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Raffaela Cipriani
- Centro de Investigaciones Biomédicas en Red (CIBERNED), Achucarro Basque Center for Neuroscience Bizkaia Science and Technology Park, Building 205, E-48170, Zamudio, Spain. .,Departamento de Neurociencias, Universidad del País Vasco, Barrio Sarriena s/n, E-48940, Leioa, Spain.
| | - Juan Carlos Chara
- Centro de Investigaciones Biomédicas en Red (CIBERNED), Achucarro Basque Center for Neuroscience Bizkaia Science and Technology Park, Building 205, E-48170, Zamudio, Spain. .,Departamento de Neurociencias, Universidad del País Vasco, Barrio Sarriena s/n, E-48940, Leioa, Spain.
| | | | - Carlos Matute
- Centro de Investigaciones Biomédicas en Red (CIBERNED), Achucarro Basque Center for Neuroscience Bizkaia Science and Technology Park, Building 205, E-48170, Zamudio, Spain. .,Departamento de Neurociencias, Universidad del País Vasco, Barrio Sarriena s/n, E-48940, Leioa, Spain.
| |
Collapse
|
22
|
Twele F, Bankstahl M, Klein S, Römermann K, Löscher W. The AMPA receptor antagonist NBQX exerts anti-seizure but not antiepileptogenic effects in the intrahippocampal kainate mouse model of mesial temporal lobe epilepsy. Neuropharmacology 2015; 95:234-42. [PMID: 25839899 DOI: 10.1016/j.neuropharm.2015.03.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 02/26/2015] [Accepted: 03/19/2015] [Indexed: 01/05/2023]
Abstract
The AMPA receptor subtype of glutamate receptors, which mediates fast synaptic excitation, is of primary importance in initiating epileptiform discharges, so that AMPA receptor antagonists exert anti-seizure activity in diverse animal models of partial and generalized seizures. Recently, the first AMPA receptor antagonist, perampanel, was approved for use as adjunctive therapy for the treatment of resistant partial seizures in patients. Interestingly, the competitive AMPA receptor antagonist NBQX has recently been reported to prevent development of spontaneous recurrent seizures (SRS) in a neonatal seizure model in rats, indicating the AMPA antagonists may exert also antiepileptogenic effects. This prompted us to evaluate competitive (NBQX) and noncompetitive (perampanel) AMPA receptor antagonists in an adult mouse model of mesial temporal lobe epilepsy. In this model, SRS develop after status epilepticus (SE) induced by intrahippocampal injection of kainate. Focal electrographic seizures in this model are resistant to several major antiepileptic drugs. In line with previous studies, phenytoin was not capable of blocking such seizures in the present experiments, while they were markedly suppressed by NBQX and perampanel. However, perampanel was less tolerable than NBQX in epileptic mice, so that only NBQX was subsequently tested for antiepileptogenic potential. When mice were treated over three days after kainate-induced SE with NBQX (20 mg/kg t.i.d.), no effect on development or frequency of seizures was found in comparison to vehicle controls. These results suggest that AMPA receptor antagonists, while being effective in suppressing resistant focal seizures, are not exerting antiepileptogenic effects in an adult mouse model of partial epilepsy.
Collapse
Affiliation(s)
- Friederike Twele
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany
| | - Marion Bankstahl
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany
| | - Sabine Klein
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany
| | - Kerstin Römermann
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Center for Systems Neuroscience, 30559 Hannover, Germany.
| |
Collapse
|
23
|
Inter-individual variation in the effect of antiepileptic drugs in the intrahippocampal kainate model of mesial temporal lobe epilepsy in mice. Neuropharmacology 2014; 90:53-62. [PMID: 25460186 DOI: 10.1016/j.neuropharm.2014.11.008] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 11/14/2014] [Accepted: 11/16/2014] [Indexed: 11/20/2022]
Abstract
Despite more than 20 clinically approved antiepileptic drugs (AEDs), there remains a substantial unmet clinical need for patients with refractory (AED-resistant) epilepsy. Animal models of refractory epilepsy are needed for at least two goals; (1) better understanding of the mechanisms underlying resistance to AEDs, and (2) development of more efficacious AEDs for patients with refractory seizures. It is only incompletely understood why two patients with seemingly identical types of epilepsy and seizures may respond differently to the same AED. Prompted by this well-known clinical phenomenon, we previously evaluated whether epileptic rats respond differently to AEDs and discovered AED responsive and resistant animals in the same models. In the present study, we used the same approach for the widely used intrahippocampal kainate mouse model of mesial temporal lobe epilepsy. In a first step, we examined anti-seizure effects of 6 AEDs on spontaneous recurrent focal electrographic seizures and secondarily generalized convulsive seizures in epileptic mice, showing that the focal nonconvulsive seizures were resistant to carbamazepine and phenytoin, whereas valproate and levetiracetam exerted moderate and phenobarbital and diazepam marked anti-seizure effects. All AEDs seemed to suppress generalized convulsive seizures. Next we investigated the inter-individual variation in the anti-seizure effects of these AEDs and, in case of focal seizures, found responders and nonresponders to all AEDs except carbamazepine. Most nonresponders were resistant to more than one AED. Our data further validate the intrahippocampal kainate mouse model as a model of difficult-to-treat focal seizures that can be used to investigate the determinants of AED efficacy.
Collapse
|
24
|
Anthony DC, Couch Y. The systemic response to CNS injury. Exp Neurol 2014; 258:105-11. [PMID: 25017891 DOI: 10.1016/j.expneurol.2014.03.013] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 03/11/2014] [Accepted: 03/21/2014] [Indexed: 12/29/2022]
Abstract
Inflammation within the brain or spinal cord has the capacity to damage neurons and is known to contribute to long-term disability in a spectrum of central nervous system (CNS) pathologies. However, there is a more profound increase in the recruitment of potentially damaging populations of leukocytes to the spinal cord than to the brain after equivalent injuries. Increased levels of inflammatory cytokines and chemokines in the spinal cord underpin this dissimilarity after injury, which also appears to be very sensitive to processes that operate within organs distant from the primary injury site such as the liver, lung and spleen. Indeed, CNS injury per se can generate profound changes in gene expression and the cellularity of these organs, which, as a consequence, gives rise to secondary organ damage. Our understanding of the local inflammatory processes that can damage neurons is becoming clearer, but our understanding of how the peripheral immune system coordinates the response to CNS injury and how any concomitant infections or injury might impact on the outcome of CNS injury is not so well developed. It is clear that the orientation of the response to peripheral challenges, be it a pro- or anti-inflammatory effect, appears to be dependent on the nature and timing of events. Here, the importance of the inter-relationship between inflammation in the CNS and the consequent inflammatory response in peripheral tissues is highlighted.
