Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Ann N Y Acad Sci ; 1278: 25-32, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23488559

ABSTRACT

Cerebral ischemia, a focal or global insufficiency of blood flow to the brain, can arise through multiple mechanisms, including thrombosis and arterial hemorrhage. Ischemia is a major driver of stroke, one of the leading causes of morbidity and mortality worldwide. While the general etiology of cerebral ischemia and stroke has been known for some time, the conditions have only recently been considered treatable. This report describes current research in this field seeking to fully understand the pathomechanisms underlying stroke; to characterize the brain's intrinsic injury, survival, and repair mechanisms; to identify putative drug targets as well as cell-based therapies; and to optimize the delivery of therapeutic agents to the damaged cerebral tissue.


Subject(s)
Brain Ischemia/therapy , Neuroprotective Agents/therapeutic use , Stroke/therapy , Brain/pathology , Brain/physiopathology , Brain Ischemia/diagnosis , Brain Ischemia/immunology , Brain Ischemia/physiopathology , Humans , Neuroimaging , Neuroprotective Agents/pharmacology , Stem Cell Transplantation , Stroke/diagnosis , Stroke/immunology , Stroke/physiopathology
2.
Neurosci Res ; 70(2): 230-7, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21300114

ABSTRACT

We examined whether chronic systemic treatment with agonists for peroxisome proliferator-activated receptor α (PPARα) influences neuroinflammation induced by lipopolysaccharide (LPS) injection into the somatosensory cortex in adult mice. Mice were pretreated with Wy-14643 or fenofibrate, both at 30 mg/kg, for 7 days. These treatment protocols increased the amount of PPARα mRNA and active form of PPARα protein in the brain. LPS injection reduced the PPARα mRNA level in the brain. On the contrary, TNFα, IL-1ß, IL-6, iNOS, COX-2, ICAM-1, VCAM-1, and PECAM-1 were elevated at 6h after LPS. Wy-14643 and fenofibrate inhibited the elevations of TNFα, IL-1ß, IL-6, COX-2, ICAM-1, and VCAM-1. Wy-14643, but not fenofibrate, also attenuated the iNOS elevation. At 3 days after LPS, Wy-14643 and fenofibrate showed similar inhibitions in these molecules. LPS injection also elevated IL-6 protein levels in the brain and serum at 6h, which was inhibited by fenofibrate. Histological analyses showed that Wy-14643 and fenofibrate profoundly attenuated microglia/macrophage activation, neutrophil recruitment, and neuronal injury at 3 days after LPS. These findings suggest that activation of PPARα attenuates neuroinflammation in the adult mouse brain, implicating that PPARα may be a potential therapeutic target for CNS diseases in which neuroinflammation plays a substantial role.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Brain/pathology , Inflammation Mediators/administration & dosage , Inflammation Mediators/toxicity , Lipopolysaccharides/toxicity , PPAR alpha/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Fenofibrate/pharmacology , Fenofibrate/therapeutic use , Fibric Acids/pharmacology , Fibric Acids/therapeutic use , Inflammation Mediators/antagonists & inhibitors , Injections, Intraventricular , Lipopolysaccharides/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , PPAR alpha/genetics , Pyrimidines/pharmacology , Pyrimidines/therapeutic use
3.
Brain Res ; 1359: 247-55, 2010 Nov 04.
Article in English | MEDLINE | ID: mdl-20813100

