Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Transl Stroke Res ; 2(1): 17-25, 2011 Mar.
Article in English | MEDLINE | ID: mdl-24323583

ABSTRACT

Although intracerebral hemorrhage (ICH) has no proven treatment, well-designed studies using animal models of ICH may lead to the development of novel therapies. We briefly review current animal models of ICH. Furthermore, we discuss how these models may be utilized and targeted to facilitate translation of preclinical findings to the clinical arena.

2.
J Neurochem ; 115(5): 1266-76, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20883517

ABSTRACT

Matrix metalloproteinases (MMPs) play an important role in reperfusion-induced brain injury following ischemia. To define the effects of peroxynitrite decomposition catalyst on MMP activation and neurovascular reperfusion injury, 5,10,15,20-tetrakis (2,4,6-trimethyl-3,5-disulfonatophenyl)-porphyrin iron (III) (FeTMPyP) was administered intravenously 30 min prior to reperfusion following a middle cerebral artery occlusion. Activation of MMP was assessed by in situ and gel zymography. Neurovascular injury was assessed using endothelial barrier antigen, collagen IV immunohistochemistry and Cresyl violet staining. Results were compared with sham and ischemia alone groups. We found that administration of FeTMPyP just before reperfusion after ischemia inhibited MMP-9 activation and total MMP-2 increases in the cortex and decreased active MMP-9 along with the total amounts of active MMP-9 and active MMP-2 in the striatum. Reperfusion-induced injury to the basal lamina of collagen IV-immunopositive microvasculature and neural cells in cortex and striatum was ameliorated by FeTMPyP. Losses of blood vessel endothelium produced by ischemia or reperfusion were also decreased in the cortex. These results suggest that administration of FeTMPy prior to reperfusion decreases MMP activation and neurovascular injury after prolonged cerebral ischemia. This strategy may be useful for future therapies targeted at preventing breakdown of the blood-brain barrier and hemorrhagic transformation.


Subject(s)
Cerebral Infarction/prevention & control , Ferric Compounds/therapeutic use , Hematinics/therapeutic use , Matrix Metalloproteinases/metabolism , Metalloporphyrins/therapeutic use , Reperfusion Injury/prevention & control , Analysis of Variance , Animals , Astrocytes/drug effects , Astrocytes/pathology , Basement Membrane/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/enzymology , Cerebral Cortex/pathology , Cerebral Infarction/etiology , Cerebral Infarction/pathology , Corpus Striatum/drug effects , Corpus Striatum/enzymology , Corpus Striatum/pathology , Disease Models, Animal , Enzyme Activation/drug effects , Ferric Compounds/pharmacology , Hematinics/pharmacology , Infarction, Middle Cerebral Artery/complications , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Metalloporphyrins/pharmacology , Neurons/drug effects , Neurons/pathology , Rats , Rats, Sprague-Dawley , Regional Blood Flow/drug effects , Reperfusion Injury/etiology , Time Factors
3.
Exp Neurol ; 216(2): 407-12, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19162014

ABSTRACT

A major complication of recanalization therapy after an acute arterial occlusion in brain is hemorrhagic transformation (HT). Although it is known that prolonged ischemia is important in the development of HT, the role of reperfusion in ischemia-reperfusion induced HT is less well studied. To address the effect of reperfusion on HT, we assessed the incidence and severity of hemorrhage in rats after 5 h of middle cerebral artery occlusion (MCAO) followed by 19-hour reperfusion compared to rats with permanent occlusion (PMCAO) at the same 24-hour time point. The incidence and amount of hemorrhage, neurological function, and mortality rates were measured. MCAO (5 h) with 19-hour reperfusion was associated with a significantly higher incidence of cortical hemorrhage compared to PMCAO (81.8% vs 18.2%, p<0.05). Hemorrhage scores were higher in the 5-hour MCAO/reperfusion group compared to PMCAO rats (17.6+/-11.5 vs 2.4+/-5.3 in cortex, 20.4+/-4.6 vs 9.7+/-4.5 in striatum, p<0.01). Neurological function was worse in the ischemia-reperfusion group compared to PMCAO (p<0.05) and mortality rates were insignificantly higher in the 5-hour MCAO/reperfusion group vs PMCAO group (54.5% vs 18.1%; p<0.08). The results suggest that reperfusion after prolonged ischemia is associated with increased hemorrhagic transformation and neurological deterioration as compared to permanent ischemia. Whether pharmacological treatments prior to reperfusion attenuate post-ischemic HT requires further study.


