1
|
Liang R, Hou X, Zhou D, Zhu L, Teng L, Song W, Tang Q. Exercise preconditioning mitigates Ischemia-Reperfusion injury in rats by enhancing mitochondrial respiration. Neuroscience 2024; 562:64-74. [PMID: 39461659 DOI: 10.1016/j.neuroscience.2024.10.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 10/29/2024]
Abstract
Cerebral ischemia and subsequent reperfusion damage are prevalent in clinical practice, linked to numerous neurodegenerative diseases. Cerebral ischemia deprives brain tissue of essential oxygen and nutrients, disrupting energy metabolism and causing cellular dysfunction. Although reperfusion theoretically aids recovery, it instead initiates complex injury responses such as oxidative stress, apoptosis, and inflammation, worsening brain damage. Recent research suggests that enhancing neuronal energy status by modulating energy metabolism pathways can effectively counter these effects. For instance, boosting mitochondrial function, improving energy provision, and decreasing harmful metabolites can mitigate oxidative stress and cellular injury. This study investigated the protective effects of exercise preconditioning against ischemia-reperfusion injury in rats. It was observed that exercise enhances energy levels and mitochondrial respiration by upregulating the expression of COX4 and NAMPT proteins and activating AMPK and mitochondrial complex V. This process facilitates metabolic reprogramming characterized by the promotion of oxidative phosphorylation (OXPHOS) and the pentose phosphate pathway (PPP), alongside a reduction in glycolysis. Such reprogramming reduces harmful metabolites, mitigating apoptosis and oxidative stress, and is a key factor in alleviating acute ischemic hypoxia-induced brain damage. These findings introduce a novel therapeutic approach for ischemic brain reperfusion injury, underscoring the crucial role of ATP production and metabolic regulation in neuroprotection.
Collapse
Affiliation(s)
- Runyu Liang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xinlei Hou
- Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Daguo Zhou
- Xiang'an Hospital of Xiamen University, China
| | - Luwen Zhu
- Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Lili Teng
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Wenjing Song
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qiang Tang
- Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China.
| |
Collapse
|
2
|
Yoshimura S, Dorok M, Mamrak U, Wehn A, Krestel E, Khalin I, Plesnila N. Reliable infarction of the middle cerebral artery territory in C57BL/6 mice using pterygopalatine artery ligation and filament optimization - The PURE-MCAo model. J Cereb Blood Flow Metab 2024:271678X241281841. [PMID: 39370987 PMCID: PMC11563556 DOI: 10.1177/0271678x241281841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/30/2024] [Accepted: 08/22/2024] [Indexed: 10/08/2024]
Abstract
Current techniques for inducing intraluminal filamentous middle cerebral artery occlusion (fMCAo) in mice produce highly variable results and often cause additional infarcts in the posterior cerebral artery (PCA) territory. The aim of the current study was to develop a novel procedure to overcome these shortcomings. Male C57BL/6 mice were subjected to 60 min of fMCAo with cerebral blood flow monitored by laser Doppler flowmetry. The influence of the length of the occlusion filament coating and the combination of common carotid artery (CCA) or pterygopalatine artery (PPA) ligation on lesion volume and functional outcome 24 h after reperfusion was evaluated. The use of appropriate filament and PPA ligation while maintaining CCA perfusion prevented the development of infarcts in the PCA area, resulted in pure MCA infarcts (68.3 ± 14.5 mm3) and reduced the variability of infarct volumes by more than half (from 26-38% to 14% standard deviation/mean). Using an improved fMCAo procedure, we were able to produce PCA area-unaffected reproducible (PURE) infarcts exclusively in the MCA territory. Thus PURE-MCAo reduced outcome variability by more than 50%. Our results may thus help to reduce the number of animals in preclinical stroke research and to increase the reproducibility of the fMCAo model.
Collapse
Affiliation(s)
- Sodai Yoshimura
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University Munich (LMU), Germany
- Department of Neurosurgery, Nihon University School of Medicine, Tokyo, Japan
| | - Maximilian Dorok
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University Munich (LMU), Germany
| | - Uta Mamrak
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University Munich (LMU), Germany
| | - Antonia Wehn
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University Munich (LMU), Germany
- Department of Neurosurgery, LMU University Hospital, Munich, Germany
| | - Eva Krestel
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University Munich (LMU), Germany
| | - Igor Khalin
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University Munich (LMU), Germany
- Normandie University, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institute Blood and Brain @ Caen-Normandie (BB@C), Caen, France
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University Munich (LMU), Germany
- Munich Cluster for Systems Neurology (Synergy), Munich, Germany
| |
Collapse
|
3
|
Jin X, Imai T, Morais A, Sasaki Y, Chung DY, Ayata C. Hippocampal infarction and generalized seizures predict early mortality after endovascular middle cerebral artery occlusion in mice. Exp Neurol 2024; 380:114903. [PMID: 39079623 PMCID: PMC11347107 DOI: 10.1016/j.expneurol.2024.114903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/08/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024]
Abstract
Endovascular middle cerebral artery occlusion (MCAO) is a widely used experimental ischemic stroke model. However, the model carries high early mortality. Our aim was to investigate the factors that influence early mortality within 48 h of reperfusion after transient MCAO. Using C57BL/6 mice, we induced 1-hour endovascular filament MCAO. To introduce heterogeneity of infarct volumes, a subset of animals had additional tandem common carotid artery occlusion (MCAO+CCAO). Continuous video monitoring was used to gain insight into the cause of death. Mortality within 48 h was 25% in the pooled cohort. All animals with early mortality suffered from infarcts in the hippocampus, sometimes accompanied by infarcts in the thalamus and midbrain, which occurred exclusively in the MCAO+CCAO group. All animals with early mortality developed convulsive seizures captured on video monitoring. None of the animals that did not develop convulsive seizures died. Among the three regions, hippocampal infarction appeared necessary for convulsive seizures and early mortality. Our data highlight seizures as the primary cause of mortality within the first 48 h after endovascular filament MCAO, linked to hippocampal infarction. Since hippocampal blood supply is mainly from the posterior cerebral artery (PCA), avoiding concurrent PCA ischemia can decrease mortality in proximal MCAO models.
Collapse
Affiliation(s)
- Xuyan Jin
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - Takahiko Imai
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - Andreia Morais
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - Yuichi Sasaki
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA
| | - David Y Chung
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA; Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Cenk Ayata
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, USA; Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
4
|
Beard DJ, Brown LS, Morris GP, Couch Y, Adriaanse BA, Karali CS, Schneider AM, Howells DW, Redzic ZB, Sutherland BA, Buchan AM. Rapamycin Treatment Reduces Brain Pericyte Constriction in Ischemic Stroke. Transl Stroke Res 2024:10.1007/s12975-024-01298-x. [PMID: 39331260 DOI: 10.1007/s12975-024-01298-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 07/18/2024] [Accepted: 09/09/2024] [Indexed: 09/28/2024]
Abstract
The contraction and subsequent death of brain pericytes may play a role in microvascular no-reflow following the reopening of an occluded artery during ischemic stroke. Mammalian target of rapamycin (mTOR) inhibition has been shown to reduce motility/contractility of various cancer cell lines and reduce neuronal cell death in stroke. However, the effects of mTOR inhibition on brain pericyte contraction and death during ischemia have not yet been investigated. Cultured pericytes exposed to simulated ischemia for 12 h in vitro contracted after less than 1 h, which was about 7 h prior to cell death. Rapamycin significantly reduced the rate of pericyte contraction during ischemia; however, it did not have a significant effect on pericyte viability at any time point. Rapamycin appeared to reduce pericyte contraction through a mechanism that is independent of changes in intracellular calcium. Using a mouse model of middle cerebral artery occlusion, we showed that rapamycin significantly increased the diameter of capillaries underneath pericytes and increased the number of open capillaries 30 min following recanalisation. Our findings suggest that rapamycin may be a useful adjuvant therapeutic to reduce pericyte contraction and improve cerebral reperfusion post-stroke.
Collapse
Affiliation(s)
- Daniel J Beard
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia.
| | - Lachlan S Brown
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Australia
| | - Gary P Morris
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Australia
| | - Yvonne Couch
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Bryan A Adriaanse
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | | | - Anna M Schneider
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - David W Howells
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Australia
| | - Zoran B Redzic
- Department of Physiology, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Brad A Sutherland
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Australia.
| | - Alastair M Buchan
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
5
|
Wang J, Li Y, Qi L, Mamtilahun M, Liu C, Liu Z, Shi R, Wu S, Yang GY. Advanced rehabilitation in ischaemic stroke research. Stroke Vasc Neurol 2024; 9:328-343. [PMID: 37788912 PMCID: PMC11420926 DOI: 10.1136/svn-2022-002285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/20/2023] [Indexed: 10/05/2023] Open
Abstract
At present, due to the rapid progress of treatment technology in the acute phase of ischaemic stroke, the mortality of patients has been greatly reduced but the number of disabled survivors is increasing, and most of them are elderly patients. Physicians and rehabilitation therapists pay attention to develop all kinds of therapist techniques including physical therapy techniques, robot-assisted technology and artificial intelligence technology, and study the molecular, cellular or synergistic mechanisms of rehabilitation therapies to promote the effect of rehabilitation therapy. Here, we discussed different animal and in vitro models of ischaemic stroke for rehabilitation studies; the compound concept and technology of neurological rehabilitation; all kinds of biological mechanisms of physical therapy; the significance, assessment and efficacy of neurological rehabilitation; the application of brain-computer interface, rehabilitation robotic and non-invasive brain stimulation technology in stroke rehabilitation.
Collapse
Affiliation(s)
- Jixian Wang
- Department of Rehabilitation Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medical, Shanghai, China
| | - Yongfang Li
- Department of Rehabilitation Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medical, Shanghai, China
| | - Lin Qi
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Muyassar Mamtilahun
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Chang Liu
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Ze Liu
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Rubing Shi
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Shengju Wu
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Guo-Yuan Yang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
6
|
Koukalova L, Chmelova M, Amlerova Z, Vargova L. Out of the core: the impact of focal ischemia in regions beyond the penumbra. Front Cell Neurosci 2024; 18:1336886. [PMID: 38504666 PMCID: PMC10948541 DOI: 10.3389/fncel.2024.1336886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 02/08/2024] [Indexed: 03/21/2024] Open
Abstract
The changes in the necrotic core and the penumbra following induction of focal ischemia have been the focus of attention for some time. However, evidence shows, that ischemic injury is not confined to the primarily affected structures and may influence the remote areas as well. Yet many studies fail to probe into the structures beyond the penumbra, and possibly do not even find any significant results due to their short-term design, as secondary damage occurs later. This slower reaction can be perceived as a therapeutic opportunity, in contrast to the ischemic core defined as irreversibly damaged tissue, where the window for salvation is comparatively short. The pathologies in remote structures occur relatively frequently and are clearly linked to the post-stroke neurological outcome. In order to develop efficient therapies, a deeper understanding of what exactly happens in the exo-focal regions is necessary. The mechanisms of glia contribution to the ischemic damage in core/penumbra are relatively well described and include impaired ion homeostasis, excessive cell swelling, glutamate excitotoxic mechanism, release of pro-inflammatory cytokines and phagocytosis or damage propagation via astrocytic syncytia. However, little is known about glia involvement in post-ischemic processes in remote areas. In this literature review, we discuss the definitions of the terms "ischemic core", "penumbra" and "remote areas." Furthermore, we present evidence showing the array of structural and functional changes in the more remote regions from the primary site of focal ischemia, with a special focus on glia and the extracellular matrix. The collected information is compared with the processes commonly occurring in the ischemic core or in the penumbra. Moreover, the possible causes of this phenomenon and the approaches for investigation are described, and finally, we evaluate the efficacy of therapies, which have been studied for their anti-ischemic effect in remote areas in recent years.
Collapse
Affiliation(s)
- Ludmila Koukalova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Martina Chmelova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| | - Zuzana Amlerova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Lydia Vargova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
7
|
Bai X, Qiu Y, Wang J, Dong Y, Zhang T, Jin H. Panax quinquefolium saponins attenuates microglia activation following acute cerebral ischemia-reperfusion injury via Nrf2/miR-103-3p/TANK pathway. Cell Biol Int 2024; 48:201-215. [PMID: 37885132 DOI: 10.1002/cbin.12100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 09/20/2023] [Accepted: 10/12/2023] [Indexed: 10/28/2023]
Abstract
Ischemic stroke is one of the leading causes of death and disability among adults worldwide. Intravenous thrombolysis is the only approved pharmacological treatment for acute ischemic stroke. However, reperfusion by thrombolysis will lead to the rapid activation of microglia cells which induces interferon-inflammatory response in the ischemic brain tissues. Panax quinquefolium saponins (PQS) has been proven to be effective in acute ischemic stroke, but there is no unified understanding about its specific mechanism. Here, we will report for the first time that PQS can significantly inhibit the activation of microglia cells in cerebral of MCAO rats via activation of Nrf2/miR-103-3p/TANK axis. Our results showed that PQS can directly bind to Nrf2 protein and inhibit its ubiquitination, which result in the indirectly enhancing the expression of TANK protein via transcriptional regulation on miR-103-3p, and finally to suppress the nuclear factor kappa-B dominated rapid activation of microglial cells induced by oxygen-glucose deprivation/reoxygenation vitro and cerebral ischemia-reperfusion injury in vivo. In conclusion, our study not only revealed the new mechanism of PQS in protecting against the inflammatory activation of microglia cells caused by cerebral ischemia-reperfusion injury, but also suggested that Nrf2 is a potential target for development of new drugs of ischemic stroke. More importantly, our study also reminded that miR-103-3p might be used as a prognostic biomarker for patients with ischemic stroke.
Collapse
Affiliation(s)
- Xuesong Bai
- Department of Pharmacy, Shanghai Pudong New Area People's Hospital, Shanghai, China
| | - Yan Qiu
- Department of Pharmacy, Shanghai Pudong New Area People's Hospital, Shanghai, China
| | - Jian Wang
- Department of Pharmacy, Shanghai Pudong New Area People's Hospital, Shanghai, China
| | - Yafen Dong
- Department of Pharmacy, Shanghai Pudong New Area People's Hospital, Shanghai, China
| | - Tao Zhang
- Department of Laboratory Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hui Jin
- Department of Pharmacy, Shanghai Pudong New Area People's Hospital, Shanghai, China
| |
Collapse
|
8
|
Huang Y, Han M, Shi Q, Li X, Mo J, Liu Y, Chu Z, Li W. Li, P HY-021068 alleviates cerebral ischemia-reperfusion injury by inhibiting NLRP1 inflammasome and restoring autophagy function in mice. Exp Neurol 2024; 371:114583. [PMID: 37884189 DOI: 10.1016/j.expneurol.2023.114583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/08/2023] [Accepted: 10/23/2023] [Indexed: 10/28/2023]
Abstract
Cerebral ischemia-reperfusion injury (CIRI) is a severe pathological condition that involves oxidative stress, inflammatory response, and neuronal damage. HY-021068 belongs to a new drug of chemical class 1, which is a potential thromboxane synthase inhibitor. Our preliminary experiment found that HY-021068 has significant anti-neuroinflammatory and neuroprotective effects. However, the protective effect and mechanism of HY-021068 in CIRI remain unclear. To investigate the protective effect and mechanism of HY-021068 in CIRI mice. In mice, CIRI was induced by bilateral common carotid artery occlusion and reperfusion. Mice were treated with HY-021068 or LV-NLRP1-shRNA (lentivirus-mediated shRNA transfection to knock down NLRP1 expression). The locomotor activity, neuronal damage, pathological changes, postsynaptic density protein-95 (PSD-95) expression, NLRP1 inflammasome activation, autophagy markers, and apoptotic proteins were assessed in CIRI mice. In this study, treatment with HY-021065 and LV-NLRP1-shRNA significantly improved motor dysfunction and neuronal damage after CIRI in mice. HY-021065 and NLRP1 knockdown significantly ameliorated the pathological damage and increased PSD-95 expression in the cortex and hippocampus CA1 and CA3 regions. The further studies showed that compared with the CIRI model group, HY-021065 and NLRP1 knockdown treatment inhibited the expressions of NLRP1, ASC, caspase-1, and IL-1β, restored the expressions of p-AMPK/AMPK, Beclin1, LC3II/LC3I, p-mTOR/m-TOR and P62, and regulated the expressions of BCL-2, Caspase3, and BAX in brain tissues of CIRI mice in CIRI mice. These results suggest that HY-021068 exerts a protective role in CIRI mice by inhibiting NLRP1 inflammasome activation and regulating autophagy function and neuronal apoptosis. HY-021068 is expected to become a new therapeutic drug for CIRI.