ABSTRACT

Fibrates are activators of peroxisome proliferator-activated receptor (PPAR) α. Pretreatment with fibrates has been shown to protect brain against ischemia in mice. We hypothesized that fibrates elevate superoxide dismutase (SOD) levels in the brain microvessels (BMVs). BMVs were isolated from male C57BL/6 and PPARα null mice that had been treated with fenofibrate or gemfibrozil for 7 days. To examine the effect of discontinuation of fenofibrate, another animal group treated with fenofibrate was examined on post-discontinuation day 3 (D-3). To examine whether SOD elevations attenuate oxidative stress in the ischemic brain, separate animals treated with fenofibrate for 7 days were subjected to 60 minutes of focal ischemia on post-discontinuation day 0 (D-0) or D-3. Fenofibrate (30 mg/kg) increased mRNA levels of all three isoforms of SOD and activity level in BMV on D-0, but these effects were not detected on D-3. The elevations were not detected in PPARα null mice. SOD levels were also elevated by gemfibrozil (30 mg/kg). Fenofibrate significantly reduced superoxide production and protein oxidation in the ischemic brain at 30 minutes after reperfusion. Fenofibrate reduced infarct size measured at 24 hours after reperfusion on D-0; however, the infarct reduction was not seen when ischemia was induced on D-3. These findings suggest that fibrates elevate SOD in BMV through PPARα, which contributes to the infarct reduction, at least in part. Further studies are needed to establish the link between the SOD elevations and the brain protection by fibrates against ischemia.


Subject(s)
Brain/drug effects , Fenofibrate/pharmacology , Gemfibrozil/pharmacology , Microvessels/drug effects , Neuroprotective Agents/pharmacology , Superoxide Dismutase/biosynthesis , Animals , Brain/enzymology , Brain/physiopathology , Brain Ischemia/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microvessels/enzymology , Microvessels/pathology , Oxidative Stress/drug effects , PPAR gamma/metabolism , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
4.
Nutr Metab (Lond) ; 7: 46, 2010 May 27.
Article in English | MEDLINE | ID: mdl-20504373

ABSTRACT

BACKGROUND: Appropriate long-term drinking of red wine is associated with a reduced risk of cardiovascular disease. Resveratrol, a well-known SIRT1 activator is considered to be one of the beneficial components contained in red wine, and also developed as a drug candidate. We previously demonstrated that resveratrol protects brain against ischemic stroke in mice through a PPARalpha-dependent mechanism. Here we report the different effects of the oligomers of resveratrol. METHODS: We evaluated the activation of PPARs by epsilon-viniferin, a resveratrol dimer, and vaticanol C, a resveratrol tetramer, in cell-based reporter assays using bovine arterial endothelial cells, as well as the activation of SIRT1. Moreover, we tested the metabolic action by administering vaticanol C with the high fat diet to wild-type and PPARalpha-knockout male mice for eight weeks. RESULTS: We show that vaticanol C activates PPARalpha and PPARbeta/delta in cell-based reporter assays, but does not activate SIRT1. epsilon-Viniferin shows a similar radical scavenging activity as resveratrol, but neither effects on PPARs and SIRT-1. Eight-week intake of vaticanol C with a high fat diet upregulates hepatic expression of PPARalpha-responsive genes such as cyp4a10, cyp4a14 and FABP1, and skeletal muscle expression of PPARbeta/delta-responsive genes, such as UCP3 and PDK4 (pyruvate dehydrogenase kinase, isoform 4), in wild-type, but not PPARalpha-knockout mice. CONCLUSION: Vaticanol C, a resveratrol tetramer, activated PPARalpha and PPARbeta/delta in vitro and in vivo. These findings indicate that activation of PPARalpha and PPARbeta/delta by vaticanol C may be a novel mechanism, affording beneficial effects against lifestyle-related diseases.

5.
J Cereb Blood Flow Metab ; 30(1): 70-8, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19724288

ABSTRACT

Fibrates, one group of peroxisome proliferator-activated receptor (PPAR) activators, are lipid lowering drugs. Fibrates have been shown to attenuate brain tissue injury after focal cerebral ischemia. In this study, we investigated the impact of fenofibrate on cerebral blood flow (CBF) in male wild type and PPARalpha-null mice. Animals were treated for 7 days with fenofibrate and subjected to 2 h of filamentous middle cerebral artery occlusion and reperfusion under isoflurane anesthesia. Cortical surface CBF was measured by laser speckle imaging. Regional CBF (rCBF) in nonischemic animals was measured by (14)C-iodoantipyrine autoradiography. Fenofibrate did not affect rCBF and mean arterial blood pressure in nonischemic animals. In ischemic animals, laser speckle imaging showed delayed expansions of ischemic area, which was attenuated by fenofibrate. Fenofibrate also enhanced CBF recovery after reperfusion. However, such effects of fenofibrate on CBF in the ischemic brain were not observed in PPARalpha-null mice. These findings show that fenofibrate improves CBF in the ischemic hemisphere. Moreover, fenofibrate requires PPARalpha expression for the cerebrovascular protective effects in the ischemic brain.