Subject(s)
Cerebral Hemorrhage/etiology , Infarction, Middle Cerebral Artery/physiopathology , Reperfusion/adverse effects , Analysis of Variance , Animals , Behavior, Animal , Brain/pathology , Brain Edema/etiology , Brain Edema/pathology , Brain Infarction/etiology , Brain Infarction/pathology , Cerebral Hemorrhage/pathology , Disease Models, Animal , Infarction, Middle Cerebral Artery/therapy , Male , Nervous System Diseases/etiology , Nervous System Diseases/pathology , Rats , Rats, Sprague-Dawley , Time Factors
4.
Neurol Res ; 31(5): 545-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19055876

ABSTRACT

OBJECTIVE: The discovery of IL-7R(alpha) polymorphisms implicated in the pathogenesis of multiple sclerosis has highlighted the importance of interleukin 7 (IL-7) in central nervous system diseases. Hypoxia affects neurological disease states in part by modulating expression of many early and late response genes. The present work used cultured PC12 cells to investigate the effect of hypoxia on IL-7 expression. METHOD: PC12 cells were cultured in Dulbecco's modified Eagle's medium (DMEM)/F12 medium. RNA was isolated and reverse transcriptase-polymerase chain reaction (RT-PCR) was run to quantify messenger RNA (mRNA) change. Western blots were used to assess IL-7 protein change in the medium. Extracellular free Ca(2+) was removed by using Ca(2+)-free DMEM/F12 with 1 mM ethylene glycol tetraacetic acid for 45 minutes before the start of hypoxia. RESULTS: Exposure of PC12 cells to 1% oxygen for 6 hours decreased IL-7 mRNA by 77% using RT-PCR (p<0.01). Exposure to 1% oxygen for 24 hours decreased IL-7 protein in the medium by 21% (p<0.05). As hypoxia duration increased (2, 4, 6 and 24 hours) or oxygen concentrations decreased (10%, 5% and 1%), IL-7 mRNA expression progressively decreased. Removal of extracellular free Ca(2+) completely prevented these hypoxia-induced decreases of IL-7 mRNA. DISCUSSION: Since IL-7 exhibits trophic properties in developing brain, down-regulation of IL-7 by hypoxia may contribute to hypoxia-induced injury to neural cells.


Subject(s)
Calcium/pharmacology , Cell Hypoxia/genetics , Interleukin-7/metabolism , Animals , Blotting, Western , Down-Regulation , Oxygen/pharmacology , PC12 Cells , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction
5.
J Cereb Blood Flow Metab ; 28(7): 1320-8, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18382470

ABSTRACT

There are no biomarkers that differentiate cardioembolic from large-vessel atherosclerotic stroke, although the treatments differ for each and approximately 30% of strokes and transient ischemic attacks have undetermined etiologies using current clinical criteria. We aimed to define gene expression profiles in blood that differentiate cardioembolic from large-vessel atherosclerotic stroke. Peripheral blood samples were obtained from healthy controls and acute ischemic stroke patients (<3, 5, and 24 h). RNA was purified, labeled, and applied to Affymetrix Human U133 Plus 2.0 Arrays. Expression profiles in the blood of cardioembolic stroke patients are distinctive from those of large-vessel atherosclerotic stroke patients. Seventy-seven genes differ at least 1.5-fold between them, and a minimum number of 23 genes differentiate the two types of stroke with at least 95.2% specificity and 95.2% sensitivity for each. Genes regulated in large-vessel atherosclerotic stroke are expressed in platelets and monocytes and modulate hemostasis. Genes regulated in cardioembolic stroke are expressed in neutrophils and modulate immune responses to infectious stimuli. This new method can be used to predict whether a stroke of unknown etiology was because of cardioembolism or large-vessel atherosclerosis that would lead to different therapy. These results have wide ranging implications for similar disorders.


