Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Brain Res ; 1678: 123-128, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29079502

ABSTRACT

12/15-Lipoxygenase (12/15-LOX) contributes to the brain damage after middle cerebral artery occlusion (MCAO) in the acute phase of stroke. The aim of this study was to investigate the effects of a 12/15-LOX inhibitor, LOXBlock-1(LB1), in mice using a FeCl3-induced permanent distal MCAO model and FeCl3-induced ischemia/thrombolysis with tPA. In order to induce permanent distal MCAO, 30% FeCl3 was used in C57BL6 mice. LB1 or DMSO treatments were applied intraperitoneally 2 h following MCAO. For FeCl3-induced ischemia/thrombolysis experiments, 10% FeCl3 was preferred so as to obtain reperfusion with tPA in CD1 mice. 4 h following ischemia either LB1 or DMSO and iv tPA was administered. Outcomes were NSS, weight loss, infarct volume, hemorrhage area and reperfusion rate. FeCl3-induced distal MCAO caused an increase in 12/15-LOX signal in the ischemic cortex with an increase in MDA2 and AIF immunoreactivity. LB1 treatment, applied 2 h after ischemia, significantly decreased the infarct volume at 24 h of permanent distal MCAO. Weight loss was also significantly reduced in LB1 treated group. Distal MCAO and tPA application with LB1 or DMSO showed that treatment significantly decreased the infarct volume and the hemorrhage area. The reperfusion rate in the LB1-treated group was surprisingly higher than in the DMSO group and NSS results were significantly improved. These data suggest that LB1 can be used as an adjuvant agent to tPA. This study not only shows the effects of LB1 treatment in distal MCAO but also confirms that FeCl3-induced MCAO model can be a useful tool to screen novel treatment options in stroke.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Brain Ischemia/drug therapy , Stroke/physiopathology , Animals , Arachidonate 12-Lipoxygenase/drug effects , Arachidonate 15-Lipoxygenase/drug effects , Brain Ischemia/metabolism , Disease Models, Animal , Fibrinolytic Agents/therapeutic use , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Lipoxygenase/drug effects , Lipoxygenase/metabolism , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Reperfusion , Stroke/drug therapy , Thrombolytic Therapy/methods , Tissue Plasminogen Activator/therapeutic use
2.
Cell Death Dis ; 5: e1084, 2014 Feb 27.
Article in English | MEDLINE | ID: mdl-24577082

ABSTRACT

Necroptosis is a newly described form of regulated necrosis that contributes to neuronal death in experimental models of stroke and brain trauma. Although much work has been done elucidating initiating mechanisms, signaling events governing necroptosis remain largely unexplored. Akt is known to inhibit apoptotic neuronal cell death. Mechanistic target of rapamycin (mTOR) is a downstream effector of Akt that controls protein synthesis. We previously reported that dual inhibition of Akt and mTOR reduced acute cell death and improved long term cognitive deficits after controlled-cortical impact in mice. These findings raised the possibility that Akt/mTOR might regulate necroptosis. To test this hypothesis, we induced necroptosis in the hippocampal neuronal cell line HT22 using concomitant treatment with tumor necrosis factor α (TNFα) and the pan-caspase inhibitor N-benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone. TNFα/zVAD treatment induced cell death within 4 h. Cell death was preceded by RIPK1-RIPK3-pAkt assembly, and phosphorylation of Thr-308 and Thr473 of AKT and its direct substrate glycogen synthase kinase-3ß, as well as mTOR and its direct substrate S6 ribosomal protein (S6), suggesting activation of Akt/mTOR pathways. Pretreatment with Akt inhibitor viii and rapamycin inhibited Akt and S6 phosphorylation events, mitochondrial reactive oxygen species production, and necroptosis by over 50% without affecting RIPK1-RIPK3 complex assembly. These data were confirmed using small inhibitory ribonucleic acid-mediated knockdown of AKT1/2 and mTOR. All of the aforementioned biochemical events were inhibited by necrostatin-1, including Akt and mTOR phosphorylation, generation of oxidative stress, and RIPK1-RIPK3-pAkt complex assembly. The data suggest a novel, heretofore unexpected role for Akt and mTOR downstream of RIPK1 activation in neuronal cell death.


