Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Brain Sci ; 11(5)2021 May 13.
Article in English | MEDLINE | ID: mdl-34068448

ABSTRACT

The authors wish to make the following corrections to this paper: ref [...].

2.
Neuroscience ; 460: 120-129, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33465414

ABSTRACT

Matrix metalloproteinase-9 (MMP-9) degrades collagen and other cellular matrix proteins. After acute ischemic stroke, increased MMP-9 levels are correlated with hemorrhage, lack of reperfusion and stroke severity. Nevertheless, definitive data that MMP-9 itself causes poor outcomes in ischemic stroke are limited. In a model of experimental ischemic stroke with reperfusion, we examined whether ischemia and recombinant tissue plasminogen activator (r-tPA) therapy affected MMP-9 expression, and we used specific inhibitors to test if MMP-9 affects brain injury and recovery. After stroke, MMP-9 expression increased significantly in the ischemic vs. non-ischemic hemisphere of the brain (p < 0.001). MMP-9 expression in the ischemic, but not the non-ischemic hemisphere, was further increased by r-tPA treatment (p < 0.001). To determine whether MMP-9 expression contributed to stroke outcomes after r-tPA treatment, we tested three different antibody MMP-9 inhibitors. When compared to treatment with r-tPA and saline, treatment with r-tPA and MMP-9 antibody inhibitors significantly reduced brain hemorrhage by 11.3 to 38.6-fold (p < 0.01), brain swelling by 2.8 to 4.3-fold (p < 0.001) and brain infarction by 2.5 to 3.9-fold (p < 0.0001). Similarly, when compared to treatment with r-tPA and saline, treatment with r-tPA and an MMP-9 antibody inhibitor significantly improved neurobehavioral outcomes (p < 0.001), decreased weight loss (p < 0.001) and prolonged survival (p < 0.01). In summary, both prolonged ischemia and r-tPA selectively enhanced MMP-9 expression in the ischemic hemisphere. When administered with r-tPA, specific MMP-9 inhibitors markedly reduced brain hemorrhage, swelling, infarction, disability and death, which suggests that blocking the deleterious effects of MMP-9 may improve outcomes after ischemic stroke.


Subject(s)
Brain Ischemia , Stroke , Animals , Brain Ischemia/drug therapy , Disease Models, Animal , Fibrinolytic Agents/therapeutic use , Ischemia/drug therapy , Matrix Metalloproteinase 9 , Stroke/drug therapy , Tissue Plasminogen Activator
3.
Front Cardiovasc Med ; 7: 608899, 2020.
Article in English | MEDLINE | ID: mdl-33426005

ABSTRACT

Alpha2-antiplasmin (α2AP), the fast-reacting, serine protease inhibitor (serpin) of plasmin, was originally thought to play a key role in protection against uncontrolled, plasmin-mediated proteolysis of coagulation factors and other molecules. However, studies of humans and mice with genetic deficiency of α2AP have expanded our understanding of this serpin, particularly in disease states. Epidemiology studies have shown an association between high α2AP levels and increased risk or poor outcome in cardiovascular diseases. Mechanistic studies in disease models indicate that α2AP stops the body's own fibrinolytic system from dissolving pathologic thrombi that cause venous thrombosis, pulmonary embolism, arterial thrombosis, and ischemic stroke. In addition, α2AP fosters the development of microvascular thrombosis and enhances matrix metalloproteinase-9 expression. Through these mechanisms and others, α2AP contributes to brain injury, hemorrhage and swelling in experimental ischemic stroke. Recent studies also show that α2AP is required for the development of stasis thrombosis by inhibiting the early activation of effective fibrinolysis. In this review, we will discuss the key role played by α2AP in controlling thrombosis and fibrinolysis and, we will consider its potential value as a therapeutic target in cardiovascular diseases and ischemic stroke.