Subject(s)
Atherosclerosis/complications , Blood Cells , Embolism/complications , Gene Expression Profiling , Stroke/etiology , Stroke/genetics , Biomarkers/blood , Blood Platelets , Case-Control Studies , Diagnosis, Differential , Gene Expression Profiling/methods , Gene Expression Profiling/standards , Humans , Monocytes , Neutrophils , Sensitivity and Specificity , Stroke/diagnosis
6.
Exp Neurol ; 210(2): 549-59, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18187134

ABSTRACT

In this study, we examine the effects of reperfusion on the activation of matrix metalloproteinase (MMP) and assess the relationship between MMP activation during reperfusion and neurovascular injury. Ischemia was produced using suture-induced middle cerebral artery occlusion in rats. The MMP activation was examined with in situ and gel zymography. Injury to cerebral endothelial cells and basal lamina was assessed using endothelial barrier antigen (EBA) and collagen IV immunohistochemistry. Injury to neurons and glial cells was assessed using Cresyl violet staining. These were examined at 3 h after reperfusion (8 h after initiation of ischemia) and compared with permanent ischemia at the same time points to assess the effects of reperfusion. A broad-spectrum MMP inhibitor, AHA (p-aminobenzoyl-Gly-Pro-D-Leu-D-Ala-hydroxamate, 50 mg/kg intravenously) was administered 30 min before reperfusion to assess the roles of MMPs in activating gelatinolytic enzymes and in reperfusion-induced injury. We found that reperfusion accelerated and potentiated MMP-9 and MMP-2 activation and injury to EBA and collagen IV immunopositive microvasculature and to neurons and glial cells in ischemic cortex and striatum relative to permanent ischemia. Administering AHA 30 min before reperfusion decreased MMP-9 activation and neurovascular injury in ischemic cerebral cortex.


Subject(s)
Basement Membrane/enzymology , Cerebrovascular Trauma/etiology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Reperfusion Injury/enzymology , Reperfusion/adverse effects , Analysis of Variance , Animals , Autoantigens/metabolism , Basement Membrane/drug effects , Basement Membrane/pathology , Collagen Type IV/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Male , Peptide Fragments/pharmacology , Rats , Rats, Sprague-Dawley , Reperfusion Injury/pathology , Time Factors
7.
Brain Res ; 1162: 130-2, 2007 Aug 08.
Article in English | MEDLINE | ID: mdl-17618608

ABSTRACT

We have developed a novel type of duplex enzyme-linked immunosorbent assay (ELISA) for the quantitation of the major plasma proteins, IgG and albumin, in edematous brain tissue. We test this duplex ELISA on our porcine intracerebral hemorrhage (ICH) model and show that it is as accurate and sensitive as independent single ELISAs. This method is useful as a marker of edema in brain tissue and the same design can be applied to other proteins and sample types.


Subject(s)
Blood Proteins/metabolism , Brain Edema/metabolism , Enzyme-Linked Immunosorbent Assay , Animals , Cerebral Hemorrhage/complications , Disease Models, Animal , Functional Laterality , Swine , Time Factors
8.
Phys Med Biol ; 52(11): 2953-67, 2007 Jun 07.
Article in English | MEDLINE | ID: mdl-17505082

ABSTRACT

Stroke is a devastating disease and a leading cause of death and disability. Currently, the only FDA approved therapy for acute ischemic stroke is the intravenous administration of the thrombolytic medication, recombinant tissue plasminogen activator (tPA). However, this treatment has many contraindications and can have dangerous side effects such as intra-cerebral hemorrhage. These treatment limitations have led to much interest in potential adjunctive therapies, such as therapeutic hypothermia (T

