51
|
Lem DW, Gierhart DL, Davey PG. Carotenoids in the Management of Glaucoma: A Systematic Review of the Evidence. Nutrients 2021; 13:1949. [PMID: 34204051 PMCID: PMC8228567 DOI: 10.3390/nu13061949] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/01/2021] [Accepted: 06/04/2021] [Indexed: 12/23/2022] Open
Abstract
Primary open-angle glaucoma (POAG) remains a leading cause of irreversible blindness globally. Recent evidence further substantiates sustained oxidative stress, and compromised antioxidant defenses are key drivers in the onset of glaucomatous neurodegeneration. Overwhelming oxidative injury is likely attributed to compounding mitochondrial dysfunction that worsens with age-related processes, causing aberrant formation of free radical species. Thus, a compromised systemic antioxidant capacity exacerbates further oxidative insult in glaucoma, leading to apoptosis, neuroinflammation, and subsequent tissue injury. The purpose of this systematic review is to investigate the neuroprotective benefits of the macular carotenoids lutein, zeaxanthin, and meso-zeaxanthin on glaucomatous neurodegeneration for the purpose of adjunctive nutraceutical treatment in glaucoma. A comprehensive literature search was conducted in three databases (PubMed, Cochrane Library, and Web of Science) and 20 records were identified for screening. Lutein demonstrated enhanced neuroprotection on retinal ganglion cell survival and preserved synaptic activity. In clinical studies, a protective trend was seen with greater dietary consumption of carotenoids and risk of glaucoma, while greater carotenoid levels in macular pigment were largely associated with improved visual performance in glaucomatous eyes. The data suggest that carotenoid vitamin therapy exerts synergic neuroprotective benefits and has the capacity to serve adjunctive therapy in the management of glaucoma.
Collapse
Affiliation(s)
- Drake W. Lem
- College of Optometry, Western University of Health Sciences, 309 E Second St, Pomona, CA 91766, USA;
| | | | - Pinakin Gunvant Davey
- College of Optometry, Western University of Health Sciences, 309 E Second St, Pomona, CA 91766, USA;
| |
Collapse
|
52
|
Yoon Y, Voloudakis G, Doran N, Zhang E, Dimovasili C, Chen L, Shao Z, Darmanis S, Tang C, Tang J, Wang VX, Hof PR, Robakis NK, Georgakopoulos A. PS1 FAD mutants decrease ephrinB2-regulated angiogenic functions, ischemia-induced brain neovascularization and neuronal survival. Mol Psychiatry 2021; 26:1996-2012. [PMID: 32541930 PMCID: PMC7736163 DOI: 10.1038/s41380-020-0812-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/29/2020] [Accepted: 06/04/2020] [Indexed: 12/12/2022]
Abstract
Microvascular pathology and ischemic lesions contribute substantially to neuronal dysfunction and loss that lead to Alzheimer disease (AD). To facilitate recovery, the brain stimulates neovascularization of damaged tissue via sprouting angiogenesis, a process regulated by endothelial cell (EC) sprouting and the EphB4/ephrinB2 system. Here, we show that in cultures of brain ECs, EphB4 stimulates the VE-cadherin/Rok-α angiogenic complexes known to mediate sprouting angiogenesis. Importantly, brain EC cultures expressing PS1 FAD mutants decrease the EphB4-stimulated γ-secretase cleavage of ephrinB2 and reduce production of the angiogenic peptide ephrinB2/CTF2, the VE-cadherin angiogenic complexes and EC sprouting and tube formation. These data suggest that FAD mutants may attenuate ischemia-induced brain angiogenesis. Supporting this hypothesis, ischemia-induced VE-cadherin angiogenic complexes, levels of neoangiogenesis marker Endoglin, vascular density, and cerebral blood flow recovery, are all decreased in brains of mouse models expressing PS1 FAD mutants. Ischemia-induced brain neuronal death and cognitive deficits also increase in these mice. Furthermore, a small peptide comprising the C-terminal sequence of peptide ephrinB2/CTF2 rescues angiogenic functions of brain ECs expressing PS1 FAD mutants. Together, our data show that PS1 FAD mutations impede the EphB4/ephrinB2-mediated angiogenic functions of ECs and impair brain neovascularization, neuronal survival and cognitive recovery following ischemia. Furthermore, our data reveal a novel brain angiogenic mechanism targeted by PS1 FAD mutants and a potential therapeutic target for ischemia-induced neurodegeneration. Importantly, FAD mutant effects occur in absence of neuropathological hallmarks of AD, supporting that such hallmarks may form downstream of mutant effects on neoangiogenesis and neuronal survival.
Collapse
Affiliation(s)
- YoneJung Yoon
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Georgios Voloudakis
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nathan Doran
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emily Zhang
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christina Dimovasili
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lei Chen
- Department of Physiology, Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, 40536, USA
| | - Zhiping Shao
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Spyros Darmanis
- Departments of Bioengineering and Applied Physics, Stanford University and Chan Zuckerberg Biohub, Stanford, CA, 94305, USA
| | - Cheuk Tang
- Department of Radiology, Neuroscience and Psychiatry Translational and Molecular Imaging Institute at Mount Sinai, New York, NY, USA
| | - Jun Tang
- Department of Radiology, Neuroscience and Psychiatry Translational and Molecular Imaging Institute at Mount Sinai, New York, NY, USA
| | - Victoria X Wang
- Department of Radiology, Translational and Molecular Imaging Institute at Mount Sinai, New York, NY, USA
| | - Patrick R Hof
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nikolaos K Robakis
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Anastasios Georgakopoulos
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
53
|
Wang P, Cui Y, Ren Q, Yan B, Zhao Y, Yu P, Gao G, Shi H, Chang S, Chang YZ. Mitochondrial ferritin attenuates cerebral ischaemia/reperfusion injury by inhibiting ferroptosis. Cell Death Dis 2021; 12:447. [PMID: 33953171 PMCID: PMC8099895 DOI: 10.1038/s41419-021-03725-5] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 02/08/2023]
Abstract
Ischaemic stroke is becoming the most common cerebral disease in aging populations, but the underlying molecular mechanism of the disease has not yet been fully elucidated. Increasing evidence has indicated that an excess of iron contributes to brain damage in cerebral ischaemia/reperfusion (I/R) injury. Although mitochondrial ferritin (FtMt) plays a critical role in iron homeostasis, the molecular function of FtMt in I/R remains unknown. We herein report that FtMt levels are upregulated in the ischaemic brains of mice. Mice lacking FtMt experience more severe brain damage and neurological deficits, accompanied by typical molecular features of ferroptosis, including increased lipid peroxidation and disturbed glutathione (GSH) after cerebral I/R. Conversely, FtMt overexpression reverses these changes. Further investigation shows that Ftmt ablation promotes I/R-induced inflammation and hepcidin-mediated decreases in ferroportin1, thus markedly increasing total and chelatable iron. The elevated iron consequently facilitates ferroptosis in the brain of I/R. In brief, our results provide evidence that FtMt plays a critical role in protecting against cerebral I/R-induced ferroptosis and subsequent brain damage, thus providing a new potential target for the treatment/prevention of ischaemic stroke.
Collapse
Affiliation(s)
- Peina Wang
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Ministry of Education Key Laboratory of Molecular and Cellular Biology, College of Life Science, Hebei Normal University, 050024, Shijiazhuang, Hebei Province, China
| | - Yanmei Cui
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Ministry of Education Key Laboratory of Molecular and Cellular Biology, College of Life Science, Hebei Normal University, 050024, Shijiazhuang, Hebei Province, China
| | - Qianqian Ren
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Ministry of Education Key Laboratory of Molecular and Cellular Biology, College of Life Science, Hebei Normal University, 050024, Shijiazhuang, Hebei Province, China
| | - Bingqi Yan
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Ministry of Education Key Laboratory of Molecular and Cellular Biology, College of Life Science, Hebei Normal University, 050024, Shijiazhuang, Hebei Province, China
| | - Yashuo Zhao
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Ministry of Education Key Laboratory of Molecular and Cellular Biology, College of Life Science, Hebei Normal University, 050024, Shijiazhuang, Hebei Province, China
- Scientific Research Center, Hebei University of Chinese Medicine, 050200, Shijiazhuang, Hebei Province, China
| | - Peng Yu
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Ministry of Education Key Laboratory of Molecular and Cellular Biology, College of Life Science, Hebei Normal University, 050024, Shijiazhuang, Hebei Province, China
| | - Guofen Gao
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Ministry of Education Key Laboratory of Molecular and Cellular Biology, College of Life Science, Hebei Normal University, 050024, Shijiazhuang, Hebei Province, China
| | - Honglian Shi
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, 1251 Wescoe Hall Drive, Malott Hall 5044, Lawrence, KS, 66045, USA
| | - Shiyang Chang
- College of basic medicine, Hebei Medical University, 050017, Shijiazhuang, Hebei Province, China.
| | - Yan-Zhong Chang
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Ministry of Education Key Laboratory of Molecular and Cellular Biology, College of Life Science, Hebei Normal University, 050024, Shijiazhuang, Hebei Province, China.
| |
Collapse
|
54
|
Usui T, Macleod MR, McCann SK, Senior AM, Nakagawa S. Meta-analysis of variation suggests that embracing variability improves both replicability and generalizability in preclinical research. PLoS Biol 2021; 19:e3001009. [PMID: 34010281 PMCID: PMC8168858 DOI: 10.1371/journal.pbio.3001009] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 06/01/2021] [Accepted: 05/04/2021] [Indexed: 01/11/2023] Open
Abstract
The replicability of research results has been a cause of increasing concern to the scientific community. The long-held belief that experimental standardization begets replicability has also been recently challenged, with the observation that the reduction of variability within studies can lead to idiosyncratic, lab-specific results that cannot be replicated. An alternative approach is to, instead, deliberately introduce heterogeneity, known as "heterogenization" of experimental design. Here, we explore a novel perspective in the heterogenization program in a meta-analysis of variability in observed phenotypic outcomes in both control and experimental animal models of ischemic stroke. First, by quantifying interindividual variability across control groups, we illustrate that the amount of heterogeneity in disease state (infarct volume) differs according to methodological approach, for example, in disease induction methods and disease models. We argue that such methods may improve replicability by creating diverse and representative distribution of baseline disease state in the reference group, against which treatment efficacy is assessed. Second, we illustrate how meta-analysis can be used to simultaneously assess efficacy and stability (i.e., mean effect and among-individual variability). We identify treatments that have efficacy and are generalizable to the population level (i.e., low interindividual variability), as well as those where there is high interindividual variability in response; for these, latter treatments translation to a clinical setting may require nuance. We argue that by embracing rather than seeking to minimize variability in phenotypic outcomes, we can motivate the shift toward heterogenization and improve both the replicability and generalizability of preclinical research.
Collapse
Affiliation(s)
- Takuji Usui
- Evolution and Ecology Research Centre and School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, Australia
- The Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, Australia
| | - Malcolm R. Macleod
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Sarah K. McCann
- QUEST Center for Transforming Biomedical Research, Berlin Institute of Health (BIH), Berlin, Germany
- Charité—Universitätsmedizin Berlin Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Alistair M. Senior
- The Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, Australia
| | - Shinichi Nakagawa
- Evolution and Ecology Research Centre and School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, Australia
- The Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, Australia
| |
Collapse
|
55
|
Qin ZS, Zheng Y, Zhou XD, Shi DD, Cheng D, Shek CS, Zhan CS, Zhang ZJ. Shexiang Baoxin Pill, a Proprietary Multi-Constituent Chinese Medicine, Prevents Locomotor and Cognitive Impairment Caused by Brain Ischemia and Reperfusion Injury in Rats: A Potential Therapy for Neuropsychiatric Sequelae of Stroke. Front Pharmacol 2021; 12:665456. [PMID: 33986688 PMCID: PMC8111446 DOI: 10.3389/fphar.2021.665456] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/14/2021] [Indexed: 12/15/2022] Open
Abstract
Ischemic stroke is a common type of cerebrovascular event and also the leading cause of disability. Post-stroke cognitive impairment occurs frequently in stroke survivors. Shexiang Baoxin Pill (SBP) is a proprietary Chinese medicine, initially used to treat cardiovascular diseases. Herein, we aim to explore the effects of SBP on oxygen glucose deprivation and reoxygenation (OGD/R) in neuronal cells (CATH.a) and cerebral ischemia/reperfusion injury induced post-stroke cognitive impairment in middle cerebral artery occlusion (MCAO) rat model. MCAO rats received two doses of oral SBP treatment (28 or 56 mg/kg) after 1 h of operation and once daily for 2 weeks continuously. Behavioral tests, immunoblotting, and immunofluorescence were examined after 14 days. Current data suggest that SBP enhanced cell viability and downregulated apoptosis via activating the PI3K/Akt signaling pathway in CATH. a cells. Furthermore, 14 days of SBP treatment promoted the recovery of learning and locomotor function in the MCAO rats. SBP up-regulated the expression of p-Akt, p-GSK3β, as well as the expression of NMDAR1, PSD-95, and AMPAR. Also, SBP down-regulated the expression of p-CaMKII. These results indicated that long-term SBP treatment might be a potential option for cognitive impairment induced by the ischemic stroke.
Collapse
Affiliation(s)
- Zong-Shi Qin
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yu Zheng
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xi-Dan Zhou
- The Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Dong-Dong Shi
- Shanghai Mental Health Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Dan Cheng
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chun Shum Shek
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chang-Sen Zhan
- Shanghai Hutchison Pharmaceuticals Ltd., Shanghai, China.,Shanghai Engineering Research Center for Innovation of Solid Preparation of TCM, Shanghai, China
| | - Zhang-Jin Zhang
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
56
|
Mavroudakis L, Stevens SL, Duncan KD, Stenzel-Poore MP, Laskin J, Lanekoff I. CpG preconditioning reduces accumulation of lysophosphatidylcholine in ischemic brain tissue after middle cerebral artery occlusion. Anal Bioanal Chem 2021; 413:2735-2745. [PMID: 33078250 PMCID: PMC8007524 DOI: 10.1007/s00216-020-02987-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/08/2020] [Accepted: 10/05/2020] [Indexed: 01/01/2023]
Abstract
Ischemic stroke is one of the major causes of death and permanent disability in the world. However, the molecular mechanisms surrounding tissue damage are complex and further studies are needed to gain insights necessary for development of treatment. Prophylactic treatment by administration of cytosine-guanine (CpG) oligodeoxynucleotides has been shown to provide neuroprotection against anticipated ischemic injury. CpG binds to Toll-like receptor 9 (TLR9) causing initialization of an inflammatory response that limits visible ischemic damages upon subsequent stroke. Here, we use nanospray desorption electrospray ionization (nano-DESI) mass spectrometry imaging (MSI) to characterize molecular effects of CpG preconditioning prior to middle cerebral artery occlusion (MCAO) and reperfusion. By doping the nano-DESI solvent with appropriate internal standards, we can study and compare distributions of phosphatidylcholine (PC) and lysophosphatidylcholine (LPC) in the ischemic hemisphere of the brain despite the large changes in alkali metal abundances. Our results show that CpG preconditioning not only reduces the infarct size but it also decreases the degradation of PC and accumulation of LPC species, which indicates reduced cell membrane breakdown and overall ischemic damage. Our findings show that molecular mechanisms of PC degradation are intact despite CpG preconditioning but that these are limited due to the initialized inflammatory response.