5.
Sci Rep ; 5: 14781, 2015 Oct 19.
Article in English | MEDLINE | ID: mdl-26477507

ABSTRACT

Stroke and vascular dementia are leading causes of morbidity and mortality. Neuroprotective therapies have been proposed but none have proven clinically tolerated and effective. While overstimulation of N-methyl-d-aspartate-type glutamate receptors (NMDARs) is thought to contribute to cerebrovascular insults, the importance of NMDARs in physiological function has made this target, at least in the view of many in 'Big Pharma,' 'undruggable' for this indication. Here, we describe novel NitroMemantine drugs, comprising an adamantane moiety that binds in the NMDAR-associated ion channel that is used to target a nitro group to redox-mediated regulatory sites on the receptor. The NitroMemantines are both well tolerated and effective against cerebral infarction in rodent models via a dual allosteric mechanism of open-channel block and NO/redox modulation of the receptor. Targeted S-nitrosylation of NMDARs by NitroMemantine is potentiated by hypoxia and thereby directed at ischemic neurons. Allosteric approaches to tune NMDAR activity may hold therapeutic potential for cerebrovascular disorders.


Subject(s)
Cerebrovascular Disorders/metabolism , Memantine/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Anura , Apoptosis/drug effects , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cerebrovascular Disorders/drug therapy , Cerebrovascular Disorders/pathology , Long-Term Potentiation/drug effects , Maze Learning/drug effects , Memantine/analogs & derivatives , Memantine/therapeutic use , Membrane Potentials/drug effects , Nitric Oxide/metabolism , Oxidation-Reduction/drug effects , Rats , Synaptic Transmission/drug effects
6.
Cell Rep ; 8(1): 217-28, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-25001280

ABSTRACT

Redox-mediated posttranslational modifications represent a molecular switch that controls major mechanisms of cell function. Nitric oxide (NO) can mediate redox reactions via S-nitrosylation, representing transfer of an NO group to a critical protein thiol. NO is known to modulate neurogenesis and neuronal survival in various brain regions in disparate neurodegenerative conditions. However, a unifying molecular mechanism linking these phenomena remains unknown. Here, we report that S-nitrosylation of myocyte enhancer factor 2 (MEF2) transcription factors acts as a redox switch to inhibit both neurogenesis and neuronal survival. Structure-based analysis reveals that MEF2 dimerization creates a pocket, facilitating S-nitrosylation at an evolutionally conserved cysteine residue in the DNA binding domain. S-Nitrosylation disrupts MEF2-DNA binding and transcriptional activity, leading to impaired neurogenesis and survival in vitro and in vivo. Our data define a molecular switch whereby redox-mediated posttranslational modification controls both neurogenesis and neurodegeneration via a single transcriptional signaling cascade.


Subject(s)
Apoptosis , MEF2 Transcription Factors/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Nitric Oxide/metabolism , Protein Processing, Post-Translational , Transcriptional Activation , Animals , Binding Sites , Cells, Cultured , DNA/metabolism , HEK293 Cells , Humans , MEF2 Transcription Factors/chemistry , MEF2 Transcription Factors/genetics , Mice , Neural Stem Cells/cytology , Oxidation-Reduction , Protein Binding
8.
Article in English | MEDLINE | ID: mdl-25584359

ABSTRACT

Beraprost sodium is a new stable, orally active Prostaglandin I2 analogue. The aim of this study was to determine the effect of beraprost on cognitive dysfunction and locomotor impairment induced by bilateral common carotid artery occlusion in mice. We investigated the ameliorating effect of beraprost through PGI2 IP receptor by studying neurologic deficit assessment and T-maze testing in young and old male C57Bl/6 wild-type (WT) and IP receptor knockout (IP KO) mice following a 12 min bilateral common carotid artery occlusion (BCCAo) and 7 days of reperfusion. Beraprost reversed BCCAo induced cognitive impairment and neurological deficit in a dose dependent manner. Immunohistochemical studies showed attenuation of neuronal cell death, astrogliosis, microglial invasion, and myeloperoxidase (MPO) activity in both young and old WT mice after post treatment with beraprost. Moreover, after BCCAo, phosphorylated cAMP response element binding protein positive cell numbers were increased with beraprost treatment over vehicle treated controls. These results show that beraprost treatment attenuated cognitive dysfunction and neurological deficits induced by BCCAo, and suggest that this effect may be mediated by the neuroprotective effects of treatment.