Subject(s)
Stroke/drug therapy , Tissue Plasminogen Activator/chemistry , Biophysics/methods , Blood Coagulation , Chemistry, Physical/methods , Diffusion , Fibrinolysis , Humans , Hypothermia/pathology , In Vitro Techniques , Models, Chemical , Models, Statistical , Stroke/pathology , Substrate Specificity , Temperature , Thrombolytic Therapy/methods , Time Factors
9.
Stroke ; 38(2 Suppl): 753-8, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17261732

ABSTRACT

A significant amount of new information has been generated in animal models of intracerebral hemorrhage during the past several years. These include findings on the pathophysiological, biochemical and molecular processes that underlie the development of brain tissue injury after intracerebral hemorrhage as well as potential new treatments. We review these various findings that include glutamate receptor activation, oxidative stress development, intracellular signaling through the transcription factor, nuclear factor-kappaB, and markedly upregulated cytokine gene expression. We also briefly review the surgical treatment for intracerebral hemorrhage and list the pharmacological treatment studies that have recently appeared.


Subject(s)
Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/physiopathology , Cytokines/physiology , Disease Models, Animal , Glutamic Acid/physiology , NF-kappa B/physiology , Signal Transduction/physiology , Animals , Humans
10.
J Neurotrauma ; 23(12): 1904-11, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17184198

ABSTRACT

Intracerebral hemorrhage (ICH) and traumatic brain injury can induce brain tissue edema (i.e., interstitial and/or vasogenic), containing high concentrations of plasma proteins. To understand biochemical processes in edema development following these insults, it would be useful to examine alterations in various proteins (e.g., transcription factors, signaling). However, determining altered protein responses in edematous brain tissue using standard immunoblotting techniques is problematic due to contaminating plasma proteins. To solve this problem, we developed an enzyme-linked immunosorbent assay (ELISA) method to quantify the two major plasma proteins, albumin and immunoglobulin G (IgG), that comprise about 80% of the total plasma proteins. We tested our method on edematous white matter samples from our porcine ICH model. To induce ICH, we infused autologous arterial whole blood (3 mL) into frontal hemispheric white matter of pentobarbital- anesthetized pigs ( approximately 20 kg) over 15 min. We froze brains in situ at various times up to 24 h post- ICH and sampled white matter adjacent and contralateral to hematomas. We prepared cytoplasmic extracts that we subjected to ELISA and immunoblotting analyses. Our results demonstrate that this ELISA method is accurate, reproducible, and enables the concentrations of albumin and IgG in edematous brain tissue samples to be accurately determined. By using this correction method, equal amounts of cellular protein can be loaded onto gels during immunoblotting procedures. This method is applicable to edematous tissue samples in brain injury models in which high plasma protein concentrations result from interstitial or vasogenic edema development.


Subject(s)
Brain Edema/metabolism , Cerebral Hemorrhage/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Immunoglobulin G/metabolism , Serum Albumin/metabolism , Animals , Brain Edema/etiology , Cerebral Hemorrhage/complications , Immunoblotting , Reproducibility of Results , Swine
11.
J Cereb Blood Flow Metab ; 26(8): 1089-102, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16395289

ABSTRACT

Ischemic brain and peripheral white blood cells release cytokines, chemokines and other molecules that activate the peripheral white blood cells after stroke. To assess gene expression in these peripheral white blood cells, whole blood was examined using oligonucleotide microarrays in 15 patients at 2.4+/-0.5, 5 and 24 h after onset of ischemic stroke and compared with control blood samples. The 2.4-h blood samples were drawn before patients were treated either with tissue-type plasminogen activator (tPA) alone or with tPA plus Eptifibatide (the Combination approach to Lysis utilizing Eptifibatide And Recombinant tPA trial). Most genes induced in whole blood at 2 to 3 h were also induced at 5 and 24 h. Separate studies showed that the genes induced at 2 to 24 h after stroke were expressed mainly by polymorphonuclear leukocytes and to a lesser degree by monocytes. These genes included: matrix metalloproteinase 9; S100 calcium-binding proteins P, A12 and A9; coagulation factor V; arginase I; carbonic anhydrase IV; lymphocyte antigen 96 (cluster of differentiation (CD)96); monocarboxylic acid transporter (6); ets-2 (erythroblastosis virus E26 oncogene homolog 2); homeobox gene Hox 1.11; cytoskeleton-associated protein 4; N-formylpeptide receptor; ribonuclease-2; N-acetylneuraminate pyruvate lyase; BCL6; glycogen phosphorylase. The fold change of these genes varied from 1.6 to 6.8 and these 18 genes correctly classified 10/15 patients at 2.4 h, 13/15 patients at 5 h and 15/15 patients at 24 h after stroke. These data provide insights into the inflammatory responses after stroke in humans, and should be helpful in diagnosis, understanding etiology and pathogenesis, and guiding acute treatment and development of new treatments for stroke.


