1
|
Warenits AM, Hatami J, Müllebner A, Ettl F, Teubenbacher U, Magnet IAM, Bauder B, Janata A, Miller I, Moldzio R, Kramer AM, Sterz F, Holzer M, Högler S, Weihs W, Duvigneau JC. Motor Cortex and Hippocampus Display Decreased Heme Oxygenase Activity 2 Weeks After Ventricular Fibrillation Cardiac Arrest in Rats. Front Med (Lausanne) 2020; 7:513. [PMID: 33015090 PMCID: PMC7511667 DOI: 10.3389/fmed.2020.00513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/24/2020] [Indexed: 01/07/2023] Open
Abstract
Heme oxygenase (HO) and biliverdin reductase (BVR) activities are important for neuronal function and redox homeostasis. Resuscitation from cardiac arrest (CA) frequently results in neuronal injury and delayed neurodegeneration that typically affect vulnerable brain regions, primarily hippocampus (Hc) and motor cortex (mC), but occasionally also striatum and cerebellum. We questioned whether these delayed effects are associated with changes of the HO/BVR system. We therefore analyzed the activities of HO and BVR in the brain regions Hc, mC, striatum and cerebellum of rats subjected to ventricular fibrillation CA (6 min or 8 min) after 2 weeks following resuscitation, or sham operation. From all investigated regions, only Hc and mC showed significantly decreased HO activities, while BVR activity was not affected. In order to find an explanation for the changed HO activity, we analyzed protein abundance and mRNA expression levels of HO-1, the inducible, and HO-2, the constitutively expressed isoform, in the affected regions. In both regions we found a tendency for a decreased immunoreactivity of HO-2 using immunoblots and immunohistochemistry. Additionally, we investigated the histological appearance and the expression of markers indicative for activation of microglia [tumor necrosis factor receptor type I (TNFR1) mRNA and immunoreactivity for ionized calcium-binding adapter molecule 1 (Iba1])], and activation of astrocytes [immunoreactivity for glial fibrillary acidic protein (GFAP)] in Hc and mC. Morphological changes were detected only in Hc displaying loss of neurons in the cornu ammonis 1 (CA1) region, which was most pronounced in the 8 min CA group. In this region also markers indicating inflammation and activation of pro-death pathways (expression of HO-1 and TNFR1 mRNA, as well as Iba1 and GFAP immunoreactivity) were upregulated. Since HO products are relevant for maintaining neuronal function, our data suggest that neurodegenerative processes following CA may be associated with a decreased capacity to convert heme into HO products in particularly vulnerable brain regions.
Collapse
Affiliation(s)
| | - Jasmin Hatami
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Andrea Müllebner
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria.,Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Florian Ettl
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria
| | - Ursula Teubenbacher
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
| | | | - Barbara Bauder
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
| | - Andreas Janata
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria
| | - Ingrid Miller
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Rudolf Moldzio
- Institute for Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | | | - Fritz Sterz
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria
| | - Michael Holzer
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria
| | - Sandra Högler
- Center for Biomedical Research, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Weihs
- Department of Emergency Medicine, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
2
|
Morgan CA, Mesquita M, Ashioti M, Beech JS, Williams SCR, Irving E, Cash D. Late changes in blood-brain barrier permeability in a rat tMCAO model of stroke detected by gadolinium-enhanced MRI. Neurol Res 2020; 42:844-852. [PMID: 32600164 DOI: 10.1080/01616412.2020.1786637] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
OBJECTIVES After cerebral ischaemia the blood-brain barrier (BBB) may be compromised and this has been observed in both clinical and preclinical studies. The timing of BBB disruption after ischaemia has long been considered to be biphasic, however some groups contest this view. Therefore, the purpose of this study was to characterize the BBB permeability timecourse in a rat model at both acute and chronic time points. METHODS Unilateral transient middle cerebral artery occlusion (tMCAO) was performed in 15 male Sprague Dawley rats. Change in T1-weighted MR signal before and after an injection of gadolinium-based contrast agent was calculated voxelwise to derive a BBB permeability index (BBBPI) at both early (6 h, 12 h, and 24 h) and late (7 and 14 days) time points. RESULTS As expected, BBBPI in the non-lesioned ROI was not significantly different from pre-occlusion baseline at any time point. However, BBBPI in the ipsilateral (lesioned) ROI was statistically different to baseline at day 7 (p < 0.001) and day 14 (p < 0.01) post-tMCAO. There was a small, but not-significant increase in BBBPI in the earlier phase (at 6 hours). DISCUSSION Our results indicate a significant late opening of the BBB. This is important as the majority of previous studies have only characterised an early acute BBB permeability in ischemia. However, the later period of increased permeability may indicate an optimal time for drug delivery across the BBB, when it is especially suited to drugs targeting delayed processes.
Collapse
Affiliation(s)
- Catherine A Morgan
- School of Psychology and Centre for Brain Research, The University of Auckland , Auckland, New Zealand.,Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London , London, UK
| | - Michel Mesquita
- School of Psychology and Centre for Brain Research, The University of Auckland , Auckland, New Zealand
| | - Maria Ashioti
- School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster , London, UK
| | - John S Beech
- School of Psychology and Centre for Brain Research, The University of Auckland , Auckland, New Zealand
| | - Steve C R Williams
- School of Psychology and Centre for Brain Research, The University of Auckland , Auckland, New Zealand
| | - Elaine Irving
- Value Evidence & Outcomes, GlaxoSmithKline R&D Ltd , Stevenage, UK
| | - Diana Cash
- School of Psychology and Centre for Brain Research, The University of Auckland , Auckland, New Zealand
| |
Collapse
|
3
|
Sandelius Å, Cullen NC, Källén Å, Rosengren L, Jensen C, Kostanjevecki V, Vandijck M, Zetterberg H, Blennow K. Transient increase in CSF GAP-43 concentration after ischemic stroke. BMC Neurol 2018; 18:202. [PMID: 30526557 PMCID: PMC6284302 DOI: 10.1186/s12883-018-1210-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 11/29/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Cerebrospinal fluid (CSF) biomarkers reflect ongoing processes in the brain. Growth-associated protein 43 (GAP-43) is highly upregulated in brain tissue shortly after experimental ischemia suggesting the CSF GAP-43 concentration may be altered in ischemic brain disorders. CSF GAP-43 concentration is elevated in Alzheimer's disease patients; however, patients suffering from stroke have not been studied previously. METHODS The concentration of GAP-43 was measured in longitudinal CSF samples from 28 stroke patients prospectively collected on days 0-1, 2-4, 7-9, 3 weeks, and 3-5 months after ischemia and cross-sectionally in 19 controls. The stroke patients were clinically evaluated using a stroke severity score system. The extent of the brain lesion, including injury size and degrees of white matter lesions and atrophy were evaluated by CT and magnetic resonance imaging. RESULTS Increased GAP-43 concentration was detected from day 7-9 to 3 weeks after stroke, compared to day 1-4 and to levels in the control group (P = 0.02 and P = 0.007). At 3-5 months after stroke GAP-43 returned to admission levels. The initial increase in GAP-43 during the nine first days was associated to stroke severity, the degree of white matter lesions and atrophy and correlated positively with infarct size (rs = 0.65, P = 0.001). CONCLUSIONS The transient increase of CSF GAP-43 is important to take into account when used as a biomarker for other neurodegenerative diseases such as Alzheimer's disease. Furthermore, GAP-43 may be a marker of neuronal responses after stroke and additional studies confirming the potential of CSF GAP-43 to reflect severity and outcome of stroke in larger cohorts are warranted.
Collapse
Affiliation(s)
- Åsa Sandelius
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden. .,Department of Psychiatry and Neurochemistry, Sahlgrenska University Hospital/Mölndal, S-431 80, Mölndal, Sweden.
| | - Nicholas C Cullen
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Åsa Källén
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Lars Rosengren
- Institute of Neuroscience and Physiology, Department of Clinical Neuroscience and Rehabilitation, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Crister Jensen
- Institute of Clinical Sciences, University of Gothenburg, Gothenburg, Sweden
| | | | | | - Henrik Zetterberg
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,UK Dementia Research Institute, WC1N, London, UK.,Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden. .,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden. .,Department of Psychiatry and Neurochemistry, Sahlgrenska University Hospital/Mölndal, S-431 80, Mölndal, Sweden.
| |
Collapse
|
4
|
Santos-Galdiano M, Pérez-Rodríguez D, Anuncibay-Soto B, Font-Belmonte E, Ugidos IF, Pérez-García CC, Fernández-López A. Celecoxib Treatment Improves Neurologic Deficit and Reduces Selective Neuronal Loss and Glial Response in Rats after Transient Middle Cerebral Artery Occlusion. J Pharmacol Exp Ther 2018; 367:528-542. [PMID: 30291174 DOI: 10.1124/jpet.118.251264] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 10/04/2018] [Indexed: 12/13/2022] Open
Abstract
Areas of selective neuronal loss (SNL) represent the first morphologic signs of damage in the penumbra region and are considered putative targets for ischemic stroke therapy. We performed a novel assessment of measuring the effects of the anti-inflammatory agent celecoxib by analyzing simultaneously the different neural populations (neurons, astrocytes, and microglia cells) in SNL and non-SNL areas. Rats were subjected to 1 hour of middle cerebral artery occlusion (MCAO) and treated with celecoxib 1 and 24 hours after ischemia. Infarct volume measurements and triple immunostaining of neurons (neuronal nuclear antigen), microglia (ionized calcium-binding adaptor molecule 1), and astroglia were performed after 12 and 48 hours of reperfusion. Motor response was tested by standard behavioral assays at 3, 12, 24, and 48 hours. Confocal analysis revealed that the percentage of SNL areas, microglia densities, and glial activation increased at 48 hours of reperfusion. Celecoxib treatment improved the neurologic deficit, reduced the infarct volume by 50% after 48 hours of reperfusion, and resulted in a reduced percentage of SNL areas and microglia and astroglia reactivity after 48 hours of reperfusion. This study proves, for the first time, that celecoxib presents postischemic neuroprotective effects in a transient MCAO model, prevents or delays the presence of SNL areas, and reduces glial activation.