9.
PLoS One ; 8(8): e71447, 2013.
Article in English | MEDLINE | ID: mdl-23940756

ABSTRACT

Neutrophils are consistently associated with arterial thrombotic morbidity in human clinical studies but the causal basis for this association is unclear. We tested the hypothesis that neutrophils modulate platelet activation and thrombus formation in vivo in a cathepsin G-dependent manner. Neutrophils enhanced aggregation of human platelets in vitro in dose-dependent fashion and this effect was diminished by pharmacologic inhibition of cathepsin G activity and knockdown of cathepsin G expression. Tail bleeding time in the mouse was prolonged by a cathepsin G inhibitor and in cathepsin G knockout mice, and formation of neutrophil-platelet conjugates in blood that was shed from transected tails was reduced in the absence of cathepsin G. Bleeding time was highly correlated with blood neutrophil count in wildtype but not cathepsin G deficient mice. In the presence of elevated blood neutrophil counts, the anti-thrombotic effect of cathepsin G inhibition was greater than that of aspirin and additive to it when administered in combination. Both pharmacologic inhibition of cathepsin G and its congenital absence prolonged the time for platelet thrombus to form in ferric chloride-injured mouse mesenteric arterioles. In a vaso-occlusive model of ischemic stroke, inhibition of cathepsin G and its congenital absence improved cerebral blood flow, reduced histologic brain injury, and improved neurobehavioral outcome. These experiments demonstrate that neutrophil cathepsin G is a physiologic modulator of platelet thrombus formation in vivo and has potential as a target for novel anti-thrombotic therapies.


Subject(s)
Cathepsin G/physiology , Neutrophils/physiology , Platelet Aggregation/genetics , Thrombosis/genetics , Adult , Animals , Blood Platelets/pathology , Blood Platelets/physiology , Female , Hemostasis/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/enzymology , Thrombosis/metabolism
10.
Brain Sci ; 3(3): 1095-108, 2013 Jul 22.
Article in English | MEDLINE | ID: mdl-24634780

ABSTRACT

Transient global cerebral ischemia causes delayed neuronal death in the hippocampal CA1 region. It also induces an up regulation of cyclooxygenase 2 (COX-2), which generates several metabolites of arachidonic acid, known as prostanoids, including Prostaglandin I2 (PGI2). The present study investigated whether the PGI2 IP receptor plays an important role in brain injury after global cerebral ischemia in aged mice. Adult young (2-3 months) and aged (12-15 months) male C57Bl/6 wild-type (WT) or IP receptor knockout (IP KO) mice underwent a 12 min bilateral common carotid artery occlusion (BCCAO) or a sham surgery. Behavior tests (neurologic deficit and T-maze) were performed 3 and 7 days after BCCAO. After seven days of reperfusion, the numbers of cells positive for markers of neurons, astrocytes, microglia, myeloperoxidase (MPO) and phosphorylated CREB (p-CREB) were evaluated immunohistochemically. Interestingly, in young and aged IP KO ischemic mice, there was a significant increase (p < 0.01) in cognitive deficit, hippocampal CA1 pyramidal neuron death, microglia and MPO activation, while p-CREB was reduced as compared to their corresponding WT controls. These data suggest that following ischemia, IP receptor deletion contributes to memory and cognitive deficits regulated by the CREB pathway and that treatment with IP receptor agonists could be a useful target to prevent harmful consequences.

11.
Free Radic Biol Med ; 52(5): 928-36, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22226832

ABSTRACT

Interest in histone deacetylase (HDAC)-based therapeutics as a potential treatment for stroke has grown dramatically. The neuroprotection of HDAC inhibition may involve multiple mechanisms, including modulation of transcription factor acetylation independent of histones. The transcription factor Nrf2 has been shown to be protective in stroke as a key regulator of antioxidant-responsive genes. Here, we hypothesized that HDAC inhibition might provide neuroprotection against mouse cerebral ischemia by activating the Nrf2 pathway. We determined that the classic HDAC inhibitor trichostatin A increased neuronal cell viability after oxygen-glucose deprivation (from an OD value of 0.10±0.01 to 0.25±0.08) and reduced infarct volume in wild-type mice with stroke (from 49.1±3.8 to 21.3±4.6%). In vitro studies showed that HDAC inhibition reduced Nrf2 suppressor Keap1 expression, induced Keap1/Nrf2 dissociation, Nrf2 nuclear translocation, and Nrf2 binding to antioxidant response elements in heme oxygenase 1 (HO1), and caused HO1 transcription. Furthermore, we demonstrated that HDAC inhibition upregulated proteins downstream of Nrf2, including HO1, NAD(P)H:quinone oxidoreductase 1, and glutamate-cysteine ligase catalytic subunit in neuron cultures and brain tissue. Finally, unlike wild-type mice, Nrf2-deficient mice were not protected by pharmacologic inhibition of HDAC after cerebral ischemia. Our studies suggest that activation of Nrf2 might be an important mechanism by which HDAC inhibition provides neuroprotection.