Collapse
Affiliation(s)
| | - Yvonne Couch
- Department of Pharmacology, University of Oxford, Oxford, UK
| |
Collapse
|
25
|
Belarbi K, Rosi S. Modulation of adult-born neurons in the inflamed hippocampus. Front Cell Neurosci 2013; 7:145. [PMID: 24046730 PMCID: PMC3764370 DOI: 10.3389/fncel.2013.00145] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 08/21/2013] [Indexed: 11/13/2022] Open
Abstract
Throughout life new neurons are continuously added to the hippocampal circuitry involved with spatial learning and memory. These new cells originate from neural precursors in the subgranular zone of the dentate gyrus, migrate into the granule cell layer, and integrate into neural networks encoding spatial and contextual information. This process can be influenced by several environmental and endogenous factors and is modified in different animal models of neurological disorders. Neuroinflammation, as defined by the presence of activated microglia, is a common key factor to the progression of neurological disorders. Analysis of the literature shows that microglial activation impacts not only the production, but also the migration and the recruitment of new neurons. The impact of microglia on adult-born neurons appears much more multifaceted than ever envisioned before, combining both supportive and detrimental effects that are dependent upon the activation phenotype and the factors being released. The development of strategies aimed to change microglia toward states that promote functional neurogenesis could therefore offer novel therapeutic opportunities against neurological disorders associated with cognitive deficits and neuroinflammation. The present review summarizes the current knowledge on how production, distribution, and recruitment of new neurons into behaviorally relevant neural networks are modified in the inflamed hippocampus.
Collapse
Affiliation(s)
- Karim Belarbi
- Brain and Spinal Injury Center, San Francisco General Hospital, University of California at San Francisco San Francisco, CA, USA ; Department of Physical Therapy and Rehabilitation Science, University of California at San Francisco San Francisco, CA, USA
| | | |
Collapse
|
26
|
Cabrera V, Ramos E, González-Arenas A, Cerbón M, Camacho-Arroyo I, Morales T. Lactation reduces glial activation induced by excitotoxicity in the rat hippocampus. J Neuroendocrinol 2013; 25:519-27. [PMID: 23356710 DOI: 10.1111/jne.12028] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Revised: 01/16/2013] [Accepted: 01/21/2013] [Indexed: 01/08/2023]
Abstract
Motherhood induces a series of adaptations in the physiology of the female, including an increase of maternal brain plasticity and a reduction of cell damage in the hippocampus caused by kainic acid (KA) excitotoxicity. We analysed the role of lactation in glial activation in the hippocampal fields of virgin and lactating rats after i.c.v. application of 100 ng of KA. Immunohistochemical analysis for glial fibrillary acidic protein (GFAP) and ionised calcium binding adaptor molecule 1 (Iba-1), which are markers for astrocytes and microglial cell-surface proteins, respectively, revealed differential cellular responses to KA in lactating and virgin rats. A significant astrocyte and microglial response in hippocampal areas of virgin rats was observed 24 h and 72 h after KA. By contrast, no increase in either GFAP- or Iba-1-positive cells was observed in response to KA in the hippocampus of lactating rats. Western blot analysis of GFAP showed an initial decrease at 24 h after KA treatment, with an increase at 72 h in the whole hippocampus of virgin but not of lactating rats. The number of GFAP-positive cells was increased by lactation in the dentate gyrus of the hippocampus but not in CA1 and CA3 areas. The present results indicate that lactating rats exhibit diminished responses of astrocyte and microglial cells in the hippocampus to damage induced by KA, supporting the notion that the maternal hippocampus is resistant to excitotoxic insults.
Collapse
Affiliation(s)
- V Cabrera
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, México
| | | | | | | | | | | |
Collapse
|
27
|
Vernet-der Garabedian B, Derer P, Bailly Y, Mariani J. Innate immunity in the Grid2Lc/+ mouse model of cerebellar neurodegeneration: glial CD95/CD95L plays a non-apoptotic role in persistent neuron loss-associated inflammatory reactions in the cerebellum. J Neuroinflammation 2013; 10:65. [PMID: 23672668 PMCID: PMC3657541 DOI: 10.1186/1742-2094-10-65] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 04/12/2013] [Indexed: 11/10/2022] Open
Abstract
Background There is growing evidence that the death receptor CD95 has a wider role in non-apoptotic functions. In the brain, it may contribute to neural death and to the associated inflammatory reaction via a non-apoptotic pathway. Brain injury triggers an inflammatory reaction in which the CD95/CD95L system acts principally through peripheral cells recruited to the lesion. In cases of inflammation within the brain, with no blood–brain barrier leakage, the role of the CD95/CD95L system is thus unclear. We investigated the possible role of CD95 and CD95L in such conditions, by studying the relationships between glial cell activation, neuron death and CD95/CD95L expression in the cerebellum of the Lurcher (Grid2Lc/+) mutant mouse, a model of cerebellar neurodegeneration. Methods Glial cells in slices of wild-type and Lurcher mouse cerebella were observed by light microscopy at various ages overlapping periods of neuron loss and of pre- and post-neurodegeneration. Subcellular organization was studied by electron microscopy. We assessed CD95 levels by western blotting, RT-PCR and glial cell cultures. The levels of CD95L and IL-6 were studied by ELISA and a biological assay, respectively. Results In the Grid2Lc/+cerebellum, neuron loss triggers a typical, but abnormally persistent, inflammatory reaction. We identified two phases of astrogliosis: an early burst of large glial cell activation, peaking at postnatal days 25 to 26, coinciding with peak cerebellar neuron loss, followed by a long period of slow decline indicating that the strength of the glial reaction is modulated by neuron mortality rates. Comparisons of time-courses of glial cell activation, cytokine production and neuron loss revealed that the number of surviving neurons decreased as CD95 increased. Thus, CD95 cannot be directly involved in neuron death, and its role must be limited to a contribution to the inflammatory reaction. The upregulation of CD95 likely on astrocytes coincides with increases in the levels of IL-6, a cytokine produced principally by astrocytes, and soluble CD95L. Conclusions These results suggest that CD95 and soluble CD95L contribute, via non-apoptotic signaling, to the inflammatory reaction initiated early in neuron death within the Grid2Lc/+ cerebellum.