Collapse
Affiliation(s)
- María Santos-Galdiano
- Área de Biología Celular, Instituto de Biomedicina (M.S.-G., D.P.-R., B.A.-S., E.F.-B., I.F.U., A.F.-L.) and Departamento de Medicina, Cirugía y Anatomía Veterinaria (C.C.P.-G.), Universidad de León and Neural Therapies SL, Edificio Institutos de Investigación (B.A.-S.), León, Spain
| | - Diego Pérez-Rodríguez
- Área de Biología Celular, Instituto de Biomedicina (M.S.-G., D.P.-R., B.A.-S., E.F.-B., I.F.U., A.F.-L.) and Departamento de Medicina, Cirugía y Anatomía Veterinaria (C.C.P.-G.), Universidad de León and Neural Therapies SL, Edificio Institutos de Investigación (B.A.-S.), León, Spain
| | - Berta Anuncibay-Soto
- Área de Biología Celular, Instituto de Biomedicina (M.S.-G., D.P.-R., B.A.-S., E.F.-B., I.F.U., A.F.-L.) and Departamento de Medicina, Cirugía y Anatomía Veterinaria (C.C.P.-G.), Universidad de León and Neural Therapies SL, Edificio Institutos de Investigación (B.A.-S.), León, Spain
| | - Enrique Font-Belmonte
- Área de Biología Celular, Instituto de Biomedicina (M.S.-G., D.P.-R., B.A.-S., E.F.-B., I.F.U., A.F.-L.) and Departamento de Medicina, Cirugía y Anatomía Veterinaria (C.C.P.-G.), Universidad de León and Neural Therapies SL, Edificio Institutos de Investigación (B.A.-S.), León, Spain
| | - Irene F Ugidos
- Área de Biología Celular, Instituto de Biomedicina (M.S.-G., D.P.-R., B.A.-S., E.F.-B., I.F.U., A.F.-L.) and Departamento de Medicina, Cirugía y Anatomía Veterinaria (C.C.P.-G.), Universidad de León and Neural Therapies SL, Edificio Institutos de Investigación (B.A.-S.), León, Spain
| | - Carlos César Pérez-García
- Área de Biología Celular, Instituto de Biomedicina (M.S.-G., D.P.-R., B.A.-S., E.F.-B., I.F.U., A.F.-L.) and Departamento de Medicina, Cirugía y Anatomía Veterinaria (C.C.P.-G.), Universidad de León and Neural Therapies SL, Edificio Institutos de Investigación (B.A.-S.), León, Spain
| | - Arsenio Fernández-López
- Área de Biología Celular, Instituto de Biomedicina (M.S.-G., D.P.-R., B.A.-S., E.F.-B., I.F.U., A.F.-L.) and Departamento de Medicina, Cirugía y Anatomía Veterinaria (C.C.P.-G.), Universidad de León and Neural Therapies SL, Edificio Institutos de Investigación (B.A.-S.), León, Spain
| |
Collapse
|
5
|
Association of Induced Hyperhomocysteinemia with Alzheimer's Disease-Like Neurodegeneration in Rat Cortical Neurons After Global Ischemia-Reperfusion Injury. Neurochem Res 2018; 43:1766-1778. [PMID: 30003389 DOI: 10.1007/s11064-018-2592-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 06/25/2018] [Accepted: 07/05/2018] [Indexed: 12/23/2022]
Abstract
Alzheimer's disease (AD) is a progressive and irreversible neurodegenerative disorder that results in massive hippocampal and neocortical neuronal loss leading to dementia and eventual death. The exact cause of Alzheimer's disease is not fully explored, although a number of risk factors have been recognized, including high plasma concentration of homocysteine (Hcy). Hyperhomocysteinemia (hHcy) is considered a strong, independent risk factor for stroke and dementia. However, the molecular background underlying these mechanisms linked with hHcy and ischemic stroke is not fully understood. Paper describes rat model of global forebrain ischemia combined with the experimentally induced hHcy. Global ischemia-reperfusion injury (IRI) was developed by 4-vessels occlusion lasting for 15 min followed by reperfusion period of 72 h. hHcy was induced by subcutaneous injection of 0.45 µmol/g of Hcy in duration of 14 days. The results showed remarkable neural cell death induced by hHcy in the brain cortex and neurodegeneration is further aggravated by global IRI. We demonstrated degeneration of cortical neurons, alterations in number and morphology of tissue astrocytes and dysregulation of oxidative balance with increased membrane protein oxidation. Complementary to, an immunohistochemical analysis of tau protein and β-amyloid peptide showed that combination of hHcy with the IRI might lead to the progression of AD-like pathological features. Conclusively, these findings suggest that combination of risk factor hHcy with IRI aggravates neurodegeneration processes and leads to development of AD-like pathology in cerebral cortex.
Collapse
|
6
|
Park JH, Cho JH, Ahn JH, Choi SY, Lee TK, Lee JC, Shin BN, Hong S, Jeon YH, Kim YM, Hwang IK, Lee YJ, Won MH, Kang IJ. Neuronal loss and gliosis in the rat striatum subjected to 15 and 30 minutes of middle cerebral artery occlusion. Metab Brain Dis 2018; 33:775-784. [PMID: 29354885 DOI: 10.1007/s11011-018-0192-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Accepted: 01/17/2018] [Indexed: 11/27/2022]
Abstract
Selective neuronal death or loss in certain brain regions has been well characterized in animal models of transient global cerebral ischemia. However, selective neuronal death in transient focal cerebral ischemia needs more investigation. Therefore, in this study, we studied selective neuronal death in the striatum (caudate putamen) of rats subjected to 15 or 30 min middle cerebral artery occlusion (MCAO). Neuronal death occurred in the dorsolateral field, not in the medial field in 30 min, not 15 min, MCAO-operated rats 5 days after MCAO using neuronal nuclear antigen immunohistochemistry and Fluoro-Jade B histofluorescence staining. In this group, immunoreactivity of glial fibrillary acidic protein in astrocytes was hardly shown in the dorsolateral field, although the immunoreactivity increased in the medial field. In addition, immunoreactivity of ionized calcium binding adapter molecule 1 in microglia was dramatically increased in the dorsolateral, not in the medial, field only in 30 min MCAO-operated rats. Briefly, these results show that at least 30 min of MCAO can evoke selective neuronal death, astrocytic dysfunction and microglial activation in the dorsolateral field of the rat striatum and suggest that a rat model of 30 min MCAO can be used to investigate mechanisms of neuronal death and gliosis following brief transient focal cerebral ischemic events for acute transient ischemic attack.
Collapse
Affiliation(s)
- Joon Ha Park
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon, 24252, Republic of Korea
| | - Jeong Hwi Cho
- Department of Neurobiology, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Ji Hyeon Ahn
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon, 24252, Republic of Korea
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon, 24252, Republic of Korea
| | - Tae-Kyeong Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Bich Na Shin
- Department of Neurobiology, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Seongkweon Hong
- Department of Surgery, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Yong Hwan Jeon
- Department of Radiology, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, Gangwon, 24341, Republic of Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, 1 Gwanak-ro, Seoul, 08826, Republic of Korea
| | - Young Joo Lee
- Department of Emergency Medicine, Seoul Hospital, College of Medicine, Sooncheonhyang University, 59 Daesagwan-ro, Seoul, 04401, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon, Gangwon, 24341, Republic of Korea.
| | - Il Jun Kang
- Department of Food Science and Nutrition, Hallym University, 1 Hallymdaehak-gil, Chuncheon, Gangwon, 24252, Republic of Korea.
| |
Collapse
|
7
|
Selective neuronal loss in ischemic stroke and cerebrovascular disease. J Cereb Blood Flow Metab 2014; 34:2-18. [PMID: 24192635 PMCID: PMC3887360 DOI: 10.1038/jcbfm.2013.188] [Citation(s) in RCA: 175] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 10/15/2013] [Accepted: 10/17/2013] [Indexed: 01/23/2023]
Abstract
As a sequel of brain ischemia, selective neuronal loss (SNL)-as opposed to pannecrosis (i.e. infarction)-is attracting growing interest, particularly because it is now detectable in vivo. In acute stroke, SNL may affect the salvaged penumbra and hamper functional recovery following reperfusion. Rodent occlusion models can generate SNL predominantly in the striatum or cortex, showing that it can affect behavior for weeks despite normal magnetic resonance imaging. In humans, SNL in the salvaged penumbra has been documented in vivo mainly using positron emission tomography and (11)C-flumazenil, a neuronal tracer validated against immunohistochemistry in rodent stroke models. Cortical SNL has also been documented using this approach in chronic carotid disease in association with misery perfusion and behavioral deficits, suggesting that it can result from chronic or unstable hemodynamic compromise. Given these consequences, SNL may constitute a novel therapeutic target. Selective neuronal loss may also develop at sites remote from infarcts, representing secondary 'exofocal' phenomena akin to degeneration, potentially related to poststroke behavioral or mood impairments again amenable to therapy. Further work should aim to better characterize the time course, behavioral consequences-including the impact on neurological recovery and contribution to vascular cognitive impairment-association with possible causal processes such as microglial activation, and preventability of SNL.
Collapse
|
8
|
Anti-inflammatory effects of total isoflavones from Pueraria lobata on cerebral ischemia in rats. Molecules 2013; 18:10404-12. [PMID: 23989686 PMCID: PMC6270189 DOI: 10.3390/molecules180910404] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 08/21/2013] [Accepted: 08/23/2013] [Indexed: 11/17/2022] Open
Abstract
Puerariae radix, the dried root of Pueraria lobata Ohwi, is one of earliest and most important edible crude herbs used for various medical purposes in Oriental medicine. The aim of the present study was to determine the anti-inflammatory effects of Total Isoflavones from P. lobata (TIPL), which contains the unique isoflavone puerarin, in ischemia in vivo models. Oral administration of TIPL (100 mg/kg) reduced the brain infarct volume and attenuated ischemia-induced cyclooxygenase-2 (COX-2) up-regulation at 2 days after middle cerebral artery occlusion (MCAo) in rats. Moreover, TIPL reduced activation of glial fibrillary acid protein (GFAP) and CD11b antibody (OX-42) at 7 days after MCAo in hippocampal CA1 region. These results show that TIPL can protect the brain from ischemic damage after MCAo. Regarding the immunohistochemical study, the effects of TIPL may be attributable to its anti-inflammatory properties by the inhibition of COX-2 expression, astrocyte expression, and microglia.
Collapse
|
9
|
Hung YC, Chou YS, Chang CH, Lin HW, Chen HY, Chen TY, Tai SH, Lee EJ. Early reperfusion improves the recovery of contralateral electrophysiological diaschisis following focal cerebral ischemia in rats. Neurol Res 2013; 32:828-34. [DOI: 10.1179/016164109x12581096870032] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
10
|
Transgenic mice over-expressing endothelial endothelin-1 show cognitive deficit with blood–brain barrier breakdown after transient ischemia with long-term reperfusion. Neurobiol Learn Mem 2013; 101:46-54. [DOI: 10.1016/j.nlm.2013.01.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 12/31/2012] [Accepted: 01/03/2013] [Indexed: 12/15/2022]
|
11
|
Ejaz S, Williamson DJ, Ahmed T, Sitnikov S, Hong YT, Sawiak SJ, Fryer TD, Aigbirhio FI, Baron JC. Characterizing infarction and selective neuronal loss following temporary focal cerebral ischemia in the rat: a multi-modality imaging study. Neurobiol Dis 2012; 51:120-32. [PMID: 23146994 DOI: 10.1016/j.nbd.2012.11.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 10/05/2012] [Accepted: 11/01/2012] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND AND PURPOSE Current models dictate that, depending on occurrence of early reperfusion, the ischemic penumbra either undergoes or escapes infarction (i.e., "pan-necrosis"). However, tissue outcome following temporary middle-cerebral artery occlusion (tMCAo) in rodents can also include selective neuronal loss (SNL), which even if subtle may impede functional recovery. In order to explore the pathophysiology of ischemic stroke, determine potential therapeutic targets and monitor effects of therapy, in vivo imaging surrogates of these varied histopathological outcomes applicable in the clinical setting would be useful. Although hyperintense signal on T(2)-weighted MRI in the chronic post-stroke stage is considered a reliable surrogate of tissue infarction, SNL is not associated with T(2)W abnormal signal. In the clinical setting, the neuron-specific PET ligand (11)C-flumazenil (FMZ) has been used to identify both pan-necrosis and peri-infarct SNL, but this inference has not been histopathological confirmed so far. Here we investigated the late tissue sequelae of tMCAo in the rodent using in vivo T(2)W MRI and FMZ-PET against post mortem immunohistochemistry as gold standard. METHODS Adult spontaneously hypertensive rats (SHRs) underwent 45 min distal-clip middle-cerebral artery occlusion and, 28 days later, FMZ-PET and T(2)W-MRI, immediately followed by immunohistochemistry for neuronal loss (NeuN), activated microglia and astrocytosis. Based on standard histopathological definitions, ischemic lesions were classified into pan-necrosis, partial infarction or SNL. NeuN changes and FMZ binding across the whole hemisphere were quantified in the same set of 44 regions-of-interest according to previously validated protocols; linear regressions between these two measures were carried out both within and across subjects. RESULTS Both cortical pan-necrosis/partial infarction and SNL were present in all rats except one, where SNL was isolated and extensive. Infarction/partial infarction, but not SNL, was associated with T(2)W hyperintense signals and cortical atrophy. In contrast, FMZ binding was decreased in all types of lesions including SNL, in proportion with NeuN staining intensity both within (p<0.05 to <0.001) and across (p<0.001) subjects, including the subject that showed pure SNL (p=0.01). CONCLUSION This novel study revealed three main facts: i) long-term histopathological cortical changes following 45 min tMCAo in SHRs included all three of SNL, partial infarction and frank infarction; ii) T2W MRI showed conspicuous high signal lesions for complete or partial infarction, but no changes for SNL; and iii) FMZ-PET was sensitive to all three types of tMCAo-induced histopathological changes, including isolated SNL, suggesting it is a valid surrogate for the histological sequelae of focal cerebral ischemia. In addition, the finding of almost universal completed cortical infarction at 28 days differed from our previous findings at 14-day survival using the same model and rat strain, where SNL was the almost exclusive outcome, possibly representing delayed infarct maturation. Prospective studies are needed to investigate this interesting possibility.