Subject(s)
Brain Ischemia/drug therapy , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/pharmacology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Brain Ischemia/pathology , Cell Hypoxia , Cell Survival/drug effects , Cells, Cultured , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Female , Gene Expression , Gene Expression Regulation/drug effects , Genes, Reporter , Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Kelch-Like ECH-Associated Protein 1 , Luciferases/biosynthesis , Luciferases/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/genetics , Neurons/drug effects , Neurons/physiology , Neuroprotective Agents/therapeutic use , Protein Binding , Response Elements
12.
Exp Transl Stroke Med ; 2(1): 12, 2010 Jul 08.
Article in English | MEDLINE | ID: mdl-20615245

ABSTRACT

BACKGROUND: The prostaglandin E2 EP2 receptor has been shown to be important in dictating outcomes in various neuroinflammatory disorders. Here, we investigated the importance of the EP2 receptor in short- and long-term ischemic outcomes by subjecting wildtype (WT) and EP2 knockout (EP2-/-) mice to two distinct and complementary stroke models [transient and permanent middle cerebral artery occlusion (tMCAO and pMCAO)] and by using the EP2 receptor agonist ONO-AE1-259-01. METHODS: First, WT and EP2-/- mice were subjected to 90-min tMCAO with a monofilament followed by 4-day reperfusion. Second, WT mice were infused intracerebroventricularly with vehicle or ONO-AE1-259-01 45-50 min before being subjected to tMCAO. Finally, WT and EP2-/- mice were subjected to pMCAO and allowed to survive for an extended period of 7 days. RESULTS: Infarct volumes in EP2-/- mice were 55.0 +/- 9.1% larger after tMCAO and 33.3 +/- 8.6% larger after pMCAO than those in WT mice. Neurobehavioral deficits also were significantly greater in the EP2-/- mice. These results suggest that EP2 is beneficial and that activation is sustained for days after the stroke. We also found that pharmacologic activation of EP2 with 1.0- and 2.0-nmol doses of ONO-AE1-259-01 was sufficient to significantly reduce the infarct volume in WT mice compared with that in vehicle-treated controls (20.1 +/- 3.9% vs. 37.1 +/- 4.6%). This reduction correlated with improved neurologic scores. No significant effect on physiologic parameters was observed. CONCLUSION: Together, our results reveal that pharmacologic stimulation of the EP2 receptor has an important beneficial role in cerebral ischemia and might be considered as an adjunct therapy for ischemic stroke.

13.
J Neurosci ; 30(1): 93-8, 2010 Jan 06.
Article in English | MEDLINE | ID: mdl-20053891

ABSTRACT

Death-associated protein kinase (DAPK) is a key player in multiple cell death signaling pathways. We report that DAPK is regulated by DANGER, a partial MAB-21 domain-containing protein. DANGER binds directly to DAPK and inhibits DAPK catalytic activity. DANGER-deficient mouse embryonic fibroblasts and neurons exhibit greater DAPK activity and increased sensitivity to cell death stimuli than do wild-type control cells. In addition, DANGER-deficient mice manifest more severe brain damage after acute excitotoxicity and transient cerebral ischemia than do control mice. Accordingly, DANGER may physiologically regulate the viability of neurons and represent a potential therapeutic target for stroke and neurodegenerative diseases.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Membrane Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Death/physiology , Cell Line , Cells, Cultured , Death-Associated Protein Kinases , Humans , Male , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Binding/physiology
14.
Behav Pharmacol ; 20(7): 567-75, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19654508