Subject(s)
Brain Ischemia/blood , Gene Expression Regulation , Monocytes/metabolism , Neutrophils/metabolism , Stroke/blood , Adult , Aged , Brain Ischemia/drug therapy , Drug Therapy, Combination , Eptifibatide , Female , Fibrinolytic Agents/therapeutic use , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Inflammation/blood , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Peptides/therapeutic use , Platelet Aggregation Inhibitors/therapeutic use , Stroke/drug therapy , Time Factors , Tissue Plasminogen Activator/therapeutic use
12.
J Cereb Blood Flow Metab ; 26(2): 230-52, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16034371

ABSTRACT

After intracerebral hemorrhage (ICH), many changes of gene transcription occur that may be important because they will contribute to understanding mechanisms of injury and recovery. Therefore, gene expression was assessed using Affymetrix microarrays in the striatum and the overlying cortex at 24 h after intracranial infusions of blood into the striatum of adult rats. Intracerebral hemorrhage regulated 369 of 8,740 transcripts as compared with saline-injected controls, with 104 regulated genes shared by the striatum and cortex. There were 108 upregulated and 126 downregulated genes in striatum, and 170 upregulated and 69 downregulated genes in the cortex. Real-time reverse transcriptase-polymerase chain reaction (RT-PCR) confirmed upregulation of IL-1-beta, Lipcortin 1 (annexin) and metallothionein 1,2, and downregulation of potassium voltage-gated channel, shaker-related subfamily, beta member 2 (Kcnab2). Of the functional groups of genes modulated by ICH, many metabolism and signal-transduction-related genes decreased in striatum but increased in adjacent cortex. In contrast, most enzyme, cytokine, chemokine, and immune response genes were upregulated in both striatum and in the cortex after ICH, likely in response to foreign proteins from the blood. A number of these genes may contribute to brain edema and cellular apoptosis caused by ICH. In addition, downregulation of growth factor pathways and the phosphatidylinositol 3-kinase (PI3K)/Akt pathway could also contribute to perihematoma cell death/apoptosis. Intracerebral hemorrhage-related downregulation of GABA-related genes and potassium channels might contribute to perihematoma cellular excitability and increased risk of post-ICH seizures. These genomic responses to ICH potentially provide new therapeutic targets for treatment.


Subject(s)
Brain/physiology , Cerebral Hemorrhage/genetics , Gene Expression Profiling , Genomics , Animals , Annexin A1/genetics , Annexin A1/metabolism , Brain/metabolism , Cluster Analysis , Disease Models, Animal , Down-Regulation , Growth Substances/genetics , Growth Substances/metabolism , Interleukin-1/genetics , Interleukin-1/metabolism , Male , Metallothionein/genetics , Metallothionein/metabolism , Oligonucleotide Array Sequence Analysis/methods , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction/methods , Transcription, Genetic , Up-Regulation , gamma-Aminobutyric Acid/genetics , gamma-Aminobutyric Acid/metabolism
13.
Curr Neurovasc Res ; 2(2): 149-55, 2005 Apr.
Article in English | MEDLINE | ID: mdl-16181107