Subject(s)
Hippocampus/enzymology , Neurons/enzymology , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Caspase Inhibitors/pharmacology , Cell Line , Dose-Response Relationship, Drug , Enzyme Activation , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hippocampus/drug effects , Hippocampus/pathology , Imidazoles/pharmacology , Indoles/pharmacology , Mice , Mitochondria/metabolism , Mitochondria/pathology , Necrosis , Neurons/drug effects , Neurons/pathology , Oligopeptides/pharmacology , Oxidative Stress , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , RNA Interference , Reactive Oxygen Species/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Ribosomal Protein S6/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , Time Factors , Transfection , Tumor Necrosis Factor-alpha/pharmacology
3.
Curr Med Chem ; 21(18): 2035-42, 2014.
Article in English | MEDLINE | ID: mdl-24372202

ABSTRACT

The neurovascular unit is now well accepted as a conceptual framework for investigating the mechanisms of ischemic stroke. From a molecular and cellular perspective, three broad mechanisms may underlie stroke pathophysiology--excitotoxicity, oxidative stress and inflammation. To date, however, most investigations of these basic mechanisms have focused on neuronal responses. In this mini-review, we ask whether these mechanisms of excitotoxicity, oxidative stress and inflammation can also be examined in terms of non-neuronal interactions in the neurovascular unit, including the release of extracellular vesicles for cell-cell signaling.


Subject(s)
Brain Ischemia/physiopathology , Stroke/physiopathology , Animals , Brain Ischemia/metabolism , Cell Communication , Extracellular Space/metabolism , Humans , Oxidative Stress , Signal Transduction , Stroke/metabolism
4.
Neurol Res ; 35(5): 448-56, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23711324

ABSTRACT

Neurovascular disease often involves multi-organ system injury. For example, patent foramen ovale (PFO) related ischemic strokes involve not just the brain, but also the heart, the lung, and the peripheral vascular circulation. For higher-risk but high-reward systemic therapy (e.g., thrombolytics, therapeutic hypothermia (TH), PFO closure) to be implemented safely, very careful patient selection and close monitoring of disease progression and therapeutic efficacy are imperative. For example, more than a decade after the approval of therapeutic hypothermic and intravenous thrombolysis treatments, they both remain extremely under-utilized, in part due to lack of clinical tools for patient selection or to follow therapeutic efficacy. Therefore, in understanding the complexity of the global effects of clinical neurovascular diseases and their therapies, a systemic approach may offer a unique perspective and provide tools with clinical utility. Clinical proteomic approaches may be promising to monitor systemic changes in complex multi-organ diseases - especially where the disease process can be 'sampled' in clinically accessible fluids such as blood, urine, and CSF. Here, we describe a 'pharmaco-proteomic' approach to three major challenges in translational neurovascular research directly at bedside - in order to better stratify risk, widen therapeutic windows, and explore novel targets to be validated at the bench - (i) thrombolytic treatment for ischemic stroke, (ii) therapeutic hypothermia for post-cardiac arrest syndrome, and (iii) treatment for PFO related paradoxical embolic stroke. In the future, this clinical proteomics approach may help to improve patient selection, ensure more precise clinical phenotyping for clinical trials, and individualize patient treatment.


Subject(s)
Biomarkers, Pharmacological/metabolism , Heart Arrest/therapy , Precision Medicine , Proteomics , Stroke/drug therapy , Stroke/metabolism , Fibrinolytic Agents/therapeutic use , Foramen Ovale, Patent/complications , Foramen Ovale, Patent/drug therapy , Foramen Ovale, Patent/surgery , Humans , Hypothermia, Induced , Intracranial Embolism/complications , Intracranial Embolism/drug therapy , Stroke/prevention & control
5.
Transl Stroke Res ; 2(4): 600-7, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22207885