Subject(s)
Cerebrovascular Circulation/drug effects , Fenofibrate/pharmacology , Hypolipidemic Agents/pharmacology , Middle Cerebral Artery/physiology , Animals , Antipyrine/analogs & derivatives , Arterial Occlusive Diseases/physiopathology , Autoradiography , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Capillaries/drug effects , Capillaries/pathology , Cerebral Infarction/pathology , Cholesterol/blood , Gene Expression/physiology , Male , Mice , Nitric Oxide Synthase Type III/biosynthesis , Nitric Oxide Synthase Type III/genetics , PPAR alpha/biosynthesis , PPAR alpha/genetics , PPAR alpha/physiology , RNA/biosynthesis , RNA/genetics , Radiopharmaceuticals , Reperfusion Injury/drug therapy , Reperfusion Injury/pathology , Reverse Transcriptase Polymerase Chain Reaction , Triglycerides/blood
6.
Brain Res ; 1294: 183-92, 2009 Oct 19.
Article in English | MEDLINE | ID: mdl-19646426

ABSTRACT

Matrix metalloproteinase (MMP)-9 has been shown to contribute to blood-brain barrier (BBB) disruption, infarct formation, and hemorrhagic transformation after ischemic stroke. The cellular source of MMP-9 detectable in the ischemic brain remains controversial since extracellular molecules in the brain may be derived from blood. We here demonstrate that bone marrow-derived cells are the major source of MMP-9 in the ischemic brain. We made bone marrow chimeric mice with MMP-9 null and wild-type as donor and recipient. After 90 min of transient focal cerebral ischemia, MMP-9 null mice receiving wild-type bone marrow showed comparable outcomes to wild-type in brain MMP-9 levels and BBB disruption (endogenous albumin extravasation) at 1 h post-reperfusion and infarct size at 24 h post-reperfusion. In contrast, wild-type animals replaced with MMP-9 null bone marrow showed barely detectable levels of MMP-9 in the ischemic brain, with attenuations in BBB disruption and infarct size. MMP-9 null mice receiving wild-type bone marrow showed enhanced Evans blue extravasation as early as 1 h post-reperfusion compared to wild-type mice replaced with MMP-9 null bone marrow. These findings suggest that MMP-9 released from bone marrow-derived cells influences the progression of BBB disruption in the ischemic brain.


Subject(s)
Blood-Brain Barrier/physiopathology , Bone Marrow Cells/physiology , Brain Infarction/physiopathology , Brain Ischemia/physiopathology , Matrix Metalloproteinase 9/metabolism , Stroke/physiopathology , Animals , Animals, Genetically Modified , Blood-Brain Barrier/pathology , Bone Marrow Transplantation , Brain/drug effects , Brain/pathology , Brain/physiopathology , Brain Infarction/pathology , Brain Ischemia/pathology , Capillary Permeability , Gelatinases/metabolism , Male , Matrix Metalloproteinase 9/genetics , Mice , Stroke/pathology , Time Factors , Transplantation Chimera
7.
Brain Res ; 1279: 121-30, 2009 Jul 07.
Article in English | MEDLINE | ID: mdl-19427843