Collapse
Affiliation(s)
- Sohail Ejaz
- Stroke Research Group, Department of Clinical Neurosciences, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Lv P, Fang W, Geng X, Yang Q, Li Y, Sha L. Therapeutic neuroprotective effects of ginkgolide B on cortex and basal ganglia in a rat model of transient focal ischemia. Eur J Pharm Sci 2011; 44:235-40. [PMID: 21855632 DOI: 10.1016/j.ejps.2011.07.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Revised: 06/28/2011] [Accepted: 07/31/2011] [Indexed: 12/26/2022]
Abstract
Cerebral ischemia and reperfusion is one of the leading causes for death and severe disabilities in the world and often lead to irreversible brain damage over later lifespan. The aim of this study was to investigate the evolution of pathological damage in cerebral cortex and basal ganglia following ischemia and to evaluate the therapeutic neuroprotective effect of ginkgolide B in a rat model of stroke induced by middle cerebral artery occlusion (MCAO). TTC stain, brain water content and Evans-Blue extravasation were used to quantify brain damage. Our results demonstrated that basal ganglia undergo progressive pathological damage earlier following MCAO, and injury was stable and irreversible after 5 h following ischemia. However, onset of ischemia injury in cerebral cortex appeared later than basal ganglia and became evident about 3 h following MCAO, and injury was stable and irreversible after 6 h following ischemia. Blood brain barrier opened progressively, and it seemed to be significantly destroyed after 4 h following MCAO comparing with 0 h. Post-ischemic treatment with ginkgolide B improved neurological function and reduced infarct size in basal ganglia within 3 h and cerebral cortex within 5 h following MCAO. The therapeutic effect of ginkgolide B on extenuate brain edema and decrease blood brain barrier permeability were extended for 5h after ischemia, and more evident reversal effect were observed when administrated at earlier time.
Collapse
Affiliation(s)
- Peng Lv
- Department of Physiology, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, PR China
| | | | | | | | | | | |
Collapse
|
13
|
Xi HJ, Zhang TH, Tao T, Song CY, Lu SJ, Cui XG, Yue ZY. Propofol improved neurobehavioral outcome of cerebral ischemia-reperfusion rats by regulating Bcl-2 and Bax expression. Brain Res 2011; 1410:24-32. [PMID: 21783180 DOI: 10.1016/j.brainres.2011.06.060] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Revised: 06/24/2011] [Accepted: 06/25/2011] [Indexed: 02/08/2023]
Abstract
Propofol is an intravenous anesthetic with neuroprotective effects against cerebral ischemia-reperfusion (I/R) injury. Few studies regarding the neuroprotective and neurobehavioral effects of propofol have been conducted, and the underlying mechanisms are still unclear. Because I/R may result in neuronal apoptosis, the apoptosis regulatory genes B-cell leukemia-2 (Bcl-2) and Bcl-2-associated X protein (Bax) may be involved in the neuroprotective process. In this study, 120 Wistar rats were randomly divided into three groups (sham, I/R-induced, and propofol-treated). Cerebral ischemia was induced by clamping the bilateral common carotid arteries for 10min. Propofol (1.0mg/kg/min) was administered intravenously for 1h before the induction of ischemia. Neuronal damage was evaluated by neurobehavioral scores and histological examination of the brain sections at the level of the dorsal hippocampus at 6h, 24h, 48h, 72h, 4days, 5days, 6days, and 7days after I/R. The apoptotic rate of hippocampal neurons was detected by flow cytometry. The expression of Bcl-2 and Bax was evaluated using immunohistochemical and Western blot methods. The results of this study showed that neurobehavioral scores were higher in propofol-treated rats compared with I/R-induced rats with no propofol treatment. Moreover, the hippocampal expression of Bcl-2 was significantly higher, while the expression of Bax was significantly lower in propofol-treated rats compared with I/R-induced rats at 24h after ischemia. Hence, this study suggests that the neuroprotective effects of propofol against neuronal apoptosis may be a consequence of the regulation of Bcl-2 and Bax.
Collapse
Affiliation(s)
- Hong-Jie Xi
- Department of Anesthesiology, China and Heilongjiang Key Laboratory for Anesthesia and Critical Care, The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | | | | | | | | | | | | |
Collapse
|
14
|
Sorimachi T, Ito Y, Morita K, Fujii Y. Long-term follow up of patients with good outcome after intra-arterial thrombolysis for major arterial occlusion in the carotid territory: clinical and magnetic resonance imaging evaluation. Neurol Med Chir (Tokyo) 2010; 50:13-9; discussion 19. [PMID: 20098019 DOI: 10.2176/nmc.50.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Long-term clinical symptoms, including extrapyramidal signs, and magnetic resonance (MR) imaging studies were retrospectively analyzed in 21 patients with good outcome (modified Rankin scale scores 0-2) after successful recanalization of occluded major arteries by intra-arterial thrombolysis with mechanical disruption. Changes in high intensity areas (HIAs) and cerebral atrophy in the ischemic hemisphere were evaluated on follow-up fluid-attenuated inversion recovery MR images. Extrapyramidal signs, short-stepped gait and/or masked face, were observed in 12 of 21 patients during the follow-up period (11 to 68 months, mean 42 months). Enlargement of HIA was demonstrated in 10 of 18 patients undergoing follow-up MR imaging. Cerebral atrophy in the ischemic hemisphere was revealed on the follow-up MR images in all 18 patients. In nine patients with small infarctions, 20 ml or less on computed tomography scans, cerebral atrophy progressed more rapidly in four patients with extrapyramidal signs compared to the other five patients without extrapyramidal signs (p < 0.05). More than half of the patients with good outcome showed extrapyramidal signs. Extrapyramidal signs in patients with small infarction may indicate rapid progression of cerebral atrophy. The occurrence of extrapyramidal signs might be related to delayed neuronal death in atrophic areas.
Collapse
|
15
|
Hughes J, Beech J, Jones P, Wang D, Menon D, Baron J. Mapping selective neuronal loss and microglial activation in the salvaged neocortical penumbra in the rat. Neuroimage 2010; 49:19-31. [DOI: 10.1016/j.neuroimage.2009.08.047] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2009] [Revised: 07/24/2009] [Accepted: 08/17/2009] [Indexed: 11/24/2022] Open
|
16
|
Morales P, Fiedler JL, Andrés S, Berrios C, Huaiquín P, Bustamante D, Cardenas S, Parra E, Herrera-Marschitz M. Plasticity of hippocampus following perinatal asphyxia: effects on postnatal apoptosis and neurogenesis. J Neurosci Res 2009; 86:2650-62. [PMID: 18512760 DOI: 10.1002/jnr.21715] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Asphyxia during delivery produces long-term deficits in brain development, including hippocampus. We investigated hippocampal plasticity after perinatal asphyxia, measuring postnatal apoptosis and neurogenesis. Asphyxia was performed by immersing rat fetuses with uterine horns removed from ready-to-deliver rats into a water bath for 20 min. Caesarean-delivered pups were used as controls. The animals were euthanized 1 week or 1 month after birth. Apoptotic nuclear morphology and DNA breaks were assessed by Hoechst and TUNEL assays. Neurogenesis was estimated by bromodeoxyuridine/MAP-2 immunocytochemistry, and the levels and expression of proteins related to apoptosis and cell proliferation were measured by Western blots and in situ hybridization, respectively. There was an increase of apoptosis in CA1, CA3, and dentate gyrus (DG) and cell proliferation and neurogenesis in CA1, DG, and hilus regions of hippocampus 1 week after asphyxia. The increase of apoptosis in CA3 and cell proliferation in the suprapyramidal band of DG was still observed 1 month following asphyxia. There was an increase of BAD, BCL-2, ERK2, and bFGF levels in whole hippocampus and bFGF expression in CA1 and CA2 and hilus at P7 and P30. There was a concomitant decrease of phosphorylated-BAD (Ser112) levels. The increase of BAD levels supports the idea of delayed cell death after perinatal asphyxia, whereas the increases of BCL-2, ERK2, and bFGF levels suggest the activation of neuroprotective and repair pathways. In conclusion, perinatal asphyxia induces short- and long-term regionally specific plastic changes, including delayed cell death and neurogenesis, involving pro- and antiapoptotic as well as mitogenic proteins, favoring hippocampal functional recovery.
Collapse
Affiliation(s)
- P Morales
- Programme of Molecular and Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Arsava EM, Gurer G, Gursoy-Ozdemir Y, Karatas H, Dalkara T. A new model of transient focal cerebral ischemia for inducing selective neuronal necrosis. Brain Res Bull 2008; 78:226-31. [PMID: 19063950 DOI: 10.1016/j.brainresbull.2008.11.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2008] [Revised: 11/06/2008] [Accepted: 11/06/2008] [Indexed: 11/28/2022]
Abstract
Brief cerebral ischemia leads to selective neuronal necrosis (SNN), which is characterized by neuronal death with sparing of glial and vascular elements of the central nervous system. Understanding the pathophysiology of SNN may help elucidating the mechanisms and consequences of neuronal injury in humans following brief ischemia. Contrary to the presence of reproducible models of transient global ischemia, animal models of transient focal ischemia producing SNN are scarce and have important limitations such as causing ischemia in a vast area and inducing additional insults. In this study, we developed a practical mouse model of SNN without these limitations, by compressing the distal middle cerebral artery (MCA) with a blunted micropipette for 15 min. The success of compression was evaluated by monitoring the regional cerebral blood flow, and conventional histopathology and immunolabeling of the brain sections. Seven/fourteen days after ischemia, intracisternally administered propidium iodide labeled numerous necrotic cells in the frontoparietal cortex, which were mostly NeuN-positive, but were not immunolabeled with astrocytic markers (GFAP and S100), and showed neuronal morphology with hematoxylin-eosin staining, indicating that the model successfully induced ischemic injury limited to neurons. The model could become an important tool for investigating the long-term effects of brief ischemic events like transient ischemic attacks and could offer convenient reversible distal MCA occlusion for studies using intravital microscopy.