Collapse
Affiliation(s)
| | - Susan L Stevens
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Kyle D Duncan
- Department of Chemistry - BMC, Uppsala University, 75123, Uppsala, Sweden
| | - Mary P Stenzel-Poore
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Julia Laskin
- Physical Sciences Division, Pacific Northwest National Laboratory, Richland, WA, 99354, USA
- Department of Chemistry, Purdue University, West Lafayette, IN, 47907, USA
| | - Ingela Lanekoff
- Department of Chemistry - BMC, Uppsala University, 75123, Uppsala, Sweden.
| |
Collapse
|
57
|
Singh D, Wasan H, Reeta KH. Preclinical Stroke Research and Translational Failure: A Bird's Eye View on Preventable Variables. Cell Mol Neurobiol 2021; 42:2003-2017. [PMID: 33786698 DOI: 10.1007/s10571-021-01083-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 03/18/2021] [Indexed: 02/08/2023]
Abstract
Despite achieving remarkable success in understanding the cellular, molecular and pathophysiological aspects of stroke, translation from preclinical research has always remained an area of debate. Although thousands of experimental compounds have been reported to be neuro-protective, their failures in clinical setting have left the researchers and stakeholders in doldrums. Though the failures described have been excruciating, they also give us a chance to refocus on the shortcomings. For better translational value, evidences from preclinical studies should be robust and reliable. Preclinical study design has a plethora of variables affecting the study outcome. Hence, this review focusses on the factors to be considered for a well-planned preclinical study while adhering to guidelines with emphasis on the study design, commonly used animal models, their limitations with special attention on various preventable attritions including comorbidities, aged animals, time of dosing, outcome measures and physiological variables along with the concept of multicentric preclinical randomized controlled trials. Here, we provide an overview of a panorama of practical aspects, which could be implemented, so that a well-defined preclinical study would result in a neuro-protectant with better translational value.
Collapse
Affiliation(s)
- Devendra Singh
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Himika Wasan
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - K H Reeta
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India.
| |
Collapse
|
58
|
Yang H, Zhang Y, Chen H, Zhu Y, Li Y, Ouyang F, Chu L, Liu D. Mir-184 Contributes to Brain Injury Through Targeting PPAP2B Following Ischemic Stroke in Male Rats. Front Mol Neurosci 2021; 14:613887. [PMID: 33833666 PMCID: PMC8021718 DOI: 10.3389/fnmol.2021.613887] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 02/10/2021] [Indexed: 12/05/2022] Open
Abstract
Our previous study revealed that miR-184 expression is significantly altered in the brain following ischemic stroke in rats. However, it is unknown whether this alteration in miR-184 expression contributes to brain injury after ischemic stroke. Here, we aim to address the potential of miR-184 to impact nerve injury following ischemia and reperfusion. Rats received ICV injection of miR-184 adenovirus or empty vector and were subjected to right middle cerebral artery occlusion (MCAO) to establish an ischemic stroke model. We cultured SH-SY5Y cells under oxygen-glucose deprivation/reoxygenation (OGD/R) and transfected them with miR-184 lentivirus to explore the primary mechanisms. To evaluate miR-184 expression, neurological function deficits, the cerebral infarct volume, cell viability, and apoptosis, qRT-PCR analysis of miR-184 expression, the modified neurological severity score (mNSS) system, TTC staining, the CCK-8 assay, flow cytometry, and dual-luciferase reporter assays were utilized. We found that miR-184 expression was downregulated and that the cerebral infarct volume and mNSSs were increased following ischemic stroke; however, increasing the level of miR-184 alleviated brain damage. Overexpression of miR-184 resulted in increased viability and reduced apoptosis of SH-SY5Y cells following OGD/R in vitro. We identified the phosphatidic acid phosphatase type 2B (PPAP2B) gene as a direct target gene of miR-184. In summary, our results reveal that attenuation of miR-184 levels in ischemic stroke contributes to ischemic injury through targeting PPAP2B mRNA-mediated apoptosis, which may be a promising therapeutic target for ischemic stroke.
Collapse
Affiliation(s)
- Huajun Yang
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China.,Department of Respiratory Medicine, The First People's Hospital of Zunyi, The Third Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yifan Zhang
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China
| | - Hongqun Chen
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China
| | - Yingwu Zhu
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China
| | - Yuan Li
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China
| | - Fu Ouyang
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China
| | - Lan Chu
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, China
| | - Daishun Liu
- Department of Respiratory Medicine, The First People's Hospital of Zunyi, The Third Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
59
|
Zheng ZY, Lu G, Xiong ZQ, Leung CK, Su XW, Li T, Poon WS, Chan WY, Wong GKC. Integrated analysis of gait parameters and gene expression profiles in a murine model of subarachnoid hemorrhage. GENES BRAIN AND BEHAVIOR 2021; 20:e12728. [PMID: 33641236 DOI: 10.1111/gbb.12728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 02/02/2021] [Accepted: 02/26/2021] [Indexed: 11/28/2022]
Abstract
Gait analysis has been widely used to examine the behavioral presentation of numerous neurological disorders. Thorough murine model evaluation of the subarachnoid hemorrhage (SAH)-associated gait deficits is missing. This study measures gait deficits using a clinically relevant murine model of SAH to examine associations between gait variability and SAH-associated gene expressions. A total of 159 dynamic and static gait parameters from the endovascular perforation murine model for simulating clinical human SAH were determined using the CatWalk system. Eighty gait parameters and the mRNA expression levels of 35 of the 88 SAH-associated genes were differentially regulated in the diseased models. Totals of 42 and 38 gait parameters correlated with the 35 SAH-associated genes positively and negatively with Pearson's correlation coefficients of >0.7 and <-0.7, respectively. p-SP1453 expression in the motor cortex in SAH animal models displays a significant correlation with a subset of gait parameters associated with muscular strength and coordination of limb movements. Our data highlights a strong correlation between gait variability and SAH-associated gene expression. p-SP1453 expression could act as a biomarker to monitor SAH pathological development and a therapeutic target for SAH.
Collapse
Affiliation(s)
- Zhi Yuan Zheng
- Division of Neurosurgery, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Gang Lu
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhi Qiang Xiong
- Bioinformatics Unit, SDIVF R&D Centre, Hong Kong Science and Technology Parks, Hong Kong, China
| | - Chi Kwan Leung
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xian Wei Su
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Tu Li
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Wai Sang Poon
- Division of Neurosurgery, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Wai Yee Chan
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - George Kwok Chu Wong
- Division of Neurosurgery, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
60
|
Trabolsi C, Takash Chamoun W, Hijazi A, Nicoletti C, Maresca M, Nasser M. Study of Neuroprotection by a Combination of the Biological Antioxidant ( Eucalyptus Extract) and the Antihypertensive Drug Candesartan against Chronic Cerebral Ischemia in Rats. Molecules 2021; 26:839. [PMID: 33562701 PMCID: PMC7915443 DOI: 10.3390/molecules26040839] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 01/28/2021] [Accepted: 01/30/2021] [Indexed: 11/17/2022] Open
Abstract
Chronic cerebral ischemia with a notable long-term cessation of blood supply to the brain tissues leads to sensorimotor defects and short- and long-term memory problems. Neuroprotective agents are used in an attempt to save ischemic neurons from necrosis and apoptosis, such as the antioxidant agent Eucalyptus. Numerous studies have demonstrated the involvement of the renin-angiotensin system in the initiation and progression of cardiovascular and neurodegenerative diseases. Candesartan is a drug that acts as an angiotensin II receptor 1 blocker. We established a rat model exhibiting sensorimotor and cognitive impairments due to chronic cerebral ischemia induced by the ligation of the right common carotid artery. Wistar male rats were randomly divided into five groups: Sham group, Untreated Ligated group, Ischemic group treated with Eucalyptus (500 mg/kg), Ischemic group treated with Candesartan (0.5 mg/kg), and Ischemic group treated with a combination of Eucalyptus and Candesartan. To evaluate the sensorimotor disorders, we performed the beam balance test, the beam walking test, and the modified sticky test. Moreover, the object recognition test and the Morris water maze test were performed to assess the memory disorders of the rats. The infarct rat brain regions were subsequently stained using the triphenyltetrazolium chloride staining technique. The rats in the Sham group had normal sensorimotor and cognitive functions without the appearance of microscopic ischemic brain lesions. In parallel, the untreated Ischemic group showed severe impaired neurological functions with the presence of considerable brain infarctions. The treatment of the Ischemic group with a combination of both Eucalyptus and Candesartan was more efficient in improving the sensorimotor and cognitive deficits (p < 0.001) than the treatment with Eucalyptus or Candesartan alone (p < 0.05), by the comparison to the non-treated Ischemic group. Our study shows that the combination of Eucalyptus and Candesartan could decrease ischemic brain injury and improve neurological outcomes.
Collapse
Affiliation(s)
- Christine Trabolsi
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut P.O. Box 6573/14, Lebanon; (C.T.); (W.T.C.)
- Rammal Hassan Rammal Research Laboratory, Physiotoxicity (PhyTox), Faculty of Sciences, Lebanese University, Beirut P.O. Box 6573/14, Lebanon
| | - Wafaa Takash Chamoun
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut P.O. Box 6573/14, Lebanon; (C.T.); (W.T.C.)
| | - Akram Hijazi
- Plateforme de recherche et d’analyse en sciences de l’environnement (EDST-PRASE), Beirut P.O. Box 6573/14, Lebanon;
| | - Cendrine Nicoletti
- Aix Marseille University, CNRS, Centrale Marseille, iSm2, 13397 Marseille, France;
| | - Marc Maresca
- Aix Marseille University, CNRS, Centrale Marseille, iSm2, 13397 Marseille, France;
| | - Mohamad Nasser
- Rammal Hassan Rammal Research Laboratory, Physiotoxicity (PhyTox), Faculty of Sciences, Lebanese University, Beirut P.O. Box 6573/14, Lebanon
- Plateforme de recherche et d’analyse en sciences de l’environnement (EDST-PRASE), Beirut P.O. Box 6573/14, Lebanon;
| |
Collapse
|
61
|
Xu ZY, Xu Y, Xie XF, Tian Y, Sui JH, Sun Y, Lin DS, Gao X, Peng C, Fan YJ. Anti-platelet aggregation of Panax notoginseng triol saponins by regulating GP1BA for ischemic stroke therapy. Chin Med 2021; 16:12. [PMID: 33468191 PMCID: PMC7816336 DOI: 10.1186/s13020-021-00424-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 01/08/2021] [Indexed: 12/20/2022] Open
Abstract
Background Panax notoginseng triol saponins (PTS) has been used clinically for ischemic stroke therapy (IST) in China for more than 17 years due to its anti-platelet aggregation and neuro-protective effects, but its mechanism of action is not fully understand. In this study, anti-platelet aggregation-related protein analysis and computer simulations of drug-protein binding interactions were performed to explore the mechanism of the effects of PTS against ischemic stroke in an ischemia reperfusion model. Methods Three oral doses of PTS were administered in a model of middle cerebral artery occlusion (MCAO) in rats. Panax notoginseng total saponins (PNS) and a combination of PTS and aspirin were chosen for comparison. To evaluate therapeutic effects and explore possible mechanisms of anti-platelet aggregation, we measured cerebral infarct size and water content in brain tissue, histomorphological changes, expression of related factors (such as arachidonic acid metabolites) and platelet receptors in serum, as well as the binding affinity of PTS for platelet adhesion receptors. Results Compared with PNS, PTS showed a stronger and more potent anti-platelet aggregation effect in MCAO model rats. The combination of PTS and aspirin could reduce adverse gastrointestinal effects by regulating the TXA2/PGI2 ratio. We demonstrated for the first time that PTS was able to regulate Glycoprotein Ib-α (GP1BA) in a model animal. The binding of ginsenoside Rg1 and GP1BA could form a stable structure. Moreover, PTS could reduce von Willebrand factor (VWF)-mediated platelet adhesion to damaged vascular endothelium, and thus enhance the probability of anti-platelet aggregation and anti-thrombosis under pathological conditions. Conclusions Our results showed that GP1BA was closely related to the anti-platelet aggregation action of PTS, which provided new scientific and molecular evidence for its clinical application.![]()
Collapse
Affiliation(s)
- Zhi-Yi Xu
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, 610064, Sichuan, China.,Chengdu Huasun Technology Group Inc., Ltd, Shuxin Avenue No.1168, Western Hi-tech Zone, Chengdu, 611731, Sichuan, China
| | - Yang Xu
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, 610064, Sichuan, China
| | - Xiao-Fang Xie
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Liutai Avenue NO. 1166, Wenjiang District, Chengdu, 611137, Sichuan, China
| | - Yin Tian
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Liutai Avenue NO. 1166, Wenjiang District, Chengdu, 611137, Sichuan, China
| | - Jun-Hui Sui
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, 610064, Sichuan, China
| | - Yong Sun
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, 610064, Sichuan, China
| | - Da-Sheng Lin
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Liutai Avenue NO. 1166, Wenjiang District, Chengdu, 611137, Sichuan, China.,Chengdu Huasun Technology Group Inc., Ltd, Shuxin Avenue No.1168, Western Hi-tech Zone, Chengdu, 611731, Sichuan, China
| | - Xing Gao
- Chengdu Huasun Technology Group Inc., Ltd, Shuxin Avenue No.1168, Western Hi-tech Zone, Chengdu, 611731, Sichuan, China
| | - Cheng Peng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Liutai Avenue NO. 1166, Wenjiang District, Chengdu, 611137, Sichuan, China.
| | - Yu-Jiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, 610064, Sichuan, China.
| |
Collapse
|
62
|
Hamdy N, Eide S, Sun HS, Feng ZP. Animal models for neonatal brain injury induced by hypoxic ischemic conditions in rodents. Exp Neurol 2020; 334:113457. [PMID: 32889009 DOI: 10.1016/j.expneurol.2020.113457] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 08/28/2020] [Accepted: 08/30/2020] [Indexed: 02/06/2023]
Abstract
Neonatal hypoxia-ischemia and resulting encephalopathies are of significant concern. Intrapartum asphyxia is a leading cause of neonatal death globally. Among surviving infants, there remains a high incidence of hypoxic-ischemic encephalopathy due to neonatal hypoxic-ischemic brain injury, manifesting as mild conditions including attention deficit hyperactivity disorder, and debilitating disorders such as cerebral palsy. Various animal models of neonatal hypoxic brain injury have been implemented to explore cellular and molecular mechanisms, assess the potential of novel therapeutic strategies, and characterize the functional and behavioural correlates of injury. Each of the animal models has individual advantages and limitations. The present review looks at several widely-used and alternative rodent models of neonatal hypoxia and hypoxia-ischemia; it highlights their strengths and limitations, and their potential for continued and improved use.