ABSTRACT

Oxidative stress plays a crucial role in the progression of cognitive decline in Alzheimer's disease (AD). Considerable attention has been focused on increasing the internal antioxidant defenses in response to AD. This study was designed to examine and compare the pretreatment effects of Pycnogenol (PYC) and vitamin E (Vit E) on cognitive deficits and oxidative damage in the hippocampus and cerebral cortex of intracerebroventricular streptozotocin (ICV-STZ)-infused rats. Rats pretreated with PYC (10 mg/kg), Vit E (100 mg/kg), and vehicle (intraperitoneal; once daily for 3 weeks) were bilaterally injected with ICV-STZ (3 mg/kg), whereas sham rats received the same volume of vehicle. After 2 weeks of ICV-STZ infusion, rats were tested for cognitive performance using passive avoidance and water maze tasks, and then killed for biochemical assays. ICV-STZ induced significant declines in cognitive performance and choline acetyltransferase activity in the hippocampus, which were significantly attenuated with PYC and Vit E. Pretreatment with PYC and Vit E produced a significantly enhanced glutathione level and Na+/K+-ATPase activity and decreased thiobarbituric acid reactive substances and protein carbonyl. These findings suggest that PYC and Vit E may provide a promising approach for the treatment of oxidative stress-related neurodegeneration in conditions such as AD.


Subject(s)
Alzheimer Disease/prevention & control , Antioxidants/therapeutic use , Cognition Disorders/prevention & control , Flavonoids/therapeutic use , Oxidative Stress/drug effects , Streptozocin/administration & dosage , Vitamin E/therapeutic use , Alzheimer Disease/metabolism , Animals , Antioxidants/pharmacology , Avoidance Learning/drug effects , Cerebral Cortex/drug effects , Choline O-Acetyltransferase/metabolism , Cognition Disorders/chemically induced , Cognition Disorders/metabolism , Disease Models, Animal , Drug Evaluation, Preclinical , Flavonoids/pharmacology , Glutathione/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Infusions, Intraventricular , Male , Maze Learning/drug effects , Plant Extracts , Protein Carbonylation/drug effects , Rats , Rats, Wistar , Sodium-Potassium-Exchanging ATPase/metabolism , Thiobarbituric Acid Reactive Substances/metabolism , Vitamin E/pharmacology
15.
Neurotox Res ; 15(1): 62-70, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19384589

ABSTRACT

Although some of the COX-2 metabolites and prostaglandins have been implicated in stroke and excitotoxicity, the role of prostaglandin F(2alpha) (PGF(2alpha)) and its FP receptor have not been elucidated in the pathogenesis of ischemic-reperfusion (I/R) brain injury. Here we investigated the FP receptor's contribution in a unilateral middle cerebral artery (MCA) occlusion model of focal cerebral ischemia in mice. The MCA in wild type (WT) and FP knockout (FP(-/-)) C57BL/6 male mice was transiently occluded with a monofilament for 90 min. After 96 h of reperfusion, the FP(-/-) mice had 25.3% less neurological deficit (P < 0.05) and 34.4% smaller infarct volumes (P < 0.05) than those of the WT mice. In a separate cohort, physiological parameters were monitored before, during, and after ischemia, and the results revealed no differences between the groups. Because excitotoxicity is an acute mediator of stroke outcome, the effect of acute NMDA-induced neurotoxicity was also tested. Forty-eight hours after unilateral intrastriatal NMDA injection, excitotoxic brain damage was 20.8% less extensive in the FP(-/-) mice (P < 0.05) than in the WT counterparts, further supporting the toxic contribution of the FP receptor in I/R injury. Additionally, we investigated the effect of post-treatment with the FP agonist latanoprost in mice subjected to MCA occlusion; such treatment resulted in an increase in neurological deficit and infarct size in WT mice (P < 0.05), though no effects were observed in the latanoprost-treated FP(-/-) mice. Together, the results suggest that the PGF(2alpha) FP receptor significantly enhances cerebral ischemic and excitotoxic brain injury and that these results are of importance when planning for potential development of therapeutic drugs to treat stroke and its acute and/or long term consequences.