ABSTRACT

Fibrates are lipid lowering drugs and found as ligands for peroxisome proliferator-activated receptors (PPARs). A clinical study has shown that one type of fibrate gemfibrozil reduces stroke incidence in men. However, it remains unknown whether gemfibrozil improves outcome after stroke. We hypothesized that prophylactic administration of gemfibrozil improves outcome after ischemic stroke. In this study, we measured the impact of gemfibrozil in two permanent middle cerebral artery occlusion (MCAO) models in young adult male mice on normal diet. First, we tested gemfibrozil in a filamentous MCAO model. Pretreatment with gemfibrozil (30 mg/kg) for 7 days moderately but significantly reduced infarct size at 24 h after MCAO. A higher dose (120 mg/kg) did not attenuate infarct size. Rather, it tended to increase brain swelling. Second, we tested in a distal MCAO model. Gemfibrozil (30 mg/kg) for 7 days before and after stroke significantly attenuated cortical lesion size at 7 days after MCAO. Cortical blood flow measured by laser speckle imaging was improved by gemfibrozil in the ischemic hemisphere. In non-stroke animals gemfibrozil also altered gene expression levels of PPARs in both the aorta and brain in organ specific manners; however, endothelial nitric oxide synthase (eNOS) was not significantly affected. These findings suggested the possibility that the observed infarct reductions and cortical blood flow improvements in ischemic brains were not through eNOS-mediated mechanisms. Further investigations may be meritorious to examine whether prophylactic usage of gemfibrozil against stroke is beneficial.


Subject(s)
Gemfibrozil/administration & dosage , Hypolipidemic Agents/administration & dosage , Infarction, Middle Cerebral Artery/drug therapy , Animals , Aorta/metabolism , Body Temperature/drug effects , Brain/blood supply , Brain/drug effects , Brain/metabolism , Cerebrovascular Circulation/drug effects , Cholesterol/metabolism , Gemfibrozil/chemistry , Gene Expression/drug effects , Hypolipidemic Agents/chemistry , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/metabolism , Peroxisome Proliferator-Activated Receptors/genetics , Peroxisome Proliferator-Activated Receptors/metabolism , RNA, Messenger/metabolism , Severity of Illness Index , Treatment Outcome , Triglycerides/metabolism
8.
Stroke ; 39(7): 2099-104, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18436885

ABSTRACT

BACKGROUND AND PURPOSE: Filamentous middle cerebral artery occlusion (fMCAO) is the most frequently used focal cerebral ischemia model in rodents. The proximity of the ophthalmic artery to the middle cerebral artery suggests that fMCAO induces retinal ischemia. We therefore tested whether fMCAO induces ischemia/reperfusion damage in retina in mice. METHODS: SV129EV mice were subjected to transient (30 or 60 minutes) fMCAO followed by reperfusion under isoflurane anesthesia. Retinal perfusion was evaluated by intravenous injection of fluorescent microspheres combined with fluorescent microscopy using flat-mounted retinas. The fluorescent density of ipsilateral retina relative to contralateral retina was determined in each animal. Retinal injury was assessed by cresyl violet staining and in situ TUNEL. RESULTS: Microsphere analysis demonstrated perfusion defect in the ipsilateral retina after 60 minutes fMCAO and effective restoration after reperfusion. Thirty minutes fMCAO did not produce evident histological changes, even after 2 days of reperfusion. Sixty minutes fMCAO followed by 2 hours reperfusion resulted in extensive cell damage in the inner nuclear (>30%) and ganglion cell (>50%) layers. TUNEL demonstrated very few positive cells, suggesting that damaged cells were mainly undergoing nonapoptotic cell death. CONCLUSIONS: Sixty minutes fMCAO produces retinal injury in SV129EV mice. Potential visual dysfunction should be considered when a particular occlusion period is selected for studying neurological outcomes after fMCAO. Because visual disturbance is often associated with thrombotic/embolic stroke in humans, fMCAO represents an appropriate model for future studies aimed at understanding and ameliorating the changes that lead to retinal damage in these patients.