ABSTRACT

Plasma infused into porcine cerebral white matter induces both acute interstitial and delayed vasogenic edema. Edematous white matter contains extracellular plasma proteins and rapidly induces oxidative stress as evidenced by increased protein carbonyl formation and heme oxygenase-1 induction. We tested the hypothesis that edematous white matter would also upregulate pro-inflammatory cytokine gene expression and develop DNA damage. We infused autologous plasma into the frontal hemispheric white matter of pentobarbital-anesthetized pigs. We monitored and controlled physiological variables and froze brains in situ at 1, 4 or 24 hrs. We determined edema volumes by computer-assisted morphometry. We measured white matter protein carbonyl formation by immunoblotting, cytokine gene expression by standard RT-PCR methods and DNA fragmentation by agarose gel electrophoresis. White matter edema developed acutely (1 hr) after plasma infusion and increased significantly in volume between 4 and 24 hrs. Protein carbonyl formation also occurred rapidly in edematous white matter with significant elevations (3 to 4-fold) already present at 1 hr. This increase remained through 24 hrs. Pro-inflammatory cytokine gene expression was also rapidly increased at 1 hr post-infusion. Evidence for DNA fragmentation began at 2 to 4 hrs, and a pattern indicative of both ongoing necrosis and apoptosis was robust by 24 hrs. Plasma protein accumulation in white matter induces acute edema development and a cascade of patho-chemical events including oxidative stress, pro-inflammatory cytokine gene expression and DNA damage. These results suggest that in diseases with increased blood-brain barrier (BBB) permeability or following intracerebral hemorrhage or traumatic brain injury, interstitial plasma can rapidly damage white matter.


Subject(s)
Blood , Brain Edema/etiology , Brain Edema/metabolism , Brain/metabolism , DNA Fragmentation , Inflammation Mediators/metabolism , Oxidative Stress , Animals , Apoptosis , Blood Proteins/metabolism , Blood-Brain Barrier , Brain/pathology , Brain Edema/pathology , Brain Edema/physiopathology , Capillary Permeability , Cytokines , Gene Expression , Necrosis , Swine , Time Factors
14.
Neurol Res ; 27(3): 238-45, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15845207

ABSTRACT

Hypothermia is well known to provide neuroprotection following various brain insults in experimental animals. Two recently completed clinical trials of whole body hypothermia in out-of-hospital cardiac arrest patients' demonstrated significantly improved survival rates and neurologic outcomes. These results provide new excitement and encouragement for clinical application of hypothermia in cerebrovascular disease. However, the intensive care challenges and adverse events (e.g. prolonged times to target temperatures, shivering and sedation, pneumonia) during the management of hypothermia, dampen enthusiasm for widespread application especially in elderly stroke patients. In this manuscript, we review recent hypothermia trials for stroke. We describe an alternate approach, i.e. local brain cooling, and discuss this new technique with reference to the extensive literature on the marked efficacy of hypothermia. We describe a new technology, the ChillerPad(TM) and ChillerStrip(TM) Systems developed by Seacoast Technologies, Inc. (Portsmouth, NH, USA). The latter device has received FDA approval and will be employed in a trial of local hypothermia for cerebral aneurysm repair. We present our experimental findings that profound local hypothermia does not damage cortical neurons. We also report that local hypothermia protects the blood-brain barrier and markedly reduces vasogenic edema development in an experimental intracerebral hemorrhage model. Lastly, we review potential mechanisms through which hypothermia provides blood-brain barrier protection and reduces edema formation. Clearly, hypothermia has a bright future for cerebrovascular disease treatment if brain cooling can be delivered in a manner that does not compromise the patient or the neurosurgical and intensive care settings. Local brain cooling may be just that new treatment approach.