ABSTRACT

There is growing evidence supporting the role of inflammation in early brain injury and cerebral vasospasm following subarachnoid hemorrhage (SAH). Matrix metalloproteinases (MMPs) are released by inflammatory cells and can mediate early brain injury via disruption of the extracellular matrix and mediate vasospasm by cleaving endothelin-1 into vasoactive fragments. We hypothesize that inflammation marked by neutrophil elevation and MMP-9 release in human SAH is associated with vasospasm and with poor clinical outcome. We enrolled consecutive SAH subjects (N = 55), banked serial blood and cerebrospinal fluid (CSF) samples, and evaluated their 3-month modified Rankin scores (mRS). Vasospasm was defined as >50% vessel caliber reduction on angiography 6-8 days post-SAH. A poor outcome was defined as mRS > 2. We compared blood leukocyte and neutrophil counts during post-SAH days 0-14 with respect to vasospasm and 3-month outcome. In a subset of SAH subjects (N = 35), we compared blood and CSF MMP-9 by enzyme-linked immunosorbent assay (ELISA) on post-SAH days 0-1, 2-3, 4-5, 6-8, and 10-14 with respect to vasospasm and to 3-month outcome. Persistent elevation of blood leukocyte (p = 0.0003) and neutrophil (p = 0.0002) counts during post-SAH days 0-14 are independently associated with vasospasm after adjustment for major confounders. In the same time period, blood neutrophil count (post-SAH days 2-3, p = 0.018), blood MMP-9 (post-SAH days 4-5, p = 0.045), and CSF MMP-9 (post-SAH days 2-3, p = 0.05) are associated with poor 3-month SAH clinical outcome. Neutrophil count correlates with blood MMP-9 (post-SAH days 6-8, R = 0.39; p = 0.055; post-SAH days 10-14, R = 0.79; p < 0.0001), and blood MMP-9 correlates with CSF MMP-9 (post-SAH days 4-5, R = 0.72; p = 0.0002). Elevation of CSF MMP-9 during post-SAH days 0-14 is associated with poor 3-month outcome (p = 0.0078). Neither CSF nor blood MMP-9 correlates with vasospasm. Early rise in blood neutrophil count and blood and CSF MMP-9 are associated with poor 3-month SAH clinical outcome. In blood, neutrophil count correlates with MMP-9 levels, suggesting that neutrophils may be an important source of blood MMP-9 early in SAH. Similarly, CSF and blood MMP-9 correlate positively early in the course of SAH, suggesting that blood may be an important source of CSF MMP-9. Blood and CSF MMP-9 are associated with clinical outcome but not with vasospasm, suggesting that MMP-9 may mediate brain injury independent of vasospasm in SAH. Future in vitro studies are needed to investigate the role of MMP-9 in SAH-related brain injury. Larger clinical studies are needed to validate blood and CSF MMP-9 as potential biomarkers for SAH outcome.

6.
Neurology ; 73(5): 393-9, 2009 Aug 04.
Article in English | MEDLINE | ID: mdl-19652144

ABSTRACT

Emerging data suggest that a wide array of measurable biomarkers in blood may provide a novel window into the pathophysiology of stroke. In this review, we survey the state of progress in the field. Three specific questions are assessed. Can biomarkers augment the clinical examination and powerful brain imaging tools to enhance the accuracy of the diagnostic process? Can biomarkers be used to help triage patients for thrombolytic therapy? Can biomarkers help predict patients who are most susceptible to malignant infarction? Many encouraging molecular candidates have been found that appear to match the known cascades of neurovascular injury after stroke. However, whether these putative biomarkers may indeed have direct clinical utility remains to be quantitatively validated. Larger clinical trials are warranted to establish the sensitivity and specificity of biomarkers for routine use in clinical stroke.


Subject(s)
Biomarkers/analysis , Biomarkers/blood , Brain/metabolism , Stroke/blood , Stroke/diagnosis , Blood Proteins/analysis , Blood Proteins/metabolism , Brain/blood supply , Brain/physiopathology , Brain Ischemia/blood , Brain Ischemia/diagnosis , Brain Ischemia/drug therapy , Cerebral Hemorrhage/chemically induced , Cerebral Hemorrhage/physiopathology , Cerebral Hemorrhage/prevention & control , Humans , Patient Selection , Predictive Value of Tests , Stroke/physiopathology , Thrombolytic Therapy/adverse effects , Thrombolytic Therapy/standards
7.
Cell Death Differ ; 16(11): 1505-14, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19609276

ABSTRACT

Accumulation of unfolded proteins triggers endoplasmic reticulum (ER) stress and is considered a part of the cellular responses to hypoxia. The nascent polypeptide-associated complex (NAC) participates in the proper maturation of newly synthesized proteins. However, thus far, there have been no comprehensive studies on NAC involvement in hypoxic stress. Here, we show that hypoxia activates glycogen synthase kinase-3beta (GSK-3beta) and that the activated GSK-3beta destabilizes alphaNAC with the subsequent apoptosis of the cell. Hypoxia of various cell types and the mouse ischemic brain was associated with rapid downregulation of alphaNAC and ER stress responses involving PERK, ATF4, gamma-taxilin, elF2alpha, Bip, and CHOP. Depletion of alphaNAC by RNA interference specifically activated ER stress responses and caused mitochondrial dysfunction, which resulted in apoptosis through caspase activation. Interestingly, we found that the hypoxic conditions activated GSK-3beta, and that GSK-3beta inhibition prevented alphaNAC protein downregulation in hypoxic cells and rescued the cells from apoptosis. In addition, alphaNAC overexpression increased the viability of hypoxic cells. Taken together, these results suggest that alphaNAC degradation triggers ER stress responses and initiates apoptotic processes in hypoxic cells, and that GSK-3beta may participate upstream in this mechanism.