Collapse
Affiliation(s)
- Ethem Murat Arsava
- Department of Neurology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | | | | | | | | |
Collapse
|
18
|
Morales P, Huaiquín P, Bustamante D, Fiedler J, Herrera-Marschitz M. Perinatal asphyxia induces neurogenesis in hippocampus: an organotypic culture study. Neurotox Res 2007; 12:81-4. [PMID: 17513202 DOI: 10.1007/bf03033903] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
There is clinical and experimental evidence indicating that neurocircuitries of the hippocampus are vulnerable to hypoxia/ischemia occurring at birth, inducing, upon re-oxygenation/re-circulation, delayed neuronal death, but also compensatory mechanisms, including neurogenesis. In the present report, perinatal asphyxia was induced by immersing foetuses-containing uterine horns removed from ready-to-deliver rats into a water bath at 37 degrees C for 20 min. Some pups were delivered immediately after the hysterectomy to be used as non-asphyxiated caesarean-delivered controls. The pups were sacrificed after seven days for preparing organotypic hippocampal cultures. The cultures were grown on a coverslip in a medium-containing culture tube inserted in a hole of a roller device standing on the internal area of a cell incubator at 35 degrees C, 10% CO2. At days in vitro (DIV) 25-27, cultures were fixed for assaying cell proliferation and neuronal phenotype with antibodies against 5-bromo-2'deoxyuridine (BrdU) and microtubule associated protein-2 (MAP-2), respectively. Confocal microscopy revealed that there was a 2-fold increase of BrdU-positive, but a 40% decrease of MAP-2-positive cells/mm3 in cultures from asphyxia-exposed, compared to that from control animals. Approximately 30% of BrdU-positive cells were also positive for MAP-2 (approximately 4800 cells), mainly seen in the dentate gyrus of the hippocampus, demonstrating a 3-fold increase of postnatal neurogenesis, when the total amount of double-labelled cells seen in cultures from asphyxia-exposed animals is compared to that from control animals.
Collapse
Affiliation(s)
- P Morales
- Programme of Molecular and Clinical Pharmacology, ICBM, Medical Faculty and Department of Biochemistry and Molecular Biology, Chemical and Pharmaceutical Sciences Faculty, University of Chile, Santiago, Chile
| | | | | | | | | |
Collapse
|
19
|
Wilde EA, Bigler ED, Hunter JV, Fearing MA, Scheibel RS, Newsome MR, Johnson JL, Bachevalier J, Li X, Levin HS. Hippocampus, amygdala, and basal ganglia morphometrics in children after moderate-to-severe traumatic brain injury. Dev Med Child Neurol 2007; 49:294-9. [PMID: 17376141 DOI: 10.1111/j.1469-8749.2007.00294.x] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
While closed head injury frequently results in damage to the frontal and temporal lobes, damage to deep cortical structures, such as the hippocampus, amygdala, and basal ganglia, has also been reported. Five deep central structures (hippocampus, amygdala, globus pallidus, putamen, and caudate) were examined in 16 children (eight males, eight females; aged 9-16y), imaged 1 to 10 years after moderate-to-severe traumatic brain injury (TBI), and in 16 individually-matched uninjured children. Analysis revealed significant volume loss in the hippocampus, amydala, and globus pallidus of the TBI group. Investigation of relative volume loss between these structures and against five cortical areas (ventromedial frontal, superomedial frontal, lateral frontal, temporal, and parieto-occipital) revealed the hippocampus to be the most vulnerable structure following TBI (i.e. greatest relative difference between the groups). In a separate analysis excluding children with focal hippocampal abnormalities (e.g. lesions), group differences in hippocampal volume were still evident, suggesting that hippocampal damage may be diffuse rather than focal.
Collapse
|
20
|
|
21
|
Miyashita K, Itoh H, Arai H, Suganami T, Sawada N, Fukunaga Y, Sone M, Yamahara K, Yurugi-Kobayashi T, Park K, Oyamada N, Sawada N, Taura D, Tsujimoto H, Chao TH, Tamura N, Mukoyama M, Nakao K. The neuroprotective and vasculo-neuro-regenerative roles of adrenomedullin in ischemic brain and its therapeutic potential. Endocrinology 2006; 147:1642-53. [PMID: 16384868 DOI: 10.1210/en.2005-1038] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Adrenomedullin (AM) is a vasodilating hormone secreted mainly from vascular wall, and its expression is markedly enhanced after stroke. We have revealed that AM promotes not only vasodilation but also vascular regeneration. In this study, we focused on the roles of AM in the ischemic brain and examined its therapeutic potential. We developed novel AM-transgenic (AM-Tg) mice that overproduce AM in the liver and performed middle cerebral artery occlusion for 20 min (20m-MCAO) to examine the effects of AM on degenerative or regenerative processes in ischemic brain. The infarct area and gliosis after 20m-MCAO was reduced in AM-Tg mice in association with suppression of leukocyte infiltration, oxidative stress, and apoptosis in the ischemic core. In addition, vascular regeneration and subsequent neurogenesis were enhanced in AM-Tg mice, preceded by increase in mobilization of CD34(+) mononuclear cells, which can differentiate into endothelial cells. The vasculo-neuro-regenerative actions observed in AM-Tg mice in combination with neuroprotection resulted in improved recovery of motor function. Brain edema was also significantly reduced in AM-Tg mice via suppression of vascular permeability. In vitro, AM exerted direct antiapoptotic and neurogenic actions on neuronal cells. Exogenous administration of AM in mice after 20m-MCAO also reduced the infarct area, and promoted vascular regeneration and functional recovery. In summary, this study suggests the neuroprotective and vasculo-neuro-regenerative roles of AM and provides basis for a new strategy to rescue ischemic brain through its multiple hormonal actions.
Collapse
Affiliation(s)
- Kazutoshi Miyashita
- Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, Sakyo-ku, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Morales P, Reyes P, Klawitter V, Huaiquín P, Bustamante D, Fiedler J, Herrera-Marschitz M. Effects of perinatal asphyxia on cell proliferation and neuronal phenotype evaluated with organotypic hippocampal cultures. Neuroscience 2006; 135:421-31. [PMID: 16112481 DOI: 10.1016/j.neuroscience.2005.05.062] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2005] [Revised: 05/24/2005] [Accepted: 05/30/2005] [Indexed: 10/25/2022]
Abstract
The present report summarizes studies combining an in vivo and in vitro approach, where asphyxia is induced in vivo at delivery time of Wistar rats, and the long term effects on hippocampus neurocircuitry are investigated in vitro with organotypic cultures plated at postnatal day seven. The cultures preserved hippocampus layering and regional subdivisions shown in vivo, and only few dying cells were observed when assayed with a viability test at day in vitro 27. When properly fixed, cultures from asphyxia-exposed animals showed a decreased amount of microtubule-associated protein-2 immunocytochemically positive cells (approximately 30%), as compared with that from controls. The decrease in microtubule-associated protein-2 immunocytochemistry was particularly prominent in Ammon's horn 1 and dentate gyrus regions (approximately 40%). 5-Bromo-2'deoxyuridine labeling revealed a two-fold increase in cellular proliferation in cultures from asphyxia-exposed, compared with that from control animals. Furthermore, confocal microscopy and quantification using the optical disector technique demonstrated that in cultures from asphyxia-exposed animals approximately 30% of 5-bromo-2'deoxyuridine-positive cells were also positive to microtubule-associated protein-2, a marker for neuronal phenotype. That proportion was approximately 20% in cultures from control animals. Glial fibrillary acidic protein-immunocytochemistry and Fast Red nuclear staining revealed that the core of the hippocampus culture was surrounded by a well-developed network of glial fibrillary acidic protein-positive cells and glial fibrillary acidic protein-processes providing an apparent protective shield around the hippocampus. That shield was less developed in cultures from asphyxia-exposed animals. The increased mitotic activity observed in this study suggests a compensatory mechanism for the long-term impairment induced by perinatal asphyxia, although it is not clear yet if that mechanism leads to neurogenesis, astrogliogenesis, or to further apoptosis.
Collapse
Affiliation(s)
- P Morales
- Programme of Molecular and Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, P.O. Box 70.000 Santiago 7, Chile
| | | | | | | | | | | | | |
Collapse
|
23
|
Adachi N. Cerebral ischemia and brain histamine. ACTA ACUST UNITED AC 2005; 50:275-86. [PMID: 16181682 DOI: 10.1016/j.brainresrev.2005.08.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2005] [Revised: 07/31/2005] [Accepted: 08/02/2005] [Indexed: 10/25/2022]
Abstract
Cerebral ischemia induces excess release of glutamate and an increase in the intracellular Ca(2+) concentration in neurons, which provokes enzymatic process leading to irreversible neuronal injury. Histamine plays a role as a neurotransmitter in the mammalian brain, and histamine release from nerve endings is enhanced in ischemia by facilitation of histaminergic activity. Dissimilar to ischemia-induced release of glutamate, histamine release is gradual and long lasting. The enhancement may contribute to neuroprotection against ischemic damage, because suppression of histaminergic activity aggravates the histologic outcome caused by ischemia. Preischemic administration of histamine (i.c.v.) suppresses ischemic release of glutamate and ameliorates neuronal damage, whereas blockade of central histamine H(2) receptors aggravates ischemic injury. These suggest that histamine provides beneficial effects against ischemic damage through histamine H(2) receptors, when administered before induction of ischemia. Postischemic loading with histidine, a precursor of histamine, alleviates both brain infarction and delayed neuronal death. Since the alleviation is abolished by blockade of central histamine H(2) receptors, facilitation of central histamine H(2) action caused by histidine may prevent reperfusion injury after ischemic events. Because the ischemia-induced increase in the glutamate level rapidly resumes after reperfusion of cerebral blood flow, beneficial effects caused by postischemic loading with histidine may be due to other mechanisms besides suppression of excitatory neurotransmitter release. Anti-inflammatory action by histamine H(2) receptor stimulation is a likely mechanism responsible for the improvement.
Collapse
Affiliation(s)
- Naoto Adachi
- Department of Anesthesiology and Resuscitology, Ehime University School of Medicine, Shitsukawa, Touon-shi, Ehime 791-0295, Japan.
| |
Collapse
|
24
|
Lee EJ, Chen HY, Lee MY, Chen TY, Hsu YS, Hu YL, Chang GL, Wu TS. Cinnamophilin reduces oxidative damage and protects against transient focal cerebral ischemia in mice. Free Radic Biol Med 2005; 39:495-510. [PMID: 16043021 DOI: 10.1016/j.freeradbiomed.2005.04.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2005] [Revised: 03/25/2005] [Accepted: 04/04/2005] [Indexed: 11/18/2022]
Abstract
Acute neuroprotective effects of cinnamophilin (CINN; (8R, 8'S)-4, 4'-dihydroxy-3, 3'-dimethoxy-7-oxo-8, 8'-neolignan), a novel antioxidant and free radical scavenger, were studied in a mouse model of transient middle cerebral artery (MCA) occlusion. CINN was administered intraperitoneally either 15 min before (pretreatment) or 2 h after the onset of MCA occlusion (postischemic treatment). Relative to vehicle-treated controls, animals pretreated with CINN, at 20-80 mg/kg, had significant reductions in brain infarction by 33-46% and improvements in neurobehavioral outcome. Postischemic administration with CINN (80 mg/kg) also significantly reduced brain infarction by 43% and ameliorated neurobehavioral deficits. Additionally, CINN administration significantly attenuated in situ accumulation of superoxide anions (O2-) in the boundary zones of infarct at 4 h after reperfusion. Consequently, CINN-treated animals exhibited significantly decreased levels of oxidative damage, as assessed by immunopositive reactions for 8-hydroxy-2'-deoxyguanosine (8-OHdG) and 4-hydroxynonenal (4-HNE), and the resultant inflammatory reactions at 24 h post-insult. It is concluded that CINN effectively reduced brain infarction and improved neurobehavioral outcome following a short-term recovery period after severe transient focal cerebral ischemia in mice. The finding of a decreased extent of reactive oxygen species and oxidative damage observed with CINN treatment highlights that its antioxidant and radical scavenging ability is contributory.