Collapse
Affiliation(s)
- Nancy Hamdy
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Sarah Eide
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Hong-Shuo Sun
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada; Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| | - Zhong-Ping Feng
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada.
| |
Collapse
|
63
|
He JW, Rabiller G, Nishijima Y, Akamatsu Y, Khateeb K, Yazdan-Shahmorad A, Liu J. Experimental cortical stroke induces aberrant increase of sharp-wave-associated ripples in the hippocampus and disrupts cortico-hippocampal communication. J Cereb Blood Flow Metab 2020; 40:1778-1796. [PMID: 31558106 PMCID: PMC7446570 DOI: 10.1177/0271678x19877889] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 07/22/2019] [Accepted: 07/25/2019] [Indexed: 11/16/2022]
Abstract
The functional consequences of ischemic stroke in the remote brain regions are not well characterized. The current study sought to determine changes in hippocampal oscillatory activity that may underlie the cognitive impairment observed following distal middle cerebral artery occlusion (dMCAO) without causing hippocampal structural damage. Local field potentials were recorded from the dorsal hippocampus and cortex in urethane-anesthetized rats with multichannel silicon probes during dMCAO and reperfusion, or mild ischemia induced by bilateral common carotid artery occlusion (CCAO). Bilateral change of brain state was evidenced by reduced theta/delta amplitude ratio and shortened high theta duration following acute dMCAO but not CCAO. An aberrant increase in the occurrence of sharp-wave-associated ripples (150-250 Hz), crucial for memory consolidation, was only detected after dMCAO reperfusion, coinciding with an increased occurrence of high-frequency discharges (250-450 Hz). dMCAO also significantly affected the modulation of gamma amplitude in the cortex coupled to hippocampal theta phase, although both hippocampal theta and gamma power were temporarily decreased during dMCAO. Our results suggest that MCAO may disrupt the balance between excitatory and inhibitory circuits in the hippocampus and alter the function of cortico-hippocampal network, providing a novel insight in how cortical stroke affects function in remote brain regions.
Collapse
Affiliation(s)
- Ji-Wei He
- Department of Neurological Surgery, UCSF, San Francisco, CA, USA
- Department of Neurological Surgery, SFVAMC, San Francisco, CA, USA
| | - Gratianne Rabiller
- Department of Neurological Surgery, UCSF, San Francisco, CA, USA
- Department of Neurological Surgery, SFVAMC, San Francisco, CA, USA
| | - Yasuo Nishijima
- Department of Neurological Surgery, UCSF, San Francisco, CA, USA
- Department of Neurological Surgery, SFVAMC, San Francisco, CA, USA
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yosuke Akamatsu
- Department of Neurological Surgery, UCSF, San Francisco, CA, USA
- Department of Neurological Surgery, SFVAMC, San Francisco, CA, USA
- Department of Neurosurgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Karam Khateeb
- Departments of Bioengineering and Electrical and Computer Engineering, University of Washington, Seattle, WA, USA
| | - Azadeh Yazdan-Shahmorad
- Departments of Bioengineering and Electrical and Computer Engineering, University of Washington, Seattle, WA, USA
- Center for Integrative Neuroscience and Department of Physiology, University of California, San Francisco, CA, USA
| | - Jialing Liu
- Department of Neurological Surgery, UCSF, San Francisco, CA, USA
- Department of Neurological Surgery, SFVAMC, San Francisco, CA, USA
| |
Collapse
|
64
|
Scheld M, Heymann F, Zhao W, Tohidnezhad M, Clarner T, Beyer C, Zendedel A. Modulatory effect of 17β-estradiol on myeloid cell infiltration into the male rat brain after ischemic stroke. J Steroid Biochem Mol Biol 2020; 202:105667. [PMID: 32407868 DOI: 10.1016/j.jsbmb.2020.105667] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/11/2020] [Accepted: 03/30/2020] [Indexed: 12/31/2022]
Abstract
Ischemic stroke is the leading cause of human disability and mortality in the world. Neuroinflammation is the main pathological event following ischemia which contributes to secondary brain tissue damage and is driven by infiltration of circulating immune cells such as macrophages. Because of neuroprotective properties against ischemic brain damage, estrogens have the potential to become of therapeutic interest. However, the exact mechanisms of neuroprotection and signaling pathways is not completely understood. In the current study, 12-week-old male Wistar rats underwent an experimental ischemia by occluding the middle cerebral artery transiently (tMCAO) for 1 h. Male rats subjected to tMCAO were randomly assigned to receive 17β-estradiol or vehicle treatment. The animals were sacrificed 72 h post tMCAO, transcardially perfused and the brains were proceeded either for TTC staining and gene analysis or for flow cytometry (CD45, CD11b, CD11c, CD40). We found that 17β-estradiol substitution significantly reduced the cortical infarct which was paralleled by an improved Garcia test scoring. Flow cytometry revealed that CD45+ cells as well as CD45+CD11b+CD11c+ cells were massively increased in tMCAO animals and numbers were nearly restored to sham levels after 17β-estradiol treatment. Gene expression analysis showed a reperfusion time-dependent upregulation of the markers CD45, CD11b and the activation marker CD40. The reduction in gene expression after 72 h of reperfusion and simultaneous 17β-estradiol substitution did not reach statistical significance. These data indicate that 17β-estradiol alleviated the cerebral ischemia-reperfusion injury and selectively suppressed the activation of the neuroinflammatory cascade via reduction of the number of activated microglia or infiltrated monocyte-derived macrophages in brain.
Collapse
Affiliation(s)
- Miriam Scheld
- University Clinic, Institute of Neuroanatomy, RWTH Aachen University, Aachen, Germany; Anatomy and Cell Biology, University of Augsburg, Augsburg, Germany.
| | - F Heymann
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Berlin, Germany.
| | - W Zhao
- University Clinic, Institute of Neuroanatomy, RWTH Aachen University, Aachen, Germany.
| | - M Tohidnezhad
- University Clinic, Institute of Anatomy and Cell Biology, RWTH Aachen University, Aachen, Germany.
| | - T Clarner
- University Clinic, Institute of Neuroanatomy, RWTH Aachen University, Aachen, Germany.
| | - C Beyer
- University Clinic, Institute of Neuroanatomy, RWTH Aachen University, Aachen, Germany.
| | - A Zendedel
- University Clinic, Institute of Neuroanatomy, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
65
|
Guo T, Liu Y, Ren X, Wang W, Liu H. Promoting Role of Long Non-Coding RNA Small Nucleolar RNA Host Gene 15 (SNHG15) in Neuronal Injury Following Ischemic Stroke via the MicroRNA-18a/CXC Chemokine Ligand 13 (CXCL13)/ERK/MEK Axis. Med Sci Monit 2020; 26:e923610. [PMID: 32862188 PMCID: PMC7480088 DOI: 10.12659/msm.923610] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Long-non-coding RNA (lncRNA) SNHG15 has been reported to be an aberrantly expressed lncRNA in patients with ischemic stroke, but its role in neuronal injury following ischemic stroke remains unclear. We hypothesized that this lncRNA is associated with the pathogenesis of ischemic stroke. Material/Methods A mouse model of ischemic stroke was established by middle cerebral artery occlusion (MCAO). A neurogenic mouse cell line Neuro-2a (N2a) was subjected to oxygen-glucose deprivation (OGD) for in vitro experiments. Expression of SNHG15, microRNA-18a (miR-18a), and CXCL13 in mouse brain and in OGD-treated N2a cells was determined. Altered expression of SNHG15 and miR-18a was introduced to detect their roles in N2a cell viability and apoptosis. Targeting relationships between miR-18a and SNHG15 or CXCL13 were validated by luciferase assays. Cells were treated with the ERK/MEK antagonist U0126 to assess the role of the ERK/MEK signaling pathway in N2a cell growth. Results SNHG15 and CXCL13 were overexpressed and miR-18a was underexpressed in MCAO-induced mice and OGD-treated N2a cells. Silencing of SNHG15 or overexpression of miR-18a promoted cell viability, while decreased cell apoptosis induced by OGD; however, subsequent disruption of the ERK/MEK signaling pathway reversed these effects. SNHG15 was found to bind to miR-18a, which could further target CXCL13. Conclusions Silencing of SNHG15 led to CXCL13 upregulation through sequestering miR-18a and the following ERK/MEK activation, thus enhancing viability while reducing apoptosis of N2a cells. SNHG15 may serve as a novel target for ischemic stroke treatment.
Collapse
Affiliation(s)
- Tiezhu Guo
- Department of Neurosurgery, Heji Hospital Affiliated with Changzhi Medical College, Changzhi, Shanxi, China (mainland)
| | - Yueting Liu
- Department of Neurosurgery, The First Hospital Affiliated with Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Xinliang Ren
- Department of Neurosurgery, Heji Hospital Affiliated with Changzhi Medical College, Changzhi, Shanxi, China (mainland)
| | - Wei Wang
- Department of Neurosurgery, Heji Hospital Affiliated with Changzhi Medical College, Changzhi, Shanxi, China (mainland)
| | - Hanrui Liu
- Department of Neurology, Heji Hospital Affiliated with Changzhi Medical College, Changzhi, Shanxi, China (mainland)
| |
Collapse
|
66
|
Acute brain injuries trigger microglia as an additional source of the proteoglycan NG2. Acta Neuropathol Commun 2020; 8:146. [PMID: 32843103 PMCID: PMC7449013 DOI: 10.1186/s40478-020-01016-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 08/10/2020] [Indexed: 01/07/2023] Open
Abstract
NG2 is a type I transmembrane glycoprotein known as chondroitin sulfate proteoglycan 4 (CSPG4). In the healthy central nervous system, NG2 is exclusively expressed by oligodendrocyte progenitor cells and by vasculature pericytes. A large body of immunohistochemical studies showed that under pathological conditions such as acute brain injuries and experimental autoimmune encephalomyelitis (EAE), a number of activated microglia were NG2 immuno-positive, suggesting NG2 expression in these cells. Alternative explanations for the microglial NG2 labeling consider the biochemical properties of NG2 or the phagocytic activity of activated microglia. Reportedly, the transmembrane NG2 proteoglycan can be cleaved by a variety of proteases to deposit the NG2 ectodomain into the extracellular matrix. The ectodomain, however, could also stick to the microglial surface. Since microglia are phagocytic cells engulfing debris of dying cells, it is difficult to identify a genuine expression of NG2. Recent studies showing (1) pericytes giving rise to microglial after stroke, and (2) immune cells of NG2-EYFP knock-in mice lacking NG2 expression in an EAE model generated doubts for the de novo expression of NG2 in microglia after acute brain injuries. In the current study, we took advantage of three knock-in mouse lines (NG2-CreERT2, CX3CR1-EGFP and NG2-EYFP) to study NG2 expression indicated by transgenic fluorescent proteins in microglia after tMCAO (transient middle cerebral artery occlusion) or cortical stab wound injury (SWI). We provide strong evidence that NG2-expressing cells, including OPCs and pericytes, did not differentiate into microglia after acute brain injuries, whereas activated microglia did express NG2 in a disease-dependent manner. A subset of microglia continuously activated the NG2 gene at least within the first week after tMCAO, whereas within 3 days after SWI a limited number of microglia at the lesion site transiently expressed NG2. Immunohistochemical studies demonstrated that these microglia with NG2 gene activity also synthesized the NG2 protein, suggesting activated microglia as an additional source of the NG2 proteoglycan after acute brain injuries.
Collapse
|
67
|
Shin TH, Lee DY, Basith S, Manavalan B, Paik MJ, Rybinnik I, Mouradian MM, Ahn JH, Lee G. Metabolome Changes in Cerebral Ischemia. Cells 2020; 9:E1630. [PMID: 32645907 PMCID: PMC7407387 DOI: 10.3390/cells9071630] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/05/2020] [Accepted: 07/06/2020] [Indexed: 12/26/2022] Open
Abstract
Cerebral ischemia is caused by perturbations in blood flow to the brain that trigger sequential and complex metabolic and cellular pathologies. This leads to brain tissue damage, including neuronal cell death and cerebral infarction, manifesting clinically as ischemic stroke, which is the cause of considerable morbidity and mortality worldwide. To analyze the underlying biological mechanisms and identify potential biomarkers of ischemic stroke, various in vitro and in vivo experimental models have been established investigating different molecular aspects, such as genes, microRNAs, and proteins. Yet, the metabolic and cellular pathologies of ischemic brain injury remain not fully elucidated, and the relationships among various pathological mechanisms are difficult to establish due to the heterogeneity and complexity of the disease. Metabolome-based techniques can provide clues about the cellular pathologic status of a condition as metabolic disturbances can represent an endpoint in biological phenomena. A number of investigations have analyzed metabolic changes in samples from cerebral ischemia patients and from various in vivo and in vitro models. We previously analyzed levels of amino acids and organic acids, as well as polyamine distribution in an in vivo rat model, and identified relationships between metabolic changes and cellular functions through bioinformatics tools. This review focuses on the metabolic and cellular changes in cerebral ischemia that offer a deeper understanding of the pathology underlying ischemic strokes and contribute to the development of new diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Tae Hwan Shin
- Department of Physiology, Ajou University School of Medicine, Suwon 16499, Korea; (T.H.S.); (D.Y.L.); (S.B.); (B.M.)
| | - Da Yeon Lee
- Department of Physiology, Ajou University School of Medicine, Suwon 16499, Korea; (T.H.S.); (D.Y.L.); (S.B.); (B.M.)
| | - Shaherin Basith
- Department of Physiology, Ajou University School of Medicine, Suwon 16499, Korea; (T.H.S.); (D.Y.L.); (S.B.); (B.M.)
| | - Balachandran Manavalan
- Department of Physiology, Ajou University School of Medicine, Suwon 16499, Korea; (T.H.S.); (D.Y.L.); (S.B.); (B.M.)
| | - Man Jeong Paik
- College of Pharmacy, Sunchon National University, Suncheon 57922, Korea;
| | - Igor Rybinnik
- Department of Neurology, Rutgers - Robert Wood Johnson Medical School, New Brunswick, NJ 08854, USA; (I.R.); (M.M.M.)
| | - M. Maral Mouradian
- Department of Neurology, Rutgers - Robert Wood Johnson Medical School, New Brunswick, NJ 08854, USA; (I.R.); (M.M.M.)
| | - Jung Hwan Ahn
- Department of Emergency Medicine, Ajou University School of Medicine, Suwon 16499, Korea
| | - Gwang Lee
- Department of Physiology, Ajou University School of Medicine, Suwon 16499, Korea; (T.H.S.); (D.Y.L.); (S.B.); (B.M.)
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea
| |
Collapse
|
68
|
Mészáros Á, Molnár K, Nógrádi B, Hernádi Z, Nyúl-Tóth Á, Wilhelm I, Krizbai IA. Neurovascular Inflammaging in Health and Disease. Cells 2020; 9:cells9071614. [PMID: 32635451 PMCID: PMC7407516 DOI: 10.3390/cells9071614] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 07/02/2020] [Indexed: 12/19/2022] Open
Abstract
Aging is characterized by a chronic low-grade sterile inflammation dubbed as inflammaging, which in part originates from accumulating cellular debris. These, acting as danger signals with many intrinsic factors such as cytokines, are sensed by a network of pattern recognition receptors and other cognate receptors, leading to the activation of inflammasomes. Due to the inflammasome activity-dependent increase in the levels of pro-inflammatory interleukins (IL-1β, IL-18), inflammation is initiated, resulting in tissue injury in various organs, the brain and the spinal cord included. Similarly, in age-related diseases of the central nervous system (CNS), inflammasome activation is a prominent moment, in which cells of the neurovascular unit occupy a significant position. In this review, we discuss the inflammatory changes in normal aging and summarize the current knowledge on the role of inflammasomes and contributing mechanisms in common CNS diseases, namely Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis and stroke, all of which occur more frequently with aging.