Subject(s)
Apoptosis , Endoplasmic Reticulum/metabolism , Molecular Chaperones/metabolism , Animals , Brain/pathology , Cell Hypoxia , Cell Line, Tumor , Down-Regulation , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Ischemia/metabolism , Mice , RNA, Small Interfering/metabolism , Signal Transduction , Ubiquitin/metabolism
8.
Neuroscience ; 162(2): 396-403, 2009 Aug 18.
Article in English | MEDLINE | ID: mdl-19401220

ABSTRACT

Neuroglobin (Ngb) is a tissue globin specifically expressed in neurons. Our laboratory and others have shown that Ngb overexpression protects neurons against hypoxia/ischemia, but the underlying mechanisms remain poorly understood. Recent studies demonstrate that hypoxia/ischemia induces a multitude of spatially and temporally regulated responses in gene expression, and initial evidence suggested that Ngb might function in altering biological processes of gene expression. In this study, we asked how Ngb may help regulate genes responsive to hypoxia. Expression of hypoxic response genes following oxygen-glucose deprivation (OGD) was examined using mRNA arrays in neuroglobin-overexpressing transgenic (Ngb-Tg) and wild type (WT) mouse neurons. From a total of 113 genes on the microarray, mRNA expression of 65 genes was detected. Under rest condition, 14 genes were downregulated in Ngb-Tg neurons compared to WT. In WT neurons, after 4-h OGD followed by 4-h reoxygenation (O4/R4), 20 genes were significantly downregulated, and only Fos mRNA was significantly increased. However, out of the 20 downregulated genes in WT neurons, 12 of them were no longer significantly changed in Ngb-Tg neurons: Add1, Arnt2, Camk2g, Cstb, Dr1, Epas1, Gna11, Hif1a, Il6st, Khsrp, Mars and Rara. Among these 12 genes, 8 (Add1, Camk2g, Cstb, Dr1, Epas1, Gna11, Hif1a, Khsrp) were already reduced in Ngb-Tg neurons compared to WT under rest conditions. Additionally, three genes that initially showed no changes in WT neurons (Ctgf, Egfr and Pea15) were downregulated after OGD in the Ngb-Tg neurons. These findings suggest that Ngb overexpression modulates mRNA expression of multiple hypoxic response genes in the early phase after OGD/reoxygenation. Further studies on these gene networks and interactions may lead to better understanding of Ngb in signaling pathways that contribute to neuroprotection.


Subject(s)
Gene Expression Profiling , Globins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Neurons/metabolism , Animals , Apoptosis/genetics , Cell Hypoxia , Cell Survival/genetics , Cells, Cultured , Cerebral Cortex/cytology , Culture Media , Gene Expression Regulation , Globins/genetics , Glucose/deficiency , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neuroglobin , Oligonucleotide Array Sequence Analysis , Oxidative Stress/genetics , Oxygen/metabolism , RNA, Messenger/metabolism , Signal Transduction , Transcription, Genetic
9.
Acta Neurochir Suppl ; 105: 173-8, 2008.
Article in English | MEDLINE | ID: mdl-19066105

ABSTRACT

Intracerebral hemorrhagic transformation is a multifactorial phenomenon in which ischemic brain tissue converts into a hemorrhagic lesion with blood vessel leakage. Hemorrhagic transformation can significantly contribute to additional brain injury after stroke. Especially threatening are the thrombolytic-induced hemorrhages after reperfusion therapy with tissue plasminogen activator (tPA), the only treatment available for ischemic stroke. In this context, it is important to understand its underlying mechanisms and identify early markers of hemorrhagic transformation, so that we can both search for new treatments as well as predict clinical outcomes in patients. In this review, we discuss the emerging mechanisms for hemorrhagic transformation after stroke, and briefly survey potential molecular, genetic, and neuroimaging markers that might be used for early detection of this challenging clinical problem.


Subject(s)
Biomarkers/metabolism , Cerebral Hemorrhage/etiology , Cerebral Hemorrhage/metabolism , Stroke/complications , Animals , Cerebral Hemorrhage/genetics , Humans , Stroke/drug therapy , Tissue Plasminogen Activator/adverse effects
10.
Magn Reson Med ; 60(4): 813-21, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18816832

ABSTRACT

Mechanisms that underlie early ischemic damages to the blood-brain-barrier (BBB) are not well understood. This study presents a novel magnetic resonance imaging (MRI) technique using a widely available pulse sequence and a long-circulating intravascular contrast agent to quantify water movements across the BBB at early stages of stroke progression. We characterized the integrity of the BBB by measuring the flip angle dependence of the water exchange-affected MRI signal intensity, to generate an efficient quantitative index of vascular permeability (WEI, or water exchange index). We performed in vivo MRI experiments to measure the transvascular WEI immediately after the permanent filament occlusion of the middle cerebral artery of mice (n = 5), in which we monitored changes in blood volume (V(b)), apparent diffusion coefficient (ADC), and intra-/extravascular WEI for 4 hours. Statistically significant elevations (P < 0.05) of WEI in the ischemic tissue were observed as early as 1 hour after ischemic onset. Initial reduction of the apparent blood volume (V(app)) in the infarct cortex was followed by a continuous increase of V(app) over time. Although the measured ADC in the ipsilesional cortex continuously decreased, the abnormally high intra-/extravascular WEI remained constant at a significantly elevated level, indicating apparent BBB injury at this early stage of stroke.