Collapse
|
28
|
Ellingson BM, Salamon N, Holly LT. Imaging techniques in spinal cord injury. World Neurosurg 2012; 82:1351-8. [PMID: 23246741 DOI: 10.1016/j.wneu.2012.12.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 11/05/2012] [Accepted: 12/07/2012] [Indexed: 10/27/2022]
Abstract
BACKGROUND Spinal imaging plays a critical role in the diagnosis, treatment, and rehabilitation of patients with spinal cord injury (SCI). In recent years there has been increasing interest in the development of advanced imaging techniques to provide pertinent microstructural and metabolic information that is not provided by conventional modalities. METHODS This review details the pathophysiological structural changes that accompany SCI, as well as their imaging correlates. The potential clinical applications of novel spinal cord imaging techniques to SCI are presented. RESULTS There are a variety of novel advanced imaging techniques that are principally focused on the microstructural and/or biochemical function of the spinal cord, and can potentially be applied to traumatic SCI, including diffusion tensor imaging, magnetic resonance spectroscopy, positron emission tomography, single-photon emission computed tomography, and functional magnetic resonance imaging. These techniques are presently in various stages of development, including some whose applications are primarily limited to laboratory investigation, whereas others are being actively used in clinical practice. CONCLUSION Advanced imaging of the spinal cord has tremendous potential to provide patient-specific physiological information about the status of cord integrity and health. Advanced spinal cord imaging is still at early stages of development and clinical implementation but is likely to play an increasingly important role in the management of spinal cord health in the foreseeable future.
Collapse
Affiliation(s)
- Benjamin M Ellingson
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA; Department of Biomedical Physics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA; Department of Bioengineering, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Noriko Salamon
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Langston T Holly
- Department of Neurosurgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA.
| |
Collapse
|
29
|
Anthony DC, Pitossi FJ. Special issue commentary: the changing face of inflammation in the brain. Mol Cell Neurosci 2012; 53:1-5. [PMID: 23147112 DOI: 10.1016/j.mcn.2012.11.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Accepted: 11/02/2012] [Indexed: 12/24/2022] Open
Abstract
The study of inflammation in the brain has been extended to include a wide range of conditions, but there remains plenty of argument over semantics and the precise definition of what constitutes inflammation in these pathologies. In this special issue, we sought to highlight the diversity of what is considered to be inflammation in the brain, and we have accepted that the presence of microglia cells with altered morphology remains a useful starting point. However, it is clear that whatever is the molecular expression profile that accompanies an activated microglial cell, it is not static and it is influenced by factors both intrinsic and extrinsic to the brain. This article is part of a Special Issue entitled 'Neuroinflammation in neurodegeneration and neurodysfunction'.
Collapse
|
30
|
Kim HS, Son TG, Park HR, Lee Y, Jung Y, Ishigami A, Lee J. Senescence marker protein 30 deficiency increases Parkinson's pathology by impairing astrocyte activation. Neurobiol Aging 2012; 34:1177-83. [PMID: 23122412 DOI: 10.1016/j.neurobiolaging.2012.10.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 07/04/2012] [Accepted: 10/07/2012] [Indexed: 10/27/2022]
Abstract
Senescence marker protein 30 (SMP30) was recently identified as gluconolactonase, which is involved in vitamin C (VC) biosynthesis. Therefore, the antioxidant property of SMP30 is thought to be mediated by its gluconolactonase function. However, pathologic effects of SMP30 deficiency independent of VC biosynthesis have not been studied in models of neurodegenerative diseases. In the present study, we evaluated the effect of SMP30 deficiency on Parkinson's disease (PD) in SMP30 knockout (KO) mice. Wild type and SMP30 KO mice supplemented with VC were treated with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Our results showed that MPTP-induced dopaminergic neuronal loss and motor function impairment were more significant in the SMP30 KO mice. Reactive oxygen species generation and microglia activation were higher in MPTP-treated SMP30 KO mice. However, SMP30 deficiency mitigated MPTP-induced astrocyte activation and glia-derived neurotrophic factor production. Cultures of astrocytes recovered from wild type and SMP30 KO mice revealed that SMP30 deficiency abolished 1-methyl-4-phenyl-pyridinium-induced astroglial activation by blocking the extracellular signal-regulated kinase pathway. Taken together, our findings demonstrate for the first time that SMP30 deficiency increases the severity of PD and suggest a beneficial role of SMP30 in protective astrocyte activation in response to neurodegeneration. The present study shows that modulation of astrocytic SMP30 can be a promising target for treating PD.
Collapse
Affiliation(s)
- Hyun Soo Kim
- College of Pharmacy and Research Institute for Drug Development, Longevity Life Science and Technology Institutes, Pusan National University, Busan, South Korea
| | | | | | | | | | | | | |
Collapse
|
31
|
Yi MH, Zhang E, Kang JW, Shin YN, Byun JY, Oh SH, Seo JH, Lee YH, Kim DW. Expression of CD200 in alternative activation of microglia following an excitotoxic lesion in the mouse hippocampus. Brain Res 2012; 1481:90-6. [PMID: 22975132 DOI: 10.1016/j.brainres.2012.08.053] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Revised: 08/10/2012] [Accepted: 08/28/2012] [Indexed: 12/23/2022]
Abstract
CD200 is a glycoprotein that is expressed on the surfaces of neurons and other cells. It interacts with its receptor, CD200R, which is expressed on cells of the myeloid lineage, including microglia. The interaction of CD200 with its receptor plays a significant role in maintaining microglia in a quiescent state; thus, a decrease in CD200 expression in the brain is associated with evidence of microglial activation. However, their roles in pathological progression remain unclear. We examined the expression of CD200 in kainic acid (KA)-induced neurodegeneration of the mouse hippocampus. Our quantitative analysis revealed that CD200 was constitutively expressed in the normal brain and transiently upregulated by KA treatment. At the cellular level, CD200 was expressed in neurons in control, and was upregulated primarily in the microglia of KA-treated mouse hippocampi. We examined the contribution of CD200 to both the classical and alternative activation of microglia in vitro using an adult microglia culture, which was exposed to interleukin-4 (IL-4) with and without lipopolysaccharide (LPS). CD200 expression was increased after exposure to IL-4, but not to LPS. These in vivo experiments demonstrated that CD200 was transiently expressed in microglia in a process mediated by the inflammatory response. Based on CD200R expression in microglia, it suggests that microglia is maintained in an activated state with autocrine signaling by interactions between microglial CD200 and its CD200R. Moreover, we suggest that CD200 may be expressed in the alternative activation of microglia and play a beneficial role in neuroinflammation.