Collapse
Affiliation(s)
- Ádám Mészáros
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (Á.M.); (K.M.); (B.N.); (Z.H.); (Á.N.-T.); (I.W.)
- Doctoral School of Biology, University of Szeged, 6726 Szeged, Hungary
| | - Kinga Molnár
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (Á.M.); (K.M.); (B.N.); (Z.H.); (Á.N.-T.); (I.W.)
- Theoretical Medicine Doctoral School, University of Szeged, 6720 Szeged, Hungary
| | - Bernát Nógrádi
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (Á.M.); (K.M.); (B.N.); (Z.H.); (Á.N.-T.); (I.W.)
- Foundation for the Future of Biomedical Sciences in Szeged, Szeged Scientists Academy, 6720 Szeged, Hungary
| | - Zsófia Hernádi
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (Á.M.); (K.M.); (B.N.); (Z.H.); (Á.N.-T.); (I.W.)
- Foundation for the Future of Biomedical Sciences in Szeged, Szeged Scientists Academy, 6720 Szeged, Hungary
| | - Ádám Nyúl-Tóth
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (Á.M.); (K.M.); (B.N.); (Z.H.); (Á.N.-T.); (I.W.)
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Oklahoma Center for Geroscience, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Imola Wilhelm
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (Á.M.); (K.M.); (B.N.); (Z.H.); (Á.N.-T.); (I.W.)
- Institute of Life Sciences, Vasile Goldiş Western University of Arad, 310414 Arad, Romania
| | - István A. Krizbai
- Institute of Biophysics, Biological Research Centre, 6726 Szeged, Hungary; (Á.M.); (K.M.); (B.N.); (Z.H.); (Á.N.-T.); (I.W.)
- Institute of Life Sciences, Vasile Goldiş Western University of Arad, 310414 Arad, Romania
- Correspondence: ; Tel.: +36-62-599-794
| |
Collapse
|
69
|
Non-coding RNAs in Ischemic Stroke: Roles in the Neuroinflammation and Cell Death. Neurotox Res 2020; 38:564-578. [DOI: 10.1007/s12640-020-00236-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 05/10/2020] [Accepted: 06/07/2020] [Indexed: 12/11/2022]
|
70
|
Lu M, Qi Y, Han Y, Yi Q, Xu L, Sun W, Ni G, Ni X, Xu C. Design and development of novel thiazolidin-4-one-1,3,5-triazine derivatives as neuro-protective agent against cerebral ischemia-reperfusion injury in mice via attenuation of NF-ĸB. Chem Biol Drug Des 2020; 96:1315-1327. [PMID: 32543026 DOI: 10.1111/cbdd.13744] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/21/2020] [Accepted: 06/05/2020] [Indexed: 11/26/2022]
Abstract
The present study enumerates the discovery and development of novel thiazolidin-4-one-1,3,5-triazine as neuro-protective agent against cerebral ischemia-reperfusion injury in mice. These compounds showed significant inhibition of NF-ĸB transcriptional activity in LPS-stimulated RAW264.7 cells, displaying compound 8k as most potent inhibitor among the tested derivative. The compound 8k was further studied in in vivo middle cerebral artery occlusion (MCAO) mice model for neuro-protective action. Results suggest that compound 8k causes attenuation of inflammation (TNF-α, IL-β, and IL-6), oxidative stress (SOD, GSH, and MDA), and apoptosis (Bcl-2, Bax, and cleaved caspase-3) in MCAO mice in concentration-dependent manner. Collectively, our results documented that compound 8k pre-treatment protects cerebral I/R. This novel lead scaffold may be helpful for investigation of new neuro-protective agent by inactivation of NF-ĸB.
Collapse
Affiliation(s)
- Min Lu
- Department of Rehabilitation Medicine, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Yujun Qi
- Department of Rehabilitation Medicine, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Yu Han
- Department of Rehabilitation Medicine, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Qiong Yi
- Department of Rehabilitation Medicine, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Lei Xu
- Department of Rehabilitation Medicine, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Wenlin Sun
- Department of Rehabilitation Medicine, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Guihua Ni
- Department of Neurology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Xiaoyu Ni
- Department of Rehabilitation Medicine, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Changsong Xu
- Department of Neurology, The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| |
Collapse
|
71
|
Mujagić A, Marushima A, Nagasaki Y, Hosoo H, Hirayama A, Puentes S, Takahashi T, Tsurushima H, Suzuki K, Matsui H, Ishikawa E, Matsumaru Y, Matsumura A. Antioxidant nanomedicine with cytoplasmic distribution in neuronal cells shows superior neurovascular protection properties. Brain Res 2020; 1743:146922. [PMID: 32504549 DOI: 10.1016/j.brainres.2020.146922] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 05/20/2020] [Accepted: 06/01/2020] [Indexed: 01/15/2023]
Abstract
This study investigated whether nitroxide radical (4-amino-TEMPOL)-containing nanoparticles (RNPs; antioxidant nanomedicine) can prevent neurovascular unit impairment caused by reactive oxygen species (ROS) after cerebral ischemia-reperfusion. C57BL/6J mice underwent transient middle cerebral artery occlusion (tMCAO). The mice were randomly divided and administered intra-arterial RNPs injection (9 mg/kg, 7 μM/kg), edaravone (3 mg/kg, 17 μM/kg), or phosphate-buffered saline (control group). Survival rate and neurological score were evaluated 24 h post-injection. RNPs distribution was determined using immunofluorescence staining and blood-brain barrier (BBB) disruption using Evans blue extravasation assay. Effect of RNPs and edaravone on microglia polarization into microglia M1 and M2 was evaluated. We also determined multiple ROS-scavenging activities in brain homogenates of RNPs- and edaravone-treated animals using an electron spin resonance-based spin-trapping method. Compared with edaravone, RNPs significantly improved the survival rate and neurological deficit, inhibited BBB disruption and supported polarization of microglia into M2 microglia. RNPs were localized in endothelial cells, the perivascular space, neuronal cell cytoplasm, astrocytes, and microglia. Scavenging capacities of hydroxyl, alkoxyl, and peroxyl radicals were significantly higher in the RNPs-treated group. RNPs show promising results as a future neuroprotective nanomedicine approach for cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Arnela Mujagić
- Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, Japan; Department of Neurosurgery, Graduate School of Comprehensive Human Science, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, Japan
| | - Aiki Marushima
- Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, Japan; Department of Neurosurgery, Graduate School of Comprehensive Human Science, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, Japan.
| | - Yukio Nagasaki
- Graduate School of Pure and Applied Sciences, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, Japan
| | - Hisayuki Hosoo
- Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, Japan; Department of Neurosurgery, Graduate School of Comprehensive Human Science, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, Japan
| | - Aki Hirayama
- Center for Integrative Medicine, Tsukuba University of Technology, Kasuga 4-12-7, Tsukuba, Ibaraki, Japan
| | - Sandra Puentes
- Graduate School of Systems and Information Engineering, University of Tsukuba, Tennodai 1-1-1, Ibaraki, Japan
| | - Toshihide Takahashi
- Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, Japan; Department of Neurosurgery, Graduate School of Comprehensive Human Science, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, Japan
| | - Hideo Tsurushima
- Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, Japan; Department of Neurosurgery, Graduate School of Comprehensive Human Science, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, Japan
| | - Kensuke Suzuki
- Department of Neurosurgery, Saitama Medical Center, Dokkyo Medical University, Minami-Koshigaya 2-1-50, Koshigaya, Saitama, Japan
| | - Hirofumi Matsui
- Department of Gastroenterology, Faculty of Medicine, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, Japan
| | - Eiichi Ishikawa
- Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, Japan; Department of Neurosurgery, Graduate School of Comprehensive Human Science, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, Japan
| | - Yuji Matsumaru
- Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, Japan; Department of Neurosurgery, Graduate School of Comprehensive Human Science, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, Japan
| | - Akira Matsumura
- Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, Japan; Department of Neurosurgery, Graduate School of Comprehensive Human Science, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, Japan
| |
Collapse
|
72
|
Jia C, Malone HM, Keasey MP, Lovins C, Elam J, Hagg T. Blood Vitronectin Induces Detrimental Brain Interleukin-6 and Correlates With Outcomes After Stroke Only in Female Mice. Stroke 2020; 51:1587-1595. [PMID: 32312218 DOI: 10.1161/strokeaha.120.029036] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background and Purpose- Women have worse stroke outcomes than men, especially after menopause. Few studies have focused on female-specific mechanisms, other than hormones. We investigated the role of the blood protein VTN (vitronectin) after ischemic stroke in mice. Methods- Adult male and female VTN knockout and wild-type littermates and C57BL/6 mice received a middle cerebral artery occlusion and the injured brain tissue analyzed 24 hours to 3 weeks later for cell loss and inflammation, as well as neurological function. Blood VTN levels were measured before and after stroke. Results- Intravenously injected VTN leaked extensively from bloodstream into brain infarct and penumbra by 24 hours after stroke. Strikingly, VTN was detrimental in female, but not male, mice, as shown by reduced brain injury (26.2±2.6% versus 13.4±3.8%; P=0.018; n=6 and 5) and forelimb dysfunction in female VTN knockout mice. Stroke increased plasma VTN 2- to 8-fold at 24 hours in females (36±4 versus 145±24 μg/mL; P<0.0001; n=10 and 7), but not males (62±8 versus 68±6; P>0.99; n=10 and 7), and returned to control levels by 7 days. Individually variable VTN levels at 24 hours correlated with stroke-induced brain injury at 7 days only in females. VTN promoted stroke-induced microglia/macrophage activation and leukocyte infiltration in females. Proinflammatory IL (interleukin)-6 greatly increased in the striatum at 24 hours in wild-type mice but was increased ≈60% less in female (739±159 versus 268±111; P=0.02; n=7 and 6), but not male (889±178 versus 1179±295; P=0.73; n=10 and 11), knockout mice. In individual wild-type females, plasma VTN levels correlated with striatal IL-6 expression at 24 hours. The female-specific effect of VTN-induced IL-6 expression following stroke was not due to gonadal hormones, as shown by ovariectomy and castration. Lastly, intrastriatal injection of IL-6 in female mice immediately before stroke reversed the VTN knockout phenotypes of reduced brain injury and microglia/macrophage activation. Conclusions- VTN plays a novel sexually dimorphic detrimental pathophysiological role in females and might ultimately be a therapeutic target to improve stroke outcomes in women.
Collapse
Affiliation(s)
- Cuihong Jia
- From the Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City
| | - Hannah M Malone
- From the Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City
| | - Matthew P Keasey
- From the Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City
| | - Chiharu Lovins
- From the Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City
| | - Jacob Elam
- From the Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City
| | - Theo Hagg
- From the Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City
| |
Collapse
|
73
|
Yang Y, Delalio LJ, Best AK, Macal E, Milstein J, Donnelly I, Miller AM, McBride M, Shu X, Koval M, Isakson BE, Johnstone SR. Endothelial Pannexin 1 Channels Control Inflammation by Regulating Intracellular Calcium. THE JOURNAL OF IMMUNOLOGY 2020; 204:2995-3007. [PMID: 32312847 PMCID: PMC7336877 DOI: 10.4049/jimmunol.1901089] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 03/27/2020] [Indexed: 12/26/2022]
Abstract
The proinflammatory cytokine IL-1β is a significant risk factor in cardiovascular disease that can be targeted to reduce major cardiovascular events. IL-1β expression and release are tightly controlled by changes in intracellular Ca2+ ([Ca2+]i), which has been associated with ATP release and purinergic signaling. Despite this, the mechanisms that regulate these changes have not been identified. The pannexin 1 (Panx1) channels have canonically been implicated in ATP release, especially during inflammation. We examined Panx1 in human umbilical vein endothelial cells following treatment with the proinflammatory cytokine TNF-α. Analysis by whole transcriptome sequencing and immunoblot identified a dramatic increase in Panx1 mRNA and protein expression that is regulated in an NF-κB-dependent manner. Furthermore, genetic inhibition of Panx1 reduced the expression and release of IL-1β. We initially hypothesized that increased Panx1-mediated ATP release acted in a paracrine fashion to control cytokine expression. However, our data demonstrate that IL-1β expression was not altered after direct ATP stimulation in human umbilical vein endothelial cells. Because Panx1 forms a large pore channel, we hypothesized it may permit Ca2+ diffusion into the cell to regulate IL-1β. High-throughput flow cytometric analysis demonstrated that TNF-α treatments lead to elevated [Ca2+]i, corresponding with Panx1 membrane localization. Genetic or pharmacological inhibition of Panx1 reduced TNF-α-associated increases in [Ca2+]i, blocked phosphorylation of the NF-κB-p65 protein, and reduced IL-1β transcription. Taken together, the data in our study provide the first evidence, to our knowledge, that [Ca2+]i regulation via the Panx1 channel induces a feed-forward effect on NF-κB to regulate IL-1β synthesis and release in endothelium during inflammation.
Collapse
Affiliation(s)
- Yang Yang
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908.,Department of Pharmacology, Dalian Medical University, Dalian 116044, China
| | - Leon J Delalio
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Angela K Best
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Edgar Macal
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Jenna Milstein
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Iona Donnelly
- British Heart Foundation Cardiovascular Research Centre, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Ashley M Miller
- British Heart Foundation Cardiovascular Research Centre, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Martin McBride
- British Heart Foundation Cardiovascular Research Centre, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Xiaohong Shu
- Department of Pharmacology, Dalian Medical University, Dalian 116044, China
| | - Michael Koval
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322.,Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322; and
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908; .,Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Scott R Johnstone
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908;
| |
Collapse
|
74
|
Electroacupuncture Pretreatment Alleviates Cerebral Ischemia-Reperfusion Injury by Increasing GSK-3 β Phosphorylation Level via Adenosine A1 Receptor. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6848450. [PMID: 32149120 PMCID: PMC7054798 DOI: 10.1155/2020/6848450] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 12/17/2019] [Accepted: 12/31/2019] [Indexed: 01/19/2023]
Abstract
Objective To observe the effect of adenosine A1 receptor in the hippocampus of mice on GSK-3β phosphorylation level and elucidate the underlying mechanisms of electroacupuncture pretreatment by activating Α1 receptor mediating cerebral ischemia-reperfusion injury. Method The model of middle cerebral artery occlusion (MCAO) was established and grouped into electroacupuncture pretreatment group (EA group), MCAO group, and sham-operated group (Sham group). The neurobehavioral manifestation, the volume of cerebral infarction, and its related protein changes in mice in each group were observed. Then, adenosine Α1 receptor antagonist and agonist were injected intraperitoneally to observe the effects of A1 receptor on the phosphorylation level of GSK-3β phosphorylation level and elucidate the underlying mechanisms of electroacupuncture pretreatment by activating Α1 receptor mediating cerebral ischemia-reperfusion injury. Results (1) Compared with the MCAO group (24 hours after reperfusion), the infarct size in the EA group decreased significantly, and the Garcia neurological score and phosphorylation level of GSK-3β phosphorylation level and elucidate the underlying mechanisms of electroacupuncture pretreatment by activating Α1 receptor mediating cerebral ischemia-reperfusion injury. β phosphorylation level and elucidate the underlying mechanisms of electroacupuncture pretreatment by activating Α1 receptor mediating cerebral ischemia-reperfusion injury. β phosphorylation level and elucidate the underlying mechanisms of electroacupuncture pretreatment by activating Α1 receptor mediating cerebral ischemia-reperfusion injury. Conclusions Electroacupuncture pretreatment can increase GSK-3β phosphorylation level via activating A1 receptor, to protect neurons in ischemia-reperfusion injury.β phosphorylation level and elucidate the underlying mechanisms of electroacupuncture pretreatment by activating Α1 receptor mediating cerebral ischemia-reperfusion injury.