Collapse
Affiliation(s)
- Ye Huang
- Department of Plastic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, Anhui, China
| | - Min Han
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei 230032, Anhui, China
| | - Qifeng Shi
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei 230032, Anhui, China
| | - Xuewang Li
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei 230032, Anhui, China
| | - Jiajia Mo
- Hefei Industrial and Pharmaceutical Co., Ltd, Hefei 230200, Anhui, China
| | - Yan Liu
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei 230032, Anhui, China
| | - Zhaoxing Chu
- Hefei Industrial and Pharmaceutical Co., Ltd, Hefei 230200, Anhui, China.
| | - Weizu Li
- Department of Pharmacology, Basic Medicine College; Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education; Anhui Medical University, Hefei 230032, Anhui, China.
| |
Collapse
|
9
|
Zhou C, Zhu X, Li J, Luo Y, Zhou Y. Dynamic assessment of brain perfusion in a middle cerebral artery occlusion rat model by contrast-enhanced ultrasound imaging: a pilot study. Acta Radiol 2023; 64:3042-3051. [PMID: 37872652 DOI: 10.1177/02841851231205163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
BACKGROUND The middle cerebral artery occlusion model (MCAo) is a commonly used animal model for cerebral ischemia studies but lacks accessible imaging techniques for the assessment of hemodynamic changes of the model. PURPOSE The study aims to explore the value of contrast-enhanced ultrasound (CEUS) in evaluating brain perfusion in the early stages after MCAo surgery. MATERIAL AND METHODS In total, 18 adult male Sprague-Dawley rats were subjected to right MCAo using an intraluminal filament model, and CEUS was performed at the three following timepoints: before (T0), immediately after (T1), and 6 h after permanent MCAo (T2). Twelve rats successfully completed the study, and their brains were removed and stained using 2, 3, 5-triphenyltetrazolium chloride (TTC). CEUS video images were visualized offline, and the time-intensity curves (TICs) were analyzed. Different cerebrovascular patterns and manifestations of the contrast enhancement in rat ischemic hemispheres were observed. Semi-quantitative parameters of TICs in ischemic areas (ROIi) and the surrounding normal- or hypo-perfused areas (ROIn) were calculated and compared between T0, T1, and T2, and also between ROIi and ROIn. RESULTS A significant correlation was found between the lesion volume (%) determined by TTC and CEUS parameters (r = -0.691, P = 0.013 for peak intensity; r = -0.742, P = 0.006 for area under the curve) at T2. After the same occlusion, there were differences in contrast perfusion in each group. CONCLUSION This study suggests that CEUS could be an effective imaging tool for studying cerebral ischemia and perfusion in small animals as long as the transcranial acoustic window allows it.
Collapse
Affiliation(s)
- Chenyun Zhou
- Department of Ultrasound, West China Hospital of Sichuan University, Chengdu, Sichuan, PR China
| | - Xiaoxia Zhu
- Department of Ultrasound, West China Hospital of Sichuan University, Chengdu, Sichuan, PR China
| | - Jin Li
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, Sichuan, PR China
| | - Yan Luo
- Department of Ultrasound, West China Hospital of Sichuan University, Chengdu, Sichuan, PR China
| | - Yuqing Zhou
- Department of Ultrasound, West China Hospital of Sichuan University, Chengdu, Sichuan, PR China
| |
Collapse
|
10
|
Seong H, Jeong D, Kim EH, Yoon KS, Na D, Yoon SZ, Cho JE. MicroRNA-323-5p Involved in Dexmedetomidine Preconditioning Impart Neuroprotection. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1518. [PMID: 37763638 PMCID: PMC10532972 DOI: 10.3390/medicina59091518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/13/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023]
Abstract
Background and Objectives: Cerebral ischemia is one of the major preoperative complications. Dexmedetomidine is a well-known sedative-hypnotic agent that has potential organ-protective effects. We examine the miRNAs associated with preconditioning effects of dexmedetomidine in cerebral ischemia. Materials and Methods: Transient infarcts were induced in mice via reperfusion after temporary occlusion of one side of the middle cerebral artery. A subset of these mice was exposed to dexmedetomidine prior to cerebral infarction and miRNA profiling of the whole brain was performed. We administered dexmedetomidine and miRNA-323-5p mimic/inhibitor to oxygen-glucose deprivation/reoxygenation astrocytes. Additionally, we administered miR-323-5p mimic and inhibitor to mice via intracerebroventricular injection 2 h prior to induction of middle cerebral artery occlusion. Results: The infarct volume was significantly lower in the dexmedetomidine-preconditioned mice. Analysis of brain samples revealed an increased expression of five miRNAs and decreased expression of three miRNAs in the dexmedetomidine-pretreated group. The viability of cells significantly increased and expression of miR-323-5p was attenuated in the dexmedetomidine-treated oxygen-glucose deprivation/reoxygenation groups. Transfection with anti-miR-323-5p contributed to increased astrocyte viability. When miRNA-323-5p was injected intraventricularly, infarct volume was significantly reduced when preconditioned with the miR-323-5p inhibitor compared with mimic and negative control. Conclusions: Dexmedetomidine has a protective effect against transient neuronal ischemia-reperfusion injury and eight specific miRNAs were profiled. Also, miRNA-323-5p downregulation has a cell protective effect under ischemic conditions both in vivo and in vitro. Our findings suggest the potential of the miR-323-5p inhibitor as a therapeutic agent against cerebral infarction.
Collapse
Affiliation(s)
- Hyunyoung Seong
- Department of Anesthesiology and Pain Medicine, Anam Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Daun Jeong
- Institute for Healthcare Service Innovation, Korea University, Seoul 02841, Republic of Korea
| | - Eung Hwi Kim
- Institute for Healthcare Service Innovation, Korea University, Seoul 02841, Republic of Korea
| | - Kyung Seob Yoon
- Department of Anesthesiology and Pain Medicine, Anam Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Donghyun Na
- Department of Anesthesiology and Pain Medicine, Anam Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Seung Zhoo Yoon
- Department of Anesthesiology and Pain Medicine, Anam Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Jang Eun Cho
- Department of Anesthesiology and Pain Medicine, Anam Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea
| |
Collapse
|
11
|
Howell JA, Gaouette N, Lopez M, Burke SP, Perkins E, Bidwell GL. Elastin-like polypeptide delivery of anti-inflammatory peptides to the brain following ischemic stroke. FASEB J 2023; 37:e23077. [PMID: 37402128 PMCID: PMC10349587 DOI: 10.1096/fj.202300309rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/15/2023] [Accepted: 06/22/2023] [Indexed: 07/05/2023]
Abstract
Inflammatory processes are activated following ischemic stroke that lead to increased tissue damage for weeks following the ischemic insult, but there are no approved therapies that target this inflammation-induced secondary injury. Here, we report that SynB1-ELP-p50i, a novel protein inhibitor of the nuclear factor kappa B (NF-κB) inflammatory cascade bound to the drug carrier elastin-like polypeptide (ELP), decreases NF-κB induced inflammatory cytokine production in cultured macrophages, crosses the plasma membrane and accumulates in the cytoplasm of both neurons and microglia in vitro, and accumulates at the infarct site where the blood-brain barrier (BBB) is compromised following middle cerebral artery occlusion (MCAO) in rats. Additionally, SynB1-ELP-p50i treatment reduces infarct volume by 11.86% compared to saline-treated controls 24 h following MCAO. Longitudinally, SynB1-ELP-p50i treatment improves survival for 14 days following stroke with no effects of toxicity or peripheral organ dysfunction. These results show high potential for ELP-delivered biologics for therapy of ischemic stroke and other central nervous system disorders and further support targeting inflammation in ischemic stroke.
Collapse
Affiliation(s)
- John Aaron Howell
- Graduate Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS 39216
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Nicholas Gaouette
- School of Medicine, University of Mississippi Medical Center, Jackson, MS 39216
| | - Mariper Lopez
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Stephen P. Burke
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Eddie Perkins
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson, MS 39216
| | - Gene L. Bidwell
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS 39216
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216
- Department of Pharmacology and Toxicology University of Mississippi Medical Center, Jackson, MS 39216
| |
Collapse
|
12
|
Li Y, Tan L, Yang C, He L, Liu L, Deng B, Liu S, Guo J. Distinctions between the Koizumi and Zea Longa methods for middle cerebral artery occlusion (MCAO) model: a systematic review and meta-analysis of rodent data. Sci Rep 2023; 13:10247. [PMID: 37353569 PMCID: PMC10290095 DOI: 10.1038/s41598-023-37187-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/17/2023] [Indexed: 06/25/2023] Open
Abstract
Ischemic stroke in rodents is usually induced by intraluminal middle cerebral artery occlusion (MCAO) via the common carotid artery plugging filament invented by Koizumi et al. (MCAO-KM), or the external carotid artery plugging filament created by Zea Longa et al. (MCAO-LG). A systematic review of the distinctions between them is currently lacking. Here, we performed a meta-analysis in terms of model establishment, cerebral blood flow (CBF), and cerebral ischemia-reperfusion injury (CIRI) between them, Weighted Mean Differences and Standardized Mean Difference were used to analyze the combined effects, Cochrane's Q test and the I2 statistic were applied to determine heterogeneity, sensitivity analysis and subgroup analysis were performed to explore the source of heterogeneity. Literature mining suggests that MCAO-KM brings shorter operation time (p = 0.007), higher probability of plugging filament (p < 0.001) and molding establishment (p = 0.006), lower possibility of subarachnoid hemorrhage (p = 0.02), larger infarct volume (p = 0.003), severer brain edema (p = 0.002), and neurological deficits (p = 0.03). Nevertheless, MCAO-LG shows a more adequate CBF after ischemia-reperfusion (p < 0.001), a higher model survival rate (p = 0.02), and a greater infarct rate (p = 0.007). In conclusion, the MCAO-KM method is simple to operate with a high modeling success rate, and is suitable for the study of brain edema under long-term hypoperfusion, while the MCAO-LG method is highly challenging for novices, and is suitable for the study of CIRI caused by complete ischemia-reperfusion. These findings are expected to benefit the selection of intraluminal filament MCAO models before undertaking ischemic stroke preclinical effectiveness trials.
Collapse
Affiliation(s)
- Yong Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Tan
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Caixia Yang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liying He
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lin Liu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bowen Deng
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Sijing Liu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinlin Guo
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
13
|
Sokolowski JD, Soldozy S, Sharifi KA, Norat P, Kearns KN, Liu L, Williams AM, Yağmurlu K, Mastorakos P, Miller GW, Kalani MYS, Park MS, Kellogg RT, Tvrdik P. Preclinical models of middle cerebral artery occlusion: new imaging approaches to a classic technique. Front Neurol 2023; 14:1170675. [PMID: 37409019 PMCID: PMC10318149 DOI: 10.3389/fneur.2023.1170675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 05/30/2023] [Indexed: 07/07/2023] Open
Abstract
Stroke remains a major burden on patients, families, and healthcare professionals, despite major advances in prevention, acute treatment, and rehabilitation. Preclinical basic research can help to better define mechanisms contributing to stroke pathology, and identify therapeutic interventions that can decrease ischemic injury and improve outcomes. Animal models play an essential role in this process, and mouse models are particularly well-suited due to their genetic accessibility and relatively low cost. Here, we review the focal cerebral ischemia models with an emphasis on the middle cerebral artery occlusion technique, a "gold standard" in surgical ischemic stroke models. Also, we highlight several histologic, genetic, and in vivo imaging approaches, including mouse stroke MRI techniques, that have the potential to enhance the rigor of preclinical stroke evaluation. Together, these efforts will pave the way for clinical interventions that can mitigate the negative impact of this devastating disease.
Collapse
Affiliation(s)
- Jennifer D. Sokolowski
- Department of Neurological Surgery, University of Virginia, Charlottesville, VA, United States
| | - Sauson Soldozy
- Department of Neurological Surgery, Westchester Medical Center, Valhalla, NY, United States
| | - Khadijeh A. Sharifi
- Department of Neurological Surgery, University of Virginia, Charlottesville, VA, United States
- Department of Neuroscience, University of Virginia, Charlottesville, VA, United States
| | - Pedro Norat
- Department of Neurological Surgery, University of Virginia, Charlottesville, VA, United States
| | - Kathryn N. Kearns
- Department of Neurological Surgery, University of Virginia, Charlottesville, VA, United States
| | - Lei Liu
- Department of Neurological Surgery and Neuroscience, Northwestern University, Chicago, IL, United States
| | - Ashley M. Williams
- School of Medicine, Morsani College of Medicine, Tampa, FL, United States
| | - Kaan Yağmurlu
- Department of Neurological Surgery, University of Tennessee, Memphis, TN, United States
| | - Panagiotis Mastorakos
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, PA, United States
| | - G. Wilson Miller
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, United States
| | - M. Yashar S. Kalani
- Department of Neurological Surgery, St. John's Neuroscience Institute, Tulsa, OK, United States
| | - Min S. Park
- Department of Neurological Surgery, University of Virginia, Charlottesville, VA, United States
| | - Ryan T. Kellogg
- Department of Neurological Surgery, University of Virginia, Charlottesville, VA, United States
| | - Petr Tvrdik
- Department of Neurological Surgery, University of Virginia, Charlottesville, VA, United States
- Department of Neuroscience, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
14
|
Won J, Khan ZA, Hong Y. Effects of isoflurane and xylazine on inducing cerebral ischemia by the model of middle cerebral artery occlusion in mice. Lab Anim Res 2023; 39:11. [PMID: 37264475 DOI: 10.1186/s42826-023-00163-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/22/2023] [Accepted: 05/30/2023] [Indexed: 06/03/2023] Open
Abstract
Preclinical ischemic stroke studies extensively utilize the intraluminal suture method of middle cerebral artery occlusion (MCAo). General anesthesia administration is an essential step for MCAo, but anesthetic agents can lead to adverse effects causing death and making a considerable impact on inducing cerebral ischemia. The purpose of this study was to comparatively assess the effect of isoflurane and xylazine on transient cerebral ischemia in a mouse model of MCAo. Twenty animals were randomly divided into four groups: sham group (no MCAo), control group (MCAo under isoflurane, no agent till reperfusion), isoflurane group (MCAo under isoflurane continued till reperfusion), xylazine group (MCAo under isoflurane, and administration of xylazine till reperfusion). The survival rate, brain infarct volume, and neurologic deficits were studied to assess the effect of isoflurane and xylazine on the stroke model. Our results showed that the body weight showed statistically significant change before and 24 h after surgery in the control and Isoflurane groups, but no difference in the Xylazine group. Also, the survival rate, brain infarct volume, and neurologic deficits were slightly reduced in the isoflurane group at 24 h after reperfusion injury. However, the xylazine and control groups showed similar BIV and neurologic deficits. Interestingly, a high survival rate was observed in the xylazine group. Our results indicate that the modified method of inhalation anesthetics combined with xylazine can reduce the risk of mortality and develop a reproducible MCAo model with predictable brain ischemia. In addition, extended isoflurane anesthesia after MCAo is associated with the risk of mortality.
Collapse
Affiliation(s)
- Jinyoung Won
- Department of Rehabilitation Science, Graduate School of Inje University, 197 Inje-ro, Gimhae, Gyeong-nam, 50834, Republic of Korea
- Research Center for Aged-life Redesign (RCAR), Inje University, Gimhae, Republic of Korea
- Biohealth Products Research Center (BPRC), Inje University, Gimhae, Republic of Korea
| | - Zeeshan Ahmad Khan
- Research Center for Aged-life Redesign (RCAR), Inje University, Gimhae, Republic of Korea
- Biohealth Products Research Center (BPRC), Inje University, Gimhae, Republic of Korea
- Department of Physical Therapy, College of Healthcare Medical Science and Engineering, Inje University, Gimhae, Republic of Korea
| | - Yonggeun Hong
- Department of Rehabilitation Science, Graduate School of Inje University, 197 Inje-ro, Gimhae, Gyeong-nam, 50834, Republic of Korea.
