1
|
He M, Wang D, Yang K, Qi H, Liu C, Sun L, Wei L, Wu Y. Animal models of epilepsy after ischemic stroke. Neuroscience 2025; 576:1-7. [PMID: 40254123 DOI: 10.1016/j.neuroscience.2025.04.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 04/10/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
Stroke ranks among the foremost causes of disability and mortality globally, with ischemic stroke (IS) being the most prevalent subtype. Post-stroke epilepsy (PSE) represents a significant and common complication following a stroke, imposing substantial burdens on patients, their families, and society at large. Establishing a reliable animal model is crucial for investigating the mechanisms and potential treatments for PSE. This article offers a review of studies pertaining to animal models of epilepsy subsequent to ischemic stroke.
Collapse
Affiliation(s)
- Min He
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, #6 Shuangyong Road, Nanning 530021 Guangxi Zhuang Autonomous Region, China.
| | - Donghui Wang
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, #6 Shuangyong Road, Nanning 530021 Guangxi Zhuang Autonomous Region, China.
| | - Kunling Yang
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, #6 Shuangyong Road, Nanning 530021 Guangxi Zhuang Autonomous Region, China.
| | - Hengchang Qi
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, #6 Shuangyong Road, Nanning 530021 Guangxi Zhuang Autonomous Region, China.
| | - Chaoning Liu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, #6 Shuangyong Road, Nanning 530021 Guangxi Zhuang Autonomous Region, China.
| | - Lanfeng Sun
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, #6 Shuangyong Road, Nanning 530021 Guangxi Zhuang Autonomous Region, China.
| | - Lei Wei
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, #6 Shuangyong Road, Nanning 530021 Guangxi Zhuang Autonomous Region, China.
| | - Yuan Wu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, #6 Shuangyong Road, Nanning 530021 Guangxi Zhuang Autonomous Region, China.
| |
Collapse
|
2
|
Boyle BR, Berghella AP, Blanco-Suarez E. Astrocyte Regulation of Neuronal Function and Survival in Stroke Pathophysiology. ADVANCES IN NEUROBIOLOGY 2024; 39:233-267. [PMID: 39190078 DOI: 10.1007/978-3-031-64839-7_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
The interactions between astrocytes and neurons in the context of stroke play crucial roles in the disease's progression and eventual outcomes. After a stroke, astrocytes undergo significant changes in their morphology, molecular profile, and function, together termed reactive astrogliosis. Many of these changes modulate how astrocytes relate to neurons, inducing mechanisms both beneficial and detrimental to stroke recovery. For example, excessive glutamate release and astrocytic malfunction contribute to excitotoxicity in stroke, eventually causing neuronal death. Astrocytes also provide essential metabolic support and neurotrophic signals to neurons after stroke, ensuring homeostatic stability and promoting neuronal survival. Furthermore, several astrocyte-secreted molecules regulate synaptic plasticity in response to stroke, allowing for the rewiring of neural circuits to compensate for damaged areas. In this chapter, we highlight the current understanding of the interactions between astrocytes and neurons in response to stroke, explaining the varied mechanisms contributing to injury progression and the potential implications for future therapeutic interventions.
Collapse
Affiliation(s)
- Bridget R Boyle
- Department of Neuroscience, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
- Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - Andrea P Berghella
- Department of Neuroscience, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
- Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - Elena Blanco-Suarez
- Department of Neuroscience, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
- Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA.