Collapse
|
75
|
Chen KY, Wu KC, Hueng DY, Huang KF, Pang CY. Anti-inflammatory effects of powdered product of Bu Yang Huan Wu decoction: Possible role in protecting against Transient Focal Cerebral Ischemia. Int J Med Sci 2020; 17:1854-1863. [PMID: 32714088 PMCID: PMC7378667 DOI: 10.7150/ijms.46581] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/17/2020] [Indexed: 12/25/2022] Open
Abstract
Bu Yang Huan Wu decoction (BYHW) is a traditional Chinese medicine (TCM) that consists of several herbs and has been used in patients with ischemic stroke for centuries. Although powdered formula of BYHW has widely been prescribed in clinic nowadays, evidence-based effectiveness and mechanism of action of BYHW powdered product in stroke remain to be characterized. Adult male Sprague-Dawley rats were subjected to middle cerebral artery occlusion (MCAO) for 90 min followed by reperfusion for 24 h (ischemia/reperfusion; I/R) or sham surgery. After I/R, the rats were then given low dose (0.5 g/kg) and high dose (2.5 g/kg) of BYHW or vehicle by oral gavage twice a day for seven consecutive days. The results showed that I/R induced obvious cerebral infarction and neurobehavioral defects, in parallel with histological aberrations and extensive signaling of proinflammatory cytokines, including tumor necrosis factor (TNF-α) and interleukin-6 (IL-6), in the stroke model. Post-I/R treatment with BYHW powdered product significantly reduced the infarct area and ameliorated neurofunctional defects in a dose-dependent manner. The dose dependence was associated with TNF-α downregulation and interleukin-10 (IL-10) induction. In summary, the present findings demonstrated that BYHW powdered product exhibited therapeutic efficacy for experimental stroke and a higher dose treatment may strengthen the effectiveness via inflammatory modulation.
Collapse
Affiliation(s)
- Kuan-Yu Chen
- Department of Surgery, New Taipei City Hospital, New Taipei city, Taiwan.,Institute of Medical Sciences, Tzu Chi University, Hualien city, Taiwan
| | - Kuo-Chen Wu
- School of Pharmacy, National Taiwan University, Taipei, Taiwan
| | - Dueng-Yuan Hueng
- Department of Neurosurgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Kuo-Feng Huang
- School of Medicine, Buddhist Tzu Chi University, Hualien, Taiwan.,Division of Neurosurgery, Department of Surgery, Taipei Tzu Chi Medical Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - Cheng-Yoong Pang
- Institute of Medical Sciences, Tzu Chi University, Hualien city, Taiwan.,Department of Medical Research, Buddhist Tzu Chi General Hospital, Hualien city, Taiwan
| |
Collapse
|
76
|
Yeh CF, Chuang TY, Hung YW, Lan MY, Tsai CH, Huang HX, Lin YY. Development of a Modified Surgical Technique for Simulating Ischemic Cerebral Cortex Injury in Rats. In Vivo 2019; 33:1175-1181. [PMID: 31280207 DOI: 10.21873/invivo.11588] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/23/2019] [Accepted: 04/24/2019] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM Middle cerebral artery occlusion (MCAO) in rodents is an essential animal model for research focusing on ischemic stroke. To date, several kinds of surgical methods for MCAO have been developed and the craniotomy method has the advantage of direct visualization of the middle cerebral artery (MCA). MCAO at a more proximal site produces better surgical results, but it is a more invasive technique. The aim of this study was to evolve the surgical technique for simulating ischemic cerebral cortex injury in rats. MATERIALS AND METHODS To approach proximal MCA with a less invasive procedure, a modified surgical technique for MCAO in rats was developed. Besides, rats receiving the modified and conventional method were compared with regard to infarct volume and by behavioral tests. RESULTS Following craniotomy, we proposed that the inferior edge of the craniotomy should be enlarged with fine forceps. This modified surgical method induces larger infarct volume, significant behavioral impairment and can induce ischemic stroke. Additionally, it does not significantly increase the operation time, and has produced no obvious complications. CONCLUSION This modified surgical technique may serve as a practical method for performing MCAO.
Collapse
Affiliation(s)
- Chien-Fu Yeh
- Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan, R.O.C.,Department of Otorhinolaryngology, National Yang-Ming University, Taipei, Taiwan, R.O.C.,Department of Otolaryngology-Head and Neck Surgery, Taipei Veterans General Hospital, Taipei, Taiwan, R.O.C
| | - Tung-Yueh Chuang
- Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, R.O.C
| | - Yu-Wen Hung
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan, R.O.C
| | - Ming-Ying Lan
- Department of Otorhinolaryngology, National Yang-Ming University, Taipei, Taiwan, R.O.C.,Department of Otolaryngology-Head and Neck Surgery, Taipei Veterans General Hospital, Taipei, Taiwan, R.O.C
| | - Ching-Han Tsai
- Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, R.O.C
| | - Hao-Xiang Huang
- Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, R.O.C
| | - Yung-Yang Lin
- Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan, R.O.C. .,Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, R.O.C.,Institute of Physiology, National Yang-Ming University, Taipei, Taiwan, R.O.C.,Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan, R.O.C.,Department of Neurology, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan, R.O.C
| |
Collapse
|
77
|
Nan L, Xie Q, Chen Z, Zhang Y, Chen Y, Li H, Lai W, Chen Y, Huang M. Involvement of PARP-1/AIF Signaling Pathway in Protective Effects of Gualou Guizhi Decoction Against Ischemia-Reperfusion Injury-Induced Apoptosis. Neurochem Res 2019; 45:278-294. [PMID: 31792665 DOI: 10.1007/s11064-019-02912-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 11/08/2019] [Accepted: 11/16/2019] [Indexed: 10/25/2022]
Abstract
Cerebral ischemia-reperfusion injury is a complex pathophysiological process. Poly(ADP-ribose) (PAR) polymerase-1 (PARP-1)/apoptosis-inducing factor (AIF) signaling pathway-mediated apoptosis is one of the non-caspase-dependent cell death programs that are widely present in neurological diseases such as stroke. In our study, we aimed to conduct further research on the effects of Gualou Guizhi decoction (GLGZD) on the PARP-1/AIF signaling pathway in cell apoptosis after ischemia-reperfusion injury caused by middle cerebral artery occlusion (MCAO). The results showed that GLGZD administration for 7 days significantly ameliorated MCAO-induced neurological damage, limb paralysis and the pathological state of the ischemic cortex. GLGZD exerted its effects by significantly reducing the volume of ischemic cerebral infarction, increasing the number of Nissl-positive cells, and reducing neuronal apoptosis. Furthermore, Western blot analysis showed that GLGZD significantly inhibited the total protein expression of PARP-1, PAR, AIF and endonuclease G (Endo G) in the ischemic cortex and significantly increased the total protein expression of heat-shock protein 70 (Hsp70). On the one hand, the expression of PARP-1, AIF and Endo G protein in the nucleus significantly decreased while the expression of PAR nucleoprotein significantly upregulated. On the other hand, compared with the MCAO model group, the GLGZD-treated group showed a significantly reduced protein expression of PAR in mitochondria and significantly increased protein expression of mitochondrial AIF and Endo G. It was concluded that GLGZD had good therapeutic effects in MCAO model rats. These effects were closely related to GLGZD-mediated inhibition of ischemia-induced neuronal apoptosis by regulation of protein expression and translocation in the PARP-1/AIF signaling pathway.
Collapse
Affiliation(s)
- Lihong Nan
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Qingqing Xie
- Hangzhou Simo Co., Ltd., Nanjing, 210001, Jiangsu, China
| | - Zheming Chen
- Pharmaceutical Preparation Section, Quanzhou First Hospital, Quanzhou, 362000, Fujian, China
| | - Yuqin Zhang
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Yaping Chen
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Huang Li
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Wenfang Lai
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Yan Chen
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Mei Huang
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China.
| |
Collapse
|
78
|
Transient versus Permanent MCA Occlusion in Mice Genetically Modified to Have Good versus Poor Collaterals. ACTA ACUST UNITED AC 2019; 4. [PMID: 31840083 PMCID: PMC6910253 DOI: 10.20900/mo.20190024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Collateral-dependent blood flow is capable of significantly lessening the severity of stroke. Unfortunately, collateral flow varies widely in patients for reasons that remain unclear. Studies in mice have shown that the number and diameter of cerebral collaterals vary widely due primarily to polymorphisms in genes, e.g., Rabep2, involved in their formation during development. However, understanding how variation in collateral abundance affects stroke progression has been hampered by lack of a method to reversibly ligate the distal middle cerebral artery (MCAO) in mice. Here we present a method and examine infarct volume 24 h after transient (tMCAO, 90 min) versus permanent occlusion (pMCAO) in mice with good versus poor collaterals. Wildtype C57BL/6 mice (have abundant collaterals) sustained small infarctions following tMCAO that increased 2.1-fold after pMCAO, reflecting significant penumbra present at 90 min. Mutant C57BL/6 mice lacking Rabep2 (have reduced collaterals) sustained a 4-fold increase in infarct volume over WT following tMCAO and a smaller additional increase (0.4-fold) after pMCAO, reflecting reduced penumbra. Wildtype BALB/cBy (have a deficient Rabep2 variant and poor collaterals) had large infarctions following tMCAO that increased less (0.6-fold) than the above wildtype C57BL/6 mice following pMCAO. Mutant BALB/cBy mice (have deficient Rabep2 replaced with the C57BL/6 variant thus increased collaterals) sustained smaller infarctions after tMCAO. However, unlike C57BL/6 versus Rabep2 mice, penumbra was not increased since infarct volume increased only 0.3-fold following pMCAO. These findings present a murine model of tMCAO and demonstrate that neuroprotective mechanisms, in addition to collaterals, also vary with genetic background and affect the evolution of stroke.
Collapse
|
79
|
Fingolimod promotes angiogenesis and attenuates ischemic brain damage via modulating microglial polarization. Brain Res 2019; 1726:146509. [PMID: 31626784 DOI: 10.1016/j.brainres.2019.146509] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/26/2019] [Accepted: 10/14/2019] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Microglial activation plays a crucial role in the pathology of ischemic stroke. Recently, we demonstrated that fingolimod (FTY720) exerted neuroprotective effects via immunomodulation in ischemic white matter damage induced by chronic cerebral hypoperfusion, which was accompanied by robust microglial activation. In this study, we assessed the pro-angiogenic potential of FTY720 in a murine model of acute cortical ischemic stroke. METHODS The photothrombotic (PT) method was used to induce cortical ischemic stroke in mice. We evaluated cortical damage, behavioral deficits, microglial polarization, and angiogenesis to identify the neuroprotective effects and possible molecular mechanisms of FTY720 in acute ischemic stroke. RESULTS In vivo, a reduction in neuronal loss and improved motor function were observed in FTY720-treated mice after PT stroke. Immunofluorescence staining revealed that robust microglial activation and the associated neuroinflammatory response in the peri-infarct area were ameliorated by FTY720 via its ability to polarize microglia toward the M2 phenotype. Furthermore, both in vivo and in vitro, angiogenesis was enhanced in the microglial M2 phenotype state. Behaviorally, a significant improvement in the FTY720-treated group compared to the control group was evident from days 7 to 14. CONCLUSIONS Our research indicated that FTY720 treatment promoted angiogenesis via microglial M2 polarization and exerted neuroprotection in PT ischemic stroke.
Collapse
|
80
|
Wang C, Wei Y, Yuan Y, Yu Y, Xie K, Dong B, Shi Y, Wang G. The role of PI3K-mediated AMPA receptor changes in post-conditioning of propofol in brain protection. BMC Neurosci 2019; 20:51. [PMID: 31570094 PMCID: PMC6771103 DOI: 10.1186/s12868-019-0532-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 09/13/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND We aimed to study the role of amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptor (AMPAR) glutamate receptor 2 (GluR2) subunit trafficking, and activity changes in short-term neuroprotection provided by propofol post-conditioning. We also aimed to determine the role of phosphoinositide-3-kinase (PI3K) in the regulation of these processes. METHODS Rats underwent 1 h of focal cerebral ischemia followed by 23 h of reperfusion were randomly divided into 6 groups (n = 36 per group): sham- operation (S), ischemia-reperfusion (IR), propofol (P group, propofol 20 mg/kg/h at the onset of reperfusion for 2 h after 60 min of occlusion), and LY294002 (PI3K non-selective antagonist) + sham (L + S, LY294002 of 1.5 mg/kg was infused 30 min before sham operation), LY294002+ ischemia-reperfusion (L + IR, LY294002 of 1.5 mg/kg was infused 30 min before middle cerebral artery occlusion), LY294002 + IR + propofol (L + P, LY294002 of 1.5 mg/kg was infused 30 min before middle cerebral artery occlusion and propofol 20 mg/kg/h at the onset of reperfusion for 2 h after 60 min of occlusion). RESULTS Compared with group IR, rats in group P had significant lower neurologic defect scores and infarct volume. Additionally, consistent with enhanced expression of PI3K-AMPAR GluR2 subunit complex substances in ipsilateral hippocampus, GluR2 subunits showed increased levels in both the plasma and postsynaptic membranes of neurons, while pGluR2 expression was reduced in group P. Furthermore, LY294002, the PI3K non-selective antagonist, blocked those effects. CONCLUSION These observations demonstrated that propofol post-conditioning revealed acute neuroprotective role against transient MCAO in rats. The short-term neuroprotective effect was contributed by enhanced GluR2 subunits trafficking to membrane and postsynaptic membranes of neurons, as well as down-regulated the expression of pGluR2 in damaged hippocampus. Finally, the above-mentioned protective mechanism might be contributed by increased combination of PI3K to AMPAR GluR2 subunit, thus maintained the expression and activation of AMPAR GluR2 in the ipsilateral hippocampus.