Subject(s)
Blood-Brain Barrier/metabolism , Body Water/metabolism , Brain Ischemia/metabolism , Gadolinium DTPA , Image Interpretation, Computer-Assisted/methods , Magnetic Resonance Imaging/methods , Stroke/metabolism , Acute Disease , Animals , Computer Simulation , Male , Mice , Mice, Inbred C57BL , Models, Biological
11.
Neuroscience ; 154(2): 551-5, 2008 Jun 23.
Article in English | MEDLINE | ID: mdl-18495350

ABSTRACT

Brain injury can often result in the subsequent appearance of seizures, suggesting an alteration in neural excitability associated with the balance between neuronal excitation and inhibition. The process by which this occurs has yet to be fully elucidated. The specific nature of the changes in excitation and inhibition is still unclear, as is the process by which the seizures appear following injury. In this study, we investigated the effects of focal cortical compression on electrically-induced localized seizure threshold in rats. Male Long Evans rats were implanted with stimulating screw electrodes in their motor cortices above the regions controlling forelimb movement. Initial seizure threshold was determined in the animals using a ramped electrical stimulation procedure prior to any compression. Following initial threshold determination, animals underwent sustained cortical compression and then following a 24 h recovery period had their seizure thresholds tested again with electrical stimulation. Reliability of threshold measurements was confirmed through repeated measurements of seizure threshold. Localized seizure threshold was significantly lowered following sustained cortical compression as compared with control cases. Taken together, the results here suggest a change in global brain excitability following localized, focal compression.


Subject(s)
Cerebral Cortex/injuries , Seizures/physiopathology , Animals , Behavior, Animal , Cell Death/drug effects , Cell Death/physiology , Electric Stimulation , Electrodes, Implanted , Electrophysiology , Fluorescent Dyes , Forelimb/physiology , In Situ Nick-End Labeling , Indoles , Male , Rats , Rats, Long-Evans , Seizures/psychology , Stereotaxic Techniques
12.
Br J Pharmacol ; 153 Suppl 1: S396-405, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18157168

ABSTRACT

Numerous failures in clinical stroke trials have led to some pessimism in the field. This short review examines the following questions: Can experimental models of stroke be validated? How can combination stroke therapies be productively pursued? Can we achieve neuroprotection without reperfusion? And finally, can we move from a pure neurobiology view of stroke towards a more integrative approach targeting all cell types within the entire neurovascular unit? Emerging data from both experimental models and clinical findings suggest that neurovascular mechanisms may provide new opportunities for treating stroke. Ultimately, both bench-to-bedside and bedside-back-to-bench interactions may be required to overcome the translational hurdles for this challenging disease.


Subject(s)
Cerebrovascular Circulation/physiology , Stroke/pathology , Stroke/therapy , Animals , Blood Vessels/drug effects , Blood Vessels/innervation , Blood Vessels/physiology , Disease Models, Animal , Humans , Neurons/drug effects , Neurons/physiology , Neuroprotective Agents/therapeutic use , Reperfusion , Stroke/drug therapy , Thrombolytic Therapy
13.
Neuroscience ; 150(1): 50-7, 2007 Nov 30.
Article in English | MEDLINE | ID: mdl-17936515

ABSTRACT

Knockout mice deficient in tissue plasminogen activator (tPA) are protected against hippocampal excitotoxicity. But it is unknown whether similar neuroprotection occurs after transient global cerebral ischemia, which is known to selectively affect the hippocampus. In this study, we tested the hypothesis that hippocampal cell death in tPA knockout mice would be reduced after transient global cerebral ischemia, and this neuroprotection would occur concomitantly with amelioration of both intra- and extracellular proteolytic cascades. Wild-type and tPA knockout mice were subjected to 20 min of transient bilateral occlusions of the common carotid arteries. Three days later, Nissl and terminal deoxynucleotidyl transferase biotin-dUTP nick end labeling staining demonstrated that hippocampal cell death was significantly reduced in tPA knockout brains compared with wild-type brains. Caspase-3 and the two major brain gelatinases (matrix metalloproteinase (MMP)-9 and MMP-2) were assessed as representative measurements of intra- and extracellular proteolysis. Post-ischemic levels of caspase-3, MMP-9 and MMP-2 were similarly reduced in tPA knockouts compared with wild-type hippocampi. Taken together, these data suggest that endogenous tPA contributes to hippocampal injury after cerebral ischemia, and these pathophysiologic pathways may involve links to aberrant activation of caspases and MMPs.