- Department of Neurological Surgery, Vickie & Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Yang T, Guo R, Ofengeim D, Hwang JY, Zukin RS, Chen J, Zhang F. Molecular and Cellular Mechanisms of Ischemia-Induced Neuronal Death. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00005-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
4
|
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
|
5
|
León-Moreno LC, Castañeda-Arellano R, Rivas-Carrillo JD, Dueñas-Jiménez SH. Challenges and Improvements of Developing an Ischemia Mouse Model Through Bilateral Common Carotid Artery Occlusion. J Stroke Cerebrovasc Dis 2020; 29:104773. [PMID: 32199775 DOI: 10.1016/j.jstrokecerebrovasdis.2020.104773] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/10/2020] [Accepted: 02/17/2020] [Indexed: 01/01/2023] Open
Abstract
Brain ischemia is one of the principal causes of death and disability worldwide in which prevention or an effective treatment does not exist. In order to develop successful treatments, an adequate and useful ischemia model is essential. Transient global cerebral ischemia is one of the most interesting pathological conditions in stroke studies because of the observed degeneration of forebrain and delayed neuronal cell death in selective vulnerable regions such as hippocampus. Transient occlusion of both common carotid arteries is the most convenient model to induce tGCI. Although there are effective rat and gerbil models using this method, the induction of a reproducible and reliable injury after global ischemia in mouse has presented higher variations, mainly because of its size and the necessary monitoring skills in order to accomplish homogeneous and reproducible results. Further, great variability among cerebral vasculature and susceptibility of the different strains and sub-strains is observed. In recent years, some modifications have been made to the model in order to normalize the heterogenic effects. Analysis of posterior communicating artery patency has been proposed as an exclusion parameter due to the direct relationship reported with the reduction of cerebral blood flow. Another method used to significantly reduce blood flow is the induction of hypotension with isoflurane. Each protocol produces distinct injury outcomes. Further improvements are needed to attain a general, simpler, reproducible and globally accepted model that allows comparisons between research groups, progress in understanding ischemia and the consequent development of therapeutic alternatives for ischemic injury.
Collapse
Affiliation(s)
| | - Rolando Castañeda-Arellano
- Department of Biomedical Sciences, University Center of Tonala, University de Guadalajara, Jalisco Mexico
| | - Jorge David Rivas-Carrillo
- Department of Physiology, Laboratory of Tissue Engineering and Transplant and cGMP Cell Processing Facility, Health Sciences Center, University de Guadalajara, Jalisco, Mexico
| | - Sergio Horacio Dueñas-Jiménez
- Department of Neuroscience, Laboratory of Neurophysiology, Health Sciences Center, University of Guadalajara, Guadalajara, C.P. 44340 Jalisco, México.
| |
Collapse
|
6
|
Tecuatl C, Herrrera-López G, Martín-Ávila A, Yin B, Weber S, Barrionuevo G, Galván EJ. TrkB-mediated activation of the phosphatidylinositol-3-kinase/Akt cascade reduces the damage inflicted by oxygen-glucose deprivation in area CA3 of the rat hippocampus. Eur J Neurosci 2018; 47:1096-1109. [PMID: 29480936 PMCID: PMC5938095 DOI: 10.1111/ejn.13880] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 02/17/2018] [Accepted: 02/20/2018] [Indexed: 12/31/2022]
Abstract
The selective vulnerability of hippocampal area CA1 to ischemia-induced injury is a well-known phenomenon. However, the cellular mechanisms that confer resistance to area CA3 against ischemic damage remain elusive. Here, we show that oxygen-glucose deprivation-reperfusion (OGD-RP), an in vitro model that mimic the pathological conditions of the ischemic stroke, increases the phosphorylation level of tropomyosin receptor kinase B (TrkB) in area CA3. Slices preincubated with brain-derived neurotrophic factor (BDNF) or 7,8-dihydroxyflavone (7,8-DHF) exhibited reduced depression of the electrical activity triggered by OGD-RP. Consistently, blockade of TrkB suppressed the resistance of area CA3 to OGD-RP. The protective effect of TrkB activation was limited to area CA3, as OGD-RP caused permanent suppression of CA1 responses. At the cellular level, TrkB activation leads to phosphorylation of the accessory proteins SHC and Gab as well as the serine/threonine kinase Akt, members of the phosphoinositide 3-kinase/Akt (PI-3-K/Akt) pathway, a cascade involved in cell survival. Hence, acute slices pretreated with the Akt antagonist MK2206 in combination with BDNF lost the capability to resist the damage inflicted with OGD-RP. Consistently, with these results, CA3 pyramidal cells exhibited reduced propidium iodide uptake and caspase-3 activity in slices pretreated with BDNF and exposed to OGD-RP. We propose that PI-3-K/Akt downstream activation mediated by TrkB represents an endogenous mechanism responsible for the resistance of area CA3 to ischemic damage.