Subject(s)
Brain Injuries/etiology , Brain Injuries/metabolism , Brain Ischemia/complications , Receptors, Prostaglandin/metabolism , Analysis of Variance , Animals , Antihypertensive Agents/pharmacology , Brain Infarction/etiology , Brain Infarction/prevention & control , Brain Injuries/genetics , Brain Injuries/prevention & control , Brain Ischemia/genetics , Disease Models, Animal , Excitatory Amino Acid Agonists/toxicity , Latanoprost , Mice , Mice, Inbred C57BL , Mice, Knockout , N-Methylaspartate/toxicity , Nervous System Diseases/etiology , Nervous System Diseases/genetics , Prostaglandins F, Synthetic/pharmacology , Receptors, Prostaglandin/genetics , Receptors, Prostaglandin E/deficiency , Receptors, Prostaglandin E/genetics
16.
J Cereb Blood Flow Metab ; 29(5): 953-64, 2009 May.
Article in English | MEDLINE | ID: mdl-19277051

ABSTRACT

Hemoproteins undergo degradation during hypoxic/ischemic conditions, but the pro-oxidant free heme that is released cannot be recycled and must be degraded. The extracellular heme associates with its high-affinity binding protein, hemopexin (HPX). Hemopexin is shown here to be expressed by cortical neurons and it is present in mouse cerebellum, cortex, hippocampus, and striatum. Using the transient ischemia model (90-min middle cerebral artery occlusion followed by 96-h survival), we provide evidence that HPX is protective in the brain, as neurologic deficits and infarct volumes were significantly greater in HPX(-/-) than in wild-type mice. Addressing the potential protective HPX cellular pathway, we observed that exogenous free heme decreased cell survival in primary mouse cortical neuron cultures, whereas the heme bound to HPX was not toxic. Heme-HPX complexes induce HO1 and, consequently, protect primary neurons against the toxicity of both heme and pro-oxidant tert-butyl hydroperoxide; such protection was decreased in HO1(-/-) neuronal cultures. Taken together, these data show that HPX protects against heme-induced toxicity and oxidative stress and that HO1 is required. We propose that the heme-HPX system protects against stroke-related damage by maintaining a tight balance between free and bound heme. Thus, regulating extracellular free heme levels, such as with HPX, could be neuroprotective.


Subject(s)
Heme/physiology , Hemopexin/physiology , Infarction, Middle Cerebral Artery/metabolism , Neurons/drug effects , Animals , Blotting, Western , Brain/drug effects , Brain/metabolism , Brain/pathology , Cell Death/drug effects , Cell Line , Disease Models, Animal , Heme/biosynthesis , Heme/pharmacology , Heme Oxygenase-1/biosynthesis , Hemopexin/biosynthesis , Hemopexin/pharmacology , Humans , Immunohistochemistry , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Neurons/metabolism , Oxidative Stress/drug effects , Reverse Transcriptase Polymerase Chain Reaction
17.
J Neuroimmunol ; 208(1-2): 87-93, 2009 Mar 31.
Article in English | MEDLINE | ID: mdl-19203800

ABSTRACT

Ischemic stroke is one of the leading causes of mortality and morbidity in humans. During brain ischemia and the subsequent reperfusion that occurs with stroke, the generation of the so-called "proinflammatory" prostaglandin E(2) (PGE(2)) increases significantly. Therefore, interest is growing regarding the differential functions of the individual PGE(2) receptors (EP1-4) and their relative contribution to brain damage following ischemic and inflammatory stimuli. Here, we address the contribution of the EP3 receptor in dictating early outcomes after transient cerebral ischemia. An oxygen-glucose deprivation (OGD)-induced in vitro model of brain ischemia was used in mouse hippocampal slice cultures. For transient ischemia, the right middle cerebral artery (MCA) of wildtype (WT) and EP3 knockout (EP3(-/-)) C57BL/6 male mice was occluded for 90 min and reperfused for 48 or 96 h, after which neurobehavioral scores and infarct volumes were determined. Mean arterial blood pressure, pH, blood gases (PaO(2) and PaCO(2)), cerebral blood flow, and body temperature were also determined before and during ischemia and reperfusion. OGD-induced cell death was significantly lower in brain slice cultures of EP3(-/-) mice than in those of WT mice. EP3(-/-) mice that underwent transient ischemia had significantly smaller infarct volumes than did WT mice at 48 h, but this difference was not sustained at 96 h. Neurological score deficits correlated with infarct volume, but no significant differences in the physiological parameters monitored were detected between the two genotypes. The results further support a role for EP3 receptors in contributing to acute ischemic stroke, but EP3 is not likely the sole contributor to the long-term detrimental consequences of PGE(2).


