Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 21
1.
Int J Stroke ; 13(4): 379-390, 2018 06.
Article En | MEDLINE | ID: mdl-28776456

Background Cornu ammonis 3 (CA3) hippocampal neurons are resistant to global ischemia, whereas cornu ammonis (CA1) 1 neurons are vulnerable. Hamartin expression in CA3 neurons mediates this endogenous resistance via productive autophagy. Neurons lacking hamartin demonstrate exacerbated endoplasmic reticulum stress and increased cell death. We investigated endoplasmic reticulum stress responses in CA1 and CA3 regions following global cerebral ischemia, and whether pharmacological modulation of endoplasmic reticulum stress or autophagy altered neuronal viability . Methods In vivo: male Wistar rats underwent sham or 10 min of transient global cerebral ischemia. CA1 and CA3 areas were microdissected and endoplasmic reticulum stress protein expression quantified at 3 h and 12 h of reperfusion. In vitro: primary neuronal cultures (E18 Wistar rat embryos) were exposed to 2 h of oxygen and glucose deprivation or normoxia in the presence of an endoplasmic reticulum stress inducer (thapsigargin or tunicamycin), an endoplasmic reticulum stress inhibitor (salubrinal or 4-phenylbutyric acid), an autophagy inducer ([4'-(N-diethylamino) butyl]-2-chlorophenoxazine (10-NCP)) or autophagy inhibitor (3-methyladenine). Results In vivo, decreased endoplasmic reticulum stress protein expression (phospho-eIF2α and ATF4) was observed at 3 h of reperfusion in CA3 neurons following ischemia, and increased in CA1 neurons at 12 h of reperfusion. In vitro, endoplasmic reticulum stress inducers and high doses of the endoplasmic reticulum stress inhibitors also increased cell death. Both induction and inhibition of autophagy also increased cell death. Conclusion Endoplasmic reticulum stress is associated with neuronal cell death following ischemia. Neither reduction of endoplasmic reticulum stress nor induction of autophagy demonstrated neuroprotection in vitro, highlighting their complex role in neuronal biology following ischemia.


Brain Ischemia/physiopathology , Endoplasmic Reticulum Stress/physiology , Animals , CA1 Region, Hippocampal/physiology , CA3 Region, Hippocampal/physiology , Cell Death/physiology , Cells, Cultured , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Hypoglycemia/physiopathology , Hypoxia/physiopathology , Male , Neurons/physiology , Neuroprotective Agents/pharmacology , Rats, Wistar , Thapsigargin/pharmacology , Tuberous Sclerosis Complex 1 Protein/metabolism , Tunicamycin/pharmacology
2.
Int J Stroke ; 12(6): 615-622, 2017 08.
Article En | MEDLINE | ID: mdl-27899743

Background The Alberta Stroke Program Early Computed Tomography Score (ASPECTS) is an established 10-point quantitative topographic computed tomography scan score to assess early ischemic changes. We performed a non-inferiority trial between the e-ASPECTS software and neuroradiologists in scoring ASPECTS on non-contrast enhanced computed tomography images of acute ischemic stroke patients. Methods In this multicenter study, e-ASPECTS and three independent neuroradiologists retrospectively and blindly assessed baseline non-contrast enhanced computed tomography images of 132 patients with acute anterior circulation ischemic stroke. Follow-up scans served as ground truth to determine the definite area of infarction. Sensitivity, specificity, and accuracy for region- and score-based analysis, receiver-operating characteristic curves, Bland-Altman plots and Matthews correlation coefficients relative to the ground truth were calculated and comparisons were made between neuroradiologists and different pre-specified e-ASPECTS operating points. The non-inferiority margin was set to 10% for both sensitivity and specificity on region-based analysis. Results In total 2640 (132 patients × 20 regions per patient) ASPECTS regions were scored. Mean time from onset to baseline computed tomography was 146 ± 124 min and median NIH Stroke Scale (NIHSS) was 11 (6-17, interquartile range). Median ASPECTS for ground truth on follow-up imaging was 8 (6.5-9, interquartile range). In the region-based analysis, two e-ASPECTS operating points (sensitivity, specificity, and accuracy of 44%, 93%, 87% and 44%, 91%, 85%) were statistically non-inferior to all three neuroradiologists (all p-values <0.003). Both Matthews correlation coefficients for e-ASPECTS were higher (0.36 and 0.34) than those of all neuroradiologists (0.32, 0.31, and 0.3). Conclusions e-ASPECTS was non-inferior to three neuroradiologists in scoring ASPECTS on non-contrast enhanced computed tomography images of acute stroke patients.