Collapse
Affiliation(s)
- Carolina Tecuatl
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Calzada de los Tenorios No. 235, México City, 14330, México
| | - Gabriel Herrrera-López
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Calzada de los Tenorios No. 235, México City, 14330, México
| | - Alejandro Martín-Ávila
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Calzada de los Tenorios No. 235, México City, 14330, México
| | - Bocheng Yin
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Stephen Weber
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Germán Barrionuevo
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | - Emilio J. Galván
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Calzada de los Tenorios No. 235, México City, 14330, México
| |
Collapse
|
7
|
Atlasi MA, Naderian H, Noureddini M, Fakharian E, Azami A. Morphology of Rat Hippocampal CA1 Neurons Following Modified Two and Four-Vessels Global Ischemia Models. ARCHIVES OF TRAUMA RESEARCH 2013; 2:124-8. [PMID: 24693522 PMCID: PMC3950915 DOI: 10.5812/atr.10240] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 07/09/2013] [Accepted: 07/10/2013] [Indexed: 11/25/2022]
Abstract
Background An appropriate animal model of ischemia stroke is essential for evaluation of different therapeutic methods. Two and four-vessel global ischemia models are one of the most common types of transient cerebral ischemia. Objectives In this study, the morphology of rat hippocampal CA1 neurons in modified models of two and four-vessel ischemia and reperfusion were evaluated. Materials and Methods In this study, 20 Wistar rats were randomly divided into five groups. In group 2 and 3, both common carotid arteries were occluded for 10 minutes in either 3 or 24 hours of reperfusions, respectively. In group 4 and 5, both common carotid and vertebral arteries were occluded for 10 minutes in either 3 or 24 hours of reperfusions, respectively. Group 1 as control, underwent the whole surgery without any arteries occlusion. Hippocampi of the rats in all groups were processed and tissue sections were stained using the Nissl method. The morphology of CA1 neurons were studied under a light microscope and compared different groups. Results In all groups ischemic changes were apparently observed in hippocampus CA1 neurons. In two-vessel occlusion model, after 3 and 24 hours of reperfusions, ischemic cells accounted for 14.9% and 23.2%, respectively. In four-vessel occlusion model, after 3 and 24 hours of reperfusions, ischemic cells accounted for 7.6% and 44.9% (P < 0.0001), respectively. Conclusions Modified four-vessel occlusion model resulted in significant ischemic changes after 24 hours of reperfusion in CA1 neurons of rat hippocampus.
Collapse
Affiliation(s)
- Mohammad Ali Atlasi
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, IR Iran
- Corresponding author: Mohammad Ali Atlasi, Anatomical Research Center, Kashan University of Medical Sciences, Kashan, IR Iran. Tel: +98-9131615603, Fax: +98-3615551112, E-mail:
| | - Homayoun Naderian
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, IR Iran
| | - Mahdi Noureddini
- Physiology Research Center, Kashan University of Medical Sciences, Kashan, IR Iran
| | - Esmaeil Fakharian
- Trauma Research Center, Kashan University of Medical Sciences, Kashan, IR Iran
| | - Abolfazl Azami
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, IR Iran
| |
Collapse
|
8
|
Decrease of synaptic plasticity associated with alteration of information flow in a rat model of vascular dementia. Neuroscience 2012; 206:136-43. [DOI: 10.1016/j.neuroscience.2011.12.050] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Revised: 12/27/2011] [Accepted: 12/28/2011] [Indexed: 11/23/2022]
|
9
|
The effect of blood pressure (37 vs 45 mmHg) and carotid occlusion duration (8 vs 10 min) on CA1-4 neuronal damage when using isoflurane in a global cerebral ischemia rat model. Brain Res Bull 2011; 86:390-4. [PMID: 21945086 DOI: 10.1016/j.brainresbull.2011.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Revised: 09/01/2011] [Accepted: 09/10/2011] [Indexed: 11/21/2022]
Abstract
This study presents our findings on the extent of neuronal damage in the hippocampal CA1-4 subfields following global (forebrain) cerebral ischemia in rats when using different blood pressure levels (37 vs 45 mmHg) and bilateral carotid occlusion durations (8 vs 10 min) under isoflurane anesthesia. We observed that global ischemia induced at a blood pressure of 37 mmHg resulted in high-grade CA1 neuron injury (>90%) at either duration of carotid occlusion. In contrast, global ischemia induced at a blood pressure of 45 mmHg resulted in either high-grade CA1 neuronal loss or a neuronal loss of ≈50% or less. We also noted that a post-reperfusion EEG recovery time (return of burst suppression spikes) of >12 min was associated with an 85% rate of high-grade CA1 neuronal injury. Neuronal loss in the other hippocampal subfields did not differ significantly between any of the 4 different model parameters tested. In these subfields ≈55% neuronal loss occurred in the CA2 subfield, and ≈30% in the CA3 and CA4 subfields. These findings highlight the need to assess different model parameters in order to achieve consistent high-grade CA1 neuronal damage, which, among other experimental outcomes, will improve the ability to uncover therapeutic effects using the least possible animals when assessing a neuroprotective treatment.