Subject(s)
Infarction, Middle Cerebral Artery/complications , Ischemia/pathology , Retina/pathology , Amaurosis Fugax , Animals , Apoptosis , In Situ Nick-End Labeling , Infarction, Middle Cerebral Artery/pathology , Isoflurane/pharmacology , Male , Mice , Microscopy, Fluorescence , Microspheres , Models, Anatomic , Oxygen Consumption , Reperfusion Injury
9.
Neurosci Lett ; 352(3): 203-6, 2003 Dec 11.
Article in English | MEDLINE | ID: mdl-14625020

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are ligand-dependent transcription factors which belong to the nuclear receptor family. We examined whether PPARalpha agonists and resveratrol, a polyphenol contained in grapes, protect the brain against ischemia. To investigate whether resveratrol activates PPARs, we performed a cell-based transfection activity assay using luciferase reporter plasmid. PPARalpha and PPARgamma were activated by resveratrol in primary cortical cultures and vascular endothelial cells. Resveratrol (20 mg/kg, 3 days) reduced infarct volume by 36% at 24 h after middle cerebral artery occlusion in wild-type mice. The PPARalpha agonists fenofibrate (30 mg/kg, 3 days) and Wy-14643 (30 mg/kg, days) exerted similar brain protection. However, resveratrol and fenofibrate failed to protect the brain in PPARalpha knockout mice. The data indicate that PPARalpha agonists protect the brain through PPARalpha.


Subject(s)
Brain/drug effects , Fenofibrate/therapeutic use , Receptors, Cytoplasmic and Nuclear/deficiency , Stilbenes/therapeutic use , Stroke/prevention & control , Transcription Factors/deficiency , Animals , Brain/metabolism , Brain/pathology , Cattle , Dose-Response Relationship, Drug , Fenofibrate/pharmacology , Haplorhini , Humans , Mice , Mice, Knockout , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/genetics , Resveratrol , Stilbenes/pharmacology , Stroke/metabolism , Transcription Factors/agonists , Transcription Factors/genetics
10.
Mol Cell Proteomics ; 2(4): 234-41, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12714567

ABSTRACT

Blood-brain barrier (BBB) failure occurs in many neurological diseases and is caused in part by activation of proinflammatory factors including matrix metalloproteinases. Counterbalancing, "BBB protective" cascades have recently been described, including NO-mediated interleukin 6 release by glia. Interleukin 6 has been shown to trigger production of matrix metalloproteinase inhibitors such as alpha2-macroglobulin (alpha2M). We hypothesized that BBB failure may result in increased alpha(2)M release by perivascular astrocytes. This was initially tested in patients undergoing iatrogenic BBB disruption by hyperosmotic mannitol for intra-arterial chemotherapy of brain tumors. Serum samples revealed significantly increased levels of alpha2M at 4 h after BBB disruption by hyperosmotic mannitol. In parallel in vitro experiments, we observed a similar increase of alpha2M release by astrocytes under conditions mimicking BBB failure and perivascular edema. For both experiments, protein analysis was initially performed by bidimensional gel electrophoresis and mass spectrometry followed by Western blotting immunodetection. We conclude that, in addition to proinflammatory changes, BBB failure may also trigger protective release of alpha2M by perivascular astrocytes as well as peripheral source.


Subject(s)
Astrocytes/metabolism , Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , alpha-Macroglobulins/metabolism , Blood-Brain Barrier/drug effects , Brain Neoplasms/drug therapy , Cells, Cultured , Chromatography, Liquid , Coculture Techniques , Electrophoresis, Gel, Two-Dimensional , Humans , Injections, Intra-Arterial , Mannitol/administration & dosage , Mannitol/adverse effects , Mass Spectrometry , Osmosis , Permeability
11.
Neurobiol Dis ; 11(1): 147-54, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12460554

ABSTRACT

Huntington's disease, with its dominant loss of striatal neurons, is triggered by an expanded glutamine tract in huntingtin. To investigate a proposed role for increased activation of the apoptotic cascade in mutant huntingtin's trigger mechanism, we examined huntingtin cleavage and lesion severity after mild ischemic injury in Hdh(Q92) mice. We found activation of calpain and caspase proteases and proteolysis of huntingtin in lesioned striatum. However, huntingtin fragments resembled products of calpain I, not caspase-3, cleavage and turnover was accompanied by augmented levels of full-length normal and mutant protein. By contrast, the number of apoptotic cells, total and striatal infarct size, and degree of neurologic deficit were similar in Hdh(Q92) and wild-type mice, indicating that the disease process neither strongly protected nor sensitized striatal neurons to apoptotic death. Thus, our findings do not support a role for increased apoptosis or caspase-3 cleavage in the mechanism by which mutant huntingtin triggers disease. However, they suggest that calpain activation and huntingtin regulation merit investigation as modifiers of disease progression in neurons injured by the harmful consequences of full-length mutant huntingtin.