Brain Ischemia/diagnostic imaging , Software , Stroke/diagnostic imaging , Adult , Aged , Brain Ischemia/therapy , Female , Humans , Male , Middle Aged , ROC Curve , Retrospective Studies , Stroke/therapy , Tomography, Emission-Computed, Single-Photon
3.
J Neurochem ; 131(2): 177-89, 2014 Oct.
Article En | MEDLINE | ID: mdl-24974727

This study investigated the effects of 2-(1-chloro-4-hydroxyisoquinoline-3-carboxamido) acetic acid (IOX3), a selective small molecule inhibitor of hypoxia-inducible factor (HIF) prolyl hydroxylases, on mouse brains subject to transient focal cerebral ischaemia. Male, 8- to 12-week-old C57/B6 mice were subjected to 45 min of middle cerebral artery occlusion (MCAO) either immediately or 24 h after receiving IOX3. Mice receiving IOX3 at 20 mg/kg 24 h prior to the MCAO had better neuroscores and smaller blood-brain barrier (BBB) disruption and infarct volumes than mice receiving the vehicle, whereas those having IOX3 at 60 mg/kg showed no significant changes. IOX3 treatment immediately before MCAO was not neuroprotective. IOX3 up-regulated HIF-1α, and increased EPO expression in mouse brains. In an in vitro BBB model (RBE4 cell line), IOX3 up-regulated HIF-1α and delocalized ZO-1. Pre-treating IOX3 on RBE4 cells 24 h before oxygen-glucose deprivation had a protective effect on endothelial barrier preservation with ZO-1 being better localized, while immediate IOX3 treatment did not. Our study suggests that HIF stabilization with IOX3 before cerebral ischaemia is neuroprotective partially because of BBB protection, while immediate application could be detrimental. These results provide information for studies aimed at the therapeutic activation of HIF pathway for neurovascular protection from cerebral ischaemia. We show that IOX3, a selective small molecule (280.66 Da) HIF prolyl hydroxylase inhibitor, could up-regulate HIF-1α and increase erythropoietin expression in mice. We further demonstrate that HIF stabilization with IOX3 before cerebral ischaemia is neuroprotective partially because of blood-brain barrier (BBB) protection, while immediate application is detrimental both in vivo and in vitro. These findings provide new insights into the role of HIF stabilization in ischaemic stroke.


Brain Ischemia/metabolism , Brain Ischemia/prevention & control , Glycine/analogs & derivatives , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Isoquinolines/administration & dosage , Neuroprotective Agents/administration & dosage , Animals , Cells, Cultured , Glycine/administration & dosage , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice , Mice, Inbred C57BL , Rats
4.
JAMA Neurol ; 71(5): 634-9, 2014 May.
Article En | MEDLINE | ID: mdl-24590416

IMPORTANCE: Preclinical stroke research has had a remarkably low translational success rate, and the clinical need for novel neuroprotective therapeutics has gone largely unmet, especially in light of the severe underuse of thrombolysis in acute ischemic stroke. OBJECTIVE: In this review, we aim to provide a brief overview of the commonly used stroke models, their merits and shortcomings, and how these have contributed to translational failures. We review some recent developments in preclinical stroke, providing examples of how improved study quality and the use of novel methods can facilitate translation into the clinical setting. EVIDENCE REVIEW: This is a narrative review of ischemic stroke neuroprotection based on electronic database searches, references of previous publications, and personal libraries. FINDINGS: The stroke research community has not been complacent in its response to criticism: preclinical stroke studies now demonstrate considerable rigor, standardization, and emphasis on minimization of experimenter bias. In addition, numerous innovative methods and strategies are providing novel avenues for investigating neuroprotection, as well as more extensive characterization of established models. CONCLUSIONS AND RELEVANCE: The improvements in preclinical stroke models and methods will make stroke research a good example for preclinical medicine, in general, and will hopefully instill greater confidence in the clinical community regarding which compounds are worthy of further investigation in a clinical setting.