Collapse
|
10
|
Li Z, Wang Y, Xie Y, Yang Z, Zhang T. Protective Effects of Exogenous Hydrogen Sulfide on Neurons of Hippocampus in a Rat Model of Brain Ischemia. Neurochem Res 2011; 36:1840-9. [DOI: 10.1007/s11064-011-0502-6] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2011] [Indexed: 12/20/2022]
|
11
|
Yuan Y, Li F, Wang YM, Zhang RQ, Wei LP, Wang HC. Respiratory face mask: a novel and cost-effective device for use during the application of myocardial ischemia in rats. J Zhejiang Univ Sci B 2009; 10:391-4. [PMID: 19434766 DOI: 10.1631/jzus.b0820216] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
To shorten operation time and improve survival rate of rats with myocardial ischemia or myocardial infarction, we use a novel device comprised of a face mask and a head/neck retainer in this study. We report the basic design of the novel respiratory face mask (RFM) and evaluate its performance in a rat model of myocardial ischemia. The device is cost-effective and easier to handle than other devices, such as tracheal intubation. Compared with conventional tracheal intubation, we found that RFM shortens operation time significantly while keeping blood indices normal; the mean operation time for rats in the mask group was (32+/-3) min, and that for the intubation group was (45+/-7) min (P<0.05). Moreover, the size and shape of the RFM can be changed according to the body weight of rats. In conclusion, RFM is an appropriate device for the establishment of myocardial infarction or ischemia-reperfusion in rats.
Collapse
Affiliation(s)
- Yuan Yuan
- Department of Cardiology, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, China
| | | | | | | | | | | |
Collapse
|
12
|
Windelborn JA, Lipton P. Lysosomal release of cathepsins causes ischemic damage in the rat hippocampal slice and depends on NMDA-mediated calcium influx, arachidonic acid metabolism, and free radical production. J Neurochem 2008; 106:56-69. [PMID: 18363826 DOI: 10.1111/j.1471-4159.2008.05349.x] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
NMDA-mediated calcium entry and reactive oxygen species (ROS) production are well-recognized perpetrators of ischemic neuronal damage. The current studies show that these events lead to the release of the protein hydrolase, cathepsin B, from lysosomes 2 h following 5-min oxygen-glucose deprivation in the rat hippocampal slice. This release reflects a lysosomal membrane permeabilization (LMP) and was measured as the appearance of diffuse immunolabeled cathepsin B in the cytosol of CA1 pyramidal neurons. Necrotic neuronal damage begins after the release of cathepsins and is prevented by inhibitors of either cathepsin B or D indicating that the release of cathepsins is an important mediator of severe damage. There was an increase in superoxide levels, measured by dihydroethidium fluorescence, at the same time as LMP and reducing ROS levels with antioxidants, Trolox or N-tert-butyl-alpha-phenyl nitrone, blocked LMP. Both LMP and ROS production were blocked by an NMDA channel blocker (MK-801) and by inhibitors of mitogen-activated protein kinase kinase (U0126), calcium-dependent/independent phospholipases A2 (methyl arachidonyl fluorophosphonate) but not calcium-independent phospholipases A2 (bromoenol lactone) and cyclooxygenase-2 (NS398). A cell-permeant specific inhibitor of calpain (PD150606) prevented LMP, but not ROS production. It is concluded that LMP results in part from calcium-initiated and extracellular signal-regulated kinase-initiated arachidonic acid metabolism, which produces free radicals; it also requires the action of calpain.