- Research Center for Aged-life Redesign (RCAR), Inje University, Gimhae, Republic of Korea.
- Biohealth Products Research Center (BPRC), Inje University, Gimhae, Republic of Korea.
- Department of Physical Therapy, College of Healthcare Medical Science and Engineering, Inje University, Gimhae, Republic of Korea.
| |
Collapse
|
15
|
Norat P, Sokolowski JD, Gorick CM, Soldozy S, Kumar JS, Chae Y, Yagmurlu K, Nilak J, Sharifi KA, Walker M, Levitt MR, Klibanov AL, Yan Z, Price RJ, Tvrdik P, Kalani MYS. Intraarterial Transplantation of Mitochondria After Ischemic Stroke Reduces Cerebral Infarction. STROKE (HOBOKEN, N.J.) 2023; 3:e000644. [PMID: 37545759 PMCID: PMC10399028 DOI: 10.1161/svin.122.000644] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 01/03/2023] [Indexed: 08/08/2023]
Abstract
Background- Transplantation of autologous mitochondria into ischemic tissue may mitigate injury caused by ischemia and reperfusion. Methods- Using murine stroke models of middle cerebral artery occlusion, we sought to evaluate feasibility of delivery of viable mitochondria to ischemic brain parenchyma. We evaluated the effects of concurrent focused ultrasound activation of microbubbles, which serves to open the blood-brain barrier, on efficacy of delivery of mitochondria. Results- Following intra-arterial delivery, mitochondria distribute through the stroked hemisphere and integrate into neural and glial cells in the brain parenchyma. Consistent with functional integration in the ischemic tissue, the transplanted mitochondria elevate concentration of adenosine triphosphate in the stroked hemisphere, reduce infarct volume and increase cell viability. Additional of focused ultrasound leads to improved blood brain barrier opening without hemorrhagic complications. Conclusions- Our results have implications for the development of interventional strategies after ischemic stroke and suggest a novel potential modality of therapy after mechanical thrombectomy.
Collapse
Affiliation(s)
- Pedro Norat
- Department of Neurological Surgery, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Jennifer D. Sokolowski
- Department of Neurological Surgery, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Catherine M. Gorick
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Sauson Soldozy
- Department of Neurological Surgery, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Jeyan S. Kumar
- Department of Neurological Surgery, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Youngrok Chae
- Department of Neurological Surgery, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Kaan Yagmurlu
- Department of Neurological Surgery, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Joelle Nilak
- Department of Neurological Surgery, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Khadijeh A. Sharifi
- Department of Neurological Surgery, University of Virginia School of Medicine, Charlottesville, Virginia
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Melanie Walker
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington
| | - Michael R. Levitt
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington
| | - Alexander L. Klibanov
- Cardiovascular Division, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Zhen Yan
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Richard J. Price
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Petr Tvrdik
- Department of Neurological Surgery, University of Virginia School of Medicine, Charlottesville, Virginia
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, Virginia
| | - M. Yashar S. Kalani
- St. John’s Neuroscience Institute and the University of Oklahoma School of Medicine, Tulsa, Oklahoma
| |
Collapse
|
16
|
Nagy SA, Ivic I, Tóth P, Komoly S, Kiss T, Pénzes M, Málnási-Csizmadia A, Dóczi T, Perlaki G, Orsi G. Post-reperfusion acute MR diffusion in stroke is a potential predictor for clinical outcome in rats. Sci Rep 2023; 13:5598. [PMID: 37019923 PMCID: PMC10076321 DOI: 10.1038/s41598-023-32679-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 03/31/2023] [Indexed: 04/07/2023] Open
Abstract
Middle cerebral artery occlusion (MCAO) models show substantial variability in outcome, introducing uncertainties in the evaluation of treatment effects. Early outcome predictors would be essential for prognostic purposes and variability control. We aimed to compare apparent diffusion coefficient (ADC) MRI data obtained during MCAO and shortly after reperfusion for their potentials in acute-phase outcome prediction. Fifty-nine male rats underwent a 45-min MCAO. Outcome was defined in three ways: 21-day survival; 24 h midline-shift and neurological scores. Animals were divided into two groups: rats surviving 21 days after MCAO (survival group, n = 46) and rats dying prematurely (non-survival/NS group, n = 13). At reperfusion, NS group showed considerably larger lesion volume and lower mean ADC of the initial lesion site (p < 0.0001), while during occlusion there were no significant group differences. At reperfusion, each survival animal showed decreased lesion volume and increased mean ADC of the initial lesion site compared to those during occlusion (p < 10-6), while NS group showed a mixed pattern. At reperfusion, lesion volume and mean ADC of the initial lesion site were significantly associated with 24 h midline-shift and neurological scores. Diffusion MRI performed soon after reperfusion has a great impact in early-phase outcome prediction, and it works better than the measurement during occlusion.
Collapse
Affiliation(s)
- Szilvia Anett Nagy
- ELKH-PTE Clinical Neuroscience MR Research Group, Ret Str. 2, 7623, Pecs, Hungary.
- Pecs Diagnostic Centre, Rét Street 2, 7623, Pecs, Hungary.
- Structural Neurobiology Research Group, Szentágothai Research Centre, University of Pecs, Ifjúság Street 20, 7624, Pecs, Hungary.
- Department of Neurology, Medical School, University of Pecs, Rét Street 2, 7623, Pecs, Hungary.
| | - Ivan Ivic
- Pecs Diagnostic Centre, Rét Street 2, 7623, Pecs, Hungary
- Selvita d.o.o., Prilaz Baruna Filipovića 29, 10000, Zagreb, Croatia
| | - Péter Tóth
- ELKH-PTE Clinical Neuroscience MR Research Group, Ret Str. 2, 7623, Pecs, Hungary
- Department of Neurosurgery, Medical School, University of Pecs, Rét Street 2, 7623, Pecs, Hungary
| | - Sámuel Komoly
- Department of Neurology, Medical School, University of Pecs, Rét Street 2, 7623, Pecs, Hungary
| | - Tamás Kiss
- Szentágothai Research Centre, University of Pecs, Ifjúság Street 20, Pecs, Hungary
| | - Máté Pénzes
- Department of Biochemistry, Eötvös Loránd University, Pázmány Péter Sétány 1/C, 1117, Budapest, Hungary
- Motorpharma Ltd., Szilágyi E. Fasor 27, 1026, Budapest, Hungary
| | - András Málnási-Csizmadia
- Motorpharma Ltd., Szilágyi E. Fasor 27, 1026, Budapest, Hungary
- ELKH-ELTE Motor Pharmacology Research Group, Department of Biochemistry, Eötvös Loránd University, Pázmány Péter Sétány 1/C, 1117, Budapest, Hungary
| | - Tamás Dóczi
- Pecs Diagnostic Centre, Rét Street 2, 7623, Pecs, Hungary
- Department of Neurosurgery, Medical School, University of Pecs, Rét Street 2, 7623, Pecs, Hungary
| | - Gábor Perlaki
- ELKH-PTE Clinical Neuroscience MR Research Group, Ret Str. 2, 7623, Pecs, Hungary
- Pecs Diagnostic Centre, Rét Street 2, 7623, Pecs, Hungary
- Department of Neurology, Medical School, University of Pecs, Rét Street 2, 7623, Pecs, Hungary
- Department of Neurosurgery, Medical School, University of Pecs, Rét Street 2, 7623, Pecs, Hungary
| | - Gergely Orsi
- ELKH-PTE Clinical Neuroscience MR Research Group, Ret Str. 2, 7623, Pecs, Hungary
- Pecs Diagnostic Centre, Rét Street 2, 7623, Pecs, Hungary
- Department of Neurology, Medical School, University of Pecs, Rét Street 2, 7623, Pecs, Hungary
- Department of Neurosurgery, Medical School, University of Pecs, Rét Street 2, 7623, Pecs, Hungary
| |
Collapse
|
17
|
López-Morales MA, Escobar I, Saul I, Jackson CW, Ferrier FJ, Fagerli EA, Raval AP, Dave KR, Perez-Pinzon MA. Resveratrol Preconditioning Mitigates Ischemia-Induced Septal Cholinergic Cell Loss and Memory Impairments. Stroke 2023; 54:1099-1109. [PMID: 36912143 DOI: 10.1161/strokeaha.122.040899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 01/10/2023] [Indexed: 03/14/2023]
Abstract
BACKGROUND Cholinergic cells originating from the nuclei of the basal forebrain (BF) are critical for supporting various memory processes, yet BF cholinergic cell viability has not been explored in the context of focal cerebral ischemia. In the present study, we examined cell survival within several BF nuclei in rodents following transient middle cerebral artery occlusion. We tested the hypothesis that a previously established neuroprotective therapy-resveratrol preconditioning-would rescue BF cell loss, deficits in cholinergic-related memory performance, and hippocampal synaptic dysfunction after focal cerebral ischemia. METHODS Adult (2-3-month old) male Sprague-Dawley rats or wild-type C57Bl/6J mice were injected intraperitoneally with a single dose of resveratrol or vehicle and subjected to transient middle cerebral artery occlusion using the intraluminal suture method 2 days later. Histopathological, behavioral, and electrophysiological outcomes were measured 1-week post-reperfusion. Animals with reduction in cerebral blood flow <30% of baseline were excluded. RESULTS Cholinergic cell loss was observed in the medial septal nucleus and diagonal band of Broca following transient middle cerebral artery occlusion. This effect was prevented by resveratrol preconditioning, which also ameliorated transient middle cerebral artery occlusion-induced deficits in cognitive performance and hippocampal long-term potentiation. CONCLUSIONS We demonstrate for the first time that focal cerebral ischemia induces cholinergic cell death within memory-relevant nuclei of the BF. The preservation of cholinergic cell viability may provide a mechanism by which resveratrol preconditioning improves memory performance and preserves functionality of memory-processing brain structures after focal cerebral ischemia.
Collapse
Affiliation(s)
- Mikahela A López-Morales
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories (M.A.L.-M., I.E., I.S., C.W.J., F.J.F., E.A.F., A.P.R., K.R.D., M.A.P.-P.), University of Miami Leonard M. Miller School of Medicine, FL
- Department of Neurology (M.A.L.-M., I.E., I.S., C.W.J., F.J.F., E.A.F., A.P.R., K.R.D., M.A.P.-P.), University of Miami Leonard M. Miller School of Medicine, FL
| | - Iris Escobar
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories (M.A.L.-M., I.E., I.S., C.W.J., F.J.F., E.A.F., A.P.R., K.R.D., M.A.P.-P.), University of Miami Leonard M. Miller School of Medicine, FL
- Department of Neurology (M.A.L.-M., I.E., I.S., C.W.J., F.J.F., E.A.F., A.P.R., K.R.D., M.A.P.-P.), University of Miami Leonard M. Miller School of Medicine, FL
- Neuroscience Program (I.E., C.W.J., F.J.F., E.A.F., A.P.R., K.R.D., M.A.P.-P.), University of Miami Leonard M. Miller School of Medicine, FL
| | - Isabel Saul
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories (M.A.L.-M., I.E., I.S., C.W.J., F.J.F., E.A.F., A.P.R., K.R.D., M.A.P.-P.), University of Miami Leonard M. Miller School of Medicine, FL
- Department of Neurology (M.A.L.-M., I.E., I.S., C.W.J., F.J.F., E.A.F., A.P.R., K.R.D., M.A.P.-P.), University of Miami Leonard M. Miller School of Medicine, FL
| | - Charles W Jackson
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories (M.A.L.-M., I.E., I.S., C.W.J., F.J.F., E.A.F., A.P.R., K.R.D., M.A.P.-P.), University of Miami Leonard M. Miller School of Medicine, FL
- Department of Neurology (M.A.L.-M., I.E., I.S., C.W.J., F.J.F., E.A.F., A.P.R., K.R.D., M.A.P.-P.), University of Miami Leonard M. Miller School of Medicine, FL
- Neuroscience Program (I.E., C.W.J., F.J.F., E.A.F., A.P.R., K.R.D., M.A.P.-P.), University of Miami Leonard M. Miller School of Medicine, FL
| | - Fernando J Ferrier
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories (M.A.L.-M., I.E., I.S., C.W.J., F.J.F., E.A.F., A.P.R., K.R.D., M.A.P.-P.), University of Miami Leonard M. Miller School of Medicine, FL
- Department of Neurology (M.A.L.-M., I.E., I.S., C.W.J., F.J.F., E.A.F., A.P.R., K.R.D., M.A.P.-P.), University of Miami Leonard M. Miller School of Medicine, FL
- Neuroscience Program (I.E., C.W.J., F.J.F., E.A.F., A.P.R., K.R.D., M.A.P.-P.), University of Miami Leonard M. Miller School of Medicine, FL
| | - Eric A Fagerli
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories (M.A.L.-M., I.E., I.S., C.W.J., F.J.F., E.A.F., A.P.R., K.R.D., M.A.P.-P.), University of Miami Leonard M. Miller School of Medicine, FL
- Department of Neurology (M.A.L.-M., I.E., I.S., C.W.J., F.J.F., E.A.F., A.P.R., K.R.D., M.A.P.-P.), University of Miami Leonard M. Miller School of Medicine, FL
- Neuroscience Program (I.E., C.W.J., F.J.F., E.A.F., A.P.R., K.R.D., M.A.P.-P.), University of Miami Leonard M. Miller School of Medicine, FL
| | - Ami P Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories (M.A.L.-M., I.E., I.S., C.W.J., F.J.F., E.A.F., A.P.R., K.R.D., M.A.P.-P.), University of Miami Leonard M. Miller School of Medicine, FL
- Department of Neurology (M.A.L.-M., I.E., I.S., C.W.J., F.J.F., E.A.F., A.P.R., K.R.D., M.A.P.-P.), University of Miami Leonard M. Miller School of Medicine, FL
- Neuroscience Program (I.E., C.W.J., F.J.F., E.A.F., A.P.R., K.R.D., M.A.P.-P.), University of Miami Leonard M. Miller School of Medicine, FL
| | - Kunjan R Dave
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories (M.A.L.-M., I.E., I.S., C.W.J., F.J.F., E.A.F., A.P.R., K.R.D., M.A.P.-P.), University of Miami Leonard M. Miller School of Medicine, FL
- Department of Neurology (M.A.L.-M., I.E., I.S., C.W.J., F.J.F., E.A.F., A.P.R., K.R.D., M.A.P.-P.), University of Miami Leonard M. Miller School of Medicine, FL
- Neuroscience Program (I.E., C.W.J., F.J.F., E.A.F., A.P.R., K.R.D., M.A.P.-P.), University of Miami Leonard M. Miller School of Medicine, FL
| | - Miguel A Perez-Pinzon
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratories (M.A.L.-M., I.E., I.S., C.W.J., F.J.F., E.A.F., A.P.R., K.R.D., M.A.P.-P.), University of Miami Leonard M. Miller School of Medicine, FL
- Department of Neurology (M.A.L.-M., I.E., I.S., C.W.J., F.J.F., E.A.F., A.P.R., K.R.D., M.A.P.-P.), University of Miami Leonard M. Miller School of Medicine, FL
- Neuroscience Program (I.E., C.W.J., F.J.F., E.A.F., A.P.R., K.R.D., M.A.P.-P.), University of Miami Leonard M. Miller School of Medicine, FL
| |
Collapse
|
18
|
Jiang Y, Liu Q, Zhao Y, Wang C, Sun M. Activation of CREB-BDNF Pathway in Pyramidal Neurons in the Hippocampus Improves the Neurological Outcome of Mice with Ischemic Stroke. Mol Neurobiol 2023; 60:1766-1781. [PMID: 36571720 DOI: 10.1007/s12035-022-03174-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 12/14/2022] [Indexed: 12/27/2022]
Abstract
Cerebral ischemia is characterized by several pathological reaction evolving over time. Hyperactivation of glutamatergic neurons is the main factor leading to excitotoxicity which potentiates oxidative stress and triggers the mechanisms of neural apoptosis after cerebral ischemia. However, it is unclear whether glutamate in the ventral hippocampal Cornus Ammonis 1 (vCA1) acts a part in neurological deficits, pain perception, anxiety, and depression induced by ischemic stroke. We investigated the effects of chemogenetic inhibition or activation of vCA1 pyramidal neurons which are mainly glutamatergic neurons on sequelae induced by cerebral ischemia. Our results revealed that inhibition of vCA1 pyramidal neurons by chemogenetics alleviated neurological deficits, pain perception, anxiety, and depression caused by cerebral ischemia in mice, but activation of vCA1 pyramidal neurons had limited effects. Moreover, we found that stroke was accompanied by decreased levels of cAMP-response element-binding protein (CREB) and brain-derived neurotrophic factor (BDNF) in vCA1, which are modulated by glutamate. In this study, overexpression of CREB protein in pyramidal neurons in vCA1 by AAV virus significantly upregulated the content of BDNF and ameliorated the dysfunction induced by ischemic stroke. Our results demonstrated activation of the CREB-BDNF pathway in vCA1 pyramidal neurons significantly improved neurological deficits, pain perception, anxiety, and depression induced by ischemic stroke.