Brain Ischemia/prevention & control , Disease Models, Animal , Neuroprotective Agents/administration & dosage , Stroke/prevention & control , Translational Research, Biomedical/trends , Animals , Brain Ischemia/diagnosis , Humans , Stroke/diagnosis
5.
Methods Mol Biol ; 1135: 111-20, 2014.
Article En | MEDLINE | ID: mdl-24510859

The four-vessel occlusion (4-VO) method of global forebrain cerebral ischemia mimics the human clinical condition of cardiac arrest. It results in selective neuronal damage and is a useful experimental system to dissect underlying mechanisms behind ischemic phenomena such as the differential susceptibility of CA1 compared to the CA3 region of the hippocampus. It also provides a "proof-of-principle" system for testing out potential agents for neuroprotection.


Brain Ischemia/etiology , Disease Models, Animal , Heart Arrest/complications , Animals , Male , Rats , Rats, Wistar
6.
Int J Stroke ; 8(6): 449-50, 2013 Aug.
Article En | MEDLINE | ID: mdl-23879750

Tuberous sclerosis complex 1 (hamartin) is an effective endogenous neuroprotectant. Understanding the endogenous mechanism for neuroprotection mediated by hamartin may afford a novel approach to effective treatment of neurological diseases such as stroke, neurodegenerative diseases, and epilepsy, with possible applications to nonneurological conditions.


Epilepsy/metabolism , Neurodegenerative Diseases/metabolism , Stroke/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Humans , Tuberous Sclerosis Complex 1 Protein
7.
Nat Med ; 19(3): 351-7, 2013 Mar.
Article En | MEDLINE | ID: mdl-23435171

Previous attempts to identify neuroprotective targets by studying the ischemic cascade and devising ways to suppress it have failed to translate to efficacious therapies for acute ischemic stroke. We hypothesized that studying the molecular determinants of endogenous neuroprotection in two well-established paradigms, the resistance of CA3 hippocampal neurons to global ischemia and the tolerance conferred by ischemic preconditioning (IPC), would reveal new neuroprotective targets. We found that the product of the tuberous sclerosis complex 1 gene (TSC1), hamartin, is selectively induced by ischemia in hippocampal CA3 neurons. In CA1 neurons, hamartin was unaffected by ischemia but was upregulated by IPC preceding ischemia, which protects the otherwise vulnerable CA1 cells. Suppression of hamartin expression with TSC1 shRNA viral vectors both in vitro and in vivo increased the vulnerability of neurons to cell death following oxygen glucose deprivation (OGD) and ischemia. In vivo, suppression of TSC1 expression increased locomotor activity and decreased habituation in a hippocampal-dependent task. Overexpression of hamartin increased resistance to OGD by inducing productive autophagy through an mTORC1-dependent mechanism.


Autophagy , CA1 Region, Hippocampal/physiology , CA3 Region, Hippocampal/physiology , Hypoxia-Ischemia, Brain/prevention & control , Neuroprotective Agents/metabolism , Tumor Suppressor Proteins/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Autophagy/drug effects , CA1 Region, Hippocampal/metabolism , CA3 Region, Hippocampal/metabolism , Cells, Cultured , Hypoxia , Hypoxia-Ischemia, Brain/metabolism , Ischemic Preconditioning , Male , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Prosencephalon/blood supply , Prosencephalon/injuries , Proteins/antagonists & inhibitors , Proteins/metabolism , RNA Interference , RNA, Small Interfering , Rats , Rats, Wistar , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Tuberous Sclerosis Complex 1 Protein , Tumor Suppressor Proteins/genetics
8.
J Neurochem ; 123 Suppl 2: 98-107, 2012 Nov.
Article En | MEDLINE | ID: mdl-23050647