Collapse
Affiliation(s)
- James A Windelborn
- Neuroscience Training Program, University of Wisconsin, Madison, Wisconsin 53706, USA
| | | |
Collapse
|
13
|
Henrich-Noack P, Gorkin AG, Reymann KG. Predictive value of changes in electroencephalogram and excitatory postsynaptic field potential for CA1 damage after global ischaemia in rats. Exp Brain Res 2007; 181:79-86. [PMID: 17431602 DOI: 10.1007/s00221-007-0907-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2005] [Accepted: 02/09/2007] [Indexed: 10/23/2022]
Abstract
Recordings of the electroencephalogram (EEG) are regularly used to asses the severity of transient global ischaemia in rats. Here, we investigated whether the EEG obtained from electrodes placed in the hippocampus does indeed give valuable information about the consequences of an ischaemic event. Furthermore, we evaluated how evoked synaptic responses from the same electrodes placed in the hippocampal CA1 area changed with time and in relation to damage. We performed transient two vessel-occlusion with hypobaric hypotension in rats to induce selective, delayed neuronal death in CA1. Beforehand, the animals had been chronically implanted with electrodes. Stimulating electrodes had been placed into the Schaffer collaterals and recording electrodes into the CA1 area. EEG was recorded from shortly before ischaemia until up to 40 min post-ischaemia. Field excitatory post-synaptic potentials (fEPSP) were recorded before ischaemia or sham-operation and 2 and 7 days afterwards. We found a significant negative correlation between the duration of the EEG amplitude decrease (flattening) and the number of surviving neurons in CA1, which were quantified by histology after 7 days post-ischaemia. However, substantial neuronal damage was only seen when the time of flattening was more than 12 min and outlasted the time of ischaemia. The impairment of synaptic function, measured as the decrease of fEPSP slope 2 days post-ischaemia correlated with the later maturated structural damage in CA1. The fEPSP remained decreased until day 7 post-ischaemia. Animals with no damage (sham condition) showed a transient decrease of the fEPSP slope. In conclusion, our data show that the duration of EEG-flattening predicts the extent of neuronal damage. However, EEG-flattening just during the period of clamping both common carotid arteries--albeit an essential precondition for substantial CA1 cell loss to occur--is not sufficient to predict damage. The degree of impairment of evoked synaptic function of CA1 neurons (fEPSP) 2 days after ischaemia predicts the final extent of damage with significant probability.
Collapse
Affiliation(s)
- Petra Henrich-Noack
- Leibniz Institute for Neurobiology, Brenneckestreet 6, 39118, Magdeburg, Germany
| | | | | |
Collapse
|
14
|
Spencer SJ, Auer RN, Pittman QJ. Rat neonatal immune challenge alters adult responses to cerebral ischaemia. J Cereb Blood Flow Metab 2006; 26:456-67. [PMID: 16094315 DOI: 10.1038/sj.jcbfm.9600206] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Infection, inflammation, and hyperthermia associated with cerebral ischaemia are known to contribute to enhanced neuronal cell loss and more severe behavioural deficits. Because neonatal exposure to an immune challenge has been shown to alter the severity of inflammatory and febrile responses to a further immune challenge experienced in adulthood, we hypothesised that this could also alter temperature responses and neuronal survival after ischaemia. Thus, male Sprague-Dawley rats were treated at postnatal day 14 with a single injection of the bacterial endotoxin lipopolysaccharide (LPS) and were examined as adults for temperature changes, behavioural deficits, and neuronal cell loss associated with global cerebral ischaemia after a two-vessel occlusion (2VO). Neonatally LPS-treated rats showed behavioural differences in a novel object exploration paradigm, as well as altered temperature responses to the 2VO compared with neonatally saline-treated controls. Interestingly, these neonatally LPS-treated rats also showed increased cell loss in the central nucleus of the amygdala, a region that is important in the processing of emotional responses, but that is not usually examined in animal models of cerebral ischaemia. No differences were seen in the CA1, CA3, or dentate gyrus regions of the hippocampus. This work shows the importance of examining brain regions other than the hippocampus in association with global ischaemia. We also highlight the importance of the early period of development in programming an animal's ability to deal with injury such as cerebral ischaemia in adulthood.