Subject(s)
Hippocampus/pathology , Ischemic Attack, Transient/genetics , Ischemic Attack, Transient/pathology , Neurons/pathology , Tissue Plasminogen Activator/deficiency , Animals , Cell Death/physiology , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/physiopathology , In Situ Nick-End Labeling/methods , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphopyruvate Hydratase/metabolism
14.
Neurology ; 66(10): 1550-5, 2006 May 23.
Article in English | MEDLINE | ID: mdl-16717217

ABSTRACT

BACKGROUND: Matrix metalloproteinase-9 (MMP9) is expressed in acute ischemic stroke and up-regulated by tissue plasminogen activator (tPA) in animal models. The authors investigated plasma MMP9 and its endogenous inhibitor, tissue inhibitor of metalloproteinase (TIMP1), in tPA-treated and -untreated stroke patients. METHODS: Nonstroke control subjects and consecutive ischemic stroke patients presenting within 8 hours of onset were enrolled. Blood was sampled within 8 hours and at 24 hours, 2 to 5 days and 4 to 6 weeks. MMP9 and TIMP1 were analyzed by ELISA and gel zymography. RESULTS: Fifty-two cases (26 tPA treated, 26 tPA untreated) and 27 nonstroke control subjects were enrolled. Hyperacute MMP9 was elevated in tPA-treated vs tPA-untreated patients (medians 43 vs 28 ng/mL; p = 0.01). tPA therapy independently predicted hyperacute MMP9 after adjustment for stroke severity, volume, and hemorrhagic transformation (p = 0.01). There was a trend toward lower hyperacute TIMP1 levels in tPA-treated vs tPA-untreated patients (p = 0.06). Hyperacute MMP9 was correlated to poor 3-month modified Rankin Scale outcome (r = 0.58, p = 0.0005). CONCLUSION: Tissue plasminogen activator independently predicted plasma matrix metalloproteinase-9 (MMP9) in the first 8 hours after human ischemic stroke. As MMP9 may be an important mediator of hemorrhagic transformation, alternative thrombolytic agents or therapeutic MMP9 inhibition may increase the safety profile of acute stroke thrombolysis.


Subject(s)
Brain Ischemia/drug therapy , Fibrinolytic Agents/adverse effects , Hemorrhagic Disorders/chemically induced , Matrix Metalloproteinase 9/blood , Thrombolytic Therapy/adverse effects , Tissue Inhibitor of Metalloproteinase-1/blood , Tissue Plasminogen Activator/adverse effects , Aged , Biomarkers , Brain/pathology , Brain Damage, Chronic/etiology , Brain Ischemia/complications , Brain Ischemia/enzymology , Brain Ischemia/pathology , Case-Control Studies , Convalescence , Echo-Planar Imaging , Enzyme Induction/drug effects , Enzyme-Linked Immunosorbent Assay , Female , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/therapeutic use , Follow-Up Studies , Hemorrhagic Disorders/enzymology , Humans , Leukocyte Count , Male , Middle Aged , Severity of Illness Index , Time Factors , Tissue Plasminogen Activator/pharmacology , Tissue Plasminogen Activator/therapeutic use , Treatment Outcome
15.
Neuroreport ; 12(18): 4117-20, 2001 Dec 21.
Article in English | MEDLINE | ID: mdl-11742249

ABSTRACT

Tissue plasminogen activator (tPA) may play a deleterious role after brain injury. Here, we compared the response to traumatic brain injury in tPA knockout (KO) and wildtype (WT) mice after controlled cortical impact. At 6 h after trauma, blood-brain barrier permeability was equally increased in all mice. However, by 24 h specific gravity measurements of brain edema were significantly worse in WT mice than in KO mice. At 1 and 2 days post-trauma, mice showed deficits in rotarod performance, but by day 7 all mice recovered motor function and there were no differences between WT and KO mice. At 7 days, cortical lesion volumes were significantly reduced in KO mice compared with WT mice. However, there were no significant differences in CA3 hippocampal neuron survival. These data suggest that tPA amplifies cortical brain damage and edema in this mouse model of traumatic brain injury.