Collapse
Affiliation(s)
- Min-Hee Yi
- Department of Anatomy, Brain Research Institute, Chungnam National University School of Medicine, Daejeon, South Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Hsieh CL, Chen CL, Tang NY, Chuang CM, Hsieh CT, Chiang SY, Lin JG, Hsu SF. Gastrodia elata BL Mediates the Suppression of nNOS and Microglia Activation to Protect Against Neuronal Damage in Kainic Acid-Treated Rats. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2012; 33:599-611. [PMID: 16173534 DOI: 10.1142/s0192415x0500320x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Our previous studies showed that Gastrodia elata (GE), an herb used in traditional Chinese medicine, has both anti-convulsive and free radical-scavenging activities in kainic acid (KA)-treated rats. The aim of the present study was to further investigate possible physiological mechanisms of GE against activities of neuronal nitric oxide synthase (nNOS) and microglia in KA-treated rats; 0.5 g/kg and 1.0 g/kg of GE extract were administered orally, whereas 20 mg/kg of N-nitro-L-arginine methyl ester (L-NAME) was administered intraperitoneally (ip), both at 30 minutes prior to KA (2 μg/2 μl) being injected into the right hippocampus region of rats. ED1-staining, apoptotic, inducible nitric oxide synthase (iNOS), and nNOS-staining cells were observed in the hippocampus region. The results indicated that 1.0 g/kg of GE and 20 mg/kg of L-NAME reduced the counts of ED1-stained cells, and 0.5 g/kg and 1.0 g/kg of GE, and 20 mg/kg of L-NAME reduced the numbers of apoptotic cells and nNOS-staining cells. In addition, 20 mg/kg of L-NAME also reduced the numbers of iNOS-staining cells, but 0.5 g/kg and 1.0 g/kg of GE did not. This study demonstrated that GE was able to reduce nNOS, microglia activation and apoptosis, suggesting that GE has a protective effect against neuronal damage in KA-treated rats.
Collapse
Affiliation(s)
- Ching-Liang Hsieh
- Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Vascular disruption and the role of angiogenic proteins after spinal cord injury. Transl Stroke Res 2011; 2:474-91. [PMID: 22448202 PMCID: PMC3296011 DOI: 10.1007/s12975-011-0109-x] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2011] [Revised: 09/20/2011] [Accepted: 09/25/2011] [Indexed: 02/06/2023]
Abstract
Spinal cord injuries (SCI) can result in devastating paralysis, for which there is currently no robustly efficacious neuroprotective/neuroregenerative treatment. When the spinal cord is subjected to a traumatic injury, the local vasculature is disrupted and the blood–spinal cord barrier is compromised. Subsequent inflammation and ischemia may then contribute to further secondary damage, exacerbating neurological deficits. Therefore, understanding the vascular response to SCI and the molecular elements that regulate angiogenesis has considerable relevance from a therapeutic standpoint. In this paper, we review the nature of vascular damage after traumatic SCI and what is known about the role that angiogenic proteins—angiopoietin 1 (Ang1), angiopoietin 2 (Ang2) and angiogenin—may play in the subsequent response. To this, we add recent work that we have conducted in measuring these proteins in the cerebrospinal fluid (CSF) and serum after acute SCI in human patients. Intrathecal catheters were installed in 15 acute SCI patients within 48 h of injury. CSF and serum samples were collected over the following 3–5 days and analysed for Ang1, Ang2 and angiogenin protein levels using a standard ELISA technique. This represents the first description of the endogenous expression of these proteins in an acute human SCI setting.
Collapse
|
34
|
Hanamsagar R, Torres V, Kielian T. Inflammasome activation and IL-1β/IL-18 processing are influenced by distinct pathways in microglia. J Neurochem 2011; 119:736-48. [PMID: 21913925 DOI: 10.1111/j.1471-4159.2011.07481.x] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Microglia are important innate immune effectors against invading CNS pathogens, such as Staphylococcus aureus (S. aureus), a common etiological agent of brain abscesses typified by widespread inflammation and necrosis. The NLRP3 inflammasome is a protein complex involved in IL-1β and IL-18 processing following exposure to both pathogen- and danger-associated molecular patterns. Although previous studies from our laboratory have established that IL-1β is a major cytokine product of S. aureus-activated microglia and is pivotal for eliciting protective anti-bacterial immunity during brain abscess development, the molecular machinery responsible for cytokine release remains to be determined. Therefore, the functional role of the NLRP3 inflammasome and its adaptor protein apoptosis-associated speck-like protein (ASC) in eliciting IL-1β and IL-18 release was examined in primary microglia. Interestingly, we found that IL-1β, but not IL-18 production, was significantly attenuated in both NLRP3 and ASC knockout microglia following exposure to live S. aureus. NLRP3 inflammasome activation was partially dependent on autocrine/paracrine ATP release and α- and γ-hemolysins produced by live bacteria. A cathepsin B inhibitor attenuated IL-β release from NLRP3 and ASC knockout microglia, demonstrating the existence of alternative inflammasome-independent mechanisms for IL-1β processing. In contrast, microglial IL-18 secretion occurred independently of cathepsin B and inflammasome action. Collectively, these results demonstrate that microglial IL-1β processing is regulated by multiple pathways and diverges from mechanisms utilized for IL-18 cleavage. Understanding the molecular events that regulate IL-1β production is important for modulating this potent proinflammatory cytokine during CNS disease.