Collapse
Affiliation(s)
- Sarah J Spencer
- Department of Physiology and Biophysics, Faculty of Medicine, Hotchkiss Brain Institute, University of Calgary, Alberta, Canada.
| | | | | |
Collapse
|
15
|
Bendel O, Bueters T, von Euler M, Ove Ogren S, Sandin J, von Euler G. Reappearance of hippocampal CA1 neurons after ischemia is associated with recovery of learning and memory. J Cereb Blood Flow Metab 2005; 25:1586-95. [PMID: 15917746 DOI: 10.1038/sj.jcbfm.9600153] [Citation(s) in RCA: 132] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The pyramidal neurons of the hippocampal CA1 region are essential for cognitive functions such as spatial learning and memory, and are selectively destroyed after cerebral ischemia. To analyze whether degenerated CA1 neurons are replaced by new neurons and whether such regeneration is associated with amelioration in learning and memory deficits, we have used a rat global ischemia model that provides an almost complete disappearance (to approximately 3% of control) of CA1 neurons associated with a robust impairment in spatial learning and memory at two weeks after ischemia. We found that transient cerebral ischemia can evoke a massive formation of new neurons in the CA1 region, reaching approximately 40% of the original number of neurons at 90 days after ischemia (DAI). Co-localization of the mature neuronal marker neuronal nuclei with 5-bromo-2'-deoxyuridine in CA1 confirmed that neurogenesis indeed had occurred after the ischemic insult. Furthermore, we found increased numbers of cells expressing the immature neuron marker polysialic acid neuronal cell adhesion molecule in the adjacent lateral periventricular region, suggesting that the newly formed neurons derive from this region. The reappearance of CA1 neurons was associated with a recovery of ischemia-induced impairments in spatial learning and memory at 90 DAI, suggesting that the newly formed CA1 neurons restore hippocampal CA1 function. In conclusion, these results show that the brain has an endogenous capacity to form new nerve cells after injury, which correlates with a restoration of cognitive functions of the brain.
Collapse
Affiliation(s)
- Olof Bendel
- Section of Clinical CNS Research, Department of Clinical Neuroscience, Karolinska Institutet, Karolinska University Hospital, Solna, Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
16
|
Bendel O, Alkass K, Bueters T, von Euler M, von Euler G. Reproducible loss of CA1 neurons following carotid artery occlusion combined with halothane-induced hypotension. Brain Res 2005; 1033:135-42. [PMID: 15694917 DOI: 10.1016/j.brainres.2004.11.033] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/24/2004] [Indexed: 11/18/2022]
Abstract
The 2-vessel occlusion approach to produce global ischemia in rats requires concomitant reduction of systemic blood pressure. We have utilized the hypotensive effect of halothane administrated by artificial respiration to prevent respiratory arrest and to ensure stable physiological conditions. Systemic blood pressure was reduced to 40-45 mmHg by instant adjustments of the halothane concentration. Bilateral occlusion of the carotid arteries caused a profound and reproducible ischemia, as analyzed by laser-Doppler flowmetry. In the rats exposed to 11, 12, or 13 min of ischemia, 5% died and 5% developed seizures. The extent of neuronal death in CA1 was highly correlated to the duration of ischemia. Following 11 min of ischemia, CA1 neuronal cell death, as analyzed by Fluoro-Jade, was absent 1 day after injury, variable at day 4, and consistent at day 7. The numbers of cresyl violet- and NeuN-positive neurons at day 7 were 8% and 20% of control, respectively. OX42 immunoreactivity was low and variable at day 4, but pronounced at day 7. In conclusion, this rat global ischemia model is relatively simple to perform, has a low mortality, and produces a profound and highly reproducible delayed cell death of hippocampal CA1 neurons.
Collapse
Affiliation(s)
- Olof Bendel
- Section of Clinical CNS Research, Department of Clinical Neuroscience, Karolinska Institutet, Karolinska University Hospital, Solna, Stockholm, Sweden
| | | | | | | | | |
Collapse
|
17
|
Grauert M, Bechtel WD, Weiser T, Stransky W, Nar H, Carter AJ. Synthesis and structure-activity relationships of 6,7-benzomorphan derivatives as use-dependent sodium channel blockers for the treatment of stroke. J Med Chem 2002; 45:3755-64. [PMID: 12166948 DOI: 10.1021/jm020875j] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We have synthesized a series of 6,7-benzomorphan derivatives and determined their ability to bind to voltage-dependent sodium channels. We have also compared the functional consequences of this blockade in vitro and in vivo. The ability of the compounds to displace [(3)H]batrachotoxin from voltage-dependent sodium channels was compared with their ability to inhibit [(3)H]glutamate release in rat brain slices and block convulsions in the maximal electroshock test in mice. We found that the hydroxyl function in the 4'-position is crucial for improving the sodium channel blocking properties. Moreover, the stereochemistry and the topology of the N-linked side chain also influence this interaction. Indeed, the affinity is improved by an aromatic substitution in the side chain. By modifying the N substituent and the substitution pattern of the hydroxyl function, we were able to discover (2R)-[2alpha,3(S),6alpha]-1,2,3,4,5,6-hexahydro-6,11,11-tri-methyl-3-[2-(phenylmethoxy)propyl]-2,6-methano-3-benzazocin-10-ol hydrochloride. This compound was chosen as the best candidate for further pharmacological investigations.