Subject(s)
Brain Edema/physiopathology , Brain Injuries/physiopathology , Cerebral Cortex/injuries , Tissue Plasminogen Activator/genetics , Animals , Blood-Brain Barrier/physiology , Brain Edema/pathology , Brain Injuries/pathology , Cell Survival/physiology , Cerebral Cortex/blood supply , Cerebral Cortex/pathology , Coloring Agents/pharmacokinetics , Evans Blue/pharmacokinetics , Extracellular Matrix/physiology , Hippocampus/blood supply , Hippocampus/injuries , Hippocampus/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Neurons/pathology , Recovery of Function
16.
Brain Res Brain Res Rev ; 38(1-2): 140-8, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11750930

ABSTRACT

Drug delivery to the brain poses unique challenges. Specialized anatomic and physiological features of the cerebrovasculature and cerebral tissue fluids result in barriers which significantly restrict delivery of a wide range of possible therapeutic agents. In addition to these normal restrictions to brain drug delivery, pathophysiological features and sequelae of acute brain injury will also impact upon the efficiency of drug delivery. This review is focused on acutely damaged brain that occurs after stroke and trauma. Pathophysiological events that may influence drug delivery include blood-brain barrier disruptions, blood flow alterations, edema and increased intracranial pressure, metabolic perturbations, and altered profiles of gene expression and protein synthesis. Careful consideration of these obstacles will provide a framework for further research into the optimization of drug delivery strategies into damaged brain. Without a rigorous assessment of these issues, it may not be possible to translate our mechanistic understanding of acute brain injury into successful clinical therapies.


Subject(s)
Blood-Brain Barrier , Brain Injuries/drug therapy , Brain/drug effects , Stroke/drug therapy , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiology , Brain/blood supply , Brain/physiopathology , Brain Injuries/metabolism , Drug Administration Routes , Humans , Pharmaceutical Preparations/administration & dosage , Stroke/metabolism
17.
Neurosci Lett ; 313(3): 121-4, 2001 Nov 09.
Article in English | MEDLINE | ID: mdl-11682142

ABSTRACT

Efflux of glutamate during cerebral ischemia is known to contribute to brain cell death via processes of excitotoxicity. However, gamma-aminobutyric acid (GABA) is also released during ischemia, and may be protective. In this study, we used in vivo microdialysis to map the efflux of glutamate and GABA from central core and peripheral zones of focal ischemia in mouse brain. We show that the temporal profiles of glutamate and GABA efflux are significantly different in core versus peripheral zones. Calculation of glutamate/GABA ratios demonstrate that, in the core, there is a significant increase above baseline ratios during the first 30 mm of ischemia, which then rapidly renormalizes. In contrast, no significant changes in glutamate/GABA ratios were seen in the ischemic periphery. These data suggest that imbalances in glutamate versus GABA efflux may be an initial trigger of excitotoxic brain damage in the core but not the peripheral zones of focal cerebral ischemia.


Subject(s)
Brain Ischemia/metabolism , Brain/metabolism , Glutamic Acid/metabolism , gamma-Aminobutyric Acid/metabolism , Animals , Blood Flow Velocity , Brain/blood supply , Brain/pathology , Brain Ischemia/pathology , Cerebrovascular Circulation , Disease Models, Animal , Laser-Doppler Flowmetry , Male , Mice , Mice, Inbred C57BL , Microdialysis , Time Factors
18.
Neuroreport ; 12(13): 3003-7, 2001 Sep 17.
Article in English | MEDLINE | ID: mdl-11588620

ABSTRACT

Matrix metalloproteinases (MMPs) may contribute to tissue damage after cerebral ischemia. In this study, wildtype and MMP-2 knockout mice were subjected to permanent and transient (2 h) occlusions of the middle cerebral artery. Gelatin zymography showed that MMP-9 levels were increased in all brains after ischemia. MMP-2 levels did not show a significant increase in wildtype mice, and were not detectable in knockout mice. Laser doppler flowmetry demonstrated equivalent ischemic reductions in perfusion in wildtype and knockout mice. In both permanent and transient occlusion paradigms, there were no statistically significant differences between wildtype and knockout mice in terms of 24 h ischemic lesion volumes. These data suggest that MMP-2 does not contribute to acute tissue damage in this model of focal ischemia.