Diphenyleneiodonium (DPI), a NADPH oxidase inhibitor, reduces production of reactive oxygen species (ROS) and confers neuroprotection to focal cerebral ischemia. Our objective was to investigate whether the neuroprotective action of DPI extends to averting the immune response. DPI-induced gene changes were analyzed by microarray analysis from rat brains subjected to 90 min of middle cerebral artery occlusion, treated with NaCl (ischemia), dimethylsulfoxide (DMSO), or DMSO and DPI (DPI), and reperfused for 48 h. The genomic expression profile was compared between groups using ingenuity pathway analysis at the pathway and network level. DPI selectively up-regulated 23 genes and down-regulated 75 genes more than twofold compared with both DMSO and ischemia. It significantly suppressed inducible nitric oxide synthase signaling and increased the expression of methionine adenosyltransferasesynthetase 2A and adenosylmethionine decarboxylase 1 genes, which are involved in increasing the production of the antioxidant glutathione. The most significantly affected gene network comprised genes implicated in the inflammatory response with an expression change indicating an overall suppression. Both integrin- and interleukin-17A-signaling pathways were also significantly associated and suppressed. In conclusion, the neuroprotective effects of DPI are mediated not only by suppressing ischemia-triggered oxidative stress but also by limiting leukocyte migration and infiltration.


Anti-Inflammatory Agents/therapeutic use , Gene Expression Regulation/drug effects , Infarction, Middle Cerebral Artery/complications , Inflammation/drug therapy , Inflammation/etiology , Onium Compounds/therapeutic use , Analysis of Variance , Animals , Anti-Inflammatory Agents/pharmacology , Cytokines/genetics , Cytokines/metabolism , Gene Expression Profiling , Integrins/genetics , Integrins/metabolism , Linear Models , Magnetic Resonance Imaging , Male , Nervous System Diseases/etiology , Nervous System Diseases/metabolism , Nervous System Diseases/prevention & control , Oligonucleotide Array Sequence Analysis , Oxidative Stress/drug effects , RNA, Messenger/metabolism , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/genetics
9.
J Physiol ; 590(16): 4079-91, 2012 Aug 15.
Article En | MEDLINE | ID: mdl-22615432

This study investigated the function of each of the hypoxia inducible factor (HIF) prolyl-4-hydroxylase enzymes (PHD1­3) in the first 24 h following transient focal cerebral ischaemia by using mice with each isoform genetically suppressed. Male, 8- to 12-week old PHD1−/−, PHD2+/− and PHD3−/− mice and their wild-type (WT) littermate were subjected to 45 min of middle cerebral artery occlusion (MCAO). During the experiments, regional cerebral blood flow (rCBF) was recorded by laser Doppler flowmetry. Behaviour was assessed at both 2 h and 24 h after reperfusion with a common neuroscore. Infarct volumes, blood­brain barrier (BBB) disruption, cerebral vascular density, apoptosis, reactive oxygen species (ROS), HIF1α, and glycogen levels were then determined using histological and immunohistochemical techniques. When compared to their WT littermates, PHD2+/− mice had significantly increased cerebral microvascular density and more effective restoration of CBF upon reperfusion. PHD2+/− mice showed significantly better functional outcomes and higher activity rates at both 2 h and 24 h after MCAO, associated with significant fewer apoptotic cells in the penumbra and less BBB disruption; PHD3−/− mice had impaired rCBF upon early reperfusion but comparable functional outcomes; PHD1−/− mice did not show any significant changes following the MCAO. Production of ROS, HIF1α staining and glycogen content in the brain were not different in any comparison. Life-long genetic inhibition of PHD enzymes produces different effects on outcome in the first 24 h after transient cerebral ischaemia. These need to be considered in optimizing therapeutic effects of PHD inhibitors, particularly when isoform specific inhibitors become available.