Collapse
Affiliation(s)
- Richa Hanamsagar
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | | | | |
Collapse
|
35
|
Markus T, Hansson SR, Cronberg T, Cilio C, Wieloch T, Ley D. β-Adrenoceptor activation depresses brain inflammation and is neuroprotective in lipopolysaccharide-induced sensitization to oxygen-glucose deprivation in organotypic hippocampal slices. J Neuroinflammation 2010; 7:94. [PMID: 21172031 PMCID: PMC3017519 DOI: 10.1186/1742-2094-7-94] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Accepted: 12/20/2010] [Indexed: 01/16/2023] Open
Abstract
Background Inflammation acting in synergy with brain ischemia aggravates perinatal ischemic brain damage. The sensitizing effect of pro-inflammatory exposure prior to hypoxia is dependent on signaling by TNF-α through TNF receptor (TNFR) 1. Adrenoceptor (AR) activation is known to modulate the immune response and synaptic transmission. The possible protective effect of α˜ and β˜AR activation against neuronal damage caused by tissue ischemia and inflammation, acting in concert, was evaluated in murine hippocampal organotypic slices treated with lipopolysaccharide (LPS) and subsequently subjected to oxygen-glucose deprivation (OGD). Method Hippocampal slices from mice were obtained at P6, and were grown in vitro for 9 days on nitrocellulose membranes. Slices were treated with β1(dobutamine)-, β2(terbutaline)-, α1(phenylephrine)- and α2(clonidine)-AR agonists (5 and 50 μM, respectively) during LPS (1 μg/mL, 24 h) -exposure followed by exposure to OGD (15 min) in a hypoxic chamber. Cell death in the slice CA1 region was assessed by propidium iodide staining of dead cells. Results Exposure to LPS + OGD caused extensive cell death from 4 up to 48 h after reoxygenation. Co-incubation with β1-agonist (50 μM) during LPS exposure before OGD conferred complete protection from cell death (P < 0.001) whereas the β2-agonist (50 μM) was partially protective (p < 0.01). Phenylephrine was weakly protective while no protection was attained by clonidine. Exposure to both β1- and β2-agonist during LPS exposure decreased the levels of secreted TNF-α, IL-6 and monocyte chemoattractant protein-1 and prevented microglia activation in the slices. Dobutamine remained neuroprotective in slices exposed to pure OGD as well as in TNFR1-/- and TNFR2-/- slices exposed to LPS followed by OGD. Conclusions Our data demonstrate that activation of both β1- and β2-receptors is neuroprotective and may offer mechanistic insights valuable for development of neuro-protective strategies in neonates.
Collapse
|
36
|
Hong J, Cho IH, Kwak KI, Suh EC, Seo J, Min HJ, Choi SY, Kim CH, Park SH, Jo EK, Lee S, Lee KE, Lee SJ. Microglial Toll-like receptor 2 contributes to kainic acid-induced glial activation and hippocampal neuronal cell death. J Biol Chem 2010; 285:39447-57. [PMID: 20923777 DOI: 10.1074/jbc.m110.132522] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Recent studies indicate that Toll-like receptors (TLRs), originally identified as infectious agent receptors, also mediate sterile inflammatory responses during tissue damage. In this study, we investigated the role of TLR2 in excitotoxic hippocampal cell death using TLR2 knock-out (KO) mice. TLR2 expression was up-regulated in microglia in the ipsilateral hippocampus of kainic acid (KA)-injected mice. KA-mediated hippocampal cell death was significantly reduced in TLR2 KO mice compared with wild-type (WT) mice. Similarly, KA-induced glial activation and proinflammatory gene expression in the hippocampus were compromised in TLR2 KO mice. In addition, neurons in organotypic hippocampal slice cultures (OHSCs) from TLR2 KO mouse brains were less susceptible to KA excitotoxicity than WT OHSCs. This protection is partly attributed to decreased expression of proinflammatory genes, such as TNF-α and IL-1β in TLR2 KO mice OHSCs. These data demonstrate conclusively that TLR2 signaling in microglia contributes to KA-mediated innate immune responses and hippocampal excitotoxicity.
Collapse
Affiliation(s)
- Jinpyo Hong
- Program in Molecular and Cellular Neuroscience, DRI, BK21, and Department of Oral Physiology, School of Dentistry, Seoul National University, Seoul 110-749, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
de Waard MC, van der Pluijm I, Zuiderveen Borgesius N, Comley LH, Haasdijk ED, Rijksen Y, Ridwan Y, Zondag G, Hoeijmakers JHJ, Elgersma Y, Gillingwater TH, Jaarsma D. Age-related motor neuron degeneration in DNA repair-deficient Ercc1 mice. Acta Neuropathol 2010; 120:461-75. [PMID: 20602234 PMCID: PMC2923326 DOI: 10.1007/s00401-010-0715-9] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2010] [Revised: 05/25/2010] [Accepted: 06/24/2010] [Indexed: 11/16/2022]
Abstract
Degeneration of motor neurons contributes to senescence-associated loss of muscle function and underlies human neurodegenerative conditions such as amyotrophic lateral sclerosis and spinal muscular atrophy. The identification of genetic factors contributing to motor neuron vulnerability and degenerative phenotypes in vivo are therefore important for our understanding of the neuromuscular system in health and disease. Here, we analyzed neurodegenerative abnormalities in the spinal cord of progeroid Ercc1Δ/− mice that are impaired in several DNA repair systems, i.e. nucleotide excision repair, interstrand crosslink repair, and double strand break repair. Ercc1Δ/− mice develop age-dependent motor abnormalities, and have a shortened life span of 6–7 months. Pathologically, Ercc1Δ/− mice develop widespread astrocytosis and microgliosis, and motor neuron loss and denervation of skeletal muscle fibers. Degenerating motor neurons in many occasions expressed genotoxic-responsive transcription factors p53 or ATF3, and in addition, displayed a range of Golgi apparatus abnormalities. Furthermore, Ercc1Δ/− motor neurons developed perikaryal and axonal intermediate filament abnormalities reminiscent of cytoskeletal pathology observed in aging spinal cord. Our findings support the notion that accumulation of DNA damage and genotoxic stress may contribute to neuronal aging and motor neuron vulnerability in human neuromuscular disorders.
Collapse
|
38
|
Yao Y, Tsirka SE. The C terminus of mouse monocyte chemoattractant protein 1 (MCP1) mediates MCP1 dimerization while blocking its chemotactic potency. J Biol Chem 2010; 285:31509-16. [PMID: 20682771 DOI: 10.1074/jbc.m110.124891] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The extracellular protease plasmin cleaves mouse MCP1 (monocyte chemoattractant protein 1) at lysine 104, releasing a 50-amino acid C-terminal domain. The cleavage event increases the chemotactic activity of MCP1 and, by doing so, promotes the progression of excitotoxic injury in the central nervous system in pathological settings. The mechanism through which the cleavage event enhances MCP1-mediated chemoattraction is unknown; to investigate it, we use wild-type and mutant forms of recombinant MCP1. Full-length MCP1 (FL-MCP1) is secreted by cells as a dimer or multimer. We show that a mutant truncated at the C terminus, K104Stop-MCP1, does not dimerize, revealing that the C terminus mediates the interaction. MCP1 interacts with the monocyte/microglia receptor CCR2. The interaction is critical to the function of MCP1 because CCR2(-/-) microglia do not undergo chemotaxis in response to MCP1 stimulation. We show that stimulation of microglia with FL-MCP1 or K104Stop-MCP1 triggers CCR2 internalization, whereas a mutant form unable to be cleaved at lysine 104 (K104A-MCP1) is relatively ineffective in this assay, suggesting that the C-terminal region interferes with the MCP1-CCR2 interaction. Moreover, FL-MCP1 and K104Stop-MCP1 stimulation leads to activation of Rac1, a small GTPase involved in cell migration. Conversely, MCP1-stimulated microglial migration is blocked by the Rac1 inhibitor, NSC23766, demonstrating the requirement for Rac1 effector pathways in this response. Taken together, we propose a model for MCP1 localization, activation, and function based on the initial presence and then removal of its C terminus, coupled with a requisite downstream signaling pathway from CCR2 stimulation to Rac1 activation.