Collapse
Affiliation(s)
- E-Jian Lee
- Neurophysiology Laboratory, Neurosurgical Service, Department of Surgery & Institute of Biomedical Engineering, National Cheng Kung University Medical Center and Medical School, Tainan, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Winter B, Juckel G, Viktorov I, Katchanov J, Gietz A, Sohr R, Balkaya M, Hörtnagl H, Endres M. Anxious and hyperactive phenotype following brief ischemic episodes in mice. Biol Psychiatry 2005; 57:1166-75. [PMID: 15866557 DOI: 10.1016/j.biopsych.2005.02.010] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2004] [Revised: 12/02/2004] [Accepted: 02/09/2005] [Indexed: 11/20/2022]
Abstract
BACKGROUND Poststroke emotional and behavioral abnormalities have an impact on outcome but have scarcely been characterized in animal models. We tested whether brief ischemic episodes induce behavioral changes in mice. METHODS 129/Sv mice were subjected to 30-min occlusion of left or right middle cerebral artery (MCAo) followed by reperfusion or sham operation (n = 9 or 10 per group). Eight to ten weeks later, mice were tested for spontaneous locomotor activity, anxiety in the elevated plus maze, and depressive behavior in the modified Porsolt forced swim test. Outcome was correlated to monoamine and amino acid levels and compared with histologic damage at 10 weeks. RESULTS Ischemia was associated with increased activity (right MCAo) and anxiety (left MCAo), but not poststroke depression. Noradrenaline increased by 30%-45% in the ischemic striatum and correlated with locomotor activity (r = .48); dopamine and homovanillinic acid were decreased compared with sham. The lesion was confined to the striatum, and scattered neuronal death was observed in a number of remote brain regions. CONCLUSION Brief ischemic episodes in the mouse induce an anxious, hyperactive but not depressive phenotype that may relate to left versus right hemispheric lesion location, alterations in brain monoamine levels, and selective neurodegeneration.
Collapse
Affiliation(s)
- Benjamin Winter
- Klinik und Poliklinik für Neurologie der Charité, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Injury to the brain usually manifests not in a diffuse uniform manner but rather with selective sites of damage indicative of differential vulnerability. This question of neuronal susceptibility has been one of major interest both in disease processes as well as damage induced by environmental factors. For experimental examination, brain structures with obvious neuronal subpopulations and organization such as the cerebellum and the hippocampus have offered the most promise. In the hippocampus distinct neuronal populations exist that demonstrate differential vulnerability to various forms of insult including ischemia, excitotoxicity, and environmental factors. The more recent data regarding the presence of neuronal progenitor cells in the subgranular zone of the dentate offers the opportunity to expand such experimental examination to the process of injury-induced neurogenesis. Thus, more recent studies have expanded the examination of the hippocampus to include models of damage to the dentate neurons in addition to the highly vulnerable pyramidal neurons. A number of these models are presented for both human disease and experimental animal conditions. Examination of the responses between these distinct cell populations offers the potential for understanding factors that are critical in neuronal death and survival.
Collapse
Affiliation(s)
- G Jean Harry
- Neurotoxicology Group, Laboratory of Molecular Toxicology, National Institute of Environmental Health Sciences, 27709, Research Triangle Park, NC, USA.
| | | |
Collapse
|
27
|
Isaacson RL, Fahey JM, Mughairbi FA. Environmental conditions unexpectedly affect the long-term extent of cell death following an hypoxic episode. Ann N Y Acad Sci 2003; 993:179-94; discussion 195-6. [PMID: 12853312 DOI: 10.1111/j.1749-6632.2003.tb07528.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Previously we reported delayed cell death, defined by clear-cut cell loss 60 days after a nitrite-induced hypoxic episode. The loss of cells was not apparent two weeks after the treatment, although some changes in cellular appearance were observed at that time. A similar delayed loss of neurons in the hippocampus after hypoxia induced by blood vessel occlusion has also been found. In addition, we reported that the amount of methemoglobinemia induced by the sodium nitrite can be reduced by the stress produced by handling and the injection of saline 2 or 24 h before the nitrite administration. The degree of methemoglobin formed is directly related to cell death in certain areas of the brain, including regions within the hippocampus. Considering the many effects that can be produced by chronic and acute stress of several kinds and the length of time during which these effects manifest themselves, we undertook to determine the histologic effects of the stresses of transport on the neuroanatomic effects of sodium nitrite administration 60 days post administration. Comparisons were made of the effects of two methods of transport from the laboratory in which the animals (male CD-1 mice) were injected with the sodium nitrite or saline (Tufts Medical School) to the laboratory in which the histologic evaluations were made (Binghamton University). The animals began their travel several hours after the injections. One transport method was by commuter airline and the other was by automobile. All animals had the same transport from the supplier to the Boston location (truck). Thus, the stress of experimental interest occurred after the nitrite administration. Upon arrival at Binghamton University, the animals were housed at the University in their own colony room for 60 days before sacrifice. After sacrifice, sections from their brains were subjected to a number of histologic staining procedures, including PTAH, the Bielschowsky silver method, GFAP, and the standard Nissl procedure. Although special attention was paid to hippocampal areas, changes in cells in the habenulae and the linings of ventricular areas were also prominent. Surprisingly, the nitrite treatment before transport to Binghamton offered partial protection against the very substantial and lasting effects of the injections, transport, and handling found in the control animals. Differential effects caused by the two methods of transport were also noted.
Collapse
Affiliation(s)
- Robert L Isaacson
- Department of Psychology, Binghamton University, New York 13902, USA
| | | | | |
Collapse
|
28
|
Kametsu Y, Osuga S, Hakim AM. Apoptosis occurs in the penumbra zone during short-duration focal ischemia in the rat. J Cereb Blood Flow Metab 2003; 23:416-22. [PMID: 12679718 DOI: 10.1097/01.wcb.0000052281.23292.7c] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
To investigate the appearance of apoptosis in short-duration focal ischemia, the authors induced left middle cerebral artery (MCA) occlusion in male rats by insertion of an intraluminal suture. The total number of apoptotic cells was determined by hematoxylin-eosin staining and TUNEL labeling and confirmed by gel electrophoresis. The data indicate that the total number of apoptotic cells increased with ischemia duration (P = 0.0006), with most apoptotic cells located in the striatum of the ischemic hemisphere. As the duration of ischemia lengthened, necrosis became more prevalent and apoptosis receded to the periphery of the infarct. Using iodo[14C]-antipyrine to correlate the distribution of apoptosis to regional CBF (rCBF), the authors found that rCBF in the ischemic dorsolateral striatum was compatible with penumbra flow and significantly lower than the ventromedial striatum and frontoparietal cortex. This difference disappeared after 45 minutes of reperfusion. The authors conclude that focal ischemia of short duration results in changes compatible with apoptosis in regions of low rCBF, and this can occur without necrosis. This model is relevant to transient ischemic attack in the human and may suggest that, in addition to being a harbinger of stroke, transient ischemic attacks may cause histopathologic changes not yet clinically detectable.
Collapse
Affiliation(s)
- Yutaka Kametsu
- Department of Neurology, School of Medicine, University of Tokai Isehara, Kanagawa, Japan
| | | | | |
Collapse
|
29
|
Sakamoto T, Kawaguchi M, Kurita N, Horiuchi T, Kakimoto M, Inoue S, Furuya H, Nakamura M, Konishi N. Long-term assessment of hind limb motor function and neuronal injury following spinal cord ischemia in rats. J Neurosurg Anesthesiol 2003; 15:104-9. [PMID: 12657995 DOI: 10.1097/00008506-200304000-00007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Recent evidence suggests that brain injury caused by ischemia is a dynamic process characterized by ongoing neuronal loss for at least 14 days after ischemia. However, long-term outcome following spinal cord ischemia has not been extensively examined. Therefore, we investigated the changes of hind limb motor function and neuronal injury during a 14-day recovery period after spinal cord ischemia. Male Sprague-Dawley rats received spinal cord ischemia (n = 64) or sham operation (n = 21). Spinal cord ischemia was induced by inflation of a 2F Fogarty catheter placed into the thoracic aorta for 6, 8, or 10 minutes. The rats were killed 2, 7, or 14 days after reperfusion. Hind limb motor function was assessed with the 21-point Basso, Beattie, and Bresnahan (BBB) scale during the recovery period. The number of normal and necrotic neurons was counted in spinal cord sections stained with hematoxylin/eosin. Longer duration of spinal cord ischemia produced severer hind limb motor dysfunction at each time point. However, BBB scores gradually improved during the 14-day recovery period. Neurologic deterioration was not observed between 7 and 14 days after reperfusion. The number of necrotic neurons peaked 2 days after reperfusion and then decreased. A small number of necrotic neurons were still observed 7 and 14 days after reperfusion in some of the animals. These results indicate that, although hind limb motor function may gradually recover, neuronal loss can be ongoing for 14 days after spinal cord ischemia.
Collapse
|
30
|
Kidwell CS, Saver JL, Starkman S, Duckwiler G, Jahan R, Vespa P, Villablanca JP, Liebeskind DS, Gobin YP, Vinuela F, Alger JR. Late secondary ischemic injury in patients receiving intraarterial thrombolysis. Ann Neurol 2002; 52:698-703. [PMID: 12447922 DOI: 10.1002/ana.10380] [Citation(s) in RCA: 149] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Although animal models have demonstrated that late secondary cerebral injury after arterial occlusion and subsequent recanalization may limit the benefit of reperfusion therapy, this phenomenon has not been well characterized in humans. Diffusion-perfusion magnetic resonance imaging studies were performed before treatment, early after treatment, and at day 7 in patients undergoing vessel recanalization with intraarterial thrombolytics. Among 18 patients studied, mean age was 71 (range, 27-94), and median entry National Institutes of Health Stroke Scale score was 13 (range, 6-25). Early after recanalization, partial or complete normalization of diffusion imaging abnormalities occurred in 8 of 18 (44%) patients. Among the eight patients with early diffusion imaging reversal, late secondary injury by day 7 occurred in 5 (63%), and sustained normalization of all reversed tissue occurred in 3 (38%). Pretreatment apparent diffusion coefficient values were lowest in regions experiencing no reversal (mean apparent diffusion coefficient, 608 microm(2)/sec), intermediate in regions with reversal and secondary decline (617 microm(2)/sec), and highest in regions with sustained reversal (663 microm(2)/sec). There was a trend toward less improvement in neurological deficit in patients with secondary injury versus patients with sustained reversal. In the future, late secondary tissue injury may become an important therapeutic target for postreperfusion neuroprotective therapies, with treatment efficacy monitored by serial diffusion magnetic resonance imaging.
Collapse
Affiliation(s)
- Chelsea S Kidwell
- Stroke Center, UCLA Medical Center, 710 Westwood Plaza, Los Angeles, CA 90095, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Snider BJ, Du C, Wei L, Choi DW. Cycloheximide reduces infarct volume when administered up to 6 h after mild focal ischemia in rats. Brain Res 2001; 917:147-57. [PMID: 11640900 DOI: 10.1016/s0006-8993(01)02822-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
We have previously described a rodent model of brief (30 min) middle cerebral artery occlusion followed by reperfusion, in which infarction develops gradually, reaching completion more than 3 days after ischemia, accompanied by morphological, biochemical, and pharmacological evidence of apoptosis. In the present study, we tested the hypotheses that delayed administration of a protein synthesis inhibitor would be effective in reducing tissue injury in this slowly evolving ischemic infarction, and that efficacy of this treatment would wane with more prolonged ischemia. Focal cerebral ischemia was induced in Long-Evans rats by occlusion of the right middle cerebral artery. Infarction volume was analyzed using triphenyl tetrazolium chloride staining, and morphology was studied using hematoxylin and eosin stained sections. Following 30 min middle cerebral artery occlusion and reperfusion, the core ischemic region exhibited vacuolization in the neuropil by 36 h after ischemia, and infarction reached full size by 7 days after ischemia. Cycloheximide reduced infarct volume when given up to 6 h after ischemia. If the duration of ischemic insult was increased to 90 min, the therapeutic window for delayed cycloheximide was only 30 min. In permanent middle cerebral artery occlusion, cycloheximide was ineffective even when given prior to ischemia onset. After mild, but not severe, ischemic insults, cerebral infarction develops slowly and may be treatable with protein synthesis inhibitors, even when treatment is delayed for up to 6 h after the onset of ischemia.