Subject(s)
Brain Injuries/metabolism , Brain Injuries/prevention & control , Brain Ischemia/metabolism , Dinoprostone/metabolism , Receptors, Prostaglandin E/deficiency , Animals , Brain Injuries/genetics , Brain Ischemia/genetics , Gene Deletion , Glucose/metabolism , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxygen/metabolism , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E, EP3 Subtype , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control
18.
Stroke ; 39(12): 3389-96, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18845796

ABSTRACT

BACKGROUND AND PURPOSE: Ginkgo biloba extracts are now prescribed in several countries for their reported health benefits, particularly for medicinal properties in the brain. The standardized Ginkgo extract, EGb761, has been reported to protect neurons against oxidative stress, but the underlying mechanisms are not fully understood. METHODS: To characterize the oral consumption of EGb761 in transient ischemia, we performed the middle cerebral artery occlusion (MCAO) filament model in wild-type and heme oxygenase 1 (HO-1) knockouts. Mice were pretreated for 7 days before the transient occlusion or posttreated acutely during reperfusion; then neurobehavioral scores and infarct volumes were assessed. Furthermore, primary cortical neuronal cultures were used to investigate the contribution of the antioxidant enzyme HO-1 in the EGb761-associated cytoprotection. RESULTS: Mice that were pretreated with EGb761 had 50.9+/-5.6% less neurological dysfunction and 48.2+/-5.3% smaller infarct volumes than vehicle-treated mice; this effect was abolished in HO-1 knockouts. In addition to the prophylactic properties of EGb761, acute posttreatment 5 minutes and 4.5 hours after reperfusion also led to significant reduction in infarct size (P<0.01). After our previous demonstration that EGb761 significantly induced HO-1 levels in a dose- and time-dependent manner in neuronal cultures, here we revealed that this de novo HO-1 induction was required for neuroprotection against free radical damage and excitotoxicity as it was significantly attenuated by the enzyme inhibitor. CONCLUSIONS: These results demonstrate that EGb761 could be used as a preventive or therapeutic agent in cerebral ischemia and suggest that HO-1 contributes, at least in part, to EGb761 neuroprotection.


Subject(s)
Antioxidants/therapeutic use , Brain Damage, Chronic/prevention & control , Brain Ischemia/drug therapy , Excitatory Amino Acid Antagonists/therapeutic use , Ginkgo biloba , Heme Oxygenase-1/physiology , Infarction, Middle Cerebral Artery/drug therapy , Membrane Proteins/physiology , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Plant Extracts/therapeutic use , Reperfusion Injury/prevention & control , Animals , Antioxidants/administration & dosage , Antioxidants/pharmacology , Brain Damage, Chronic/etiology , Brain Ischemia/enzymology , Cells, Cultured/drug effects , Cells, Cultured/enzymology , Cerebrovascular Circulation/drug effects , Cyclopentanes/pharmacology , Drug Evaluation, Preclinical , Enzyme Induction/drug effects , Excitatory Amino Acid Antagonists/administration & dosage , Excitatory Amino Acid Antagonists/pharmacology , Furans/pharmacology , Ginkgolides/pharmacology , Glutamic Acid/pharmacology , Heme Oxygenase-1/biosynthesis , Heme Oxygenase-1/deficiency , Heme Oxygenase-1/genetics , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/enzymology , Male , Membrane Proteins/biosynthesis , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/physiology , Neurons/enzymology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Plant Extracts/administration & dosage , Plant Extracts/pharmacology , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Response Elements/drug effects
19.
Basic Clin Pharmacol Toxicol ; 101(4): 246-53, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17845506