Subject(s)
Brain Ischemia/pathology , Corpus Striatum/pathology , Huntington Disease/genetics , Huntington Disease/pathology , Neurons/pathology , Animals , Calpain/metabolism , Caspase 3 , Caspases/metabolism , Cell Death , Huntingtin Protein , Infarction, Middle Cerebral Artery/pathology , Mice , Mice, Mutant Strains , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/enzymology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism
12.
J Cereb Blood Flow Metab ; 22(7): 780-4, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12142563

ABSTRACT

The effect of a nonpeptide CC chemokine receptor antagonist, TAK-779, on ischemic brain injury resulting from 1-hour middle cerebral artery occlusion followed by 48-hour reperfusion was examined in ddY mice. On intracerebroventricular injection of vehicle or TAK-779, infarct volume in the vehicle-treated group was 44.2 +/- 13.2% of the contralateral hemispheric volume, and TAK-779 (25 and 250 ng/mouse) dose-dependently reduced the infarct volume to 35.0 +/- 12.2% and 31.1 +/- 12.9%, respectively. On intravenous injection, infarct volume in the vehicle-treated group was 32.0 +/- 16.1%, and TAK-779 (5 microg per 20 g body weight) significantly reduced this to 22.0 +/- 10.5%. The results showed for the first time that a nonpeptide chemokine receptor antagonist is protective against ischemic brain injury.


Subject(s)
Amides/therapeutic use , CCR5 Receptor Antagonists , Ischemic Attack, Transient/prevention & control , Quaternary Ammonium Compounds/therapeutic use , Amides/administration & dosage , Animals , Brain/pathology , Chemokine CCL4 , Immunohistochemistry , Injections, Intravenous , Injections, Intraventricular , Ischemic Attack, Transient/etiology , Ischemic Attack, Transient/pathology , Leukocytes/pathology , Macrophage Inflammatory Proteins/analysis , Male , Mice , Middle Cerebral Artery/surgery , Peroxidase/analysis , Quaternary Ammonium Compounds/administration & dosage
13.
Neurosci Lett ; 324(2): 117-20, 2002 May 17.
Article in English | MEDLINE | ID: mdl-11988341

ABSTRACT

Excessively released glutamate is neurotoxic. Glutamate transporters maintain the extracellular level of glutamate by uptake into glia or neurons. We examined the role of GLT-1, a glial glutamate transporter, in brain damage resulting from transient focal ischemia in mice. Heterozygous gene deletion of GLT-1 significantly augmented brain swelling resulting from 1 h of middle cerebral artery occlusion and 24 h reperfusion. In addition, this gene deletion significantly increased brain water contents in ischemic hemisphere at 6 h after reperfusion. Moreover, intraperitoneal injection of dihydrokainate (10 mg/kg), a specific inhibitor of GLT-1, augmented brain swelling. These data suggest that GLT-1 limits brain edema resulting from ischemia.


Subject(s)
Brain Edema/genetics , Brain Edema/metabolism , Brain Ischemia/complications , Excitatory Amino Acid Transporter 2/deficiency , Glutamic Acid/metabolism , Kainic Acid/analogs & derivatives , Neuroglia/metabolism , Reperfusion Injury/complications , Animals , Brain/drug effects , Brain/metabolism , Brain/physiopathology , Brain Edema/physiopathology , Excitatory Amino Acid Transporter 2/antagonists & inhibitors , Excitatory Amino Acid Transporter 2/genetics , Extracellular Space/metabolism , Female , Infarction, Middle Cerebral Artery/complications , Kainic Acid/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurotoxins/metabolism , Neurotransmitter Uptake Inhibitors/pharmacology , Water-Electrolyte Balance/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...