Collapse
Affiliation(s)
- B J Snider
- Center for the Study of Nervous System Injury and Department of Neurology, Washington University School of Medicine, Campus Box 8111, 660 S. Euclid, St. Louis, MO 63110, USA
| | | | | | | |
Collapse
|
32
|
Shiozaki T, Akai H, Taneda M, Hayakata T, Aoki M, Oda J, Tanaka H, Hiraide A, Shimazu T, Sugimoto H. Delayed hemispheric neuronal loss in severely head-injured patients. J Neurotrauma 2001; 18:665-74. [PMID: 11497093 DOI: 10.1089/089771501750357618] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Recent experimental studies have revealed that traumatic brain injury as well as ischemic brain injury can cause chronic progressive neuronal damage. In the present study, we demonstrate previously unreported delayed cerebral atrophy on computerized tomography (CT) scans in severely head-injured patients. Seventeen severely head-injured patients who required mild hypothermia to control intracranial hypertension after the failure of conventional therapies were retrospectively analyzed. All 17 patients survived more than 1 year. Delayed neuronal loss (DNL) was observed in only eight of the 17 patients. Eight patients with DNL required longer durations of mild hypothermia to control intracranial hypertension than nine patients without DNL. Six of these eight patients with DNL achieved functional recovery despite progressive atrophic changes demonstrated on CT scans. On CT scans, DNL was characterized by (1) the sudden appearance at several months postinjury of a low-density area in the hemisphere ipsilateral to the injury; (2) the preservation of essential cortical structure although related white matter structures showed severe atrophic changes; and (3) no spread of the low-density area to the contiguous territory of the other main cerebral artery. It is concluded that focal primary injury to underlying brain, if severe enough, can result in delayed hemispheric atrophy.
Collapse
Affiliation(s)
- T Shiozaki
- Department of Traumatology, Osaka University Medical School, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Li F, Silva MD, Liu KF, Helmer KG, Omae T, Fenstermacher JD, Sotak CH, Fisher M. Secondary decline in apparent diffusion coefficient and neurological outcomes after a short period of focal brain ischemia in rats. Ann Neurol 2001. [DOI: 10.1002/1531-8249(200008)48:2<236::aid-ana14>3.0.co;2-7] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
34
|
Abstract
The nonreceptor tyrosine kinase PYK2 represents a stress-sensitive mediator of c-Jun N-terminal kinase and p38 mitogen-activated protein kinase (MAPK) signaling pathways in many cell types. In the present study, we assessed the tyrosine phosphorylation of PYK2 under normal and pathological conditions in the CNS. We generated a polyclonal antibody that selectively recognizes tyrosine-phosphorylated PYK2 at its major autophosphorylation site. By using this antibody, we demonstrate that the phosphorylation profile of PYK2 after focal cerebral ischemia is biphasic. The first phase occurs within 1 hr, when most of the phospho-PYK2 immunoreactivity was observed in cortical neurons, whereas 24-72 hr after ischemia, a striking induction of phospho-PYK2 immunoreactivity was evident in microglia around the necrotic infarcted area. Double-immunostaining analysis using both anti-phospho-PYK2 antibody and antibody against the double-phosphorylated active form of p38MAPK revealed that the two phosphorylated protein kinases exhibit strikingly similar distribution patterns after ischemia. A short time after ischemia, phosphorylation of p38MAPK was evident in the cortical neurons as demonstrated by both immunohistochemistry and immunoblotting analysis, whereas 24-72 hr after ischemia, phospho-p38MAPK was found in activated microglia and colocalized with phospho-PYK2. In contrast to cortical neurons, basal phospho-PYK2 immunoreactivity was observed in hippocampal pyramidal neurons, which was markedly decreased after kainate acid-induced status epilepticus. However, 24 hr after the epileptic onset, a pronounced upregulation of PYK2 and phospho-PYK2 immunoreactivities was evident in microglial cells, as demonstrated by double-immunostaining with the microglial marker OX42. These results provide, for the first time, in situ localization of tyrosine-phosphorylated PYK2 in neuronal stress pathways in the adult rat brain and are consistent with the role of PYK2 as an upstream regulator of p38MAPK signaling cascades in response to stress signals.
Collapse
|
35
|
Abstract
Neuronal death following ischemic insults has been thought to reflect necrosis. However, recent evidence from several labs suggests that programmed cell death, leading to apoptosis, might additionally contribute to this death. We have used both in vitro and in vivo models to study the role of apoptosis in ischemic cell death. Some features of apoptosis (TUNEL staining, internucleosomal DNA fragmentation, sensitivity to cycloheximide) were observed following transient focal ischemia in rats. Brief transient focal ischemia was followed by delayed infarction more than 3 days later; this delayed infarction was sensitive to cycloheximide. A cycloheximide-sensitive component of neuronal cell death was also observed in cultured murine neocortical neurons deprived of oxygen-glucose in the presence of glutamate receptor antagonists. This presumed ischemic apoptosis was attenuated by caspase inhibitors, or by homozygous deletion of the bax gene. Neurons may undergo both apoptosis and necrosis after ischemic insults, and thus it may be therapeutically desirable to block both processes.
Collapse
Affiliation(s)
- B J Snider
- Center for the Study of Nervous System Injury, Washington University School of Medicine, St. Louis, Missouri 63110, USA.
| | | | | |
Collapse
|
36
|
Heim C, Pardowitz I, Sieklucka M, Kolasiewicz W, Sontag T, Sontag KH. The analysis system COGITAT for the study of cognitive deficiencies in rodents. BEHAVIOR RESEARCH METHODS, INSTRUMENTS, & COMPUTERS : A JOURNAL OF THE PSYCHONOMIC SOCIETY, INC 2000; 32:140-56. [PMID: 10758672 DOI: 10.3758/bf03200796] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
COGITAT is an automated hole board system that, following minimal experimental interventions, makes it possible to measure a variety of parameters associated with learning, memory, relearning, cognition, and cognitive shifts, but also changes in exploratory and sensorimotor performance in rodent models. The individual parameters--that is, overall exploratory activity, number of visits (deep in the hole) into or inspections of (at the upper surface) holes, number of baited, unbaited, or previously baited holes visited or inspected, reinspections of or revisits into any holes, number of pellets eaten, time to find pellets, serial order collection (without intermediate inspections or visits), and reference and working memory errors (visits, inspections, or total)--are obtained simultaneously, and the results are immediately available after the end of each experiment. The system appears to be well suited to neurophysiological, neuropharmacological, and gene-technological investigations in rodent models.
Collapse
Affiliation(s)
- C Heim
- Department of Neuropharmacology and -physiology, Max Planck Institute for Experimental Medicine, Göttingen, Germany
| | | | | | | | | | | |
Collapse
|
37
|
Fresta M, Puglisi G. Reduction of maturation phenomenon in cerebral ischemia with CDP-choline-loaded liposomes. Pharm Res 1999; 16:1843-9. [PMID: 10644072 DOI: 10.1023/a:1018999225435] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
PURPOSE Cerebral ischemia represents a serious therapeutic challenge. We investigated the therapeutic efficacy of CDP-choline-loaded liposomes against cerebral ischemia. The determination of post-ischemic brain recovery by EEG analysis was carried out to evaluate the effect of CDP-choline-loaded liposomes with respect to the free drug on the maturation of ischemic injury. METHODS Long-circulating unilamellar liposomes were prepared by a freeze and thaw procedure followed by an extrusion through polycarbonate membranes. Wistar rats were ischemized by bilateral clamping of the common carotid arteries. Free or liposomally entrapped drug was administered (20 mg/kg) just after ischemia and thereafter once a day for six days. Post-ischemic survival, neuronal membrane peroxidation and brain recovery (EEG analysis) were evaluated. RESULTS The post-ischemic reperfused rats treated with CDP-choline-loaded liposomes showed a higher survival rate than animals treated with the free drug. The delayed cerebral neurodegenerative injury due to an ischemic event, referred to as maturation phenomenon, was substantially reduced with the administration of the liposomal formulation. The liposomal carrier showed a marked protection against lipoperoxidative damage. CONCLUSIONS Liposomes ensured a rapid recovery of the damaged membranous structure of the neuronal cells, allowing a significant improvement of brain functionality. The reduction of the maturation phenomenon may probably be of particular importance in humans, where a fundamental problem is the quality of life after an ischemic event.
Collapse
Affiliation(s)
- M Fresta
- Department of Pharmaceutical Sciences, University of Catania, Italy.
| | | |
Collapse
|
38
|
Li F, Han SS, Tatlisumak T, Liu KF, Garcia JH, Sotak CH, Fisher M. Reversal of acute apparent diffusion coefficient abnormalities and delayed neuronal death following transient focal cerebral ischemia in rats. Ann Neurol 1999; 46:333-42. [PMID: 10482264 DOI: 10.1002/1531-8249(199909)46:3<333::aid-ana9>3.0.co;2-x] [Citation(s) in RCA: 114] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Twenty-two rats were subjected to 8, 15, 30, or 60 minutes of temporary middle cerebral artery occlusion (n = 5 per group) or sham occlusion (n = 2) in the magnetic resonance imaging unit. Diffusion-, perfusion-, and T2-weighted imaging were acquired before and during occlusion, and after reperfusion. A coregistration method was used to correlate the acute changes of the average apparent diffusion coefficient (ADCav) with the histology after 72 hours at the same topographic sites. The initially reduced ADCav values recovered completely in both the lateral caudoputamen and upper frontoparietal cortex in the 8-, 15-, and 30-minute groups, partially in the cortex, and not at all in the caudoputamen in the 60-minute group. The histology showed that the caudoputamen was either normal or had mild neuronal injury in the 8-minute group and invariably had some degree of neuronal death in the 15-, 30-, and 60-minute groups, whereas the cortex was either normal or had varying degrees of neuronal injury in all groups. No histological abnormalities were seen in the sham-operated rats. Our data suggest that acute ADCav reversal does not always predict tissue recovery from ischemic injury and that temporary focal ischemia for even 8-minute duration can cause delayed neuronal death that is more severe in the caudoputamen where the initial ADCav decline was greater than in the cortex.
Collapse
Affiliation(s)
- F Li
- Department of Neurology, UMass Memorial Health Care, Worcester, MA 01605, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Fujioka M, Taoka T, Hiramatsu KI, Sakaguchi S, Sakaki T. Delayed ischemic hyperintensity on T1-weighted MRI in the caudoputamen and cerebral cortex of humans after spectacular shrinking deficit. Stroke 1999; 30:1038-42. [PMID: 10229741 DOI: 10.1161/01.str.30.5.1038] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Transient internal carotid artery (ICA)-middle cerebral artery (MCA) occlusion caused by cardiogenic embolus can lead to spectacular shrinking deficit (SSD): sudden hemispheric stroke syndrome followed by rapid improvement. The aim of this study was to investigate sequential neuroradiological changes in the brains of patients after SSD compared with those after brief cardiac arrest and hypoglycemia, which we previously studied with the same methods. METHODS We serially studied CT scans and MR images obtained at 1.5 T in 4 patients with SSD. All 4 patients suffered from transient neurological deficits due to cardiogenic embolus in ICA-MCA. The symptoms began to disappear from 25 to 50 minutes after onset. RESULTS Repeated CT scans demonstrated no abnormal findings in the affected cerebral hemisphere in 3 of the 4 patients and a small cortical infarct in the remaining 1. In each patient, repeated MRI between day 7 and month 23 after stroke showed basal ganglionic and cortical lesions. These lesions were hyperintense on T1-weighted and relatively hypointense on T2-weighted imaging. These ischemic lesions of hyperintensity on T1-weighted MRI subsided with time. CONCLUSIONS Transient ICA-MCA occlusion leading to SSD produces a specific ischemic change with delayed onset in the basal ganglia and cerebral cortex in humans on MRI but not CT scans. We speculate that the lesions represent incomplete ischemic injury, including selective neuronal death, proliferation of glial cells, paramagnetic substance deposition, and/or lipid accumulation. Unlike brief cardiac arrest or hypoglycemia, the localized lesions on MRI of patients after SSD seem to be incomplete and to differ from infarction or hemorrhage.