Collapse
Affiliation(s)
- Yingying Jiang
- Department of Neuropharmacology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Qingying Liu
- Department of Pain Management, the First Affiliated Hospital, Zhengzhou University, Henan, China
| | - Yumei Zhao
- Department of Neuropharmacology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Chunyang Wang
- Department of Neuropharmacology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Ming Sun
- Department of Neuropharmacology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
| |
Collapse
|
19
|
Howell JA, Gaouette N, Lopez M, Burke SP, Perkins E, Bidwell GL. Elastin-like polypeptide delivery of anti-inflammatory peptides to the brain following ischemic stroke. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.15.532834. [PMID: 36993686 PMCID: PMC10055169 DOI: 10.1101/2023.03.15.532834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Inflammatory processes are activated following ischemic strokes and lead to increased tissue damage for weeks following the ischemic insult, but there are no approved therapies that target this inflammation-induced secondary injury. Here, we report that SynB1-ELP-p50i, a novel protein inhibitor of the nuclear factor kappa B (NF-κB) inflammatory cascade bound to drug carrier elastin-like polypeptide (ELP), is able to enter both neurons and microglia, cross the blood-brain barrier, localize exclusively in the ischemic core and penumbra in Wistar-Kyoto and spontaneously hypertensive rats (SHRs), and reduce infarct volume in male SHRs. Additionally, in male SHRs, SynB1-ELP-p50i treatment improves survival for 14 days following stroke with no effects of toxicity or peripheral organ dysfunction. These results show high potential for ELP-delivered biologics for therapy of ischemic stroke and other central nervous system disorders and further support targeting inflammation in ischemic stroke.
Collapse
Affiliation(s)
- John Aaron Howell
- Graduate Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS 39216
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Nicholas Gaouette
- School of Medicine, University of Mississippi Medical Center, Jackson, MS 39216
| | - Mariper Lopez
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Stephen P. Burke
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Eddie Perkins
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson, MS 39216
| | - Gene L. Bidwell
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS 39216
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS 39216
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216
| |
Collapse
|
20
|
Keshavarz S, Nemati M, Saied Salehi M, Naseh M. The impact of anesthetic drugs on hemodynamic parameters and neurological outcomes following temporal middle cerebral artery occlusion in rats. Neuroreport 2023; 34:199-204. [PMID: 36789841 PMCID: PMC10516172 DOI: 10.1097/wnr.0000000000001863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/21/2022] [Indexed: 02/16/2023]
Abstract
The induction of ischemic stroke in the experimental model requires general anesthesia. One of the factors that can be effective in the size of ischemic brain lesions and neurological outcomes is the type of anesthesia. So, the current study was designed to compare the impacts of the most important and widely used anesthetics including halothane, isoflurane, and chloral hydrate on the transient middle cerebral artery occlusion (MCAO) outcomes. Adult Male Sprague-Dawley rats were randomly divided into three groups as follows: (1) MCAO + halothane group, (2) MCAO + isoflurane group, and (3) MCAO + chloral hydrate group. After 24 h, the mortality rate, infarct size, tissue swelling, neurological function, hemodynamic, and arterial blood gas parameters were assessed. Our finding showed that 60 min MCAO rats anesthetized with chloral hydrate significantly increased mortality rate, infarct size, tissue swelling, and neurological deficits compared with halothane and isoflurane anesthetics after 24 h of MCAO. Also, chloral hydrate caused a significant decrease in mean arterial pressure and arterial pO2 compared to halothane and isoflurane anesthetics. On the basis of the current data, we concluded that chloral hydrate increased cerebral infarct volume and neurological outcomes and reduced hemodynamic and metabolic parameters compared with halothane and isoflurane-anesthetized rats temporal MCAO.
Collapse
Affiliation(s)
- Somaye Keshavarz
- Histomorphometry and Stereology Research Center
- Department of Physiology
| | | | - Mohammad Saied Salehi
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | |
Collapse
|
21
|
Beccari S, Sierra-Torre V, Valero J, Pereira-Iglesias M, García-Zaballa M, Soria FN, De Las Heras-Garcia L, Carretero-Guillen A, Capetillo-Zarate E, Domercq M, Huguet PR, Ramonet D, Osman A, Han W, Dominguez C, Faust TE, Touzani O, Pampliega O, Boya P, Schafer D, Mariño G, Canet-Soulas E, Blomgren K, Plaza-Zabala A, Sierra A. Microglial phagocytosis dysfunction in stroke is driven by energy depletion and induction of autophagy. Autophagy 2023:1-30. [PMID: 36622892 DOI: 10.1080/15548627.2023.2165313] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Microglial phagocytosis of apoptotic debris prevents buildup damage of neighbor neurons and inflammatory responses. Whereas microglia are very competent phagocytes under physiological conditions, we report their dysfunction in mouse and preclinical monkey models of stroke (macaques and marmosets) by transient occlusion of the medial cerebral artery (tMCAo). By analyzing recently published bulk and single cell RNA sequencing databases, we show that the phagocytosis dysfunction was not explained by transcriptional changes. In contrast, we demonstrate that the impairment of both engulfment and degradation was related to energy depletion triggered by oxygen and nutrient deprivation (OND), which led to reduced process motility, lysosomal exhaustion, and the induction of a protective macroautophagy/autophagy response in microglia. Basal autophagy, in charge of removing and recycling intracellular elements, was critical to maintain microglial physiology, including survival and phagocytosis, as we determined both in vivo and in vitro using pharmacological and transgenic approaches. Notably, the autophagy inducer rapamycin partially prevented the phagocytosis impairment induced by tMCAo in vivo but not by OND in vitro, where it even had a detrimental effect on microglia, suggesting that modulating microglial autophagy to optimal levels may be a hard to achieve goal. Nonetheless, our results show that pharmacological interventions, acting directly on microglia or indirectly on the brain environment, have the potential to recover phagocytosis efficiency in the diseased brain. We propose that phagocytosis is a therapeutic target yet to be explored in stroke and other brain disorders and provide evidence that it can be modulated in vivo using rapamycin.Abbreviations: AIF1/IBA1: allograft inflammatory factor 1; AMBRA1: autophagy/beclin 1 regulator 1; ATG4B: autophagy related 4B, cysteine peptidase; ATP: adenosine triphosphate; BECN1: beclin 1, autophagy related; CASP3: caspase 3; CBF: cerebral blood flow; CCA: common carotid artery; CCR2: chemokine (C-C motif) receptor 2; CIR: cranial irradiation; Csf1r/v-fms: colony stimulating factor 1 receptor; CX3CR1: chemokine (C-X3-C motif) receptor 1; DAPI: 4',6-diamidino-2-phenylindole; DG: dentate gyrus; GO: Gene Ontology; HBSS: Hanks' balanced salt solution; HI: hypoxia-ischemia; LAMP1: lysosomal-associated membrane protein 1; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MCA: medial cerebral artery; MTOR: mechanistic target of rapamycin kinase; OND: oxygen and nutrient deprivation; Ph/A coupling: phagocytosis-apoptosis coupling; Ph capacity: phagocytic capacity; Ph index: phagocytic index; SQSTM1: sequestosome 1; RNA-Seq: RNA sequencing; TEM: transmission electron microscopy; tMCAo: transient medial cerebral artery occlusion; ULK1: unc-51 like kinase 1.
Collapse
Affiliation(s)
- Sol Beccari
- Glial Cell Biology Labb, Department of Biochemistry and Molecular Biology, Achucarro Basque Center for Neuroscience, 48940, Leioa, Bizkaia, Spain.,Department of Neuroscience, University of the Basque Country UPV/EHU, 48940, Leioa, Bizkaia, Spain
| | - Virginia Sierra-Torre
- Glial Cell Biology Labb, Department of Biochemistry and Molecular Biology, Achucarro Basque Center for Neuroscience, 48940, Leioa, Bizkaia, Spain.,Department of Neuroscience, University of the Basque Country UPV/EHU, 48940, Leioa, Bizkaia, Spain
| | - Jorge Valero
- Glial Cell Biology Labb, Department of Biochemistry and Molecular Biology, Achucarro Basque Center for Neuroscience, 48940, Leioa, Bizkaia, Spain.,Department of Neuroscience, University of the Basque Country UPV/EHU, 48940, Leioa, Bizkaia, Spain.,Neural Plasticity and Neurorepair Group, Laboratory of Neuronal Plasticity and Neurorepair, Institute for Neuroscience of Castilla y León (INCyL), and Institute for Biomedical Research of Salamanca, University of Salamanca, 37007, Salamanca, Spain
| | - Marta Pereira-Iglesias
- Glial Cell Biology Labb, Department of Biochemistry and Molecular Biology, Achucarro Basque Center for Neuroscience, 48940, Leioa, Bizkaia, Spain.,Department of Neuroscience, University of the Basque Country UPV/EHU, 48940, Leioa, Bizkaia, Spain
| | - Mikel García-Zaballa
- Glial Cell Biology Labb, Department of Biochemistry and Molecular Biology, Achucarro Basque Center for Neuroscience, 48940, Leioa, Bizkaia, Spain.,Department of Neuroscience, University of the Basque Country UPV/EHU, 48940, Leioa, Bizkaia, Spain
| | - Federico N Soria
- Glial Cell Biology Labb, Department of Biochemistry and Molecular Biology, Achucarro Basque Center for Neuroscience, 48940, Leioa, Bizkaia, Spain.,Department of Neuroscience, University of the Basque Country UPV/EHU, 48940, Leioa, Bizkaia, Spain.,Ikerbasque Foundation, 48009, Bilbao, Bizkaia, Spain
| | - Laura De Las Heras-Garcia
- Glial Cell Biology Labb, Department of Biochemistry and Molecular Biology, Achucarro Basque Center for Neuroscience, 48940, Leioa, Bizkaia, Spain.,Department of Neuroscience, University of the Basque Country UPV/EHU, 48940, Leioa, Bizkaia, Spain
| | - Alejandro Carretero-Guillen
- Glial Cell Biology Labb, Department of Biochemistry and Molecular Biology, Achucarro Basque Center for Neuroscience, 48940, Leioa, Bizkaia, Spain
| | - Estibaliz Capetillo-Zarate
- Glial Cell Biology Labb, Department of Biochemistry and Molecular Biology, Achucarro Basque Center for Neuroscience, 48940, Leioa, Bizkaia, Spain.,Department of Neuroscience, University of the Basque Country UPV/EHU, 48940, Leioa, Bizkaia, Spain.,Ikerbasque Foundation, 48009, Bilbao, Bizkaia, Spain.,Centro de Investigación en Red de Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Maria Domercq
- Glial Cell Biology Labb, Department of Biochemistry and Molecular Biology, Achucarro Basque Center for Neuroscience, 48940, Leioa, Bizkaia, Spain.,Department of Neuroscience, University of the Basque Country UPV/EHU, 48940, Leioa, Bizkaia, Spain
| | - Paloma R Huguet
- Glial Cell Biology Labb, Department of Biochemistry and Molecular Biology, Achucarro Basque Center for Neuroscience, 48940, Leioa, Bizkaia, Spain.,Department of Neuroscience, University of the Basque Country UPV/EHU, 48940, Leioa, Bizkaia, Spain
| | - David Ramonet
- INSERM U1060 CarMeN, Université Claude Bernard Lyon 1 - IRIS team, CarMeN, bat. B13, gpt hosp. Est, 59 bld Pinel, 69500, Bron, Auvergne-Rhône-Alpes, France
| | - Ahmed Osman
- Department of Women and Children´s Health, Karolisnka Institute, 17164, Stockholm, Södermanland and Uppland, Sweden
| | - Wei Han
- Department of Women and Children´s Health, Karolisnka Institute, 17164, Stockholm, Södermanland and Uppland, Sweden
| | - Cecilia Dominguez
- Department of Women and Children´s Health, Karolisnka Institute, 17164, Stockholm, Södermanland and Uppland, Sweden
| | - Travis E Faust
- Department of Neurobiology, University of Massachusetts Medical School, 01605, Worcester, MA, USA
| | - Omar Touzani
- Normandie-Univ, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, 14000, Caen, Normandie, France
| | - Olatz Pampliega
- Glial Cell Biology Labb, Department of Biochemistry and Molecular Biology, Achucarro Basque Center for Neuroscience, 48940, Leioa, Bizkaia, Spain.,Department of Neuroscience, University of the Basque Country UPV/EHU, 48940, Leioa, Bizkaia, Spain
| | - Patricia Boya
- Laboratory of Autophagy, Centro de Investigaciones Biológicas Margarita Salas, Madrid 28040, Spain.,Department of Medicine, University of Fribourg, 1700, Freiburg, Switzerland
| | - Dorothy Schafer
- Department of Neurobiology, University of Massachusetts Medical School, 01605, Worcester, MA, USA
| | - Guillermo Mariño
- Department of Medicine, University of Fribourg, 1700, Freiburg, Switzerland.,Department of Functional Biology, University of Oviedo, 33003, Oviedo, Asturias, Spain
| | - Emmanuelle Canet-Soulas
- INSERM U1060 CarMeN, Université Claude Bernard Lyon 1 - IRIS team, CarMeN, bat. B13, gpt hosp. Est, 59 bld Pinel, 69500, Bron, Auvergne-Rhône-Alpes, France
| | - Klas Blomgren
- Department of Women and Children´s Health, Karolisnka Institute, 17164, Stockholm, Södermanland and Uppland, Sweden.,Department of Pediatric Oncology, Karolinska University Hospital, 171 64, Stockholm, Södermanland and Uppland, Sweden
| | - Ainhoa Plaza-Zabala
- Glial Cell Biology Labb, Department of Biochemistry and Molecular Biology, Achucarro Basque Center for Neuroscience, 48940, Leioa, Bizkaia, Spain.,Department of Pharmacology, University of the Basque Country UPV/EHU, 48940, Leioa, Bizkaia, Spain
| | - Amanda Sierra
- Glial Cell Biology Labb, Department of Biochemistry and Molecular Biology, Achucarro Basque Center for Neuroscience, 48940, Leioa, Bizkaia, Spain.,Department of Neuroscience, University of the Basque Country UPV/EHU, 48940, Leioa, Bizkaia, Spain.,Ikerbasque Foundation, 48009, Bilbao, Bizkaia, Spain
| |
Collapse
|
22
|
Optimizing intraluminal monofilament model of ischemic stroke in middle-aged Sprague-Dawley rats. BMC Neurosci 2022; 23:75. [PMID: 36494808 PMCID: PMC9733327 DOI: 10.1186/s12868-022-00764-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Intraluminal monofilament model of middle cerebral artery occlusion (MCAO) is widely adopted for ischemic stroke; and Sprague-Dawley (SD) rats are commonly used rodents for preclinical research. Due to the paucity of information on the appropriate monofilament size for inducing MCAO in SD rats and the importance of including middle-aged models in ischemic stroke studies, we aimed to: (i). determine an appropriate Doccol® monofilament size for middle-aged male SD rats which weighed > 500 g following 24-h transient MCAO survival as well as (ii). demonstrate the optimal Doccol® filament size for middle-aged males (≤ 500 g) and females (273-300 g) while using young adult male SD rats (372-472 g) as control for severity of infarct volume following 7-days post-MCAO. All rats were subjected to 90-min transient MCAO. We show that 0.43 mm Doccol® monofilament size is more appropriate to induce large infarct lesion and optimal functional deficit when compared to 0.45 mm and 0.47 mm at 24 h post-MCAO. Our data on infarct volumes at 7 days post-MCAO as well as the observed weight loss and functional deficits at post-MCAO days 1, 3 and 7 demonstrate that 0.41 mm, 0.37 mm and 0.39 mm are optimal Doccol® filament sizes for middle-aged male (477.3 ± 39.61 g) and female (302.6 ± 26.28 g) as well as young-adult male (362.2 ± 28.38 g) SD rats, respectively.