Collapse
Affiliation(s)
- Chenxu Wang
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052 People’s Republic of China
| | - Ying Wei
- Department of Anesthesiology, Tianjin People’s Hospital, Tianjin Union Medical Center, Tianjin, 300191 China
| | - Yuan Yuan
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052 People’s Republic of China
| | - Yonghao Yu
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052 People’s Republic of China
| | - Keliang Xie
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052 People’s Republic of China
| | - Beibei Dong
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052 People’s Republic of China
| | - Yuan Shi
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052 People’s Republic of China
| | - Guolin Wang
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, General Hospital of Tianjin Medical University, No. 154 Anshan Road, Heping District, Tianjin, 300052 People’s Republic of China
| |
Collapse
|
81
|
Yang P, Tian YM, Deng WX, Cai X, Liu WH, Li L, Huang HY. Sijunzi decoction may decrease apoptosis via stabilization of the extracellular matrix following cerebral ischaemia-reperfusion in rats. Exp Ther Med 2019; 18:2805-2812. [PMID: 31572528 PMCID: PMC6755478 DOI: 10.3892/etm.2019.7878] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 06/13/2019] [Indexed: 12/19/2022] Open
Abstract
Neurons undergo degeneration, apoptosis and death due to ischaemic stroke. The present study investigated the effect of Sijunzi decoction (SJZD), a type of traditional Chinese medicine known as invigorating spleen therapy, on anoikis (a type of apoptosis) in rat brains following cerebral ischaemia-reperfusion. Rats were randomly divided into sham, model, nimodipine and SJZD low/medium/high dose groups. A middle cerebral artery occlusion model was established. Neurobehavioural scores were evaluated after administration for 14 days using a five-grade scale. Blood-brain barrier permeability and apoptotic rate were detected using Evans blue (EB) extravasation and TUNEL staining, respectively. Tissue inhibitor of metalloproteinase 1 (TIMP-1), matrix metalloproteinase 9 (MMP-9) and collagen IV (COL IV) were determined using immunohistochemistry. Neurobehavioural scores decreased remarkably in all SJZD and nimodipine groups compared to the model group (P<0.05). Compared with the sham group, EB extravasation was higher in the model group (P<0.01). The amount of EB extravasation decreased in the SJZD high dose and nimodipine groups compared to the model group (P<0.01), and extravasation in the SJZD high dose group was lower than the SJZD low and medium dose groups (P<0.01). TIMP-1 and MMP-9 expression and apoptotic rate increased, but COL IV decreased significantly in the hippocampus of the model group compared to the sham group (P<0.01). TIMP-1 and COL IV expression increased significantly and MMP-9 and apoptotic rate decreased remarkably in all SJZD and nimodipine groups compared to the model group (P<0.01). TIMP-1 and COL IV expression decreased, but MMP-9 expression and apoptotic rate increased in the SJZD low and medium dose groups compared to the SJZD high dose group (P<0.01). SJZD rescued neurons and improved neurobehavioural function in rats following cerebral ischaemia-reperfusion, especially when used at a high dose. The mechanism may be related to protection of the extracellular matrix followed by anti-apoptotic effects.
Collapse
Affiliation(s)
- Ping Yang
- Department of Psychiatry, Brains Hospital of Hunan Province, Clinical Medical School, Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Ye-Mei Tian
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Wen-Xiang Deng
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Xiong Cai
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Wang-Hua Liu
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Liang Li
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China.,Key Discipline of Anatomy and Histoembryology, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Hui-Yong Huang
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| |
Collapse
|
82
|
Shadman J, Sadeghian N, Moradi A, Bohlooli S, Panahpour H. Magnesium sulfate protects blood-brain barrier integrity and reduces brain edema after acute ischemic stroke in rats. Metab Brain Dis 2019; 34:1221-1229. [PMID: 31037556 DOI: 10.1007/s11011-019-00419-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 04/16/2019] [Indexed: 11/27/2022]
Abstract
Brain edema is a fatal complication of acute ischemic stroke and associated with worse outcomes in patients. This study was designed to evaluate the effects of magnesium sulfate on vasogenic brain edema formation and blood-brain barrier (BBB) disruption caused by ischemia-reperfusion (IR) in a rat model of ischemic stroke. A total of 72 male Sprague-Dawley rats were categorized into the following three primary groups: sham, control ischemic, magnesium-sulfate-treated (300 mg/kg loading dose, followed by an additional 100 mg/kg) ischemic (n = 24 in each group). Transient focal cerebral ischemia was induced by 60-min-long occlusion of the left middle cerebral artery, followed by 24-h-long reperfusion. Sensorimotor deficits, infarct volume, and brain edema were evaluated at the end of the reperfusion period. The BBB permeability was assessed by Evans Blue extravasation technique. Lipid peroxidation levels were assessed by measuring the malondialdehyde content in the brain tissue homogenate, and the activities of the antioxidant enzymes superoxide dismutase, catalase, and glutathione peroxidase were detected according to the technical manual of the assay kits. Induction of cerebral ischemia in the control group produced considerable BBB damage in conjunction with severe brain edema formation. Treatment with magnesium sulfate significantly attenuated brain edema and protected BBB integrity in the ischemic lesioned hemisphere. In addition, magnesium sulfate reduced lipid peroxidation and increased antioxidant protection of brain tissue by upregulating the activities of antioxidant enzymes. Treatment with magnesium sulfate protected BBB integrity against IR-induced damage and reduced vasogenic edema formation partly via antioxidant mechanisms in a rat model of acute ischemic stroke.
Collapse
Affiliation(s)
- Javad Shadman
- Department of Physiology, Medical School, Ardabil University of Medical Sciences, Ardabil, 56197, Iran
- Physiological Studies Research Center, Medical School, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Nooshin Sadeghian
- Department of Physiology, Medical School, Ardabil University of Medical Sciences, Ardabil, 56197, Iran
- Physiological Studies Research Center, Medical School, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Alireza Moradi
- Department of Physiology, Medical School, Ardabil University of Medical Sciences, Ardabil, 56197, Iran
- Physiological Studies Research Center, Medical School, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Shahab Bohlooli
- Department of Pharmacology, Pharmacy School, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hamdollah Panahpour
- Department of Physiology, Medical School, Ardabil University of Medical Sciences, Ardabil, 56197, Iran.
- Physiological Studies Research Center, Medical School, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
83
|
Xie P, Deng M, Sun QG, Ma YG, Zhou Y, Ming JH, Chen Q, Liu SQ, Liu JQ, Cai J, Wu F. Therapeutic effect of transplantation of human bone marrow‑derived mesenchymal stem cells on neuron regeneration in a rat model of middle cerebral artery occlusion. Mol Med Rep 2019; 20:3065-3074. [PMID: 31432152 PMCID: PMC6755237 DOI: 10.3892/mmr.2019.10536] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 05/31/2019] [Indexed: 12/15/2022] Open
Abstract
Human bone marrow-derived mesenchymal stromal cells (hBMSCs) have been revealed to be beneficial for the regeneration of tissues and cells in several diseases. The present study aimed to elucidate the mechanisms underlying the effect of hBMSC transplantation on neuron regeneration in a rat model of middle cerebral artery occlusion (MCAO). The hBMSCs were isolated, cultured and identified. A rat model of MCAO was induced via the modified Longa method. Neurological severity scores (NSS) were adopted for the evaluation of neuronal function in the model rats after cell transplantation. Next, the expression levels of nestin, β-III-tubulin (β-III-Tub), glial fibrillary acidic protein (GFAP), HNA and neuronal nuclear antigen (NeuN) were examined, as well as the positive expression rates of human neutrophil alloantigen (HNA), nestin, NeuN, β-III-Tub and GFAP. The NSS, as well as the mRNA and protein expression of nestin, decreased at the 1st, 2nd, 4 and 8th weeks, while the mRNA and protein expression of NeuN, β-III-Tub and GFAP increased with time. In addition, after treatment, the MCAO rats showed decreased NSS and mRNA and protein expression of nestin, but elevated mRNA and protein expression of NeuN, β-III-Tub and GFAP at the 2nd, 4 and 8th weeks, and decreased positive expression of HNA and nestin with enhanced expression of NeuN, β-III-Tub and GFAP. Therefore, the present findings demonstrated that hBMSC transplantation triggered the formation of nerve cells and enhanced neuronal function in a rat model of MCAO.
Collapse
Affiliation(s)
- Ping Xie
- Department of Chinese Traditional Medicine, Tongren Hospital of Wuhan University (Wuhan Third Hospital), Wuhan, Hubei 430060, P.R. China
| | - Ming Deng
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qin-Guo Sun
- Department of Chinese Traditional Medicine, Tongren Hospital of Wuhan University (Wuhan Third Hospital), Wuhan, Hubei 430060, P.R. China
| | - Yong-Gang Ma
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yan Zhou
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jiang-Hua Ming
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qing Chen
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Shi-Qing Liu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jun-Qi Liu
- Department of Radiation Oncology, The First of Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 45003, P.R. China
| | - Jun Cai
- Department of Emergency and Trauma Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Fei Wu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
84
|
Ip Z, Rabiller G, He JW, Yao Z, Akamatsu Y, Nishijima Y, Liu J, Yazdan-Shahmorad A. Cortical stroke affects activity and stability of theta/delta states in remote hippocampal regions .. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2019; 2019:5225-5228. [PMID: 31947036 PMCID: PMC8523210 DOI: 10.1109/embc.2019.8857679] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cognitive impairment is a common outcome of ischemic stroke. Our previous work has shown that an experimental stroke in the cortex reduces activity in remote hippocampal layers in rats. This study seeks to uncover the underlying functional connections between these areas by analyzing changes to oscillatory activity, signal power, and communication. We induced an ischemic stroke in the left somatosensory cortex of rats and used linear micro-electrode arrays to simultaneously record from cortex and hippocampus under urethane anesthesia at two weeks and one month after stroke. We found significant increase in signal power, as well as an increase in the number of brain state changes in response to stroke. Our results suggest that the cortex modulates the activity and stability of hippocampal oscillations, which is disrupted following cortical stroke that can lead to cognitive impairment.
Collapse
|
85
|
Gudasheva TA, Povarnina PY, Volkova AA, Kruglov SV, Antipova TA, Seredenin SB. A Nerve Growth Factor Dipeptide Mimetic Stimulates Neurogenesis and Synaptogenesis in the Hippocampus and Striatum of Adult Rats with Focal Cerebral Ischemia. Acta Naturae 2019; 11:31-37. [PMID: 31720014 PMCID: PMC6826148 DOI: 10.32607/20758251-2019-11-3-31-37] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 06/06/2019] [Indexed: 11/20/2022] Open
Abstract
The nerve growth factor (NGF) and its mimetics, which have neuroprotective and neuroregenerative properties, are attractive candidates for developing new drugs for brain injury therapy. A dipeptide mimetic of NGF loop 4, bis(N-succinyl-L-glutamyl-L-lysine) hexamethylenediamide (GK-2), developed at the Zakusov Research Institute of Pharmacology, has the NGF-like ability to activate TrkA receptors, but unlike NGF, GK-2 activates mainly the PI3K/AKT pathway associated with neuroprotection and has no effect on the MAPK cascade associated with hyperalgesia, the main side effect of NGF. That GK-2 possesses neuroprotective activity has been observed in various models of cerebral ischemia. GK-2 was found to statistically significantly reduce the cerebral infarct volume in experimental stroke, even at treatment onset 24 h after injury. This suggests that GK-2 possesses neuroregenerative properties, which may be associated with the activation of neurogenesis and/or synaptogenesis. We studied the effect of GK-2 on neurogenesis and synaptogenesis in experimental ischemic stroke caused by transient occlusion of the middle cerebral artery in rats. GK-2 was administered 6 or 24 h after surgery and then once a day for 7 days. One day after the last administration, proliferative activity in the hippocampus and striatum of the affected hemisphere was assessed using Ki67 and synaptogenesis in the striatum was evaluated using synaptophysin and PSD-95. Ki67 immunoreactivity, both in the striatum and in the hippocampus of the ischemic rats, was found to have dropped by approximately 30% compared to that in the sham-operated controls. Synaptic markers - synaptophysin and PSD-95 - were also statistically significantly reduced, by 14 and 29%, respectively. GK-2 in both administration schedules completely restored the level of Ki67 immunoreactivity in the hippocampus and promoted its increase in the striatum. In addition, GK-2 restored the level of the postsynaptic marker PSD-95, with the therapeutic effect amounting to 70% at the start of its administration after 6 h, and promoted restoration of the level of this marker at the start of administration 24 h after an experimental stroke. GK-2 had no effect on the synaptophysin level. These findings suggest that the neurotrophin mimetic GK-2, which mainly activates one of the main Trk receptor signaling pathways PI3K/ AKT, has a stimulating effect on neurogenesis (and, probably, gliogenesis) and synaptogenesis in experimental cerebral ischemia. This effect may explain the protective effect observed at the start of dipeptide administration 24 h after stroke simulation.
Collapse
Affiliation(s)
- T. A. Gudasheva
- Federal State Budgetary Institution “Zakusov Research Institute of Pharmacology”, Baltiyskay Str. 8 , Moscow, 125315, Russia
| | - P. Yu. Povarnina
- Federal State Budgetary Institution “Zakusov Research Institute of Pharmacology”, Baltiyskay Str. 8 , Moscow, 125315, Russia
| | - A. A. Volkova
- Federal State Budgetary Institution “Zakusov Research Institute of Pharmacology”, Baltiyskay Str. 8 , Moscow, 125315, Russia
| | - S. V. Kruglov
- Federal State Budgetary Institution “Zakusov Research Institute of Pharmacology”, Baltiyskay Str. 8 , Moscow, 125315, Russia
| | - T. A. Antipova
- Federal State Budgetary Institution “Zakusov Research Institute of Pharmacology”, Baltiyskay Str. 8 , Moscow, 125315, Russia
| | - S. B. Seredenin
- Federal State Budgetary Institution “Zakusov Research Institute of Pharmacology”, Baltiyskay Str. 8 , Moscow, 125315, Russia
| |
Collapse
|
86
|
Shah FA, Kury LA, Li T, Zeb A, Koh PO, Liu F, Zhou Q, Hussain I, Khan AU, Jiang Y, Li S. Polydatin Attenuates Neuronal Loss via Reducing Neuroinflammation and Oxidative Stress in Rat MCAO Models. Front Pharmacol 2019; 10:663. [PMID: 31293416 PMCID: PMC6606791 DOI: 10.3389/fphar.2019.00663] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 05/23/2019] [Indexed: 12/14/2022] Open
Abstract
Ischemic stroke is characterized by permanent or transient obstruction of blood flow, which initiates a cascading pathological process, starting from acute ATP loss and ionic imbalance to subsequent membrane depolarization, glutamate excitotoxicity, and calcium overload. These initial events are followed by neuroinflammation and oxidative stress, eventually causing neuronal neurosis and apoptosis. Complicated interplays exist between these steps happening across various stages, which not only represent the complicated nature of ischemic pathology but also warrant a detailed delineation of the underlying molecular mechanisms to develop better therapeutic options. In the present study, we examined the neuroprotective effects of polydatin against ischemic brain injury using a rat model of permanent middle cerebral artery occlusion (MCAO). Our results demonstrated that polydatin treatment reduced the infarction volume and mitigated the neurobehavioral deficits, sequentially rescued neuronal apoptosis. Ischemic stroke induced an elevation of neuroinflammation and reactive oxygen species, which could be attenuated by polydatin via the reduced activation of p38 mitogen-activated protein kinase and c-Jun N-terminal kinase. In addition, polydatin upregulated the endogenous antioxidant nuclear factor erythroid 2-related factor 2, heme oxygenase-1, the thioredoxin pathway, and eventually reversed ischemic-stroke-induced elevation of ROS and inflammation in ischemic cortical tissue. The diverse and broad actions of polydatin suggested that it could be a multiple targeting neuroprotective agent in ameliorating the detrimental effects of MCAO, such as neuroinflammation, oxidative stress, and neuronal apoptosis. As repetitive clinical trials of neuroprotectants targeting a single step of stroke pathological process have failed previously, our results suggested that a neuroprotective strategy of acting at different stages may be more advantageous to intervene in the vicious cycles in MCAO.