Subject(s)
Brain Ischemia/enzymology , Cerebral Infarction/enzymology , Matrix Metalloproteinase 2/deficiency , Nerve Degeneration/enzymology , Animals , Brain/blood supply , Brain/enzymology , Brain/pathology , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cerebral Infarction/pathology , Cerebral Infarction/physiopathology , Cerebrovascular Circulation/physiology , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Knockout , Nerve Degeneration/pathology , Nerve Degeneration/physiopathology
19.
J Neurosci ; 21(19): 7724-32, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11567062

ABSTRACT

Deleterious processes of extracellular proteolysis may contribute to the progression of tissue damage after acute brain injury. We recently showed that matrix metalloproteinase-9 (MMP-9) knock-out mice were protected against ischemic and traumatic brain injury. In this study, we examined the mechanisms involved by focusing on relevant MMP-9 substrates in blood-brain barrier, matrix, and white matter. MMP-9 knock-out and wild-type mice were subjected to transient focal ischemia. MMP-9 levels increased after ischemia in wild-type brain, with expression primarily present in vascular endothelium. Western blots showed that the blood-brain barrier-associated protein and MMP-9 substrate zonae occludens-1 was degraded after ischemia, but this was reduced in knock-out mice. There were no detectable changes in another blood-brain barrier-associated protein, occludin. Correspondingly, blood-brain barrier disruption assessed via Evans Blue leakage was significantly attenuated in MMP-9 knock-out mice compared with wild types. In white matter, ischemic degradation of the MMP-9 substrate myelin basic protein was significantly reduced in knock-out mice compared with wild types, whereas there was no degradation of other myelin proteins that are not MMP substrates (proteolipid protein and DM20). There were no detectable changes in the ubiquitous structural protein actin or the extracellular matrix protein laminin. Finally, 24 hr lesion volumes were significantly reduced in knock-out mice compared with wild types. These data demonstrate that the protective effects of MMP-9 gene knock-out after transient focal ischemia may be mediated by reduced proteolytic degradation of critical blood-brain barrier and white matter components.


Subject(s)
Blood-Brain Barrier , Ischemic Attack, Transient/metabolism , Matrix Metalloproteinase 9/deficiency , Nerve Fibers, Myelinated/metabolism , Peptide Hydrolases/metabolism , Actins/metabolism , Animals , Blood-Brain Barrier/physiology , Blotting, Western , Brain/blood supply , Brain/metabolism , Brain/pathology , Cell Survival/physiology , Disease Models, Animal , Extracellular Matrix/metabolism , Immunohistochemistry , Ischemic Attack, Transient/pathology , Laminin/metabolism , Male , Matrix Metalloproteinase 9/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Myelin Basic Protein/metabolism , Phosphoproteins/metabolism , Zonula Occludens-1 Protein
20.
J Cereb Blood Flow Metab ; 21(8): 964-71, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11487732

ABSTRACT

The authors characterized effects of late recombinant tissue plasminogen activator (rt-PA) administration in a rat embolic stroke model with magnetic resonance imaging (MRI), to assess potential MRI correlates, or predictors, or both, of rt-PA-induced hemorrhage. Diffusion-, perfusion-, and postcontrast T1-weighted MRI were performed between 4 and 9 hours and at 24 hours after embolic stroke in spontaneously hypertensive rats. Treatment with either rt-PA or saline was started 6 hours after stroke. A spectrophotometric hemoglobin assay quantified hemorrhage severity. Before treatment, relative cerebral blood flow index (rCBFi) and apparent diffusion coefficient (ADC) in the ischemic territory were 30% +/- 23% and 60% +/- 5% (of contralateral), respectively, which increased to 45% +/- 39% and 68% +/- 4% 2 hours after rt-PA. After 24 hours, rCBFi and ADC were 27% +/- 27% and 59 +/- 5%. Hemorrhage volume after 24 hours was significantly greater in rt-PA-treated animals than in controls (8.7 +/- 3.7 microL vs. 5.1 +/- 2.4 microL, P < 0.05). Before rt-PA administration, clear postcontrast T1-weighted signal intensity enhancement was evident in areas of subsequent bleeding. These areas had lower rCBFi levels than regions without hemorrhage (23% +/- 22% vs. 36% +/- 29%, P < 0.05). In conclusion, late thrombolytic therapy does not necessarily lead to successful reperfusion. Hemorrhage emerged in areas with relatively low perfusion levels and early blood-brain barrier damage. Magnetic resonance imaging may be useful for quantifying effects of thrombolytic therapy and predicting risks of hemorrhagic transformation.


Subject(s)
Stroke/drug therapy , Tissue Plasminogen Activator/therapeutic use , Animals , Brain Ischemia/diagnostic imaging , Brain Ischemia/pathology , Cerebral Hemorrhage/diagnostic imaging , Cerebral Hemorrhage/pathology , Intracranial Embolism , Magnetic Resonance Imaging , Male , Prognosis , Radiography , Rats , Rats, Inbred SHR , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Stroke/diagnostic imaging , Stroke/pathology , Time Factors , Tissue Plasminogen Activator/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...