Subject(s)
Aneurysm/prevention & control , Hypothermia , Stroke/prevention & control , Animals , Body Temperature/physiology , Brain/physiopathology , Brain Infarction/prevention & control , Clinical Trials as Topic , Cold Temperature , Humans , Hypothermia, Induced/methods , Stroke/physiopathology , Time Factors
15.
J Neurosurg ; 101(6): 1026-9, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15597764

ABSTRACT

OBJECT: A model of subarachnoid hemorrhage (SAH) in pigs was developed to investigate bilirubin concentration in cerebrospinal fluid (CSF) as a potential marker of sentinel SAH. METHODS: Seven male Yorkshire pigs received a 250-microl injection of either whole autologous arterial blood (four animals) or isotonic saline (three animals) into the cisternae magna in an effort to produce volumetrically a model of sentinel SAH and a control injection model, respectively. Cerebrospinal fluid volumes of 100 microl were then collected from both the lumbar cistern and cisternae magna at 1 to 2-hour intervals for a total of 24 hours postinjection. The CSF was then tested for bilirubin. Mean concentrations of bilirubin (+/- standard deviation [SD]) obtained from the lumbar cistern 24 hours following the injection of blood or saline were 4.38 +/- 1.04 microM in the SAH animals and 1.02 +/- 0.05 microM in the controls. At 24 hours postinjection, mean concentrations (+/- SD) of cisternae magna bilirubin were 7.29 +/- 1.33 microM and 1.33 +/- 0.14 microM in the SAH animals and controls, respectively. In the SAH group, both the lumbar cistern and cisternae magna bilirubin concentrations differed significantly from baseline values 12 hours following SAH. CONCLUSIONS: Elevated concentrations of CSF bilirubin can be detected following a low-volume SAH, and the production of bilirubin occurred over a predictable time course. Twelve hours after hemorrhage, an elevated CSF bilirubin concentration was an indicator of hemolysis occurring in the subarachnoid spaces. The presence of bilirubin in CSF is a potential marker for differentiating SAHs from traumatic lumbar punctures in humans.


Subject(s)
Bilirubin/cerebrospinal fluid , Subarachnoid Hemorrhage/cerebrospinal fluid , Subarachnoid Hemorrhage/diagnosis , Animals , Biomarkers , Diagnosis, Differential , Disease Models, Animal , Male , Pilot Projects , Spinal Puncture/adverse effects , Sus scrofa
16.
Stroke ; 35(11): 2587-91, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15375303

ABSTRACT

BACKGROUND AND PURPOSE: Intracerebral hemorrhage has no effective treatment. The delayed appearance of edema, apoptosis, and inflammation in perihematomal brain suggests that these events may be targets for therapeutic intervention. To develop successful treatments, we must learn more about the effects of hemorrhage on brain tissue. In this study, we investigated the acute metabolic effects of intrastriatal hemorrhage in rat brain. METHODS: Lysed blood or saline (50 microL each) was injected into the striatum of male Sprague-Dawley rats. The rats recovered for 1 to 72 hours before injection of [14C]-2-deoxyglucose (intraperitoneally) 30 minutes before decapitation. Animals were pretreated with the N-methyl-D-aspartate (NMDA) and alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) glutamate receptor antagonists dizolcilpine maleate (MK-801; 1 mg/kg) or 2,3-dihydroxy-6-nitro-7-sulfamoylbenzo[f]quinoxaline (NBQX; 30 mg/kg), or saline vehicle. Additional animals received intrastriatal injections of glutamate (1.0 mmol/L), NMDA (1.0 mmol/L), or AMPA (0.1 mmol/L) in the place of blood. Semiquantitative autoradiographs from the brains were analyzed to determine the effects of hemorrhage on relative glucose metabolism. RESULTS: We found an acute phase of increased [14C]-2-deoxyglucose uptake in the perihematomal region that peaks 3 hours after lysed blood injection. Saline injections had no effect on striatal glucose utilization. The increased [14C]-2-deoxyglucose uptake produced by the hemorrhages was blocked by pretreatment with MK-801 and NBQX. Glutamate injections alone had no effect on striatal metabolism, whereas NMDA and AMPA injections increased [14C]-2-deoxyglucose uptake. CONCLUSIONS: The data imply that glutamate activation of NMDA or AMPA receptors increases glucose metabolism in perihematomal brain at early times after intracerebral hemorrhage. This may provide a possible target for the treatment of intracerebral hemorrhage.