Collapse
Affiliation(s)
- Yao Yao
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, New York 11794-8651, USA
| | | |
Collapse
|
39
|
Motor neuron-immune interactions: the vicious circle of ALS. J Neural Transm (Vienna) 2010; 117:981-1000. [PMID: 20552235 DOI: 10.1007/s00702-010-0429-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Accepted: 05/22/2010] [Indexed: 12/12/2022]
Abstract
Because microglial cells, the resident macrophages of the CNS, react to any lesion of the nervous system, they have for long been regarded as potential players in the pathogenesis of several neurodegenerative disorders including amyotrophic lateral sclerosis, the most common motor neuron disease in the adult. In recent years, this microglial reaction to motor neuron injury, in particular, and the innate immune response, in general, has been implicated in the progression of the disease, in mouse models of ALS. The mechanisms by which microglial cells influence motor neuron death in ALS are still largely unknown. Microglial activation increases over the course of the disease and is associated with an alteration in the production of toxic factors and also neurotrophic factors. Adding to the microglial/macrophage response to motor neuron degeneration, the adaptive immune system can likewise influence the disease process. Exploring these motor neuron-immune interactions could lead to a better understanding in the physiopathology of ALS to find new pathways to slow down motor neuron degeneration.
Collapse
|
40
|
Costa-Ferro ZSM, Vitola AS, Pedroso MF, Cunha FB, Xavier LL, Machado DC, Soares MBP, Ribeiro-dos-Santos R, DaCosta JC. Prevention of seizures and reorganization of hippocampal functions by transplantation of bone marrow cells in the acute phase of experimental epilepsy. Seizure 2010; 19:84-92. [PMID: 20080419 DOI: 10.1016/j.seizure.2009.12.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2009] [Revised: 12/11/2009] [Accepted: 12/17/2009] [Indexed: 12/24/2022] Open
Abstract
In this study, we investigated the therapeutic potential of bone marrow mononuclear cells (BMCs) in a model of epilepsy induced by pilocarpine in rats. BMCs obtained from green fluorescent protein (GFP) transgenic mice or rats were transplanted intravenously after induction of status epilepticus (SE). Spontaneous recurrent seizures (SRS) were monitored using Racine's seizure severity scale. All of the rats in the saline-treated epileptic control group developed SRS, whereas none of the BMC-treated epileptic animals had seizures in the short term (15 days after transplantation), regardless of the BMC source. Over the long-term chronic phase (120 days after transplantation), only 25% of BMC-treated epileptic animals had seizures, but with a lower frequency and duration compared to the epileptic control group. The density of hippocampal neurons in the brains of animals treated with BMCs was markedly preserved. At hippocampal Schaeffer collateral-CA1 synapses, long-term potentiation was preserved in BMC-transplanted rats compared to epileptic controls. The donor-derived GFP(+) cells were rarely found in the brains of transplanted epileptic rats. In conclusion, treatment with BMCs can prevent the development of chronic seizures, reduce neuronal loss, and influence the reorganization of the hippocampal neuronal network.
Collapse
Affiliation(s)
- Zaquer S M Costa-Ferro
- Departamento de Fisiologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Cunningham O, Campion S, Perry VH, Murray C, Sidenius N, Docagne F, Cunningham C. Microglia and the urokinase plasminogen activator receptor/uPA system in innate brain inflammation. Glia 2010; 57:1802-14. [PMID: 19459212 PMCID: PMC2816357 DOI: 10.1002/glia.20892] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The urokinase plasminogen activator (uPA) receptor (uPAR) is a GPI-linked cell surface protein that facilitates focused plasmin proteolytic activity at the cell surface. uPAR has been detected in macrophages infiltrating the central nervous system (CNS) and soluble uPAR has been detected in the cerebrospinal fluid during a number of CNS pathologies. However, its expression by resident microglial cells in vivo remains uncertain. In this work, we aimed to elucidate the murine CNS expression of uPAR and uPA as well as that of tissue plasminogen activator and plasminogen activator inhibitor 1 (PAI-1) during insults generating distinct and well-characterized inflammatory responses; acute intracerebral lipopolysaccharide (LPS), acute kainate-induced neurodegeneration, and chronic neurodegeneration induced by prion disease inoculation. All three insults induced marked expression of uPAR at both mRNA and protein level compared to controls (naïve, saline, or control inoculum-injected). uPAR expression was microglial in all cases. Conversely, uPA transcription and activity was only markedly increased during chronic neurodegeneration. Dissociation of uPA and uPAR levels in acute challenges is suggestive of additional proteolysis-independent roles for uPAR. PAI-1 was most highly expressed upon LPS challenge, whereas tissue plasminogen activator mRNA was constitutively present and less responsive to all insults studied. These data are novel and suggest much wider involvement of the uPAR/uPA system in CNS function and pathology than previously supposed.
Collapse
|
42
|
Cabrera V, Cantú D, Ramos E, Vanoye-Carlo A, Cerbón M, Morales T. Lactation is a natural model of hippocampus neuroprotection against excitotoxicity. Neurosci Lett 2009; 461:136-9. [PMID: 19539698 DOI: 10.1016/j.neulet.2009.06.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Revised: 06/02/2009] [Accepted: 06/12/2009] [Indexed: 02/02/2023]
Abstract
Lactation is a temporary but complex physiological condition in which hormones and neurogenic stimulation from suckling cause maternal brain plasticity. It has been shown that lactation prevents cell damage induced by excitotoxicity in the dorsal hippocampus of the dam after peripheral administration of kainic acid (KA). The aim of this study was to determine whether lactation protects the maternal hippocampus against damage induced by intracerebral application (ICV) of KA and if lactation decreases, or only delays, this damaging effect of KA. Cell damage was assessed by Fluoro-Jade C staining in the hippocampus of virgin and lactating rats 24 or 72 h after ICV KA. Lactation prevented cell damage of the pyramidal layers of the hippocampus (CA1, CA3, and CA4), as compared to virgin rats. The longer period of KA exposure increased the difference in cell damage between these two conditions. The present results confirm that lactation is a natural model for neuroprotection, since it effectively prevents acute and chronic cell damage of the hippocampus induced by exposure to KA.