ABSTRACT

Majun Baladar (MB), a traditional herbal formulation of the Unani system of medicine, was studied for its efficacy against cerebral ischaemia-induced oxidative damage in hippocampus and associated neurobehavioural deficits. Adult male Wistar rats were divided into four groups. The first group was sham, the second group was ischaemic (MCAO: middle cerebral artery occluded) and the third group was a MB pre-treated ischaemic group (MCAO + MB). The fourth group was given MB (1.05 g/kg) orally for 15 days as a drug control. The middle cerebral artery was occluded for 2 hr and reperfused for 22 hr in the ischaemic as well as the drug pre-treated group. The activity of the various enzymatic antioxidants like glutathione peroxidase, glutathione reductase, glutathione S-transferase and non-enzymatic antioxidants, glutathione along with levels of lipid peroxidation were evaluated. Cerebral ischaemic rats showed elevated level of lipid peroxidation and decreased levels of various antioxidants significantly over sham values. As a result of MB pre-treatment, the level of lipid peroxidation was found to be significantly depleted as compared to the ischaemic group. Furthermore, depleted levels of glutathione and the activity of glutathione peroxidase, glutathione S-transferase and glutathione reductase were restored significantly in MB treated group. Majun Baladar exhibited a significant improvement in neurobehavioural activities in the drug pre-treated animals as compared to the ischaemic group as evidenced by the grip strength test, Rota-Rod and video path analysis. The results of the present study provide baseline information regarding the neuroprotective efficacy of MB and also open a window for a potent therapeutic use of this traditional herbal Unani medicine.


Subject(s)
Antioxidants/therapeutic use , Ischemic Attack, Transient/drug therapy , Neuroprotective Agents/therapeutic use , Plant Extracts/therapeutic use , Animals , Antioxidants/administration & dosage , Antioxidants/pharmacology , Behavior, Animal/drug effects , Glutathione/analysis , Glutathione Peroxidase/analysis , Glutathione Reductase/analysis , Glutathione Transferase/analysis , Hippocampus/drug effects , Hippocampus/physiopathology , Ischemic Attack, Transient/physiopathology , Lipid Peroxidation/drug effects , Male , Medicine, Traditional , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Phytotherapy , Plant Extracts/administration & dosage , Plant Extracts/pharmacology , Psychomotor Performance/drug effects , Rats
20.
J Neurosci Res ; 85(11): 2433-40, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17600836

ABSTRACT

The lipid mediator prostaglandin E2 (PGE2) exhibits diverse biologic activity in a variety of tissues. Four PGE2 receptor subtypes (EP1-4) are involved in various physiologic and pathophysiologic conditions, but differ in tissue distribution, ligand-binding affinity, and coupling to intracellular signaling pathways. To characterize the role of the EP1 receptor, physiologic parameters (mean arterial blood pressure, pH, blood gases PaO2 and PaCO2, and body temperature), cerebral blood flow (CBF), and neuronal cell death were studied in a middle cerebral artery occlusion model of ischemic stroke in wild-type (WT) and EP1 knockout (EP1-/-) mice. The right middle cerebral artery was occluded for 60 min, and absolute CBF was measured by [14C] iodoantipyrine autoradiography. The effect of EP1 receptor on oxidative stress in neuronal cultures was investigated. Although no differences were observed in the physiologic parameters, CBF was significantly (P < 0.01) higher in EP1-/- mice than in WT mice, suggesting a role for this receptor in physiologic and pathophysiologic control of vascular tone. Similarly, neuronal cultures derived from EP1-/- mice were more resistant (90.6 +/- 5.8% viability) to tert-butyl hydroperoxide-induced oxidative stress than neurons from WT mice (39.6 +/- 17.2% viability). The EP1 receptor antagonist SC-51089 and calcium channel blocker verapamil each attenuated the neuronal cell death induced by PGE2. Thus, the prostanoid EP1 receptor plays a significant role in regulating CBF and neuronal cell death. These findings suggest that pharmacologic modulation of the EP1 receptor might be a means to improve CBF and neuronal survival during ischemic stroke.


Subject(s)
Brain/blood supply , Cerebrovascular Circulation/physiology , Infarction, Middle Cerebral Artery/metabolism , Receptors, Prostaglandin E/metabolism , Stroke/metabolism , Animals , Brain/metabolism , Brain/pathology , Cell Survival/physiology , Cells, Cultured , Female , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/pathology , Mice , Mice, Knockout , Neurons/metabolism , Neurons/pathology , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E, EP1 Subtype , Stroke/etiology , Stroke/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...