Subject(s)
Cerebral Hemorrhage/metabolism , Glucose/metabolism , Receptors, Glutamate/metabolism , Animals , Autoradiography , Brain/metabolism , Cerebrovascular Circulation , Disease Models, Animal , Glutamic Acid/metabolism , Hematoma , Male , Rats , Rats, Sprague-Dawley , Receptors, AMPA , Receptors, N-Methyl-D-Aspartate
18.
Ann N Y Acad Sci ; 1012: 237-51, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15105270

ABSTRACT

The hemorrhagic strokes, intracerebral (ICH) and subarachnoid hemorrhage (SAH), often have poor outcomes. Indeed, the most common hemorrhagic stroke, ICH, has the highest mortality and morbidity rates of any stroke subtype. In this report, we discuss the evidence for the staging of red blood cell removal after ICH and the significance of control of this process. The protective effects of clinically relevant metalloporphyrin heme oxygenase inhibitors in experimental models of ICH and in superficial siderosis are also discussed. We also examine literature paradoxes related to both heme and heme oxygenase in various disorders of the central nervous system. Last, new data are presented that support the concept that heme, although primarily a pro-oxidant, can also have antioxidant properties.


Subject(s)
Brain Ischemia/complications , Hematoma/surgery , Heme Oxygenase (Decyclizing)/metabolism , Heme/metabolism , Intracranial Hemorrhages/etiology , Stroke/etiology , Animals , Dose-Response Relationship, Drug , Heme/therapeutic use , Hemoglobins , Humans , Intracranial Hemorrhages/enzymology , Intracranial Hemorrhages/metabolism , Intracranial Hemorrhages/prevention & control , Metalloporphyrins/therapeutic use , Stroke/blood , Stroke/physiopathology , Stroke/prevention & control , Time Factors
19.
J Cereb Blood Flow Metab ; 23(6): 629-52, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12796711

ABSTRACT

Heme and iron metabolism are of considerable interest and importance in normal brain function as well as in neurodegeneration and neuropathologically following traumatic injury and hemorrhagic stroke. After a cerebral hemorrhage, large numbers of hemoglobin-containing red blood cells are released into the brain's parenchyma and/or subarachnoid space. After hemolysis and the subsequent release of heme from hemoglobin, several pathways are employed to transport and metabolize this heme and its iron moiety to protect the brain from potential oxidative stress. Required for these processes are various extracellular and intracellular transporters and storage proteins, the heme oxygenase isozymes and metabolic proteins with differing localizations in the various brain-cell types. In the past several years, additional new genes and proteins have been discovered that are involved in the transport and metabolism of heme and iron in brain and other tissues. These discoveries may provide new insights into neurodegenerative diseases like Alzheimer's, Parkinson's, and Friedrich's ataxia that are associated with accumulation of iron in specific brain regions or in specific organelles. The present review will examine the uptake and metabolism of heme and iron in the brain and will relate these processes to blood removal and to the potential mechanisms underlying brain injury following cerebral hemorrhage.


Subject(s)
Brain/metabolism , Cerebral Hemorrhage/metabolism , Heme/metabolism , Iron/metabolism , Animals , Humans
20.
Neurosurg Clin N Am ; 13(3): 349-54, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12486924

ABSTRACT

The efficacy of surgical treatment of ICH remains unproven and controversial [40]. Although open surgery does not appear to improve the patient's outcome [2], less invasive methods of hematoma evacuation seem to show promising results in improving patient outcome and survival. To date, the only two clinical trials that have demonstrated benefit from surgical treatment over medical therapy for ICH have used minimally invasive techniques [27,38]. Randomized controlled clinical trials comparing minimally invasive surgical techniques versus best medical treatment are needed to determine the best management of ICH.


Subject(s)
Cerebral Hemorrhage/surgery , Minimally Invasive Surgical Procedures/methods , Cerebral Hemorrhage/diagnostic imaging , Humans , Tomography, X-Ray Computed
SELECTION OF CITATIONS
SEARCH DETAIL
...