Collapse
Affiliation(s)
- Matthias Grauert
- Department of Medicinal Chemistry, Boehringer Ingelheim Pharma KG, 88397 Biberach an der Riss, and Corporate Development, Boehringer Ingelheim GmbH, 55216 Ingelheim am Rhein, Germany.
| | | | | | | | | | | |
Collapse
|
18
|
Read SJ, Hirano T, Davis SM, Donnan GA. Limiting neurological damage after stroke: a review of pharmacological treatment options. Drugs Aging 1999; 14:11-39. [PMID: 10069406 DOI: 10.2165/00002512-199914010-00002] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Acute ischaemic stroke is a leading cause of death and a major cause of long term disability worldwide. Effective treatments for limiting the neurological damage after stroke have proven elusive. An improved understanding of the complicated cascade of cellular events following the onset of cerebral ischaemia has led to exploration of a number of avenues for early intervention. Reperfusion of the ischaemic territory using thrombolytic drugs has shown promise in clinical trials as a method for achieving tissue salvage. Antithrombotic and antiplatelet agents have not demonstrated efficacy as acute therapies, although the early use of aspirin (acetylsalicylic acid) appears to produce a reduction in early stroke recurrence. A wide variety of drugs which interfere at various points in the ischaemic cascade, so-called 'neuroprotective agents', have also been studied, but with mixed success. Of these, antagonists of voltage-gated calcium channels, antagonists at the N-methyl-D-aspartate (NMDA) receptor and scavengers of free radicals have been most extensively studied. Despite proving effective in animal models of cerebral ischaemia, these drugs have largely failed to fulfil their promise in clinical trials. While individual compounds have proven ineffective, combinations of drugs with different mechanisms of action may yet provide the best treatment for acute ischaemic stroke.
Collapse
Affiliation(s)
- S J Read
- Department of Neurology, Austin and Repatriation Medical Centre, Heidelberg, Victoria, Australia
| | | | | | | |
Collapse
|
19
|
Abstract
Strategies for the treatment of thromboembolic stroke are based on restoring the blood flow as soon as possible and protecting the neurons from the deleterious consequences of cerebral ischaemia. Interest has focused on blockers of voltage-dependent Na+ channels as potential neuroprotective agents because they prevent neuronal death in various experimental models of cerebral ischaemia and act cytoprotectively in models of white matter damage. Although several Na+ blockers are currently being tested in various phases of clinical development, most of these agents are relatively weak and unspecific. I therefore consider it worthwhile to search for molecules which specifically block voltage-dependent Na+ channels for the treatment of cerebral ischaemia.
Collapse
Affiliation(s)
- A J Carter
- Department of Central Nervous System Research, Ingelheim am Rhein, Federal Republic of Germany
| |
Collapse
|
20
|
Overexpression of SOD1 in transgenic rats protects vulnerable neurons against ischemic damage after global cerebral ischemia and reperfusion. J Neurosci 1998. [PMID: 9763473 DOI: 10.1523/jneurosci.18-20-08292.1998] [Citation(s) in RCA: 249] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Transient global cerebral ischemia resulting from cardiac arrest is known to cause selective death in vulnerable neurons, including hippocampal CA1 pyramidal neurons. It is postulated that oxygen radicals, superoxide in particular, are involved in cell death processes. To test this hypothesis, we first used in situ imaging of superoxide radical distribution by hydroethidine oxidation in vulnerable neurons. We then generated SOD1 transgenic (Tg) rats with a five-fold increase in copper zinc superoxide dismutase activity. The Tg rats and their non-Tg wild-type littermates were subjected to 10 min of global ischemia followed by 1 and 3 d of reperfusion. Neuronal damage, as assessed by cresyl violet staining and DNA fragmentation analysis, was significantly reduced in the hippocampal CA1 region, cortex, striatum, and thalamus in SOD1 Tg rats at 3 d, as compared with the non-Tg littermates. There were no changes in the hippocampal CA3 subregion and dentate gyrus, resistant areas in both SOD1 Tg and non-Tg rats. Quantitative analysis of the damaged CA1 subregion showed marked neuroprotection against transient global cerebral ischemia in SOD1 Tg rats. These results suggest that superoxide radicals play a role in the delayed ischemic death of hippocampal CA1 neurons. Our data also indicate that SOD1 Tg rats are useful tools for studying the role of oxygen radicals in the pathogenesis of neuronal death after transient global cerebral ischemia.