Collapse
Affiliation(s)
- Verónica Cabrera
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro 76230, Mexico
| | | | | | | | | | | |
Collapse
|
43
|
Huang L, Smith A, Cummings P, Kendall EJ, Obenaus A. Neuroimaging assessment of memory-related brain structures in a rat model of acute space-like radiation. J Magn Reson Imaging 2009; 29:785-92. [PMID: 19306400 DOI: 10.1002/jmri.21661] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
PURPOSE To investigate the acute effects on the central nervous system (CNS) of (56)Fe radiation, a component of high-energy charged particles (HZE) in space radiation, using quantitative magnetic resonance imaging (MRI) noninvasively. MATERIALS AND METHODS Sprague-Dawley rats were exposed to whole-brain (56)Fe (0, 1, 2, and 4 Gy). At 1 week postirradiation, MRI scans were made using T2-weighted (T2WI), diffusion-weighted (DWI), and contrast enhanced T1-(CET1) imaging. T2 relaxation time and apparent diffusion coefficient (ADC) values were obtained from memory-related brain regions of interest (ROIs). Histopathology was correlated using ex vivo tissues. RESULTS No overt abnormalities were visualized using T2WI and DWI at 1 week postradiation. CET1 values did not differ significantly between the irradiated and control animals. Compared to 0 Gy, there were significant prolongations in T2 values and reductions in ADC after irradiation. In the absence of evident neuronal pathology, immunohistochemistry revealed astrocytic activation in 4 Gy animals. CONCLUSION At 1 week after whole-brain (56)Fe exposure, T2 and ADC values can differentiate radiosensitivity in regions critical for hippocampal-related memory. MRI may provide noninvasive assessment of the initial molecular/cellular disturbances in vivo after HZE irradiation.
Collapse
Affiliation(s)
- Lei Huang
- Department of Radiation Medicine, Loma Linda University School of Medicine, Loma Linda, California, USA
| | | | | | | | | |
Collapse
|
44
|
Chen ZL, Yu H, Yu WM, Pawlak R, Strickland S. Proteolytic fragments of laminin promote excitotoxic neurodegeneration by up-regulation of the KA1 subunit of the kainate receptor. ACTA ACUST UNITED AC 2009; 183:1299-1313. [PMID: 19114596 PMCID: PMC2606967 DOI: 10.1083/jcb.200803107] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Degradation of the extracellular matrix (ECM) protein laminin contributes to excitotoxic cell death in the hippocampus, but the mechanism of this effect is unknown. To study this process, we disrupted laminin gamma1 (lamgamma1) expression in the hippocampus. Lamgamma1 knockout (KO) and control mice had similar basal expression of kainate (KA) receptors, but the lamgamma1 KO mice were resistant to KA-induced neuronal death. After KA injection, KA1 subunit levels increased in control mice but were unchanged in lamgamma1 KO mice. KA1 levels in tissue plasminogen activator (tPA)-KO mice were also unchanged after KA, indicating that both tPA and laminin were necessary for KA1 up-regulation after KA injection. Infusion of plasmin-digested laminin-1 into the hippocampus of lamgamma1 or tPA KO mice restored KA1 up-regulation and KA-induced neuronal degeneration. Interfering with KA1 function with a specific anti-KA1 antibody protected against KA-induced neuronal death both in vitro and in vivo. These results demonstrate a novel pathway for neurodegeneration involving proteolysis of the ECM and KA1 KA receptor subunit up-regulation.
Collapse
Affiliation(s)
- Zu-Lin Chen
- Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY 10065, USA
| | | | | | | | | |
Collapse
|
45
|
Markus T, Cronberg T, Cilio C, Pronk C, Wieloch T, Ley D. Tumor necrosis factor receptor-1 is essential for LPS-induced sensitization and tolerance to oxygen-glucose deprivation in murine neonatal organotypic hippocampal slices. J Cereb Blood Flow Metab 2009; 29:73-86. [PMID: 18728678 DOI: 10.1038/jcbfm.2008.90] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Inflammation and ischemia have a synergistic damaging effect in the immature brain. The role of tumor necrosis factor (TNF) receptors 1 and 2 in lipopolysaccharide (LPS)-induced sensitization and tolerance to oxygen-glucose deprivation (OGD) was evaluated in neonatal murine hippocampal organotypic slices. Hippocampal slices from balb/c, C57BL/6 TNFR1(-/-), TNFR2(-/-), and wild-type (WT) mice obtained at P6 were grown in vitro for 9 days. Preexposure to LPS immediately before OGD increased propidium iodide-determined cell death in regions CA1, CA3, and dentate gyrus from 4 up to 48 h after OGD (P<0.001). Extending the time interval between LPS exposure and OGD to 72 h resulted in tolerance, that is reduced neuronal cell death after OGD (P<0.05). Slices from TNFR1(-/-) mice showed neither LPS-induced sensitization nor LPS-induced tolerance to OGD, whereas both effects were present in slices from TNFR2(-/-) and WT mice. Cytokine secretion (TNFalpha and interleukin-6) during LPS exposure was decreased in TNFR1(-/-) slices and increased in TNFR2(-/-) as compared with WT slices. We conclude that LPS induces sensitization or tolerance to OGD depending on the time interval between exposure to LPS and OGD in murine hippocampal slice cultures. Both paradigms are dependent on signaling through TNFR1.
Collapse
Affiliation(s)
- Tina Markus
- Department of Pediatrics, Lund University, Lund, Sweden.