Brain Ischemia/enzymology , Procollagen-Proline Dioxygenase/metabolism , Animals , Body Temperature Regulation , Body Weight , Brain Ischemia/metabolism , Cerebral Arteries , Cerebral Cortex/blood supply , Gene Expression Regulation/physiology , Genotype , Hypoxia-Inducible Factor-Proline Dioxygenases , Male , Mice , Mice, Knockout , Procollagen-Proline Dioxygenase/genetics , Protein Isoforms
10.
Exp Neurol ; 233(2): 815-20, 2012 Feb.
Article En | MEDLINE | ID: mdl-22193110

Intracranial collaterals provide residual blood flow to penumbral tissue in acute ischemic stroke and contribute to infarct size variability in humans. In the present study, hemodynamic monitoring of the borderzone territory between the leptomeningeal branches of middle cerebral artery and anterior cerebral artery was compared to lateral middle cerebral artery territory, during common carotid artery occlusion and middle cerebral artery occlusion in rats. The functional performance of intracranial collaterals, shown by perfusion deficit in the territory of leptomeningeal branches either during common carotid artery occlusion or middle cerebral artery occlusion, showed significant variability among animals and consistently predicted infarct size and functional deficit. Our findings indicate that leptomeningeal collateral flow is a strong predictor of stroke severity in rats, similarly to humans. Monitoring of collateral blood flow in experimental stroke is essential for reducing variability in neuroprotection studies and accelerating the development of collateral therapeutics.


Brain Ischemia/physiopathology , Cerebrovascular Circulation/physiology , Collateral Circulation/physiology , Hemodynamics/physiology , Recovery of Function/physiology , Stroke/physiopathology , Animals , Blood Flow Velocity/physiology , Brain Ischemia/diagnosis , Brain Ischemia/pathology , Male , Predictive Value of Tests , Rats , Rats, Wistar , Stroke/diagnosis , Stroke/pathology , Treatment Outcome
11.
J Long Term Eff Med Implants ; 21(3): 193-6, 2011.
Article En | MEDLINE | ID: mdl-22150351

Intramedullary interlocking nailing is the gold standard method for treatment of tibial shaft fractures. Thus, the growing use of the intramedullary nailing resulted in an increased number of tibial nails removal procedures in daily clinical practice. Despite adequate surgeon experience, the removal of tibial intramedullary nails is not without complications. One of the commonly used nails is the ACE (DePuy Orthopaedics, Inc., Warsaw, IN, USA). The purpose of this paper is to report such a complication following the removal procedure and review the pertinent literature. A 39-year-old female who had sustained a lower-third tibial fracture was treated with an intramedullary nail 5 years ago. Despite her unremarkable follow-up, for personal reasons, a removal procedure was planned. Two undisplaced fractures were observed in the postoperative radiological evaluation. The potential complications of the removal of intramedullary nailing of the tibia should be considered by both physician and patient.


Device Removal/adverse effects , Fracture Fixation, Intramedullary/instrumentation , Tibial Fractures/etiology , Adult , Bone Nails , Female , Humans , Reoperation , Tibial Fractures/diagnostic imaging , Tomography, X-Ray Computed
12.
J Physiol ; 589(17): 4105-14, 2011 Sep 01.
Article En | MEDLINE | ID: mdl-21708904

The best neuroprotectant for acute ischaemic stroke would always be the rapid return of oxygen and glucose to physiological levels. This is currently provided by thrombolysis which restores blood flow to the ischaemic region. The attempt to confer neuroprotection by targeting the brain parenchyma has shown promise in experimental stroke models, but has unequivocally failed to translate to the clinic. Neuroprotective therapy primarily targets the biochemical cascade that produces cell death following cerebral ischaemia. However, these agents may also alter signal transduction that controls cerebral blood flow, for example glutamate, which may affect the outcome after ischaemia. In these cases, neuroprotection may potentially be due to the improved access to oxygen and glucose rather than biochemical prevention of cell death. Improvement in cerebral blood flow is an important but often overlooked effect of neuroprotective therapy, analogous to the protective effects of drug-induced hypothermia. This short review will discuss cerebral blood flow alteration and protection of the brain in the context of ischaemic preconditioning, oxygen sensing and thrombolysis. Future neuroprotection studies in cerebral ischaemia require stringent monitoring of cerebral blood flow, plus other physiological parameters. This will increase the chances that any protection observed may be able to translate to human therapy.