Collapse
|
23
|
Onufriev MV, Stepanichev MY, Moiseeva YV, Zhanina MY, Nedogreeva OA, Kostryukov PA, Lazareva NA, Gulyaeva NV. A Comparative Study of Two Models of Intraluminal Filament Middle Cerebral Artery Occlusion in Rats: Long-Lasting Accumulation of Corticosterone and Interleukins in the Hippocampus and Frontal Cortex in Koizumi Model. Biomedicines 2022; 10:biomedicines10123119. [PMID: 36551875 PMCID: PMC9775077 DOI: 10.3390/biomedicines10123119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/20/2022] [Accepted: 11/29/2022] [Indexed: 12/10/2022] Open
Abstract
Recently, we have shown the differences in the early response of corticosterone and inflammatory cytokines in the hippocampus and frontal cortex (FC) of rats with middle cerebral artery occlusion (MCAO), according to the methods of Longa et al. (LM) and Koizumi et al. (KM) which were used as alternatives in preclinical studies to induce stroke in rodents. In the present study, corticosterone and proinflammatory cytokines were assessed 3 months after MCAO. The most relevant changes detected during the first days after MCAO became even more obvious after 3 months. In particular, the MCAO-KM (but not the MCAO-LM) group showed significant accumulation of corticosterone and IL1β in both the ipsilateral and contralateral hippocampus and FC. An accumulation of TNFα was detected in the ipsilateral hippocampus and FC in the MCAO-KM group. Thus, unlike the MCAO-LM, the MCAO-KM may predispose the hippocampus and FC of rats to long-lasting bilateral corticosterone-dependent distant neuroinflammatory damage. Unexpectedly, only the MCAO-LM rats demonstrated some memory deficit in a one-trial step-through passive avoidance test. The differences between the two MCAO models, particularly associated with the long-lasting increase in glucocorticoid and proinflammatory cytokine accumulation in the limbic structures in the MCAO-KM, should be considered in the planning of preclinical experiments, and the interpretation and translation of received results.
Collapse
Affiliation(s)
- Mikhail V. Onufriev
- Laboratory of Functional Biochemistry of the Nervous System, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 117485 Moscow, Russia
- Research and Clinical Center for Neuropsychiatry of Moscow Healthcare Department, 115419 Moscow, Russia
| | - Mikhail Y. Stepanichev
- Laboratory of Functional Biochemistry of the Nervous System, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 117485 Moscow, Russia
| | - Yulia V. Moiseeva
- Laboratory of Functional Biochemistry of the Nervous System, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 117485 Moscow, Russia
| | - Marina Y. Zhanina
- Laboratory of Functional Biochemistry of the Nervous System, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 117485 Moscow, Russia
- Research and Clinical Center for Neuropsychiatry of Moscow Healthcare Department, 115419 Moscow, Russia
| | - Olga A. Nedogreeva
- Laboratory of Functional Biochemistry of the Nervous System, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 117485 Moscow, Russia
| | - Pavel A. Kostryukov
- Laboratory of Functional Biochemistry of the Nervous System, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 117485 Moscow, Russia
| | - Natalia A. Lazareva
- Laboratory of Functional Biochemistry of the Nervous System, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 117485 Moscow, Russia
| | - Natalia V. Gulyaeva
- Laboratory of Functional Biochemistry of the Nervous System, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 117485 Moscow, Russia
- Research and Clinical Center for Neuropsychiatry of Moscow Healthcare Department, 115419 Moscow, Russia
- Correspondence: ; Tel.: +7-(495)-952-4007
| |
Collapse
|
24
|
Apelin-13 attenuates cerebral ischemia/reperfusion injury through regulating inflammation and targeting the JAK2/STAT3 signaling pathway. J Chem Neuroanat 2022; 126:102171. [DOI: 10.1016/j.jchemneu.2022.102171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 10/07/2022] [Accepted: 10/19/2022] [Indexed: 11/07/2022]
|
25
|
Justić H, Barić A, Šimunić I, Radmilović M, Ister R, Škokić S, Dobrivojević Radmilović M. Redefining the Koizumi model of mouse cerebral ischemia: A comparative longitudinal study of cerebral and retinal ischemia in the Koizumi and Longa middle cerebral artery occlusion models. J Cereb Blood Flow Metab 2022; 42:2080-2094. [PMID: 35748043 PMCID: PMC9580169 DOI: 10.1177/0271678x221109873] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cerebral and retinal ischemia share similar pathogenesis and epidemiology, each carrying both acute and prolonged risk of the other and often co-occurring. The most used preclinical stroke models, the Koizumi and Longa middle cerebral artery occlusion (MCAO) methods, have reported retinal damage with great variability, leaving the disruption of retinal blood supply via MCAO poorly investigated, even providing conflicting assumptions on the origin of the ophthalmic artery in rodents. The aim of our study was to use longitudinal in vivo magnetic resonance assessment of cerebral and retinal vascular perfusion after the ischemic injury to clarify whether and how the Koizumi and Longa methods induce retinal ischemia and how they differ in terms of cerebral and retinal lesion evolution. We provided anatomical evidence of the origin of the ophthalmic artery in mice from the pterygopalatine artery. Following the Koizumi surgery, retinal responses to ischemia overlapped with those in the brain, resulting in permanent damage. In contrast, the Longa method produced only extensive cerebral lesions, with greater tissue loss than in the Koizumi method. Additionally, our data suggests the Koizumi method should be redefined as a model of ischemia with chronic hypoperfusion rather than of ischemia and reperfusion.
Collapse
Affiliation(s)
- Helena Justić
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Department of Histology and Embryology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Anja Barić
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Department of Histology and Embryology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Iva Šimunić
- Department of Histology and Embryology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Marin Radmilović
- Department of Ophthalmology, Sestre Milosrdnice University Hospital Center, Zagreb, Croatia *These authors contributed equally to this work
| | - Rok Ister
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Department of Histology and Embryology, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Siniša Škokić
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Marina Dobrivojević Radmilović
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Department of Histology and Embryology, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
26
|
Sharma R, Chischolm A, Parikh M, Qureshi AI, Sahota P, Thakkar MM. Ischemic Stroke Disrupts Sleep Homeostasis in Middle-Aged Mice. Cells 2022; 11:2818. [PMID: 36139392 PMCID: PMC9497108 DOI: 10.3390/cells11182818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/30/2022] [Accepted: 09/05/2022] [Indexed: 11/16/2022] Open
Abstract
Sleep disturbances, including insomnia and excessive daytime sleepiness, are highly prevalent in patients with ischemic stroke (IS), which severely impacts recovery and rehabilitation efforts. However, how IS induces sleep disturbances is unclear. Three experiments were performed on middle-aged C57BL/6J mice, instrumented with sleep recording electrodes and/or subjected to 1 h of middle cerebral artery (MCAO; Stroke group) or sham (Sham group) occlusion to induce IS. After 48 h of reperfusion (a) experiment 1 verified sensorimotor deficit (using Garcia scale) and infarction (using TTC staining) in this mouse model; (b) experiment 2 examined the effects of IS on the quality (sleep latency and NREM delta power) and quantity (duration) of sleep; and (c) experiment 3 determined the effects of IS on sleep homeostasis using sleep deprivation (SD) and recovery sleep (RS) paradigm. Stroke mice display (a) a significant correlation between sensorimotor deficit and cerebral infarction; (b) insomnia-like symptoms (increased sleep latency, reduced NREM duration and delta power) during the light (inactive) period and daytime sleepiness-like symptoms during the dark (active) period mimicking sleep in IS patients; and (c) impairments in the markers of sleep pressure (during SD) and sleep dissipation (during RS). Our results suggest that IS disrupts sleep homeostasis to cause sleep disturbances.
Collapse
Affiliation(s)
- Rishi Sharma
- Harry S. Truman Memorial Veterans Hospital and Department of Neurology, School of Medicine, University of Missouri, Columbia, MO 65201, USA
| | | | | | | | | | | |
Collapse
|
27
|
Hemodynamics and Tissue Optical Properties in Bimodal Infarctions Induced by Middle Cerebral Artery Occlusion. Int J Mol Sci 2022; 23:ijms231810318. [PMID: 36142225 PMCID: PMC9499323 DOI: 10.3390/ijms231810318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/24/2022] Open
Abstract
Various infarct sizes induced by middle cerebral artery occlusion (MCAO) generate inconsistent outcomes for stroke preclinical study. Monitoring cerebral hemodynamics may help to verify the outcome of MCAO. The aim of this study was to investigate the changes in brain tissue optical properties by frequency-domain near-infrared spectroscopy (FD-NIRS), and establish the relationship between cerebral hemodynamics and infarct variation in MCAO model. The rats were undergone transient MCAO using intraluminal filament. The optical properties and hemodynamics were measured by placing the FD-NIRS probes on the scalp of the head before, during, and at various time-courses after MCAO. Bimodal infarction severities were observed after the same 90-min MCAO condition. Significant decreases in concentrations of oxygenated hemoglobin ([HbO]) and total hemoglobin ([HbT]), tissue oxygenation saturation (StO2), absorption coefficient (μa) at 830 nm, and reduced scattering coefficient (μs’) at both 690 and 830 nm were detected during the occlusion in the severe infarction but not the mild one. Of note, the significant increases in [HbO], [HbT], StO2, and μa at both 690 and 830 nm were found on day 3; and increases in μs’ at both 690 and 830 nm were found on day 2 and day 3 after MCAO, respectively. The interhemispheric correlation coefficient (IHCC) was computed from low-frequency hemodynamic oscillation of both hemispheres. Lower IHCCs standing for interhemispheric desynchronizations were found in both mild and severe infarction during occlusion, and only in severe infarction after reperfusion. Our finding supports that sequential FD-NIRS parameters may associated with the severity of the infarction in MCAO model, and the consequent pathologies such as vascular dysfunction and brain edema. Further study is required to validate the potential use of FD-NIRS as a monitor for MCAO verification.
Collapse
|
28
|
The impacts of anesthetic regimens on the middle cerebral artery occlusion outcomes in male rats. Neuroreport 2022; 33:561-568. [DOI: 10.1097/wnr.0000000000001816] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
29
|
Zhou H, Yang X, Yu J, Xu J, Zhang R, Zhang T, Wang X, Ma J. Reference gene identification for normalisation of RT-qPCR analysis in plasma samples of the rat middle cerebral artery occlusion model. Vet Med Sci 2022; 8:2076-2085. [PMID: 35894780 PMCID: PMC9514484 DOI: 10.1002/vms3.879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
OBJECTIVE In quantitative reverse transcription-polymerase chain reaction (RT-qPCR) studies, the selection and validation of reference genes are crucial for the accurate analysis of MicroRNAs (miRNAs) expression. In this work, the optimal reference genes for RT-qPCR normalisation in plasma samples of rat middle cerebral artery occlusion (MCAO) models were identified. METHODS Six rat MCAO models were established. Blood samples were collected before modelling and approximately 16-24 h after modelling. Two commonly used reference genes (U6 and 5S) and three miRNAs (miR-24, miR-122 and miR-9a) were selected as candidate reference genes, and the expression of these genes was detected with RT-qPCR. The acquired data were analysed using geNorm, Normfinder, BestKeeper, RefFinder and comparative delta threshold cycle statistical models. RESULTS The analysed results consistently showed that miR-24 was the most stably expressed reference gene. The 'optimal combination' calculated by geNorm was miR-24, U6 and5S. The expression level of the target gene miR124 was similar when the most stable reference gene miR-24 or the 'optimal combination' was used as a reference gene. However, compared with miR24 or the 'optimal combination', the less stable reference genes influenced the fold change and the data accuracy with a large standard deviation. CONCLUSION These results confirmed the importance of selecting suitable reference genes for normalisation to obtain reliable results in RT-qPCR studies and demonstrated that the identified reference gene miR-24 or the 'optimal combination' could be used as an internal control for gene expression analysis in the rat MCAO model.
Collapse
Affiliation(s)
- Hui Zhou
- Shanghai Innostar Bio‐tech Co. Ltd.China State Institute of Pharmaceutical IndustryShanghaiPeople's Republic of China
| | - Xin Yang
- Shanghai Innostar Bio‐tech Co. Ltd.China State Institute of Pharmaceutical IndustryShanghaiPeople's Republic of China
| | - Jiayi Yu
- Shanghai Innostar Bio‐tech Co. Ltd.China State Institute of Pharmaceutical IndustryShanghaiPeople's Republic of China
| | - Jingyi Xu
- Shanghai Innostar Bio‐tech Co. Ltd.China State Institute of Pharmaceutical IndustryShanghaiPeople's Republic of China
| | - Ruiwen Zhang
- Shanghai Innostar Bio‐tech Co. Ltd.China State Institute of Pharmaceutical IndustryShanghaiPeople's Republic of China
| | - Ting Zhang
- Shanghai Innostar Bio‐tech Co. Ltd.China State Institute of Pharmaceutical IndustryShanghaiPeople's Republic of China
| | - Xijie Wang
- Shanghai Innostar Bio‐tech Co. Ltd.China State Institute of Pharmaceutical IndustryShanghaiPeople's Republic of China
| | - Jing Ma
- Shanghai Innostar Bio‐tech Co. Ltd.China State Institute of Pharmaceutical IndustryShanghaiPeople's Republic of China
| |
Collapse
|
30
|
Chen J, Zhang J, Yang DD, Li ZC, Zhao B, Chen Y, He Z. Clonidine ameliorates cerebral ischemia-reperfusion injury by up-regulating the GluN3 subunits of NMDA receptor. Metab Brain Dis 2022; 37:1829-1841. [PMID: 35727521 DOI: 10.1007/s11011-022-01028-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/04/2022] [Indexed: 10/18/2022]
Abstract
This study aimed to investigate the protective effects of the alpha-2 adrenergic receptor (α2-AR) agonist, clonidine, on the cerebral ischemia-reperfusion (I/R) injury and elaborate the underlying mechanisms. Cerebral I/R model was established by middle cerebral artery occlusion (MCAO) for 2 h followed by reperfusion for 4 h in adult male SD rats. Saline, clonidine and yohimbine (an α2-AR antagonist) were intraperitoneally administered each day for one week before surgery. Neurological deficit was evaluated just before decapitation. TTC staining was applied for correlation of cerebral infarction volume. HE staining was performed to observe the neuron morphology. Immunohistochemical staining was performed to detect the localization and expression of GluN3 proteins. Western blot analysis also was used to detect the expression levels of GluN3 proteins. Our data showed that clonidine ameliorated neurological deficit and reduced the cerebral infarction volume of the rats with cerebral I/R. It is worth noting that treatment with clonidine up-regulated the protein expression of GluN3 in the rats with the cerebral I/R, especially in the cell membrane. Moreover, clonidine also up-regulated the transposition from cytoplasm to cell membrane of GluN3 after cerebral I/R. In addition, yohimbine abolished the neuroprotective effects of clonidine. The results indicated that clonidine played a protective role in cerebral I/R injury through regulation of the protein expression of GluN3 subunits of N-methyl-D-aspartate (NMDA) receptor.