| | | | | | | | | | | |
Collapse
|
46
|
Cho IH, Hong J, Suh EC, Kim JH, Lee H, Lee JE, Lee S, Kim CH, Kim DW, Jo EK, Lee KE, Karin M, Lee SJ. Role of microglial IKKbeta in kainic acid-induced hippocampal neuronal cell death. ACTA ACUST UNITED AC 2008; 131:3019-33. [PMID: 18819987 DOI: 10.1093/brain/awn230] [Citation(s) in RCA: 136] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Microglial cells are activated during excitotoxin-induced neurodegeneration. However, the in vivo role of microglia activation in neurodegeneration has not yet been fully elucidated. To this end, we used Ikkbeta conditional knockout mice (LysM-Cre/Ikkbeta(F/F)) in which the Ikkbeta gene is specifically deleted in cells of myeloid lineage, including microglia, in the CNS. This deletion reduced IkappaB kinase (IKK) activity in cultured primary microglia by up to 40% compared with wild-type (Ikkbeta(F/F)), and lipopolysaccharide-induced proinflammatory gene expression was also compromised. Kainic acid (KA)-induced hippocampal neuronal cell death was reduced by 30% in LysM-Cre/Ikkbeta(F/F) mice compared with wild-type mice. Reduced neuronal cell death was accompanied by decreased KA-induced glial cell activation and subsequent expression of proinflammatory genes such as tumour necrosis factor (TNF)-alpha and interleukin (IL)-1beta. Similarly, neurons in organotypic hippocampal slice cultures (OHSCs) from LysM-Cre/Ikkbeta(F/F) mouse brain were less susceptible to KA-induced excitotoxicity compared with wild-type OHSCs, due in part to decreased TNF-alpha and IL-1beta expression. Based on these data, we concluded that IKK/nuclear factor-kappaB dependent microglia activation contributes to KA-induced hippocampal neuronal cell death in vivo through induction of inflammatory mediators.
Collapse
Affiliation(s)
- Ik-Hyun Cho
- Program in Molecular and Cellular Neuroscience, DRI, and Department of Oral Physiology, School of Dentistry, Seoul National University, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Gröticke I, Hoffmann K, Löscher W. Behavioral alterations in a mouse model of temporal lobe epilepsy induced by intrahippocampal injection of kainate. Exp Neurol 2008; 213:71-83. [DOI: 10.1016/j.expneurol.2008.04.036] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2008] [Revised: 04/29/2008] [Accepted: 04/30/2008] [Indexed: 10/22/2022]
|
48
|
Kim GH, Kellner CP, Hahn DK, Desantis BM, Musabbir M, Starke RM, Rynkowski M, Komotar RJ, Otten ML, Sciacca R, Schmidt JM, Mayer SA, Connolly ES. Monocyte chemoattractant protein–1 predicts outcome and vasospasm following aneurysmal subarachnoid hemorrhage. J Neurosurg 2008; 109:38-43. [DOI: 10.3171/jns/2008/109/7/0038] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Object
Despite efforts to elucidate both the molecular mechanism and the clinical predictors of vasospasm after aneurysmal subarachnoid hemorrhage (ASAH), its pathogenesis remains unclear. Monocyte chemoattractant protein–1 (MCP-1) is a chemokine that has been firmly implicated in the pathophysiology of vasospasm and in neural tissue injury following focal ischemia in both animal models and human studies. The authors hypothesized that MCP-1 would be found in increased concentrations in the blood and cerebrospinal fluid (CSF) of patients with ASAH and would correlate with both outcome and the occurrence of vasospasm.
Methods
Seventy-seven patients who presented with ASAH were prospectively enrolled in this study between July 2001 and May 2002. Using an enzyme-linked immunosorbent assay, MCP-1 levels were measured in serum daily and in CSF when available. The mean serum and CSF MCP-1 concentrations were calculated for each patient throughout the entire hospital stay. Neurological outcome was evaluated at discharge or 14 days posthemorrhage using the modified Rankin Scale. Vasospasm was evaluated on angiography.
Results
The serum MCP-1 concentrations correlated with negative outcome such that a 10% increase in concentration predicted a 25% increase in the probability of a poor outcome, whereas the serum MCP-1 levels did not correlate with vasospasm. Concentrations of MCP-1 in the CSF, however, proved to be significantly higher in patients with angiographically demonstrated vasospasm.
Conclusions
These findings suggest a role for MCP-1 in neurological injury and imply that it may act as a biomarker of poor outcome in the serum and of vasospasm in the CSF.
Collapse
|
49
|
Shin EJ, Ko KH, Kim WK, Chae JS, Yen TPH, Kim HJ, Wie MB, Kim HC. Role of glutathione peroxidase in the ontogeny of hippocampal oxidative stress and kainate seizure sensitivity in the genetically epilepsy-prone rats. Neurochem Int 2008; 52:1134-47. [DOI: 10.1016/j.neuint.2007.12.003] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2007] [Revised: 11/20/2007] [Accepted: 12/04/2007] [Indexed: 01/30/2023]
|
50
|
Carare RO, Bernardes-Silva M, Newman TA, Page AM, Nicoll JAR, Perry VH, Weller RO. Solutes, but not cells, drain from the brain parenchyma along basement membranes of capillaries and arteries: significance for cerebral amyloid angiopathy and neuroimmunology. Neuropathol Appl Neurobiol 2008; 34:131-44. [PMID: 18208483 DOI: 10.1111/j.1365-2990.2007.00926.x] [Citation(s) in RCA: 452] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
UNLABELLED Elimination of interstitial fluid and solutes plays a role in homeostasis in the brain, but the pathways are unclear. Previous work suggests that interstitial fluid drains along the walls of arteries. AIMS to define the pathways within the walls of capillaries and arteries for drainage of fluid and solutes out of the brain. METHODS Fluorescent soluble tracers, dextran (3 kDa) and ovalbumin (40 kDa), and particulate fluospheres (0.02 microm and 1.0 microm in diameter) were injected into the corpus striatum of mice. Brains were examined from 5 min to 7 days by immunocytochemistry and confocal microscopy. RESULTS soluble tracers initially spread diffusely through brain parenchyma and then drain out of the brain along basement membranes of capillaries and arteries. Some tracer is takenf up by vascular smooth muscle cells and by perivascular macrophages. No perivascular drainage was observed when dextran was injected into mouse brains following cardiac arrest. Fluospheres expand perivascular spaces between vessel walls and surrounding brain, are ingested by perivascular macrophages but do not appear to leave the brain even following an inflammatory challenge with lipopolysaccharide or kainate. CONCLUSIONS capillary and artery basement membranes act as 'lymphatics of the brain' for drainage of fluid and solutes; such drainage appears to require continued cardiac output as it ceases following cardiac arrest. This drainage pathway does not permit migration of cells from brain parenchyma to the periphery. Amyloid-beta is deposited in basement membrane drainage pathways in cerebral amyloid angiopathy, and may impede elimination of amyloid-beta and interstitial fluid from the brain in Alzheimer's disease. Soluble antigens, but not cells, drain from the brain by perivascular pathways. This atypical pattern of drainage may contribute to partial immune privilege of the brain and play a role in neuroimmunological diseases such as multiple sclerosis.
Collapse
Affiliation(s)
- R O Carare
- Clinical Neurosciences, University of Southampton, UK
| | | | | | | | | | | | | |
Collapse
|