Collapse
Affiliation(s)
- M Fujioka
- Department of Neurosurgery, Nara Medical University, Nara, Japan.
| | | | | | | | | |
Collapse
|
40
|
Affiliation(s)
- W D Dietrich
- Department of Neurology, University of Miami of School of Medicine, Florida 33101, USA
| |
Collapse
|
41
|
Garcia JH, Liu KF, Ye ZR, Gutierrez JA. Incomplete infarct and delayed neuronal death after transient middle cerebral artery occlusion in rats. Stroke 1997; 28:2303-9; discussion 2310. [PMID: 9368580 DOI: 10.1161/01.str.28.11.2303] [Citation(s) in RCA: 113] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND PURPOSE The clinical syndrome of transient ischemic attacks is accompanied in a significant percentage of patients by brain lesions or neuroimaging abnormalities whose structural counterparts have not been defined. The objective of this study was to analyze, in an experimental model of short-term (< 25 minutes) focal ischemia and long-term (< or = 28 days) reperfusion, the extent and nature of the structural abnormalities affecting neurons and glia located within the territory of the transiently occluded artery. METHODS Adult Wistar rats (n = 121) had the origin of one middle cerebral artery (MCA) occluded with a nylon monofilament for periods of 10 to 25 minutes. Experiments of transient MCA occlusion were terminated at variable periods ranging from 1 day to 4 weeks. Control experiments consisted of (1) MCA occlusion without reperfusion (n = 7) lasting 7 to 14 days and (2) sham operations (n = 2) followed by 1- to 4-day survival. After in situ fixation, brain specimens were serially sectioned and subjected to detailed morphometric evaluations utilizing light and electron microscopes. The statistical method used to evaluate the results was based on ANOVA followed by Bonferroni's corrected t test and Student's t test comparisons. RESULTS Brain lesions were not detectable in the sham-operated controls. All brains with permanent MCA occlusion (7 to 14 days) had large infarctions with abundant macrophage infiltration and early cavitation. Forty-five (37%) of the experiments involving transient MCA occlusion had no detectable brain lesions after 4 weeks. Selective neuronal necrosis was found in 76 of 121 rats (63%) with transient MCA occlusion. Neuronal necrosis always involved the striatum, and in 29% of the brains with ischemic injury, necrosis also included a short segment of the cortex. In the striatum, the length of the arterial occlusion was the main determinant of the number of necrotic neurons (20 minutes [22.6 +/- 19] is worse than 10 minutes [4.9 +/- 7]) (P < .0001). In the cortex, the length of reperfusion determined the number of necrotic neurons appearing in layer 3. Experiments with reperfusion of 4 to 7 days' duration yielded more necrotic neurons per microscopic field (2.02 +/- 3) than those lasting fewer days (0.04 +/- 0.1) (P < .05). The histological features of these lesions underwent continuous change until the end of the fourth week, at which time necrotic neurons were still visible both in the striatum and in the cortex. CONCLUSIONS Arterial occlusions of short duration (< 25 minutes) produced, in 76 of 121 experiments (63%), brain lesions characterized by selective neuronal necrosis and various glial responses (or incomplete infarction). This lesion is entirely different from the pannecrosis/cavitation typical of an infarction that appears 3 to 4 days after a prolonged arterial occlusion. Delayed neuronal necrosis, secondary to a transient arterial occlusion or increasing numbers of necrotic neurons in experiments with variable periods of reperfusion, was a response observed only at a predictable segment of the frontoparietal cortex.
Collapse
Affiliation(s)
- J H Garcia
- Department of Pathology (Neuropathology), Henry Ford Hospital, Detroit, Mich. 48202, USA
| | | | | | | |
Collapse
|
42
|
Abstract
The influence of post-insult temperature modulation on ischemic injury in immature brain was studied in 7-day-old rats that underwent a unilateral carotid artery ligation followed by exposure to hypoxia in 8% oxygen at an ambient temperature of 36.5 degrees C. After the hypoxic exposure, the animals were separated into three groups and placed for 3 h in temperature-controlled incubators set at 32 degrees C, 35 degrees C, and 38 degrees C. In Study 1, the influence of post-insult temperature modulation was assessed after graded cerebral hypoxic-ischemic injury. Brain damage was assessed 1 week after the insult by comparison of wet weights in the cerebral hemispheres ipsilateral and contralateral to the carotid artery ligation. Rectal temperatures of the animals significantly correlated with extent of brain injury after 60 min (Spearman correlation coefficient, p = 0.44, P = 0.005) and 90 min (p = 0.46, P = 0.004) but not 120 min of hypoxia (p = 0.18, P = 0.46). In Study 2, animals were exposed to 75 min hypoxia, and injury was assessed morphometrically and histologically at 1 and 4 weeks after the injury. Rectal temperatures significantly correlated with the extent of ischemic injury in the cerebral cortex (p = 0.3, P = 0.046) and striatum (p = 0.3, P = 0.048) at 1 week, but not 4 weeks, after the insult. The findings indicate that post-insult hypothermia delayed the expression of mild to moderate brain damage by more than a week, after which the damage was as severe as in normothermic animals. The results indicate that the events that determine the final expression of a neonatal hypoxic-ischemic insult can be extended over a long interval by post-insult hypothermia.
Collapse
Affiliation(s)
- W H Trescher
- Department of Neurology, Johns Hopkins University School of Medicine, Kennedy Krieger Research Institute, Baltimore, MD 21205, USA.
| | | | | |
Collapse
|
43
|
Yamasaki Y, Matsuo Y, Zagorski J, Matsuura N, Onodera H, Itoyama Y, Kogure K. New therapeutic possibility of blocking cytokine-induced neutrophil chemoattractant on transient ischemic brain damage in rats. Brain Res 1997; 759:103-11. [PMID: 9219868 DOI: 10.1016/s0006-8993(97)00251-5] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Earlier we indicated that neutrophilic invasion into cerebral parenchyma is an important step in rat cerebral ischemia-reperfusion injury and the production of chemotactic factors, cytokine-induced neutrophil chemoattractant (CINC) precede the neutrophilic invasion. The aim of the present study was to evaluate the role of CINC production and the therapeutic possibility of blocking CINC activity in the transient ischemic brain damage in rats. Focal transient ischemia was produced by intraluminal occlusion of the right middle cerebral artery for 60 min. An enzyme immunoassay was used to measure the brain concentration of CINC and myeloperoxidase activity in ischemic areas was measured as a marker of neutrophilic accumulation. An immunohistochemical staining technique was used to detect the immunopositive cells for anti-CINC antibody. Further, application of anti-CINC antibody or anti-neutrophil antibody to rats was used to evaluate the role of CINC production. In ischemic areas, CINC production was detected and peaked 12 h after reperfusion, which followed 60 min of ischemia. Intraperitoneal injection of anti-neutrophil antibody 24 h before and immediately after reperfusion significantly reduced the brain water content and partially reduced the CINC production in ischemic areas. Further, immunohistochemical staining showed that anti-CINC antibody was found on the endothelial surface of venules and on parts of neutrophils that had invaded the ischemic area 6 to 24 h after reperfusion. Also, treatment with anti-CINC antibody reduced ischemic edema formation 24 h after reperfusion and the size of infarction areas 7 days after reperfusion. It thus appears that CINC, mainly produced by endothelium activated by factors released from neutrophils, plays an important role in ischemic brain damage. Furthermore, the blocking of CINC activity with antibody suggests an immuno-therapeutic approach to the treatment of stroke patients.
Collapse
Affiliation(s)
- Y Yamasaki
- Hanno Research Center, Taiho Pharmaceutical Co. Ltd., Hanno City, Saitama, Japan
| | | | | | | | | | | | | |
Collapse
|
44
|
Yamada K, Goto S, Yoshikawa M, Okamura A, Nagahiro S, Ushio Y. Intraventricular administration of GABAAreceptor agonist muscimol attenuates the exo-focal change of the substantia nigra neurons following transient middle cerebral artery occlusion in rats. Neuropathology 1997. [DOI: 10.1111/j.1440-1789.1997.tb00021.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
45
|
Nelson A, Sowinski P, Hodges H. Differential effects of global ischemia on delayed matching- and non-matching-to-position tasks in the water maze and Skinner box. Neurobiol Learn Mem 1997; 67:228-47. [PMID: 9159761 DOI: 10.1006/nlme.1996.3758] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In order to assess effects of global ischemia in tasks of spatial learning and working memory, male Wistar rats were subjected to four vessel occlusion (4 VO) for periods of 5, 10, and 20 min and compared with sham-operated controls over four test phases, from 6 to 54 weeks after surgery. Rats were assessed on acquisition in the water maze, a task that is sensitive to ischemic impairments, before testing in Skinner box and water maze working memory tasks, which both require the short-term storage of information, but make different demands on spatial information processing. Phases 1 and 3 assessed spatial learning in a standard water maze procedure (12 and 10 training days, 2 trials/day with a 10-min intertrial interval: ITI). Phase 2 involved training and testing in delayed non-matching-to-position task in the Skinner box, with delays of 2-10 s between the information and choice stages. Phase 4 examined working memory in a water maze delayed matching-to-position task with 4 trials/day, an ITI of 30 s, and a novel platform position on each day. Ischemic rats showed duration-related impairments in water maze acquisition and working memory, but not in the less spatially demanding Skinner box task. Since water maze acquisition deficits were seen both before and after testing in the Skinner box the lack of effect cannot be attributed to time or to prior training. Ischemic deficits were more marked in Phase 3 than in Phase 1 of acquisition, suggesting that impairment may be progressive. Histological assessment showed that cell loss was largely confined to the hippocampal CA1 field and was linearly related to duration of occlusion. At the maximal level of loss (5.7 mm before the interaural line) the 20-min group showed 90% loss, the 10-min group 60% loss, and the 5-min group, which did not differ from controls, less than 10% loss. Only the 20-min group showed significant damage beyond the CA1 field, ranging from 30-40% loss in the CA3 field to 5% loss in one striatal area. No cortical damage was seen. The extent of CA1 cell loss correlated modestly with water maze acquisition (Phase 3) and working memory scores, but not with trials to criterion in the Skinner box task. There were significant correlations between different measures both within and between water maze tasks, but not Skinner box tasks, suggesting that the two types of procedure engaged different cognitive processes. The results indicate that the intrahippocampal damage induced by 4 VO impaired tasks which required processing of allocentric spatial information, but did not impair the storage of limited spatial information in working memory.