Collapse
Affiliation(s)
- Jing Chen
- Third-Grade Pharmacological Laboratory On Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, People's Republic of China
- Medical College, China Three Gorges University, Yichang, 443002, People's Republic of China
| | - Juan Zhang
- The First People's Hospital of Yichang, Yichang, 443000, People's Republic of China
| | - Dan-Dan Yang
- The Second People's Hospital of China Three Gorges University, Yichang, 443000, People's Republic of China
| | - Zi-Cheng Li
- Third-Grade Pharmacological Laboratory On Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, People's Republic of China
- Medical College, China Three Gorges University, Yichang, 443002, People's Republic of China
| | - Bo Zhao
- Third-Grade Pharmacological Laboratory On Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, People's Republic of China
- Medical College, China Three Gorges University, Yichang, 443002, People's Republic of China
| | - Yue Chen
- Third-Grade Pharmacological Laboratory On Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, People's Republic of China
- Medical College, China Three Gorges University, Yichang, 443002, People's Republic of China
| | - Zhi He
- Third-Grade Pharmacological Laboratory On Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, People's Republic of China.
- Medical College, China Three Gorges University, Yichang, 443002, People's Republic of China.
| |
Collapse
|
31
|
Optimisation of a Mouse Model of Cerebral Ischemia-Reperfusion to Address Issues of Survival and Model Reproducibility and Consistency. COMPUTATIONAL INTELLIGENCE AND NEUROSCIENCE 2022; 2022:7594969. [PMID: 35845875 PMCID: PMC9279060 DOI: 10.1155/2022/7594969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/09/2022] [Accepted: 06/13/2022] [Indexed: 11/17/2022]
Abstract
Middle cerebral artery occlusion (MCAO) induced brain ischemia-reperfusion model in Mice is essential for understanding the pathology of stroke and investigating potential treatments, in which a variety of methods may be employed to block the middle cerebral artery (MCA), the most common being through the insertion of a monofilament; however, in vivo ischemia-reperfusion models are associated, particularly in mice, with high variability in lesion volume and high mortality. We aimed to optimise a mouse model of cerebral ischemia-reperfusion, addressing issues of mouse survival, model reproducibility, and consistency. The model was optimised in two ways: first, insert the monofilament directly through the internal carotid artery rather than through the external or common carotid artery, and second, by extending the length of the silicone coating on the monofilament, the length of the silicone coating enables embolization of the beginning of the middle cerebral artery, as well as the anterior cerebral artery and part of the posterior communicating artery. Results: We assessed various parameters, including blood flow changes in the middle cerebral artery, stability of the infarct area, correlation between infarct volume percentages and neurological deficit scores, mortality, weight changes, and wellbeing. We found that optimisation of the surgical procedure may improve mouse wellbeing and reduce mortality, through reduced weight loss and decrease the variability. In conclusion, we suggest that the optimisation of the model is superior for the study of both short and long-term outcomes of ischemic stroke. These results have considerable implications on stroke model selection for researchers.
Collapse
|
32
|
Development of a Carotid Artery Thrombolysis (iCAT) Stroke Model in Mice. Blood Adv 2022; 6:5449-5462. [PMID: 35767737 PMCID: PMC9631707 DOI: 10.1182/bloodadvances.2021006008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 06/21/2022] [Indexed: 11/20/2022] Open
Abstract
Development of a mouse carotid artery thrombolysis model of stroke. iCAT enables assessment of adjunctive antithrombotic therapies on arterial recanalization, cerebral perfusion, and stroke outcomes.
Recanalization with restored cerebral perfusion is the primary goal of thrombolytic therapy in acute ischemic stroke. The identification of adjunctive therapies that can be safely used to enhance thrombolysis in stroke remains an elusive goal. We report here the development of a mouse in situ carotid artery thrombolysis (iCAT) stroke model involving graded cerebral ischemia to induce unihemispheric infarction after thrombotic occlusion of the common carotid artery (CCA). Electrolytic-induced thrombotic occlusion of the left CCA enabled real-time assessment of recanalization and rethrombosis events after thrombolysis with recombinant tissue-type plasminogen activator (rtPA). Concurrent transient stenosis of the right CCA induced unihemispheric hypoperfusion and infarction in the left middle cerebral artery territory. Real-time assessment of thrombolysis revealed recanalization rates <30% in rtPA-treated animals with high rates of rethrombosis. Addition of the direct thrombin inhibitor argatroban increased recanalization rates to 50% and reduced rethrombosis. Paradoxically, this was associated with increased cerebral ischemia and stroke-related mortality (25%-42%). Serial analysis of carotid and cerebral blood flow showed that coadministration of argatroban with rtPA resulted in a marked increase in carotid artery embolization, leading to distal obstruction of the middle cerebral artery. Real-time imaging of carotid thrombi revealed that adjunctive anticoagulation destabilized platelet-rich thrombi at the vessel wall, leading to dislodgement of large platelet emboli. These studies confirm the benefits of anticoagulants in enhancing thrombolysis and large artery recanalization; however, at high levels of anticoagulation (∼3-fold prolongation of activated partial thromboplastin time), this effect is offset by increased incidence of carotid artery embolization and distal middle cerebral artery occlusion. The iCAT stroke model should provide important new insight into the effects of adjunctive antithrombotic agents on real-time thrombus dynamics during thrombolysis and their correlation with stroke outcomes.
Collapse
|
33
|
Ocular Ischemic Syndrome and Its Related Experimental Models. Int J Mol Sci 2022; 23:ijms23095249. [PMID: 35563640 PMCID: PMC9100201 DOI: 10.3390/ijms23095249] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 04/28/2022] [Accepted: 05/06/2022] [Indexed: 12/17/2022] Open
Abstract
Ocular ischemic syndrome (OIS) is one of the severe ocular disorders occurring from stenosis or occlusion of the carotid arteries. As the ophthalmic artery is derived from the branch of the carotid artery, stenosis or occlusion of the carotid arteries could induce chronic ocular hypoperfusion, finally leading to the development of OIS. To date, the pathophysiology of OIS is still not clearly unraveled. To better explore the pathophysiology of OIS, several experimental models have been developed in rats and mice. Surgical occlusion or stenosis of common carotid arteries or internal carotid arteries was conducted bilaterally or unilaterally for model development. In this regard, final ischemic outcomes in the eye varied depending on the surgical procedure, even though similar findings on ocular hypoperfusion could be observed. In the current review, we provide an overview of the pathophysiology of OIS from various experimental models, as well as several clinical cases. Moreover, we cover the status of current therapies for OIS along with promising preclinical treatments with recent advances. Our review will enable more comprehensive therapeutic approaches to prevent the development and/or progression of OIS.
Collapse
|
34
|
Nirwane A, Yao Y. SMA low/undetectable pericytes differentiate into microglia- and macrophage-like cells in ischemic brain. Cell Mol Life Sci 2022; 79:264. [PMID: 35482211 PMCID: PMC11073453 DOI: 10.1007/s00018-022-04322-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 12/23/2022]
Abstract
Pericytes are multipotent perivascular cells that play important roles in CNS injury. However, controversial findings exist on how pericytes change and whether they differentiated into microglia-like cells after ischemic stroke. This discrepancy is mainly due to the lack of pericyte-specific markers: the "pericyte" population identified in previous studies contained vascular smooth muscle cells (vSMCs) and/or fibroblasts. Therefore, it remains unclear which cell type differentiates into microglia-like cells after stroke. In this study, lineage-tracing technique was used to mark α-smooth muscle actin (SMA)low/undetectable pericytes, vSMCs, and fibroblasts, and their fates were analyzed after ischemic stroke. We found that SMAlow/undetectable pericytes and fibroblasts but not vSMCs substantially proliferated at the subacute phase after injury, and that SMAlow/undetectable pericyte but not vSMCs or fibroblasts differentiated into Iba1+ cells after ischemic stroke. Further imaging flow cytometry analysis revealed that SMAlow/undetectable pericytes differentiated into both microglia and macrophages at day 7 after stroke. These results demonstrate that SMAlow/undetectable pericytes rather than vSMCs or fibroblasts differentiate into both microglia-like and macrophage-like cells after stroke, suggesting that these pericytes may be targeted in the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Abhijit Nirwane
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B. Downs Blvd., MDC8, Tampa, FL, 33612, USA
| | - Yao Yao
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B. Downs Blvd., MDC8, Tampa, FL, 33612, USA.
| |
Collapse
|
35
|
Antioxidant Polyphenols of Antirhea borbonica Medicinal Plant and Caffeic Acid Reduce Cerebrovascular, Inflammatory and Metabolic Disorders Aggravated by High-Fat Diet-Induced Obesity in a Mouse Model of Stroke. Antioxidants (Basel) 2022; 11:antiox11050858. [PMID: 35624723 PMCID: PMC9138119 DOI: 10.3390/antiox11050858] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/22/2022] [Accepted: 04/26/2022] [Indexed: 12/12/2022] Open
Abstract
Metabolic disorders related to obesity and type 2 diabetes are associated with aggravated cerebrovascular damages during stroke. In particular, hyperglycemia alters redox and inflammatory status, leading to cerebral endothelial cell dysfunction, blood–brain barrier (BBB) disruption and brain homeostasis loss. Polyphenols constitute the most abundant dietary antioxidants and exert anti-inflammatory effects that may improve cerebrovascular complications in stroke. This study evaluated the effects of the characterized polyphenol-rich extract of Antirhea borbonica medicinal plant and its major constituent caffeic acid on a high-fat diet (HFD)-induced obesity mouse model during ischemic stroke, and murine bEnd3 cerebral endothelial cells in high glucose condition. In vivo, polyphenols administered by oral gavage for 12 weeks attenuated insulin resistance, hyperglycemia, hyperinsulinemia and dyslipidemia caused by HFD-induced obesity. Polyphenols limited brain infarct, hemorrhagic transformation and BBB disruption aggravated by obesity during stroke. Polyphenols exhibited anti-inflammatory and antioxidant properties by reducing IL-1β, IL-6, MCP-1, TNF-α and Nrf2 overproduction as well as total SOD activity elevation at the cerebral or peripheral levels in obese mice. In vitro, polyphenols decreased MMP-2 activity that correlated with MCP-1 secretion and ROS intracellular levels in hyperglycemic condition. Protective effects of polyphenols were linked to their bioavailability with evidence for circulating metabolites including caffeic acid, quercetin and hippuric acid. Altogether, these findings show that antioxidant polyphenols reduced cerebrovascular, inflammatory and metabolic disorders aggravated by obesity in a mouse model of stroke. It will be relevant to assess polyphenol-based strategies to improve the clinical consequences of stroke in the context of obesity and diabetes.
Collapse
|
36
|
Ernst L, Kümmecke AM, Zieglowski L, Liu W, Schulz M, Czigany Z, Tolba RH. Severity Assessment in Rats Undergoing Subarachnoid Hemorrhage Induction by Endovascular Perforation or Corresponding Sham Surgery. Eur Surg Res 2022; 64:120-138. [PMID: 35385845 PMCID: PMC9808704 DOI: 10.1159/000524432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 03/29/2022] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Animal models for preclinical research of subarachnoid hemorrhage (SAH) are widely used as much of the pathophysiology remains unknown. However, the burden of these models inflicted on the animals is not well characterized. The European directive requires severity assessment-based allocation to categories. Up to now, the classification into predefined categories is rather subjective and often without underlying scientific knowledge. We therefore aimed at assessing the burden of rats after SAH or the corresponding sham surgery to provide a scientific assessment. METHODS We performed a multimodal approach, using different behavior tests, clinical and neurological scoring, and biochemical markers using the common model for SAH of intracranial endovascular filament perforation in male Wistar rats. Up to 7 days after surgery, animals with SAH were compared to sham surgery and to a group receiving only anesthesia and analgesia. RESULTS Sham surgery (n = 15) and SAH (n = 16) animals showed an increase in the clinical score the first days after surgery, indicating clinical deterioration, while animals receiving only anesthesia without surgery (n = 5) remained unaffected. Body weight loss occurred in all groups but was more pronounced and statistically significant only after surgery. The analysis of burrowing, open field (total distance, erections), balance beam, and neuroscore showed primarily an effect of the surgery itself in sham surgery and SAH animals. Only concerning balance beam and neuroscore, a difference was visible between sham surgery and SAH. The outcome of the analysis of systemic and local inflammatory parameters and of corticosterone in blood and its metabolites in feces was only robust in animals suffering from larger bleedings. Application of principal component analysis resulted in a clear separation of sham surgery and SAH animals from their respective baseline as well as from the anesthesia-only group at days 1 and 3, with the difference between sham surgery and SAH being not significant. DISCUSSION/CONCLUSION To our knowledge, we are the first to publish detailed clinical score sheet data combined with advanced behavioral assessment in the endovascular perforation model for SAH in rats. The tests chosen here clearly depict an impairment of the animals within the first days after surgery and are consequently well suited for assessment of the animals' suffering in the model. A definitive classification into one of the severity categories named by the EU directive is yet pending and has to be performed in the future by including the assessment data from different neurological and nonneurological disease models.
Collapse
Affiliation(s)
- Lisa Ernst
- Institute for Laboratory Animal Science & Experimental Surgery, Faculty of Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Anna Maria Kümmecke
- Institute for Laboratory Animal Science & Experimental Surgery, Faculty of Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Leonie Zieglowski
- Institute for Laboratory Animal Science & Experimental Surgery, Faculty of Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Wenjia Liu
- Department of Surgery and Transplantation, Faculty of Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Mareike Schulz
- Institute for Laboratory Animal Science & Experimental Surgery, Faculty of Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Zoltan Czigany
- Department of Surgery and Transplantation, Faculty of Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - René H. Tolba
- Institute for Laboratory Animal Science & Experimental Surgery, Faculty of Medicine, University Hospital RWTH Aachen, Aachen, Germany
| |
Collapse
|
37
|
Pinto R, Magalhães A, Sousa M, Melo L, Lobo A, Barros P, Gomes JR. Bridging the Transient Intraluminal Stroke Preclinical Model to Clinical Practice: From Improved Surgical Procedures to a Workflow of Functional Tests. Front Neurol 2022; 13:846735. [PMID: 35359638 PMCID: PMC8963503 DOI: 10.3389/fneur.2022.846735] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/07/2022] [Indexed: 12/18/2022] Open
Abstract
Acute ischemic stroke (AIS) remains a leading cause of mortality, despite significant advances in therapy (endovascular thrombectomy). Failure in developing novel effective therapies is associated with unsuccessful translation from preclinical studies to clinical practice, associated to inconsistent and highly variable infarct areas and lack of relevant post-stroke functional evaluation in preclinical research. To outreach these limitations, we optimized the intraluminal transient middle cerebral occlusion, a widely used mouse stroke model, in two key parameters, selection of appropriate occlusion filaments and time of occlusion, which show a significant variation in the literature. We demonstrate that commercially available filaments with short coating length (1–2 mm), together with 45-min occlusion, results in a consistent affected brain region, similar to what is observed in most patients with AIS. Importantly, a dedicated post-stroke care protocol, based on clinical practice applied to patients who had stroke, resulted in lower mortality and improved mice welfare. Finally, a battery of tests covering relevant fine motor skills, sensory functions, and learning/memory behaviors revealed a significant effect of tMCAO brain infarction, which is parallel to patient symptomatology as measured by relevant clinical scales (NIH Stroke Scale, NIHSS and modified Rankin Scale, mRS). Thus, in order to enhance translation to clinical practice, future preclinical stroke research must consider the methodology described in this study, which includes improved reproducible surgical procedure, postoperative care, and the battery of functional tests. This will be a major step s closing the gap from bench to bedside, rendering the development of novel effective therapeutic approaches.