Brain Ischemia , Neuroprotection , Cerebrovascular Circulation/drug effects , Humans , Neuroprotective Agents/pharmacology , Stroke
13.
J Cereb Blood Flow Metab ; 31(1): 132-43, 2011 Jan.
Article En | MEDLINE | ID: mdl-20407463

Dimethyloxalylglycine (DMOG) is an inhibitor of prolyl-4-hydroxylase domain (PHD) enzymes that regulate the stability of hypoxia-inducible factor (HIF). We investigated the effect of DMOG on the outcome after permanent and transient middle cerebral artery occlusion (p/tMCAO) in the rat. Before and after pMCAO, rats were treated with 40 mg/kg, 200 mg/kg DMOG, or vehicle, and with 40 mg/kg or vehicle after tMCAO. Serial magnetic resonance imaging (MRI) was performed to assess infarct evolution and regional cerebral blood flow (rCBF). Both doses significantly reduced infarct volumes, but only 40 mg/kg improved the behavior after 24 hours of pMCAO. Animals receiving 40 mg/kg were more likely to maintain rCBF values above 30% from the contralateral hemisphere within 24 hours of pMCAO. DMOG after tMCAO significantly reduced the infarct volumes and improved behavior at 24 hours and 8 days and also improved the rCBF after 24 hours. A consistent and significant upregulation of both mRNA and protein levels of vascular endothelial growth factor (VEGF) and endothelial nitric oxide synthase (eNOS) was associated with the observed neuroprotection, although this was not consistently related to HIF-1α levels at 24 hours and 8 days. Thus, DMOG afforded neuroprotection both at 24 hours after pMCAO and at 24 hours and 8 days after tMCAO. This effect was associated with an increase of VEGF and eNOS and was mediated by improved rCBF after DMOG treatment.


Amino Acids, Dicarboxylic/pharmacology , Brain Ischemia/drug therapy , Ischemic Attack, Transient/drug therapy , Neuroprotective Agents , Animals , Behavior, Animal/drug effects , Blood Gas Analysis , Blotting, Western , Brain Chemistry/drug effects , Brain Chemistry/physiology , Brain Ischemia/pathology , Brain Ischemia/psychology , Chronic Disease , Gene Expression/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Infarction, Middle Cerebral Artery/pathology , Ischemic Attack, Transient/pathology , Ischemic Attack, Transient/psychology , Magnetic Resonance Imaging , Male , Nitric Oxide Synthase Type III/biosynthesis , Nitric Oxide Synthase Type III/genetics , RNA/genetics , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics
14.
Transl Stroke Res ; 1(4): 304-14, 2010 Dec.
Article En | MEDLINE | ID: mdl-24323556

The aim of this study was to investigate the quality and reproducibility of mass spectra derived from a matrix-assisted laser desorption ionisation time-of-flight mass spectrometry (MALDI-TOF MS) platform in a patient population undergoing carotid endarterectomy. Plasma samples were either digested with trypsin or left undigested, fractionated with either C18 or weak cation exchange (WCX) columns and analysed by MALDI-TOF MS. Quality of mass spectra for each method was assessed by baseline correction (lower area under the curve ratio indicating higher quality) and signal-to-noise ratio. Mean coefficient of variation (CV%) assessed reproducibility between repeated experiments and methods. Identified mass peak intensity differences were assessed for consistency across repeated experiments. Plasma from six patients was analysed. The quality of mass spectra was significantly better when derived from digested plasma fractionated by either WCX or C18 methods compared to undigested plasma fractionated by WCX (analysis of variance, p < 0.05). Mean CV% for repeated experiments was 18% and 28% for WCX and C18 fractionated digested plasma, respectively. A small number of differences in mass peak intensities were consistently observed in repeated experiments. Repeated experiments are required to confidently identify non-random mass peak intensity differences as putative plasma biomarkers that merit further investigation.