Collapse
Affiliation(s)
- A Nelson
- Department of Psychology, Institute of Psychiatry, London, United Kingdom
| | | | | |
Collapse
|
46
|
Zoli M, Grimaldi R, Ferrari R, Zini I, Agnati LF. Short- and long-term changes in striatal neurons and astroglia after transient forebrain ischemia in rats. Stroke 1997; 28:1049-58; discussion 1059. [PMID: 9158649 DOI: 10.1161/01.str.28.5.1049] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND PURPOSE The striatum is one of the regions most sensitive to transient forebrain ischemia. After 30-minute ischemia, areas of massive neuronal degeneration are clearly detectable a few hours after the insult and attain their maximal extension 24 hours after the insult. However, for most cellular and neurochemical parameters it is not known whether some recovery occurs at later times. We examined certain cell populations in the caudate putamen at different times after transient ischemia. METHODS Adult male Sprague-Dawley rats were subjected to 30-minute forebrain ischemia (four-vessel occlusion model). Six experimental groups were considered: control animals and ischemic animals killed 4 hours, 1 day, 7 days, 40 days, and 8 months after reperfusion. Three striatal cell populations were examined by means of immunocytochemistry coupled to computer-assisted image analysis: vulnerable medium spiny neurons, resistant aspiny neurons, and reactive astrocytes, labeled for their content of dopamine- and cAMP-regulated phosphoprotein mr32 (DARPP-32), somatostatin and neuropeptide Y, and glial fibrillary acidic protein, respectively. RESULTS (1) The area containing DARPP-32 immunoreactive neurons was markedly decreased (15% to 20% of control caudate putamen area) at 1 day after reperfusion and partially recovered at the following times (40% to 50% at 7 days and 50% to 60% at 40 days and 8 months after reperfusion). (2) The appearance of reactive astrocytes was precocious (4 hours to 1 day after ischemia) in the medial caudate putamen, the region in which DARPP-32 recovered within 40 days after ischemia, and late (7 to 40 days after ischemia) in the lateral caudate putamen, where no DARPP-32 recovery was detected. (3) Neuropeptide Y/somatostatin-containing neurons resisted the ischemic insult and could be detected in areas devoid of DARPP-32 immunoreactive neurons as long as 8 months after reperfusion. CONCLUSIONS The present results show a marked recovery of DARPP-32-positive neurons within 40 days after 30-minute forebrain ischemia in the medial, but not the lateral, caudate putamen. Medial caudate putamen also contains a high density of reactive astrocytes on the first day after ischemia, suggesting that astrocytic support has an important role in the spontaneous recovery of ischemic neurons.
Collapse
Affiliation(s)
- M Zoli
- Department of Biomedical Sciences, University of Modena, Italy
| | | | | | | | | |
Collapse
|
47
|
Heiss WD, Graf R, Löttgen J, Ohta K, Fujita T, Wagner R, Grond M, Weinhard K. Repeat positron emission tomographic studies in transient middle cerebral artery occlusion in cats: residual perfusion and efficacy of postischemic reperfusion. J Cereb Blood Flow Metab 1997; 17:388-400. [PMID: 9143221 DOI: 10.1097/00004647-199704000-00004] [Citation(s) in RCA: 97] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The wider clinical acceptance of thrombolytic therapy for ischemic stroke has focused more attention on experimental models of reversible focal ischemia. Such models enable the study of the effect of ischemia of various durations and of reperfusion on the development of infarctions. We used high-resolution positron emission tomography (PET) to assess cerebral blood flow (CBF), cerebral metabolic rate of oxygen (CMRO2), oxygen extraction fraction (OEF), and cerebral metabolic rate of glucose (CMRglc) before, during, and up to 24 h after middle cerebral artery occlusion (MCAO) in cats. After determination of resting values, the MCA was occluded by a transorbital device. The MCA was reopened after 30 min in five, after 60 min in 11, and after 120 min in two cats. Whereas all cats survived 30-min MCAO, six died after 60-min and one after 120-min MCAO during 6-20 h of reperfusion. In those cats surviving the first day, infarct size was determined on serial histologic sections. The arterial occlusion immediately reduced CBF in the MCA territory to < 40% of control, while CMRO2 was less affected, causing an increase in OEF. Whereas in the cats surviving 24 h of reperfusion after 60- and 120-min MCAO, OEF remained elevated throughout the ischemic episode, the initial OEF increase had already disappeared during the later period of ischemia in those cats that died during the reperfusion period. After 30-min MCAO, the reperfusion period was characterized by a transient reactive hyperemia and fast normalization of CBF, CMRO2, and CMRglc, and no or only small infarcts in the deep nuclei were found in histology. After 60- and 120-min MCAO, the extent of hyperperfusion was related to the severity of ischemia, decreased CMRO2 and CMRglc persisted, and cortical/subcortical infarcts of varying sizes developed. A clear difference was found in the flow/metabolic pattern between surviving and dying cats: In cats dying during the observation period, extended postischemic hyperperfusion accompanied large defects in CMRO2 and CMRglc, large infarcts developed, and intracranial pressure increased fatally. In those surviving the day after MCAO, increased OEF persisted over the ischemic episode, postischemic hyperperfusion was less severe and shorter, and the perfusional and metabolic defects as well as the final infarcts were smaller. These results stress the importance of the severity of ischemia for the further course after reperfusion and help to explain the diverging outcome after thrombolysis, where a relation between the residual flow and the effectiveness of reperfusion was also observed.
Collapse
Affiliation(s)
- W D Heiss
- Max Planck Institute for Neurological Research, Cologue, Germany
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Hodges H, Nelson A, Virley D, Kershaw TR, Sinden JD. Cognitive deficits induced by global cerebral ischaemia: prospects for transplant therapy. Pharmacol Biochem Behav 1997; 56:763-80. [PMID: 9130304 DOI: 10.1016/s0091-3057(96)00424-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Global ischaemia induced by interruption of cerebral blood flow results in damage to vulnerable cells, notably in the CA1 and hilar hippocampal fields, and is frequently associated with memory deficits. This review examines cognitive deficits that occur in animal models of global ischaemia in rats and monkeys, the extent to which these deficits are associated with CA1 cell loss, and the evidence for functional recovery following transplants of foetal CA1 cells and grafts of conditionally immortalised precursor cells. In rats, impairments are seen most consistently in tasks of spatial learning and spatial working memory dependent on use of allocentric environmental cues. In monkeys, ischaemic deficits have been shown to a moderate extent in delayed object recognition tasks, but animals with a selective excitotoxic CA1 lesion show a profound impairment in conditional discrimination tasks, suggesting that these may be a more sensitive measure of ischaemic impairments. Several studies have reported correlational links between the extent of CA1 cell loss following two or four vessel occlusion (2 VO, 4 VO) in rats and behavioural impairments, but recent findings indicate that at intermediate levels of damage these relationships are weak and variable, and emerge clearly only when animals with maximal CA1 cell loss are included, suggesting that the deficits involve more than damage to the CA1 field. Nevertheless, ischaemic rats and CA1-lesioned marmosets with grafts of foetal CA1 cells show substantial improvements; in rats these are not found with grafts from other hippocampal fields. Conditionally immortalised cell lines and trophic grafts are currently being assessed for their functional potential in animal models, because clinical use of foetal cells will not be practicable. Recent findings suggest that an expanded population of neuroepithelial cells derived from the conditionally immortalised H-2Kb-tsA58 transgenic mouse improve spatial learning as effectively as CA1 foetal grafts in rats subjected to 4 VO, and clonal lines from the same source show similar promise. Lines derived from precursor cells have the potential to develop into different types of cell (neuronal or glial) depending on signals from the host brain. These cell lines may therefore have the capacity to repair damaged host circuits more precisely than is possible with foetal grafts, and offer a promising, approach both to functional recovery and to elucidating graft-host interactions.
Collapse
Affiliation(s)
- H Hodges
- Department of Psychology, Institute of Psychiatry, London, UK
| | | | | | | | | |
Collapse
|
49
|
Schmidt-Kastner R, Fliss H, Hakim AM. Subtle neuronal death in striatum after short forebrain ischemia in rats detected by in situ end-labeling for DNA damage. Stroke 1997; 28:163-9; discussion 169-70. [PMID: 8996506 DOI: 10.1161/01.str.28.1.163] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND AND PURPOSE Neuronal cell death after global brain ischemia occurs predominantly by necrosis, whereas only a minor fraction of cell death may occur through apoptosis. Brief or moderate insults are thought to facilitate apoptosis, which is associated with DNA fragmentation. After 10 minutes of four-vessel occlusion in rats, conventional neuropathological analysis shows neuronal cell death in hippocampal CA1 but not in the striatum. Thus, we compared hippocampus and striatum for occurrence of cells with DNA fragmentation. METHODS A brief insult of 10 minutes of forebrain ischemia was induced in rats using four-vessel occlusion, and groups of brains were studied at 1, 3, 6, and 12 hours and at 1, 3, and 7 days after ischemia. In situ end-labeling (ISEL) was used to detect neurons undergoing DNA fragmentation. The hippocampal CA1 area was compared with the striatum. Conventional staining and immunohistochemical markers served to exclude ischemic neuronal cell death in the striatum. RESULTS Hippocampal CA1 neurons were ISEL-positive by 3 days after ischemia. In contrast, positive cells became evident in the striatum between 3 hours to 3 days after ischemia. The ISEL-positive cells were scattered throughout the striatum with a preference for the dorsomedial areas and accounted for about 0.2% of the neurons per striatal area at 1 day. Conventional staining and immunohistochemical markers failed to reveal areas of overt cell damage in the striatum. CONCLUSIONS The scattered cell damage in the striatum after brief forebrain ischemia suggests the occurrence of an apoptotic process. The striatum therefore may be prone to subtle cell death due to metabolic insults.
Collapse
Affiliation(s)
- R Schmidt-Kastner
- Neuroscience Research Institute, Faculty of Medicine, University of Ottawa, Canada
| | | | | |
Collapse
|
50
|
Coimbra C, Drake M, Boris-Möller F, Wieloch T. Long-lasting neuroprotective effect of postischemic hypothermia and treatment with an anti-inflammatory/antipyretic drug. Evidence for chronic encephalopathic processes following ischemia. Stroke 1996; 27:1578-85. [PMID: 8784133 DOI: 10.1161/01.str.27.9.1578] [Citation(s) in RCA: 161] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND AND PURPOSE It has been recognized that postischemic pharmacological interventions may delay the evolution of neuronal damage rather than provide long-lasting neuroprotection. Also, fever complicates recovery after stroke in humans. Here we report the effects of late postischemic treatment with hypothermia and an antipyretic/anti-inflammatory drug, dipyrone, on cell damage at 1 week and 2 months of survival. METHODS Rats were subjected to 10 minutes of forebrain ischemia. Hypothermia (33 degrees C) was induced at 2 hours of recovery and maintained for 7 hours. Dipyrone (100 mg.kg-1IP) was given every 3 hours from 14 to 72 hours of recovery. Temperature was measured every 6 hours for 60 days. Neuronal damage was assessed at 7 days and 2 months of recovery. RESULTS From 17 to 72 hours of recovery, a period of hyperthermia was observed, which dipyrone abolished but postischemic hypothermia treatment did not. Dipyrone treatment diminished neuronal damage by 43% at 7 days, and at 2 months of survival, a minor (16%) protection was seen. Postischemic hypothermia treatment alone delayed neuronal damage. In contrast, combined treatment of hypothermia followed by dipyrone markedly diminished neuronal damage by more than 50% at both 7 days and 2 months of recovery. CONCLUSIONS Neuronal degeneration may be ongoing for months after a transient ischemic insult, and prolonged protective measures need to be instituted for long-lasting neuroprotective effects. Hyperthermia during recovery worsens ischemic damage, and processes associated with inflammation may contribute to the development of neuronal damage. An early and extended period of postischemic hypothermia provides a powerful and long-lasting protection if followed by treatment with anti-inflammatory/ antipyretic drug.
Collapse
Affiliation(s)
- C Coimbra
- Laboratory for Experimental Brain Research, Lund University, Sweden
| | | | | | | |
Collapse
|