Collapse
Affiliation(s)
- Raquel Pinto
- Molecular Neurobiology Unit, IBMC-Instituto de Biologia Molecular e Celular, Porto, Portugal.,I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Ana Magalhães
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Addiction Biology Unit, IBMC-Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Mafalda Sousa
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Advanced Light Microscopy Unit, I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Lúcia Melo
- Molecular Neurobiology Unit, IBMC-Instituto de Biologia Molecular e Celular, Porto, Portugal.,I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Andrea Lobo
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Addiction Biology Unit, IBMC-Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Pedro Barros
- Neurology Department, Centro Hospitalar de Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal.,Stroke Unit, Centro Hospitalar de Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
| | - João R Gomes
- Molecular Neurobiology Unit, IBMC-Instituto de Biologia Molecular e Celular, Porto, Portugal.,I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| |
Collapse
|
38
|
Jiao Y, Wang J, Jia Y, Xue M. Remote ischemic preconditioning protects against cerebral ischemia injury in rats by upregulating miR-204-5p and activating the PINK1/Parkin signaling pathway. Metab Brain Dis 2022; 37:945-959. [PMID: 35067796 DOI: 10.1007/s11011-022-00910-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 01/12/2022] [Indexed: 11/25/2022]
Abstract
Remote ischemic preconditioning (RiPC) is the process where preconditioning ischemia protects the organs against the subsequent index ischemia. RiPC is a protective method for brain damage. This study is to explore the effect and mechanism of RiPC in cerebral ischemia injury in rats through regulation of miR-204-5p/BRD4 expression. Middle cerebral artery occlusion (MCAO) rat model and glucose deprivation (OGD) neuron model were established. The effect of RiPC on neurological deficits, cerebral infarct size, autophagy marker, inflammatory cytokines and apoptosis was evaluated. miR-204-5p expression was analyzed using RT-qPCR, and then downregulated using miR-204-5p antagomir to estimate its effect on MCAO rats. The downstream mechanism of miR-204-5p was explored. RiPC promoted autophagy, reduced cerebral infarct volume and neurological deficit score, and alleviated apoptosis and cerebral ischemia injury in rats, with no significant effects on healthy rat brains. RiPC up-regulated miR-204-5p expression in MCAO rats. miR-204-5p knockdown partially reversed the effect of RiPC. RiPC promoted autophagy in OGD cells, and attenuated inflammation and apoptosis. miR-204-5p targeted BRD4, which partially reversed the effect of miR-204-5p on OGD cells. RiPC activated the PINK1/Parkin pathway via the miR-204-5p/BRD4 axis. In conclusion, RiPC activated the PINK1/Parkin pathway and prevented cerebral ischemia injury by up-regulating miR-204-5p and inhibiting BRD4.
Collapse
Affiliation(s)
- Yiming Jiao
- The Departments of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, 2 Jingba Road, Zhengzhou, 450001, Henan, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, Henan, China
| | - Jinlan Wang
- The Departments of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, 2 Jingba Road, Zhengzhou, 450001, Henan, China
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, Henan, China
| | - Yanjie Jia
- The Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengzhou Xue
- The Departments of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, 2 Jingba Road, Zhengzhou, 450001, Henan, China.
- Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, Henan, China.
| |
Collapse
|
39
|
Yang Z, Li X, Cao Z, Wang P, Warner DS, Sheng H. Post-ischemia common carotid artery occlusion worsens memory loss, but not sensorimotor deficits, in long-term survived stroke mice. Brain Res Bull 2022; 183:153-161. [PMID: 35304288 DOI: 10.1016/j.brainresbull.2022.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 03/07/2022] [Accepted: 03/11/2022] [Indexed: 11/02/2022]
Abstract
Ischemic stroke in rodents is usually induced by intraluminal occlusion of the middle cerebral artery (MCA) via the external carotid artery (ECA) or the common carotid artery (CCA). The latter route requires permanent CCA occlusion after ischemia, and here, we assess its effects on long-term outcomes. Transient occlusion of MCA and CCA was performed at normal body temperature. After 90minutes of ischemia, mice were randomized to permanent CCA occlusion or no occlusion (control group). Body weight, and motor and sensory functions, ie, pole test, adhesive tape removal, and elevated plus maze, were evaluated at 24hours, and at 7 and 28 days after stroke. Infarct volume, apoptosis, and activation of astrocytes and microglia were assessed at 4 weeks by an investigator blinded to groups. The Morris water maze test was performed at 3 weeks in the second experiment. One mouse died at 4 days, and the other mice survived with persistent neurologic deficits. CCA-occluded mice exhibited delayed turn on the pole at 24hours and decreased responses to the von Frey filament, and spent more time on the pole at 7 and 28 days than the control group. Infarction, hemispheric atrophy, glial activation, and apoptotic neuronal death were present in all mice, and no intra-group difference was found. However, CCA-occluded mice had a significantly poorer performance in the Morris water maze compared to the control group, which showed an adverse effect of post-ischemia CCA occlusion on cognition. Thus, the model selection should be well considered in preclinical efficacy studies on stroke-induced vascular dementia and stroke with Alzheimer's disease.
Collapse
Affiliation(s)
- Zhong Yang
- Multidisciplinary Neuroprotection Laboratories, Center of Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Orthopedics, The Fifth Central Hospital of Tianjin, Tanggu District, Tianjin, 300450, China
| | - Xuan Li
- Multidisciplinary Neuroprotection Laboratories, Center of Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| | - Zhipeng Cao
- Multidisciplinary Neuroprotection Laboratories, Center of Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA; School of Forensic Medicine, China Medical University, Shenyang Liaoning, 110122, China
| | - Peng Wang
- Multidisciplinary Neuroprotection Laboratories, Center of Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Anesthesiology, The Fifth Central Hospital of Tianjin, Tanggu District, Tianjin, 300450, China
| | - David S Warner
- Multidisciplinary Neuroprotection Laboratories, Center of Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA; Surgery, Duke University Medical Center, Durham, NC 27710, USA; Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Huaxin Sheng
- Multidisciplinary Neuroprotection Laboratories, Center of Perioperative Organ Protection, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
40
|
Cramer T, Gill R, Thirouin ZS, Vaas M, Sampath S, Martineau F, Noya SB, Panzanelli P, Sudharshan TJJ, Colameo D, Chang PKY, Wu PY, Shi R, Barker PA, Brown SA, Paolicelli RC, Klohs J, McKinney RA, Tyagarajan SK. Cross-talk between GABAergic postsynapse and microglia regulate synapse loss after brain ischemia. SCIENCE ADVANCES 2022; 8:eabj0112. [PMID: 35245123 PMCID: PMC8896802 DOI: 10.1126/sciadv.abj0112] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Microglia interact with neurons to facilitate synapse plasticity; however, signal(s) contributing to microglia activation for synapse elimination in pathology are not fully understood. Here, using in vitro organotypic hippocampal slice cultures and transient middle cerebral artery occlusion (MCAO) in genetically engineered mice in vivo, we report that at 24 hours after ischemia, microglia release brain-derived neurotrophic factor (BDNF) to downregulate glutamatergic and GABAergic synapses within the peri-infarct area. Analysis of the cornu ammonis 1 (CA1) in vitro shows that proBDNF and mBDNF downregulate glutamatergic dendritic spines and gephyrin scaffold stability through p75 neurotrophin receptor (p75NTR) and tropomyosin receptor kinase B (TrkB) receptors, respectively. After MCAO, we report that in the peri-infarct area and in the corresponding contralateral hemisphere, similar neuroplasticity occurs through microglia activation and gephyrin phosphorylation at serine-268 and serine-270 in vivo. Targeted deletion of the Bdnf gene in microglia or GphnS268A/S270A (phospho-null) point mutations protects against ischemic brain damage, neuroinflammation, and synapse downregulation after MCAO.
Collapse
Affiliation(s)
- Teresa Cramer
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, CH 8057 Zürich, Switzerland
| | - Raminder Gill
- Department of Pharmacology and Therapeutics, McGill University, 3649 Prom. Sir-William-Osler, Montreal, QC H3G 0B1, Canada
| | - Zahra S. Thirouin
- Research Institute of the McGill University Health Centre, 1650 Cedar Avenue, Montreal, QC H3G1A4, Canada
| | - Markus Vaas
- Clinical Trials Center, University Hospital Zurich, Rämistrasse 100/MOU2, CH 8044 Zürich, Switzerland
| | - Suchita Sampath
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, CH 8057 Zürich, Switzerland
| | - Fanny Martineau
- Department of Biomedical Sciences, University of Lausanne, Rue du Bugnon 7, CH 1005 Lausanne, Switzerland
| | - Sara B. Noya
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, CH 8057 Zürich, Switzerland
| | - Patrizia Panzanelli
- Department of Neuroscience Rita Levi Montalcini, University of Turin, Turin, Italy
| | - Tania J. J. Sudharshan
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, CH 8057 Zürich, Switzerland
| | - David Colameo
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, CH 8057 Zürich, Switzerland
| | - Philip K.-Y. Chang
- Department of Pharmacology and Therapeutics, McGill University, 3649 Prom. Sir-William-Osler, Montreal, QC H3G 0B1, Canada
| | - Pei You Wu
- Department of Pharmacology and Therapeutics, McGill University, 3649 Prom. Sir-William-Osler, Montreal, QC H3G 0B1, Canada
| | - Roy Shi
- Department of Pharmacology and Therapeutics, McGill University, 3649 Prom. Sir-William-Osler, Montreal, QC H3G 0B1, Canada
| | - Philip A. Barker
- Department of Biology, University of British Columbia, 3187 University Way, ASC 413, Kelowna, BC V1V 1V7, Canada
| | - Steven A. Brown
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, CH 8057 Zürich, Switzerland
| | - Rosa C. Paolicelli
- Department of Biomedical Sciences, University of Lausanne, Rue du Bugnon 7, CH 1005 Lausanne, Switzerland
| | - Jan Klohs
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Wolfgang-Pauli-Strasse 27, CH 8093 Zürich, Switzerland
| | - Rebecca Anne McKinney
- Department of Pharmacology and Therapeutics, McGill University, 3649 Prom. Sir-William-Osler, Montreal, QC H3G 0B1, Canada
- Corresponding author. (S.K.T.); (R.A.M.)
| | - Shiva K. Tyagarajan
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, CH 8057 Zürich, Switzerland
- Department of Pharmacology and Therapeutics, McGill University, 3649 Prom. Sir-William-Osler, Montreal, QC H3G 0B1, Canada
- Corresponding author. (S.K.T.); (R.A.M.)
| |
Collapse
|
41
|
Multiparametric monitoring of early pathophysiological changes in a porcine model of sequential focal and global cerebral ischemia. World Neurosurg 2022; 161:e473-e481. [DOI: 10.1016/j.wneu.2022.02.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 02/10/2022] [Indexed: 11/23/2022]
|
42
|
Gulyaeva NV, Onufriev MV, Moiseeva YV. Ischemic Stroke, Glucocorticoids, and Remote Hippocampal Damage: A Translational Outlook and Implications for Modeling. Front Neurosci 2021; 15:781964. [PMID: 34955730 PMCID: PMC8695719 DOI: 10.3389/fnins.2021.781964] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 11/22/2021] [Indexed: 01/16/2023] Open
Abstract
Progress in treating ischemic stroke (IS) and its delayed consequences has been frustratingly slow due to the insufficient knowledge on the mechanism. One important factor, the hypothalamic-pituitary-adrenocortical (HPA) axis is mostly neglected despite the fact that both clinical data and the results from rodent models of IS show that glucocorticoids, the hormones of this stress axis, are involved in IS-induced brain dysfunction. Though increased cortisol in IS is regarded as a biomarker of higher mortality and worse recovery prognosis, the detailed mechanisms of HPA axis dysfunction involvement in delayed post-stroke cognitive and emotional disorders remain obscure. In this review, we analyze IS-induced HPA axis alterations and supposed association of corticoid-dependent distant hippocampal damage to post-stroke brain disorders. A translationally important growing point in bridging the gap between IS pathogenesis and clinic is to investigate the involvement of the HPA axis disturbances and related hippocampal dysfunction at different stages of SI. Valid models that reproduce the state of the HPA axis in clinical cases of IS are needed, and this should be considered when planning pre-clinical research. In clinical studies of IS, it is useful to reinforce diagnostic and prognostic potential of cortisol and other HPA axis hormones. Finally, it is important to reveal IS patients with permanently disturbed HPA axis. Patients-at-risk with high cortisol prone to delayed remote hippocampal damage should be monitored since hippocampal dysfunction may be the basis for development of post-stroke cognitive and emotional disturbances, as well as epilepsy.
Collapse
Affiliation(s)
- Natalia V Gulyaeva
- Laboratory of Functional Biochemistry of Nervous System, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, Russia.,Research and Clinical Center for Neuropsychiatry of Moscow Healthcare Department, Moscow, Russia
| | - Mikhail V Onufriev
- Laboratory of Functional Biochemistry of Nervous System, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, Russia.,Research and Clinical Center for Neuropsychiatry of Moscow Healthcare Department, Moscow, Russia
| | - Yulia V Moiseeva
- Laboratory of Functional Biochemistry of Nervous System, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
43
|
Onufriev MV, Moiseeva YV, Zhanina MY, Lazareva NA, Gulyaeva NV. A Comparative Study of Koizumi and Longa Methods of Intraluminal Filament Middle Cerebral Artery Occlusion in Rats: Early Corticosterone and Inflammatory Response in the Hippocampus and Frontal Cortex. Int J Mol Sci 2021; 22:13544. [PMID: 34948340 PMCID: PMC8703333 DOI: 10.3390/ijms222413544] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/11/2021] [Accepted: 12/14/2021] [Indexed: 02/08/2023] Open
Abstract
Two classical surgical approaches for intraluminal filament middle cerebral artery occlusion (MCAO), the Longa et al. (LM) and Koizumi et al. methods (KM), are used as alternatives in preclinical studies to induce stroke in rodents. Comparisons of these MCAO models in mice showed critical differences between them along with similarities (Smith et al. 2015; Morris et al. 2016). In this study, a direct comparison of MCAO-KM and MCAO-LM in rats was performed. Three days after MCAO, infarct volume, mortality rate, neurological deficit, and weight loss were similar in these models. MCAO-LM rats showed an increase in ACTH levels, while MCAO-KM rats demonstrated elevated corticosterone and interleukin-1β in blood serum. Corticosterone accumulation was detected in the frontal cortex (FC) and the hippocampus of the MCAO-KM group. IL1β beta increased in the ipsilateral hippocampus in the MCAO-KM group and decreased in the contralateral FC of MCAO-LM rats. Differences revealed between MCAO-KM and MCAO-LM suggest that corticosterone and interleukin-1β release as well as hippocampal accumulation is more expressed in MCAO-KM rats, predisposing them to corticosterone-dependent distant neuroinflammatory hippocampal damage. The differences between two models, particularly, malfunction of the hypothalamic-pituitary-adrenal axis, should be considered in the interpretation, comparison, and translation of pre-clinical experimental results.
Collapse
Affiliation(s)
- Mikhail V. Onufriev
- Laboratory of Functional Biochemistry of the Nervous System, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 5A Butlerov Str., 117485 Moscow, Russia; (M.V.O.); (Y.V.M.); (M.Y.Z.); (N.A.L.)
- Research and Clinical Center for Neuropsychiatry of Moscow Healthcare Department, 43 Donskaya Str., 115419 Moscow, Russia
| | - Yulia V. Moiseeva
- Laboratory of Functional Biochemistry of the Nervous System, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 5A Butlerov Str., 117485 Moscow, Russia; (M.V.O.); (Y.V.M.); (M.Y.Z.); (N.A.L.)
| | - Marina Y. Zhanina
- Laboratory of Functional Biochemistry of the Nervous System, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 5A Butlerov Str., 117485 Moscow, Russia; (M.V.O.); (Y.V.M.); (M.Y.Z.); (N.A.L.)
- Research and Clinical Center for Neuropsychiatry of Moscow Healthcare Department, 43 Donskaya Str., 115419 Moscow, Russia
| | - Natalia A. Lazareva
- Laboratory of Functional Biochemistry of the Nervous System, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 5A Butlerov Str., 117485 Moscow, Russia; (M.V.O.); (Y.V.M.); (M.Y.Z.); (N.A.L.)
| | - Natalia V. Gulyaeva
- Laboratory of Functional Biochemistry of the Nervous System, Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 5A Butlerov Str., 117485 Moscow, Russia; (M.V.O.); (Y.V.M.); (M.Y.Z.); (N.A.L.)