Collapse
Affiliation(s)
- Fawad Ali Shah
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School, Peking University, Shenzhen, China.,Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Lina Al Kury
- College of Natural and Health Sciences, Zayed University, Abu Dhabi, United Arab Emirates
| | - Tao Li
- Department of Forensic Medicine, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Alam Zeb
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Phil Ok Koh
- Department of Anatomy, College of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, Jinju, South Korea
| | - Fang Liu
- Campbell Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Qiang Zhou
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School, Peking University, Shenzhen, China
| | | | - Arif Ullah Khan
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Yuhua Jiang
- Cancer Centre, The Second Hospital of Shandong University, Jinan, China
| | - Shupeng Li
- State Key Laboratory of Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School, Peking University, Shenzhen, China.,Campbell Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada.,Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
87
|
Neuroprotective Effects of Musk of Muskrat on Transient Focal Cerebral Ischemia in Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:9817949. [PMID: 31341507 PMCID: PMC6614976 DOI: 10.1155/2019/9817949] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 06/11/2019] [Indexed: 11/18/2022]
Abstract
Musk of musk deer has been one of the most precious traditional medicinal materials for treatment of stroke, but trading is prohibited. Musk of muskrat, Ondatra zibethicus, is an accessible substitute for musk of musk deer. However, neuroprotective effects of the musk of muskrat on stroke model are so far unclear. Aim of the study is to determine neuroprotective effects of the musk of muskrat on focal cerebral ischemia. The protective effects against focal cerebral ischemia were evaluated using a model of middle cerebral artery occlusion (90-minute occlusion followed by 24-hour reperfusion). Musk of muskrat was collected from scent bag of muskrat and orally administered at doses of 100 and 300 mg/kg twice at times of 0 and 90 min after occlusion. The effects on sensorimotor dysfunction were investigated by using balance beam test and rotarod test after brain ischemia. The expression of cyclooxygenase-2 (COX-2) was investigated by immunohistochemistry. Oral administration of musk at 300 mg/kg significantly reduced (p<0.001) the infarct volume by 32.4% compared with a vehicle-treated group. Oral administration of musk at 300 mg/kg also ameliorated ischemia-induced spontaneous and vestibule sensorimotor dysfunction in balance beam test and rotarod test compared with control group and COX-2 upregulation. Musk of muskrat may have neuroprotective effects against transient focal cerebral ischemia with recovery of sensorimotor dysfunction. Regarding the immunohistochemistry, the effects of muskrat may be due to anti-inflammatory properties through inhibition of COX-2 expressions.
Collapse
|
88
|
Wang Z, Wu L, Fang Q, Shen M, Zhang L, Liu X. Effects of capsaicin on swallowing function in stroke patients with dysphagia: A randomized controlled trial. J Stroke Cerebrovasc Dis 2019; 28:1744-1751. [DOI: 10.1016/j.jstrokecerebrovasdis.2019.02.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/05/2019] [Accepted: 02/11/2019] [Indexed: 12/19/2022] Open
|
89
|
Shi XF, Ai H, Lu W, Cai F. SAT: Free Software for the Semi-Automated Analysis of Rodent Brain Sections With 2,3,5-Triphenyltetrazolium Chloride Staining. Front Neurosci 2019; 13:102. [PMID: 30809120 PMCID: PMC6379447 DOI: 10.3389/fnins.2019.00102] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 01/28/2019] [Indexed: 11/22/2022] Open
Abstract
Ischemic stroke places an increasing burden on individuals, families, and societies around the world. However, effective therapies or drugs for ischemic stroke are lacking. Therefore, animal models mimicking ischemic stroke in humans are of great value for preclinical experiments. middle cerebral artery occlusion (MCAO) in mice or rats and subsequent 2,3,5-triphenyltetrazolium chloride (TTC) staining of brain sections are common methods in the study of experimental animal ischemic stroke. In this study, we present and assess the utility of the semi-automated analysis of the TTC staining (SAT) software program, a novel, small, user-friendly, and free software program, in the quantification of the infarct size in rodent brain sections, with TTC staining, by analyzing images captured by cell phones or scan systems. We performed MCAO and TTC staining in adult mice. We then utilized the SAT software and Image J to analyze the infarct size in the brain sections with TTC staining and compared the findings of the two analysis methods. We found that the data on infarct size from SAT and from Image J were comparable, suggesting that the SAT software could be an alternative option to Image J in the evaluation of ischemic stroke.
Collapse
Affiliation(s)
- Xiao-Fang Shi
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, China
| | - Heng Ai
- Department of Physiology, Hangzhou Medical College, Hangzhou, China
| | - Wen Lu
- Department of Biochemistry and Molecular Biology, Hainan Medical University, Haikou, China
| | - Fuhong Cai
- Department of Electrical Engineering, Mechanical and Electrical Engineering College, Hainan University, Haikou, China
| |
Collapse
|
90
|
Gandhi R, Tsoumpas C. Preclinical Imaging Biomarkers for Postischaemic Neurovascular Remodelling. CONTRAST MEDIA & MOLECULAR IMAGING 2019; 2019:3128529. [PMID: 30863220 PMCID: PMC6378027 DOI: 10.1155/2019/3128529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/22/2018] [Accepted: 12/04/2018] [Indexed: 11/30/2022]
Abstract
In the pursuit of understanding the pathological alterations that underlie ischaemic injuries, such as vascular remodelling and reorganisation, there is a need for recognising the capabilities and limitations of in vivo imaging techniques. Thus, this review presents contemporary published research of imaging modalities that have been implemented to study postischaemic neurovascular changes in small animals. A comparison of the technical aspects of the various imaging tools is included to set the framework for identifying the most appropriate methods to observe postischaemic neurovascular remodelling. A systematic search of the PubMed® and Elsevier's Scopus databases identified studies that were conducted between 2008 and 2018 to explore postischaemic neurovascular remodelling in small animal models. Thirty-five relevant in vivo imaging studies are included, of which most made use of magnetic resonance imaging or positron emission tomography, whilst various optical modalities were also utilised. Notably, there is an increasing trend of using multimodal imaging to exploit the most beneficial properties of each imaging technique to elucidate different aspects of neurovascular remodelling. Nevertheless, there is still scope for further utilising noninvasive imaging tools such as contrast agents or radiotracers, which will have the ability to monitor neurovascular changes particularly during restorative therapy. This will facilitate more successful utility of the clinical imaging techniques in the interpretation of neurovascular reorganisation over time.
Collapse
Affiliation(s)
- Richa Gandhi
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 9NL, West Yorkshire, UK
| | - Charalampos Tsoumpas
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 9NL, West Yorkshire, UK
| |
Collapse
|
91
|
Liu Q, Radwanski R, Babadjouni R, Patel A, Hodis DM, Baumbacher P, Zhao Z, Zlokovic B, Mack WJ. Experimental chronic cerebral hypoperfusion results in decreased pericyte coverage and increased blood-brain barrier permeability in the corpus callosum. J Cereb Blood Flow Metab 2019; 39:240-250. [PMID: 29192539 PMCID: PMC6365610 DOI: 10.1177/0271678x17743670] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Murine chronic cerebral hypoperfusion (CCH) results in white matter (WM) injury and behavioral deficits. Pericytes influence blood-brain barrier (BBB) integrity and cerebral blood flow. Under hypoxic conditions, pericytes detach from perivascular locations increasing vessel permeability and neuronal injury. This study characterizes the time course of BBB dysfunction and pericyte coverage following murine experimental CCH secondary to bilateral carotid artery stenosis (BCAS). Mice underwent BCAS or sham operation. On post-procedure days 1, 3, 7 and 30, corpus callosum BBB permeability was characterized using Evans blue (EB) extravasation and IgG staining and pericyte coverage/count was calculated. The BCAS cohort demonstrated increased EB extravasation on postoperative days 1 ( p = 0.003) 3 ( p = 0.002), and 7 ( p = 0.001) when compared to sham mice. Further, EB extravasation was significantly greater ( p = 0.05) at day 3 than at day 30 in BCAS mice. BCAS mice demonstrated a nadir in pericyte coverage and count on post-operative day 3 ( p < 0.05, compared to day 7, day 30 and sham). Decreased pericyte coverage/count and increased BBB permeability are most pronounced on postoperative day 3 following murine CCH. This precedes any notable WM injury or behavioral deficits.
Collapse
Affiliation(s)
- Qinghai Liu
- 1 Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Ryan Radwanski
- 1 Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Robin Babadjouni
- 1 Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Arati Patel
- 1 Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Drew M Hodis
- 1 Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Peter Baumbacher
- 1 Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Zhen Zhao
- 1 Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Berislav Zlokovic
- 1 Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - William J Mack
- 1 Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,2 Department of Neurosurgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
92
|
Zhang H, Lin S, Chen X, Gu L, Zhu X, Zhang Y, Reyes K, Wang B, Jin K. The effect of age, sex and strains on the performance and outcome in animal models of stroke. Neurochem Int 2018; 127:2-11. [PMID: 30291954 DOI: 10.1016/j.neuint.2018.10.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/01/2018] [Accepted: 10/02/2018] [Indexed: 12/26/2022]
Abstract
Stroke is one of the leading causes of death worldwide, and the majority of cerebral stroke is caused by occlusion of cerebral circulation, which eventually leads to brain infarction. Although stroke occurs mainly in the aged population, most animal models for experimental stroke in vivo almost universally rely on young-adult rodents for the evaluation of neuropathological, neurological, or behavioral outcomes after stroke due to their greater availability, lower cost, and fewer health problems. However, it is well established that aged animals differ from young animals in terms of physiology, neurochemistry, and behavior. Stroke-induced changes are more pronounced with advancing age. Therefore, the overlooked role of age in animal models of stroke could have an impact on data quality and hinder the translation of rodent models to humans. In addition to aging, other factors also influence functional performance after ischemic stroke. In this article, we summarize the differences between young and aged animals, the impact of age, sex and animal strains on performance and outcome in animal models of stroke and emphasize age as a key factor in preclinical stroke studies.
Collapse
Affiliation(s)
- Hongxia Zhang
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Siyang Lin
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xudong Chen
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Lei Gu
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xiaohong Zhu
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yinuo Zhang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Kassandra Reyes
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Brian Wang
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Kunlin Jin
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
93
|
Morihara R, Yamashita T, Liu X, Nakano Y, Fukui Y, Sato K, Ohta Y, Hishikawa N, Shang J, Abe K. Protective effect of a novel sigma-1 receptor agonist is associated with reduced endoplasmic reticulum stress in stroke male mice. J Neurosci Res 2018; 96:1707-1716. [PMID: 30102416 DOI: 10.1002/jnr.24270] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 05/22/2018] [Accepted: 05/29/2018] [Indexed: 12/22/2022]
Abstract
Sigma-1 receptor (Sig-1R) is expressed at endoplasmic reticulum (ER) membranes, where it regulates a variety of specific physiological functions. However, the profile and exact roles of ER stress-related molecules after Sig-1R agonist treatment in an in vivo stroke model are largely unknown. The aim of this study is to investigate the effect of a novel Sig-1R agonist, aniline derivative compound (Comp-AD), on the ER stress response following ischemic stroke. Male C57BL/6J mice received transient middle cerebral artery occlusion for 90 min, and were then treated with vehicle saline or Comp-AD at reperfusion. At 3 hr, 1 day, and 7 days after reperfusion, immunohis- tochemistry was performed for Sig-1R and ER stress-related proteins including phospho protein kinase RNA-like endoplasmic reticulum kinase (p-PERK), phospho inositol requiring enzyme 1α (p- IRE1α), and activating transcription factor 6 (ATF6). Neurobehavioral analysis showed improved functional recovery at 1 day and 7 days after reperfusion, and the infarct volume was significantly smaller at 7 days (p < .05), in the Comp-AD group compared with the vehicle group. Comp-AD treatment upregulated Sig-1R immunoreactivity at 3 hr and 1 day (p < .05), and reduced p-PERK and p-IRE1α expression at 1 day (p < .05, respectively), in the peri-ischemic region compared with the vehicle group. Treatment with the novel Sig-1R agonist Comp-AD was neuroprotective after transient middle cerebral artery occlusion, and was associated with upregulation of Sig-1R and a reduction of ER stress.
Collapse
Affiliation(s)
- Ryuta Morihara
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Toru Yamashita
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Xia Liu
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yumiko Nakano
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yusuke Fukui
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kota Sato
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yasuyuki Ohta
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Nozomi Hishikawa
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Jingwei Shang
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Koji Abe
- Department of Neurology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
94
|
Dergunova LV, Filippenkov IB, Stavchansky VV, Denisova AE, Yuzhakov VV, Mozerov SA, Gubsky LV, Limborska SA. Genome-wide transcriptome analysis using RNA-Seq reveals a large number of differentially expressed genes in a transient MCAO rat model. BMC Genomics 2018; 19:655. [PMID: 30185153 PMCID: PMC6125876 DOI: 10.1186/s12864-018-5039-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 08/27/2018] [Indexed: 01/29/2023] Open
Abstract
Background The transient middle cerebral artery occlusion (tMCAO) model is used for studying the molecular mechanisms of ischemic damage and neuroprotection. Numerous studies have demonstrated the role of individual genes and associated signaling pathways in the pathogenesis of ischemic stroke. Here, the tMCAO model was used to investigate the genome-wide response of the transcriptome of rat brain tissues to the damaging effect of ischemia and subsequent reperfusion. Results Magnetic resonance imaging and histological examination showed that the model of focal ischemia based on endovascular occlusion of the right middle cerebral artery for 90 min using a monofilament, followed by restoration of the blood flow, led to reproducible localization of ischemic damage in the subcortical structures of the brain. High-throughput RNA sequencing (RNA-Seq) revealed the presence of differentially expressed genes (DEGs) in subcortical structures of rat brains resulting from hemisphere damage by ischemia after tMCAO, as well as in the corresponding parts of the brains of sham-operated animals. Real-time reverse transcription polymerase chain reaction expression analysis of 20 genes confirmed the RNA-Seq results. We identified 469 and 1939 genes that exhibited changes in expression of > 1.5-fold at 4.5 and 24 h after tMCAO, respectively. Interestingly, we found 2741 and 752 DEGs under ischemia–reperfusion and sham-operation conditions at 24 h vs. 4.5 h after tMCAO, respectively. The activation of a large number of genes involved in inflammatory, immune and stress responses, apoptosis, ribosome function, DNA replication and other processes was observed in ischemia–reperfusion conditions. Simultaneously, massive down-regulation of the mRNA levels of genes involved in the functioning of neurotransmitter systems was recorded. A Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis showed that dozens of signaling pathways were associated with DEGs in ischemia–reperfusion conditions. Conclusions The data obtained revealed a global profile of gene expression in the rat brain sub-cortex under tMCAO conditions that can be used to identify potential therapeutic targets in the development of new strategies for the prevention and treatment of ischemic stroke. Electronic supplementary material The online version of this article (10.1186/s12864-018-5039-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lyudmila V Dergunova
- Human Molecular Genetics Department, Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, Russian Federation. .,Research Institute of Cerebrovascular Pathology and Stroke, Pirogov Russian National Research Medical University, Moscow, Russian Federation.