Collapse
|
21
|
Chan PH, Kawase M, Murakami K, Chen SF, Li Y, Calagui B, Reola L, Carlson E, Epstein CJ. Overexpression of SOD1 in transgenic rats protects vulnerable neurons against ischemic damage after global cerebral ischemia and reperfusion. J Neurosci 1998; 18:8292-9. [PMID: 9763473 PMCID: PMC6792858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Transient global cerebral ischemia resulting from cardiac arrest is known to cause selective death in vulnerable neurons, including hippocampal CA1 pyramidal neurons. It is postulated that oxygen radicals, superoxide in particular, are involved in cell death processes. To test this hypothesis, we first used in situ imaging of superoxide radical distribution by hydroethidine oxidation in vulnerable neurons. We then generated SOD1 transgenic (Tg) rats with a five-fold increase in copper zinc superoxide dismutase activity. The Tg rats and their non-Tg wild-type littermates were subjected to 10 min of global ischemia followed by 1 and 3 d of reperfusion. Neuronal damage, as assessed by cresyl violet staining and DNA fragmentation analysis, was significantly reduced in the hippocampal CA1 region, cortex, striatum, and thalamus in SOD1 Tg rats at 3 d, as compared with the non-Tg littermates. There were no changes in the hippocampal CA3 subregion and dentate gyrus, resistant areas in both SOD1 Tg and non-Tg rats. Quantitative analysis of the damaged CA1 subregion showed marked neuroprotection against transient global cerebral ischemia in SOD1 Tg rats. These results suggest that superoxide radicals play a role in the delayed ischemic death of hippocampal CA1 neurons. Our data also indicate that SOD1 Tg rats are useful tools for studying the role of oxygen radicals in the pathogenesis of neuronal death after transient global cerebral ischemia.
Collapse
Affiliation(s)
- P H Chan
- CNS Injury and Edema Research Center, University of California, School of Medicine, San Francisco, California 94143, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
McBean DE, Kelly PA. Rodent models of global cerebral ischemia: a comparison of two-vessel occlusion and four-vessel occlusion. GENERAL PHARMACOLOGY 1998; 30:431-4. [PMID: 9522158 DOI: 10.1016/s0306-3623(97)00284-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
1. Human stroke is a complex and heterogeneous phenomenon that may defy attempts to develop a unitary animal model with which to address all of the relevant issues. 2. Focal models are regarded by many to be the approach of choice, but both global and focal models of cerebral ischemia can be sources of useful and complementary insight. 3. Of the global models, four-vessel occlusion requires a preparatory operative procedure that may increase the risk of extraneous factors confounding the response to the ischemic insult itself. The procedures are only partly reversible, with the vertebral arteries remaining permanently occluded. 4. The two-vessel occlusion model is easier to perform in a single procedure, and the less-intrusive surgical intervention allows greater scope for recovery experiments. The occlusion is fully reversible. 5. Many classes of compounds with therapeutic potential have been identified in the laboratory, often on the basis of success in one class of animal model, but translating these successes into a clinical context has proved singularly difficult. If, in future, compounds of interest are tested across a range of the available models, the likelihood of subsequent clinical success may be enhanced.
Collapse
Affiliation(s)
- D E McBean
- Department of Clinical Neurosciences, University of Edinburgh, Western General Hospitals NHS Trust, Scotland
| | | |
Collapse
|
23
|
|
24
|
Obrenovitch TP. Sodium and potassium channel modulators: their role in neuroprotection. INTERNATIONAL REVIEW OF NEUROBIOLOGY 1996; 40:109-35. [PMID: 8989619 DOI: 10.1016/s0074-7742(08)60718-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- T P Obrenovitch
- Department of Neurochemistry, Institute of Neurology, London, UK
| |
Collapse
|