- Research and Clinical Center for Neuropsychiatry of Moscow Healthcare Department, 43 Donskaya Str., 115419 Moscow, Russia
| |
Collapse
|
44
|
Kang EJ, Cho S, Lim C, Lee B, Kim YK, Kim KM. Effects of the methanol fraction of modified Seonghyangjeongki-san water extract on transient ischaemic brain injury in mice. PHARMACEUTICAL BIOLOGY 2021; 59:840-853. [PMID: 34184969 PMCID: PMC8245089 DOI: 10.1080/13880209.2021.1941130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 06/04/2021] [Accepted: 06/04/2021] [Indexed: 06/13/2023]
Abstract
CONTEXT Recently in Korean medicine, the antioxidant and anti-inflammatory activities of Seonghyangjeongki-san (SHJKS) were reported. However, studies on the specific mechanisms of action of SHJKS for the treatment of ischaemic stroke are still lacking. OBJECTIVE This study investigates the mechanism of action of the water extract methanol fraction of modified SHJKS (SHJKSmex) on cerebral ischaemic injury. MATERIALS AND METHODS C57BL/6 male mice were orally administered SHJKSmex (30, 100, or 300 mg/kg) for 3 consecutive days (2 days, 1 day, and 1 h, respectively) before middle cerebral artery occlusion (MCAO). Twenty-four hours after MCAO, the infarct volumes were measured, brain edoema indices were calculated, and neurological deficit scores were determined. Inflammation-related substances in the ipsilateral hemisphere were determined by western blotting, dichlorofluorescin diacetate, thiobarbituric acid-reactive substances assay, and enzyme-linked immunosorbent assay. RESULTS SHJKSmex pre-treatment at 300 mg/kg decreased infarct volume by 87% and mean brain water content by 90% of the MCAO control group. Moreover, SHJKSmex effectively suppressed the expression of inducible nitric oxide synthase, reactive oxygen species, interleukin 1, and caspases-8 and -9 and increased the B-cell lymphoma 2/Bcl-2-associated X protein ratio (Bcl-2/Bax) in ischaemic mouse brain. The hippocampal pyramidal cell densities were significantly increased in the 300 mg/kg SHJKSmex-administered group compared to the MCAO control group. DISCUSSION AND CONCLUSIONS SHJKSmex protected the brain from ischaemic stroke in mice through its antioxidant, anti-inflammatory, and antiapoptotic activities. Our findings suggest that SHJKSmex is a promising therapeutic candidate for the development of a new formulation for ischaemia-induced brain damage.
Collapse
Affiliation(s)
- Eun-Jin Kang
- College of Korean Medicine, Dong-Eui University, Busan, Republic of Korea
| | - Suin Cho
- School of Korean Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Chiyeon Lim
- College of Medicine, Dongguk University, Gyeonggi-do, Republic of Korea
| | - Byoungho Lee
- Inju Hospital of Korean Medicine, Incheon, Republic of Korea
| | - Young Kyun Kim
- College of Korean Medicine, Dong-Eui University, Busan, Republic of Korea
| | - Kyoung-Min Kim
- College of Korean Medicine, Dong-Eui University, Busan, Republic of Korea
| |
Collapse
|
45
|
Nishie H, Nakano-Doi A, Sawano T, Nakagomi T. Establishment of a Reproducible Ischemic Stroke Model in Nestin-GFP Mice with High Survival Rates. Int J Mol Sci 2021; 22:ijms222312997. [PMID: 34884811 PMCID: PMC8657611 DOI: 10.3390/ijms222312997] [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: 10/16/2021] [Revised: 11/22/2021] [Accepted: 11/26/2021] [Indexed: 11/24/2022] Open
Abstract
An accumulation of evidence shows that endogenous neural stem/progenitor cells (NSPCs) are activated following brain injury such as that suffered during ischemic stroke. To understand the expression patterns of these cells, researchers have developed mice that express an NSPC marker, Nestin, which is detectable by specific reporters such as green fluorescent protein (GFP), i.e., Nestin-GFP mice. However, the genetic background of most transgenic mice, including Nestin-GFP mice, comes from the C57BL/6 strain. Because mice from this background strain have many cerebral arterial branches and collateral vessels, they are accompanied by several major problems including variable ischemic areas and high mortality when subjected to ischemic stroke by occluding the middle cerebral artery (MCA). In contrast, CB-17 wild-type mice are free from these problems. Therefore, with the aim of overcoming the aforementioned defects, we first crossed Nestin-GFP mice (C57BL/6 background) with CB-17 wild-type mice and then developed Nestin-GFP mice (CB-17 background) by further backcrossing the generated hybrid mice with CB-17 wild-type mice. Subsequently, we investigated the phenotypes of the established Nestin-GFP mice (CB-17 background) following MCA occlusion; these mice had fewer blood vessels around the MCA compared with the number of blood vessels in Nestin-GFP mice (C57BL/6 background). In addition, TTC staining showed that infarcted volume was variable in Nestin-GFP mice (C57BL/6 background) but highly reproducible in Nestin-GFP mice (CB-17 background). In a further investigation of mice survival rates up to 28 days after MCA occlusion, all Nestin-GFP mice (CB-17 background) survived the period, whereas Nestin-GFP mice (C57BL/6 background) frequently died within 1 week and exhibited a higher mortality rate. Immunohistochemistry analysis of Nestin-GFP mice (CB-17 background) showed that GFP+ cells were mainly obverted in not only conventional neurogenic areas, including the subventricular zone (SVZ), but also ischemic areas. In vitro, cells isolated from the ischemic areas and the SVZ formed GFP+ neurosphere-like cell clusters that gave rise to various neural lineages including neurons, astrocytes, and oligodendrocytes. However, microarray analysis of these cells and genetic mapping experiments by Nestin-CreERT2 Line4 mice crossed with yellow fluorescent protein (YFP) reporter mice (Nestin promoter-driven YFP-expressing mice) indicated that cells with NSPC activities in the ischemic areas and the SVZ had different characteristics and origins. These results show that the expression patterns and fate of GFP+ cells with NSPC activities can be precisely investigated over a long period in Nestin-GFP mice (CB-17 background), which is not necessarily possible with Nestin-GFP mice (C57BL/6 background). Thus, Nestin-GFP mice (CB-17 background) could become a useful tool with which to investigate the mechanism of neurogenesis via the aforementioned cells under pathological conditions such as following ischemic stroke.
Collapse
Affiliation(s)
- Hideaki Nishie
- Institute for Advanced Medical Sciences, Hyogo College of Medicine, 1-1 Mukogawacho, Nishinomiya 663-8501, Japan; (H.N.); (A.N.-D.)
| | - Akiko Nakano-Doi
- Institute for Advanced Medical Sciences, Hyogo College of Medicine, 1-1 Mukogawacho, Nishinomiya 663-8501, Japan; (H.N.); (A.N.-D.)
- Department of Therapeutic Progress in Brain Diseases, Hyogo College of Medicine, 1-1 Mukogawacho, Nishinomiya 663-8501, Japan
| | - Toshinori Sawano
- Department of Biomedical Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu 525-8577, Japan;
| | - Takayuki Nakagomi
- Institute for Advanced Medical Sciences, Hyogo College of Medicine, 1-1 Mukogawacho, Nishinomiya 663-8501, Japan; (H.N.); (A.N.-D.)
- Department of Therapeutic Progress in Brain Diseases, Hyogo College of Medicine, 1-1 Mukogawacho, Nishinomiya 663-8501, Japan
- Correspondence: ; Tel.: +81-798-45-6821; Fax: +81-798-45-6823
| |
Collapse
|
46
|
Scalp Acupuncture and Treadmill Training Inhibits Neuronal Apoptosis through Activating cIAP1 in Cerebral Ischemia Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:1418616. [PMID: 34804173 PMCID: PMC8604578 DOI: 10.1155/2021/1418616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 10/02/2021] [Accepted: 10/26/2021] [Indexed: 12/20/2022]
Abstract
Stroke is the leading cause of long-term disability in developed countries. Multitudinous evidence suggests that treadmill training treatment is beneficial for balance and stroke rehabilitation; however, the need for stroke therapy remains unmet. In the present study, a cerebral ischemia rat model was established by permanent middle cerebral artery occlusion (pMCAO) to explore the therapeutic effect and mechanism of scalp acupuncture combined with treadmill training on ischemic stroke. Terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling and neuronal nuclear protein (NeuN) double staining and cellular inhibitor of apoptosis protein-1 (cIAP1) and NeuN immunofluorescence double staining were used to detect the short-term and long-term neuroprotective effects of scalp acupuncture combined with treadmill training on pMCAO rats. In addition, the antiapoptotic effect of the combined treatment was evaluated in pMCAO rats transfected with cIAP1 shRNA. Western blotting was used to detect the relative protein expression in the caspase-8/-9/-3 activation pathway downstream of cIAP1 to further clarify its regulatory mechanism. Our results showed that scalp acupuncture combined with treadmill training successfully achieved short-term and long-term functional improvement within 14 days after stroke, significantly inhibited neuronal apoptosis, and upregulated the expression of cIAP1 protein in the ischemic penumbra area of the ischemic brain. However, no significant functional improvement and antiapoptotic effect were found in pMCAO rats transfected with cIAP1 shRNA. Western blotting results showed that the combined therapy markedly inhibited the activation of the caspase-8/-9/-3 pathway. These findings indicate that scalp acupuncture combined with treadmill training therapy may serve as a more effective alternative modality in the treatment of ischemic stroke, playing an antiapoptotic role by upregulating the expression of cIAP1 and inhibiting the activation of the caspase-8/-9/-3 pathway.
Collapse
|
47
|
Electroacupuncture Preconditioning Reduces Oxidative Stress in the Acute Phase of Cerebral Ischemia-Reperfusion in Rats by Regulating Iron Metabolism Pathways. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:3056963. [PMID: 34790244 PMCID: PMC8592755 DOI: 10.1155/2021/3056963] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 10/08/2021] [Accepted: 10/25/2021] [Indexed: 12/14/2022]
Abstract
Background Oxidative stress is an important mechanism of cerebral ischemia-reperfusion injury. Ferroptosis caused by iron overload after cerebral ischemia-reperfusion is considered a common cause of oxidative stress. Many recent studies have shown that electroacupuncture (EA) can regulate the expression of inflammatory factors, and the use of electroacupuncture preconditioning can produce a protective effect, which can reduce injury after cerebral ischemia and reperfusion. We aimed to assess whether EA could be used to reduce oxidative stress. Methods The oxidative stress level of rats during the acute phase of cerebral ischemia and reperfusion was assessed with and without preconditioning with EA. Molecular biology methods were used to detect iron metabolism and oxidative stress-related proteins. Results Rats that had EA preconditioning had lower infarct volumes than rats in the control group. Furthermore, western blot analysis showed that the expression of iron metabolism-related protein FPN-1 was higher in the intervention group than in the model group after reperfusion. In this regard, further investigation also demonstrated higher expression of glutathione and glutathione peroxidase-4, and lower reactive oxygen species values in the brain tissue of the EA group were compared with those of the control group rats. Conclusions Electroacupuncture preconditioning can reduce oxidative stress after cerebral ischemia-reperfusion by regulating iron overload.
Collapse
|
48
|
Wang Y, Liu X, Hu T, Li X, Chen Y, Xiao G, Huang J, Chang Y, Zhu Y, Zhang H, Wang Y. Astragalus saponins improves stroke by promoting the proliferation of neural stem cells through phosphorylation of Akt. JOURNAL OF ETHNOPHARMACOLOGY 2021; 277:114224. [PMID: 34044075 DOI: 10.1016/j.jep.2021.114224] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 05/01/2021] [Accepted: 05/19/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As one of major components of Buyang Huanwu decoction, Astragali Radix is broadly used for stroke treatment. Astragalus saponins (AST), the main active compound from Astragali Radix has the potentials for neuroprotection and improving spatial memory without clear pharmacological mechanism. AIM OF THE STUDY The aim of this study was to investigate that pretreatment of AST is beneficial to protect against focal ischemic stroke in mouse model and its related underlying mechanism. MATERIALS AND METHODS The neurological and motor function of MCAO mice were assessed by TTC staining and CatWalk gait analysis. The effect of AST on proliferation of NSCs was showed by the expression of Ki67 of MCAO mice and the number and size of primary neurospheres cultured from adult SVZ. The intersection of stroke-related targets, neurogenesis targets and drug-related targets were identified by the online website (https://www.omicstudio.cn/index). Then GO functional annotation and KEGG pathway enrichment analysis were performed. Candidate target Akt was confirmed to increase proliferation of cultured NSCs from adult SVZ by CCK8 assay and Western blot. RESULTS We found that with the prolongation of administration time, AST improved neurological and motor function of MCAO mice, by promoting the proliferation of NSCs both in vivo and in vitro. Then, the primary network among drug, genes and biological pathway was established by using compound-target-disease & function-pathway analysis of astragalus membranaceus. PI3K/Akt which plays a key role in cell proliferation was among the top 10 most significant GO terms from above three aspects. Further analysis using cultured NSCs from adult SVZ confirmed that AST, astragaloside I (A1) and astragaloside III (A3) increased the proliferation of NSCs through targeting Akt. CONCLUSION The present study elucidated that Astragalus saponins pretreatment could provide a protective effect on experimental stroke mainly by enhancing proliferation of NSCs through targeting Akt. The findings provided a basis for the development of novel strategies for the treatment of stroke.
Collapse
Affiliation(s)
- Yu Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, People's Republic of China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Xu Liu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Tingdong Hu
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China
| | - Xin Li
- Zhongxin Pharma, Tianjin No.6 Traditional Chinese Medicine Factory, Tianjin, 300401, China
| | - Yuru Chen
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, People's Republic of China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Guangxu Xiao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Juyang Huang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin 301617, People's Republic of China
| | - Yanxu Chang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Yan Zhu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Han Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| | - Ying Wang
- Jiangsu Key Laboratory of Xenotransplantation, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, People's Republic of China.
| |
Collapse
|
49
|
Abstract
Stroke is a devastating disease with high morbidity and mortality. Animal models are indispensable tools that can mimic stroke processes and can be used for investigating mechanisms and developing novel therapeutic regimens. As a heterogeneous disease with complex pathophysiology, mimicking all aspects of human stroke in one animal model is impossible. Each model has unique strengths and weaknesses. Models such as transient or permanent intraluminal thread occlusion middle cerebral artery occlusion (MCAo) models and thromboembolic models are the most commonly used in simulating human ischemic stroke. The endovascular filament occlusion model is characterized by easy manipulation and accurately controllable reperfusion and is suitable for studying the pathogenesis of focal ischemic stroke and reperfusion injury. Although the reproducibility of the embolic model is poor, it is more convenient for investigating thrombolysis. Rats are the most frequently used animal model for stroke. This review mainly outlines the stroke models of rats and discusses their strengths and shortcomings in detail.
Collapse
Affiliation(s)
- Yanyu Li
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Diseases of Guangdong Medical UniversityZhanjiangChina
| | - Jingjing Zhang
- Affiliated Hospital of Guangdong Medical University & Key Laboratory of Zebrafish Model for Development and Diseases of Guangdong Medical UniversityZhanjiangChina
| |
Collapse
|
50
|
Faillot M, Chaillet A, Palfi S, Senova S. Rodent models used in preclinical studies of deep brain stimulation to rescue memory deficits. Neurosci Biobehav Rev 2021; 130:410-432. [PMID: 34437937 DOI: 10.1016/j.neubiorev.2021.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 11/28/2022]
Abstract
Deep brain stimulation paradigms might be used to treat memory disorders in patients with stroke or traumatic brain injury. However, proof of concept studies in animal models are needed before clinical translation. We propose here a comprehensive review of rodent models for Traumatic Brain Injury and Stroke. We systematically review the histological, behavioral and electrophysiological features of each model and identify those that are the most relevant for translational research.
Collapse
Affiliation(s)
- Matthieu Faillot
- Neurosurgery department, Henri Mondor University Hospital, APHP, DMU CARE, Université Paris Est Créteil, Mondor Institute for Biomedical Research, INSERM U955, Team 15, Translational Neuropsychiatry, France
| | - Antoine Chaillet
- Laboratoire des Signaux et Systèmes (L2S-UMR8506) - CentraleSupélec, Université Paris Saclay, Institut Universitaire de France, France
| | - Stéphane Palfi
- Neurosurgery department, Henri Mondor University Hospital, APHP, DMU CARE, Université Paris Est Créteil, Mondor Institute for Biomedical Research, INSERM U955, Team 15, Translational Neuropsychiatry, France
| | - Suhan Senova
- Neurosurgery department, Henri Mondor University Hospital, APHP, DMU CARE, Université Paris Est Créteil, Mondor Institute for Biomedical Research, INSERM U955, Team 15, Translational Neuropsychiatry, France.
| |
Collapse
|