| | - Ivan B Filippenkov
- Human Molecular Genetics Department, Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, Russian Federation
| | - Vasily V Stavchansky
- Human Molecular Genetics Department, Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, Russian Federation
| | - Alina E Denisova
- Research Institute of Cerebrovascular Pathology and Stroke, Pirogov Russian National Research Medical University, Moscow, Russian Federation
| | - Vadim V Yuzhakov
- A. Tsyb Medical Radiological Research Center - branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russian Federation
| | - Sergey A Mozerov
- A. Tsyb Medical Radiological Research Center - branch of the National Medical Research Radiological Center of the Ministry of Health of the Russian Federation, Obninsk, Russian Federation
| | - Leonid V Gubsky
- Research Institute of Cerebrovascular Pathology and Stroke, Pirogov Russian National Research Medical University, Moscow, Russian Federation
| | - Svetlana A Limborska
- Human Molecular Genetics Department, Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, Russian Federation.,Research Institute of Cerebrovascular Pathology and Stroke, Pirogov Russian National Research Medical University, Moscow, Russian Federation
| |
Collapse
|
95
|
King ZA, Sheth KN, Kimberly WT, Simard JM. Profile of intravenous glyburide for the prevention of cerebral edema following large hemispheric infarction: evidence to date. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:2539-2552. [PMID: 30147301 PMCID: PMC6101021 DOI: 10.2147/dddt.s150043] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Glyburide (also known as glibenclamide) is a second-generation sulfonylurea drug that inhibits sulfonylurea receptor 1 (Sur1) at nanomolar concentrations. Long used to target KATP (Sur1–Kir6.2) channels for the treatment of diabetes mellitus type 2, glyburide was recently repurposed to target Sur1–transient receptor potential melastatin 4 (Trpm4) channels in acute central nervous system injury. Discovered nearly two decades ago, SUR1–TRPM4 has emerged as a critical target in stroke, specifically in large hemispheric infarction, which is characterized by edema formation and life-threatening brain swelling. Following ischemia, SUR1–TRPM4 channels are transcriptionally upregulated in all cells of the neurovascular unit, including neurons, astrocytes, microglia, oligodendrocytes and microvascular endothelial cells. Work by several independent laboratories has linked SUR1–TRPM4 to edema formation, with blockade by glyburide reducing brain swelling and death in preclinical models. Recent work showed that, following ischemia, SUR1–TRPM4 co-assembles with aquaporin-4 to mediate cellular swelling of astrocytes, which contributes to brain swelling. Additionally, recent work linked SUR1–TRPM4 to secretion of matrix metalloproteinase-9 (MMP-9) induced by recombinant tissue plasminogen activator in activated brain endothelial cells, with blockade of SUR1–TRPM4 by glyburide reducing MMP-9 and hemorrhagic transformation in preclinical models with recombinant tissue plasminogen activator. The recently completed GAMES (Glyburide Advantage in Malignant Edema and Stroke) clinical trials on patients with large hemispheric infarctions treated with intravenous glyburide (RP-1127) revealed promising findings with regard to brain swelling (midline shift), MMP-9, functional outcomes and mortality. Here, we review key elements of the basic science, preclinical experiments and clinical studies, both retrospective and prospective, on glyburide in focal cerebral ischemia and stroke.
Collapse
Affiliation(s)
- Zachary A King
- Department of Neurology, Division of Neurocritical Care and Emergency Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Kevin N Sheth
- Department of Neurology, Division of Neurocritical Care and Emergency Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - W Taylor Kimberly
- Department of Neurology, Division of Neurocritical Care and Emergency Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - J Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, USA,
| |
Collapse
|
96
|
Gubskiy IL, Namestnikova DD, Cherkashova EA, Chekhonin VP, Baklaushev VP, Gubsky LV, Yarygin KN. MRI Guiding of the Middle Cerebral Artery Occlusion in Rats Aimed to Improve Stroke Modeling. Transl Stroke Res 2018; 9:417-425. [PMID: 29178027 PMCID: PMC6061245 DOI: 10.1007/s12975-017-0590-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 11/11/2017] [Accepted: 11/17/2017] [Indexed: 01/30/2023]
Abstract
The middle cerebral artery occlusion (MCAO) model in rats closely imitates ischemic stroke and is widely used. Existing instrumental methods provide a certain level of MCAO guidance, but monitoring of the MCA-occluding intraluminal filament position and possible complications can be improved. The goal of this study was to develop a MRI-based method of simultaneous control of the filament position, blood flow in the intracranial vessels, and hemorrhagic complications. Rats were subjected to either MRI-guided MCAO (group 1, n = 51) or MCAO without MRI control (group 2, n = 38). After operation, group 1 rats were transferred into a MRI scanner for the control of the filament position and possible complications. Ninety minutes after the onset of MCAO, the filament was removed in rats of both groups and MRI control of the infarct volume and hemorrhagic complications performed. High-resolution T1- and T2-weighted imaging performed immediately after filament insertion provided visualization of the filament position, blood flow in brain arteries, and complications related to inappropriate filament insertion. It permitted replacement of wrongly positioned filaments and exclusion of animals with complications from the experiment. MRI-based MCAO guiding provided real-time intra-operational monitoring of crucial parameters determining MCAO suitability for stroke modeling, including better assessment of the operation outcomes in individual animals and significant enhancement of the model success rate. The possibility of simultaneous visualization of the filament, blood flow in the arteries, brain tissue, and hemorrhagic complications is the principal advantage of the proposed method over other instrumental methods of MCAO quality control. Graphical Abstract MRI-guided middle cerebral artery occlusion technique permits intra-operational monitoring via direct non-invasive simultaneous visualization of the filament, blood flow in the arteries, brain tissue, and hemorrhagic complications. It provides better assessment of MCAO outcomes in individual animals and significant enhancement of MCAO success rate.
Collapse
Affiliation(s)
- Ilya L Gubskiy
- Research Institute of Cerebrovascular Pathology and Stroke, Pirogov Russian National Research Medical University, Moscow, Russia.
| | - Daria D Namestnikova
- Department of Neurology, Neurosurgery and Medical Genetics, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Elvira A Cherkashova
- Department of Neurology, Neurosurgery and Medical Genetics, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Vladimir P Chekhonin
- Serbsky Federal Medical Research Centre of Psychiatry and Narcology, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Vladimir P Baklaushev
- Federal Research Clinical Center of Specialized Medical Care and Medical Technologies of the FMBA of Russia, Moscow, Russia
| | - Leonid V Gubsky
- Research Institute of Cerebrovascular Pathology and Stroke, Pirogov Russian National Research Medical University, Moscow, Russia
| | | |
Collapse
|
97
|
Drieu A, Levard D, Vivien D, Rubio M. Anti-inflammatory treatments for stroke: from bench to bedside. Ther Adv Neurol Disord 2018; 11:1756286418789854. [PMID: 30083232 PMCID: PMC6066814 DOI: 10.1177/1756286418789854] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 06/19/2018] [Indexed: 12/11/2022] Open
Abstract
So far, intravenous tissue-type plasminogen activator (tPA) and mechanical
removal of arterial blood clot (thrombectomy) are the only available treatments
for acute ischemic stroke. However, the short therapeutic window and the lack of
specialized stroke unit care make the overall availability of both treatments
limited. Additional agents to combine with tPA administration or thrombectomy to
enhance efficacy and improve outcomes associated with stroke are needed.
Stroke-induced inflammatory processes are a response to the tissue damage due to
the absence of blood supply but have been proposed also as key contributors to
all the stages of the ischemic stroke pathophysiology. Despite promising results
in experimental studies, inflammation-modulating treatments have not yet been
translated successfully into the clinical setting. This review will (a) describe
the timing of the stroke immune pathophysiology; (b) detail the immune responses
to stroke sift-through cell type; and (c) discuss the pitfalls on the
translation from experimental studies to clinical trials testing the therapeutic
pertinence of immune modulators.
Collapse
Affiliation(s)
- Antoine Drieu
- Pathophysiology and Imaging of Neurological Disorders, Normandy University, Caen, France
| | - Damien Levard
- Pathophysiology and Imaging of Neurological Disorders, Normandy University, Caen, France
| | - Denis Vivien
- Pathophysiology and Imaging of Neurological Disorders, Normandy University, Caen, France Pathophysiology and Imaging of Neurological Disorders, Centre Hospitalier Universitaire de Caen, Caen, France
| | - Marina Rubio
- Pathophysiology and Imaging of Neurological Disorders, Normandy University, Boulevard Henri Becquerel BP 5229, Caen Cedex, 14000, France
| |
Collapse
|
98
|
Abstract
OBJECTIVE Stroke patients often suffer from delayed disturbances of mood and cognition. In rodents, the prefrontal cortex (PFC) is involved in both higher order cognition and emotion. Our objective was to determine if bilateral focal ischaemic lesions restricted to the medial prefrontal cortex (mPFC) could be used to model post-stroke anxiety and/or cognitive deficits. METHODS Groups of adult male Sprague-Dawley rats (n=9) received bilateral injections of either endothelin-1 (ET-1) (400 pmol) or vehicle (artificial cerebrospinal fluid) into the mPFC and were tested at various times using both a test of temporal order memory and in an elevated plus maze. Lesions were verified histologically. RESULTS ET-1 lesioned rats had reduced mobility on post-surgery day 8 that had resolved by day 29 at which time they spent significantly more time in the closed arm of the plus maze CONCLUSION: We conclude that ischaemic lesions localised to the mPFC can be used to model post-stroke anxiety in rats.
Collapse
|
99
|
Kotoda M, Furukawa H, Miyamoto T, Korai M, Shikata F, Kuwabara A, Xiong X, Rutledge C, Giffard RG, Hashimoto T. Role of Myeloid Lineage Cell Autophagy in Ischemic Brain Injury. Stroke 2018; 49:1488-1495. [PMID: 29748423 DOI: 10.1161/strokeaha.117.018637] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 04/06/2018] [Accepted: 04/12/2018] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND PURPOSE Inflammatory cells play a significant role in secondary injury after ischemic stroke. Recent studies have suggested that a lack of autophagy in myeloid cells causes augmented proinflammatory cytokine release and prolonged inflammation after tissue injury. In this study, we investigated the roles of myeloid cell autophagy in ischemic brain injury. METHODS Focal cerebral ischemia was induced via transient middle cerebral artery occlusion in mice with autophagy-deficient myeloid lineage cells (Atg5flox/flox LysMCre+) and in their littermate controls (Atg5flox/flox). Infarct volume, neurological function, inflammatory cell infiltration, and proinflammatory cytokine expression levels were evaluated. RESULTS Mice lacking autophagy in myeloid lineage cells had a lower survival rate for 14 days than control mice (20% versus 70%; P<0.05). Although there was no difference in infarct volume at 12 hours between the 2 groups, mice lacking autophagy in myeloid lineage cells had larger infarct volumes at later time points (3 and 7 days after reperfusion) with worse neurological deficit scores and lower grip test scores. There were a higher number of ionized calcium binding adaptor molecule 1-positive cells and cells expressing M1 marker CD16/32 in mice lacking autophagy in myeloid cells at the later time points. Moreover, these mice had higher expression levels of proinflammatory cytokines at later time points; however, there was no difference in ionized calcium binding adaptor molecule 1-positive cells or mRNA levels of proinflammatory cytokines at the earlier time point (12 hours after reperfusion). CONCLUSIONS These data suggest that the lack of myeloid cell autophagy aggravates secondary injury by augmenting and prolonging inflammation after ischemic stroke without affecting the initial injury.
Collapse
Affiliation(s)
- Masakazu Kotoda
- From the Departments of Anesthesia and Perioperative Care (M.K., H.F., T.M., M.K., F.S., A.K., T.H.)
| | - Hajime Furukawa
- From the Departments of Anesthesia and Perioperative Care (M.K., H.F., T.M., M.K., F.S., A.K., T.H.)
| | - Takeshi Miyamoto
- From the Departments of Anesthesia and Perioperative Care (M.K., H.F., T.M., M.K., F.S., A.K., T.H.)
- Department of Neurosurgery and Neurobiology, Barrow Neurological Institute, Phoenix, AZ (T.M., T.H.)
| | - Masaaki Korai
- From the Departments of Anesthesia and Perioperative Care (M.K., H.F., T.M., M.K., F.S., A.K., T.H.)
| | - Fumiaki Shikata
- From the Departments of Anesthesia and Perioperative Care (M.K., H.F., T.M., M.K., F.S., A.K., T.H.)
| | - Atsushi Kuwabara
- From the Departments of Anesthesia and Perioperative Care (M.K., H.F., T.M., M.K., F.S., A.K., T.H.)
| | - Xiaoxing Xiong
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, CA (X.X., R.G.G.)
| | - Caleb Rutledge
- Neurological Surgery (C.R., T.H.), University of California, San Francisco
| | - Rona G Giffard
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, CA (X.X., R.G.G.)
| | - Tomoki Hashimoto
- From the Departments of Anesthesia and Perioperative Care (M.K., H.F., T.M., M.K., F.S., A.K., T.H.)
- Neurological Surgery (C.R., T.H.), University of California, San Francisco
- Department of Neurosurgery and Neurobiology, Barrow Neurological Institute, Phoenix, AZ (T.M., T.H.)
| |
Collapse
|
100
|
Indole-3-carbinol improves neurobehavioral symptoms in a cerebral ischemic stroke model. Naunyn Schmiedebergs Arch Pharmacol 2018; 391:613-625. [PMID: 29602953 DOI: 10.1007/s00210-018-1488-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 03/19/2018] [Indexed: 12/27/2022]
Abstract
Stroke is one of the most common causes of death worldwide and also responsible for permanent disability. Ischemic stroke has been found to affect 80% of stroke patients. Recombinant tissue plasminogen activator (rtPA) is the widely used drug for the ischemic stroke with narrow therapeutic window. Indole-3-carbinol (I3C) is a natural compound obtained from brassica species having antithrombotic activity. Middle cerebral artery occlusion (MCAO) model was used followed by reperfusion after 2 h of ischemia for the evaluation of the I3C against ischemic stroke. After reperfusion, I3C (12.5, 25, and 50 mg/kg) was given by oral route once daily and continued up to the 14th day. Behavioral studies including postural reflex, forelimb placing, and cylinder tests showed I3C attenuated the MCAO-induced increase in average score and asymmetry score efficiently. Mean cerebral blood flow (CBF) was improved by treatment with I3C (12.5 mg/kg) by 60% of baseline at 6 h. I3C inhibited ADP-induced platelet aggregation and reduced ischemic volume significantly. It also inhibited in vitro the ADP-induced platelet aggregation in healthy human volunteers. I3C improves behavioral scores and mean CBF after focal cerebral ischemia in rats. Furthermore, I3C showed prophylactic anti-thrombotic activity against carrageenan induced tail thrombosis. Therefore, preclinical evidence points to I3C as a potential candidate for use in cerebral ischemic stroke.
Collapse
|