16.
Expert Rev Neurother ; 8(8): 1255-68, 2008 Aug.
Article En | MEDLINE | ID: mdl-18671669

Experimental evidence shows that therapeutic hypothermia (TH) protects the brain from cerebral injury in multiple ways. In different models of focal and global cerebral ischemia, mild-to-moderate hypothermia reduces mortality and neuronal injury and improves neurological outcome. In models of experimental intracerebral hemorrhage (ICH), TH reduces edema formation but does not show consistent benefi cial effects on functional outcome parameters. However, the number of studies of hypothermia on ICH is still limited. TH is most effective when applied before or during the ischemic event, and its neuroprotective properties vary according to species, strains and the model of ischemia used. Intrinsic changes in body and brain temperature frequently occur in experimental models of focal and global cerebral ischemia, and may have infl uenced studies on other neuroprotectants. This might be one explanation for the failure of a large amount of translational clinical neuroprotective trials. Hypothermia is the only neuroprotective therapeutic agent for cerebral ischemia that has successfully managed the transfer from bench to bedside, and it is an approved therapy for patients after cardiac arrest and children with hypoxic-ischemic encephalopathy. However, the implementation of hypothermia in the treatment of stroke patients is still far from routine clinical practice. In this article, the authors describe the development of TH in different models of focal and global cerebral ischemia, point out why hypothermia is so efficient in experimental cerebral ischemia, explain why temperature regulation is essential for further neuroprotective studies and discuss why TH for acute ischemic stroke still remains a promising but controversial therapeutic option.


Brain Ischemia/physiopathology , Brain Ischemia/therapy , Cerebral Hemorrhage/physiopathology , Cerebral Hemorrhage/therapy , Disease Models, Animal , Hypothermia, Induced/methods , Animals , Humans , Treatment Outcome
17.
J Neurochem ; 104(4): 903-13, 2008 Feb.
Article En | MEDLINE | ID: mdl-18233995

NMDA receptors are a subclass of ionotropic glutamate receptors. They are trafficked and/or clustered at synapses by the post-synaptic density (PSD)-95 membrane associated guanylate kinase (MAGUK) family of scaffolding proteins that associate with NMDA receptor NR2 subunits via their C-terminal glutamate serine (aspartate/glutamate) valine motifs. We have carried out a systematic study investigating in a heterologous expression system, the association of the four major NMDA receptor subtypes with the PSD-95 family of MAGUK proteins, chapsyn-110, PSD-95, synapse associated protein (SAP) 97 and SAP102. We report that although each PSD-95 MAGUK was shown to co-immunoprecipitate with NR1/NR2A, NR1/NR2B, NR1/NR2C and NR1/NR2D receptor subtypes, they elicited differential effects with regard to the enhancement of total NR2 subunit expression which then results in an increased cell surface expression of NMDA receptor subtypes. PSD-95 and chapsyn-110 enhanced NR2A and NR2B total expression which resulted in increased NR1/NR2A and NR1/NR2B receptor cell surface expression whereas SAP97 and SAP102 had no effect on total or cell surface expression of these subtypes. PSD-95, chapsyn-110, SAP97 and SAP102 had no effect on either total NR2C and NR2D subunit expression or cell surface NR1/NR2C and NR1/NR2D expression. A comparison of PSD-95alpha, PSD-95beta and PSD-95alpha(C3S,C5S) showed that PSD-95-enhanced cell surface expression of NR1/NR2A receptors was dependent upon the PSD-95 N-terminal C3,C5 cysteines. These observations support differential interaction of NMDA receptor subtypes with different PSD-95 MAGUK scaffolding proteins. This has implications for the stabilisation, turnover and compartmentalisation of NMDA receptor subtypes in neurones during development and in the mature brain.


Guanylate Kinases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Protein Subunits/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Cell Line , Disks Large Homolog 4 Protein , Guanylate Kinases/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Protein Subunits/genetics , Receptors, N-Methyl-D-Aspartate/genetics
18.
Brain Res ; 1121(1): 231-7, 2006 Nov 22.
Article En | MEDLINE | ID: mdl-17010948

INTRODUCTION: Ischemic preconditioning (IPC) induces protection to cerebral ischemia. However, it was previously unclear whether this protection resulted from altered susceptibility to ischemia. The current study examines the effects of late phase ischemic preconditioning in a mouse model of middle cerebral artery occlusion (MCAO). Specific examination of the regional cerebral blood flow (rCBF) was conducted. EXPERIMENTAL PROCEDURE: Intra-abdominal radiofrequency probes were implanted in animals and core temperature was regulated. Mice were subjected to MCAO: (1) brief 15 min duration (preconditioning ischemia) and (2) 45 min MCAO (injurious ischemia). Naive (i.e. not preconditioned) animals were compared with preconditioned animals (preconditioning ischemia plus injurious ischemia at 72 h reperfusion). rCBF was measured using laser Doppler flowmetry (LDF) and magnetic resonance cerebral perfusion (MRP) arterial spin labeling. Percentage of brain infarcted was compared between groups. RESULTS: rCBF was significantly improved in the preconditioned cohorts of mice. Naive animals showed flow reductions to 16+/-3.59% (MCAO_45; injurious, unpreconditioned) and 17.1+/-8.6% (MCAO_15; preconditioning ischemia alone) of baseline, while preconditioned animals had flows 33.9+/-13.2% (IPC_45; preconditioned animals with injurious ischemia at 72 h reperfusion) of baseline (p=0.001). Percentage of brain infarcted was 17.2+/-6.2% in naive animals, while it was 5.1+/-4.6% in the preconditioned animals (p=0.003). MRP of the perfusion to the ischemic hemisphere, in a striatal coronal slice of the brain was 26.7+/-5.8% of the contralateral hemisphere in naive animals while preconditioned mice had flows of 38.7+/-6.8% of contralateral (p=0.04). CONCLUSIONS: Improved rCBF is an important factor in the protection of IPC, during injurious MCAO in the mouse. Stringent monitoring of rCBF is required in future studies of IPC.


Brain Ischemia/prevention & control , Cerebrovascular Circulation/physiology , Ischemic Attack, Transient/prevention & control , Ischemic Preconditioning/methods , Animals , Body Temperature , Disease Models, Animal , Ischemic Attack, Transient/physiopathology , Mice , Mice, Inbred C57BL , Middle Cerebral Artery , Regional Blood Flow , Telemetry
20.
Brain Res ; 1075(1): 201-12, 2006 Feb 23.
Article En | MEDLINE | ID: mdl-16480690

Hippocampal CA1 pyramidal neurons undergo delayed neurodegeneration after transient forebrain ischemia, and the phenomenon is dependent upon hyperactivation of l-alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA) subtype of glutamate receptors, resulting in aberrant intracellular calcium influx. The GluR2 subunit of AMPA receptors is critical in limiting the influx of calcium. The CA1 pyramidal neurons are very sensitive to ischemic damage and attempts to achieve neuroprotection, mediated by drugs, have been unsuccessful. Moreover, receptor antagonism strategies in the past have failed to provide long-term protection against ischemic injury. Long-term protection against severe forebrain ischemia can be conferred by fimbria-fornix (FF) deafferentation, which interrupts the afferent input to CA1. Our study evaluated the long-term protective effect of FF deafferentation, 12 days prior to induction of ischemia, on vulnerable CA1 neurons. Our results indicate that at 7 and 28 days post-ischemia, prior FF deafferentation protected 60% of neurons against ischemic cell death. Furthermore, we sought to evaluate whether FF deafferentation also sustained GluR2 levels in these neurons. GluR2 protein and mRNA expression were sustained by deafferentation at 70% of control following ischemia. Correlation studies revealed a positive correlation between GluR2 protein and mRNA level. These results demonstrate that protection conferred by FF deafferentation was long-term and related to sustained GluR2 expression.


Afferent Pathways/physiology , Fornix, Brain/physiology , Hippocampus/physiopathology , Ischemic Attack, Transient/physiopathology , Prosencephalon/physiology , Pyramidal Cells/physiology , Receptors, AMPA/genetics , Afferent Pathways/physiopathology , Animals , Carotid Arteries/physiopathology , Hippocampus/injuries , Ischemic Attack, Transient/genetics , Male , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
...