Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Neurochem Res ; 44(4): 897-904, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30656593

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a severe neurodegenerative disease caused by the loss of upper and lower motor neurons resulting in muscle weakness and paralysis. Recently, VGF, a neuropeptide that is a precursor of bioactive polypeptides, was found to be decreased in ALS patients, and its inducer exerted protective effects in models of ALS. These findings suggested that VGF was involved in the pathology of ALS. Here, we investigated the neuroprotective effects of various VGF-derived peptides in an in vitro ALS model. We applied seven VGF-derived peptides (TLQP-21, AQEE-30, AQEE-11, LQEQ-19, QEEL-16, LENY-13, and HVLL-7) to the motor neuron-derived cell line, NSC-34, expressing SOD1G93A, which is one of the mutated proteins responsible for familial ALS. Nuclear staining revealed that AQEE-30 and LQEQ-19, which are derived from the C-terminal polypeptide of the VGF precursor protein, attenuated neuronal cell death. Furthermore, immunoblot analysis demonstrated that LQEQ-19 promoted the phosphorylation of Akt and extracellular signal-regulated kinase (ERK) 1/2, and inhibiting these mitogen-activated MAP kinases (MAPKs) with phosphoinositide 3-kinase or MEK/ERK inhibitors, eliminated the neuroprotective effects of LQEQ-19. In conclusion, these results suggest that VGF C-terminal peptides exert their neuroprotective effects via activation of MAPKs such as Akt and ERK1/2. Furthermore, these findings indicate that VGF-derived peptides have potential application in ALS therapy.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Neuropeptides/therapeutic use , Neuroprotective Agents/pharmacology , Amyotrophic Lateral Sclerosis/prevention & control , Animals , Cell Line , Dose-Response Relationship, Drug , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase Kinases/metabolism , Nerve Growth Factors , Neuropeptides/pharmacology , Neuroprotective Agents/therapeutic use , Rats , Treatment Outcome
2.
Neuroscience ; 277: 123-31, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25010402

ABSTRACT

Glycoprotein nonmetastatic melanoma protein B (GPNMB) is a type I transmembrane protein reported to have neuroprotective effects in the neurodegenerative disease amyotrophic lateral sclerosis (ALS). We investigated whether GPNMB is also neuroprotective against brain ischemia-reperfusion injury (IRI). Focal ischemia/reperfusion injury was induced via filament middle cerebral artery occlusion (MCAO) for 2h, followed by reperfusion upon withdrawal of the filament. We assessed the neuroprotective effects of GPNMB using transgenic (Tg) mice which over expressing GPNMB or recombinant GPNMB which has the sequence of human extracellular GPNMB. The results showed that GPNMB was up-regulated after IRI, and that genomic over-expression of GPNMB significantly ameliorated infarct volume. Next, we investigated the protective mechanisms of GPNMB via Western blotting and immunohistochemistry (IHC). Phosphorylation of extracellular signal-regulated kinase 1 and 2 (ERK1/2), and protein kinase B (Akt), were increased in the GPNMB Tg group according to Western blotting data. IHC analysis showed that GPNMB was expressed not only in neurons, but also in astrocytes, produced labeling patterns similar to that in human brain ischemia. Furthermore, recombinant GPNMB also decreased infarction volume. These results indicate that GPNMB protected neurons against IRI, and phosphor-Akt and phosphor-ERK might be a part of the protective mechanisms, and that the neuroprotective effect of GPNMB was seemingly induced by the extracellular sequence of GPNMB. In conclusion, these findings indicate that GPNMB has neuroprotective effects against IRI, via phosphorylation of ERK1/2 and Akt, suggesting that GPNMB may be a therapeutic target for ischemia-reperfusion injuries.


Subject(s)
Brain Ischemia/metabolism , Eye Proteins/metabolism , Membrane Glycoproteins/metabolism , Reperfusion Injury/metabolism , Adult , Aged , Animals , Astrocytes/metabolism , Astrocytes/pathology , Brain/metabolism , Brain/pathology , Brain Ischemia/pathology , Disease Models, Animal , Epilepsy/metabolism , Extracellular Space/metabolism , Eye Proteins/genetics , Female , Humans , Infarction, Middle Cerebral Artery , MAP Kinase Signaling System/physiology , Male , Membrane Glycoproteins/genetics , Mice , Mice, Transgenic , Neurons/metabolism , Neurons/pathology , Proto-Oncogene Proteins c-akt/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Reperfusion Injury/pathology
3.
Br J Pharmacol ; 170(2): 341-51, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23763343

ABSTRACT

BACKGROUND AND PURPOSE: Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder with no effective treatment. Fasudil hydrochloride (fasudil), a potent rho kinase (ROCK) inhibitor, is useful for the treatment of ischaemic diseases. In previous reports, fasudil improved pathology in mouse models of Alzheimer's disease and spinal muscular atrophy, but there is no evidence in that it can affect ALS. We therefore investigated its effects on experimental models of ALS. EXPERIMENTAL APPROACH: In mice motor neuron (NSC34) cells, the neuroprotective effect of hydroxyfasudil (M3), an active metabolite of fasudil, and its mechanism were evaluated. Moreover, the effects of fasudil, 30 and 100 mg·kg(-1), administered via drinking water to mutant superoxide dismutase 1 (SOD1(G93A)) mice were tested by measuring motor performance, survival time and histological changes, and its mechanism investigated. KEY RESULTS: M3 prevented motor neuron cell death induced by SOD1(G93A). Furthermore, M3 suppressed both the increase in ROCK activity and phosphorylated phosphatase and tensin homologue deleted on chromosome 10 (PTEN), and the reduction in phosphorylated Akt induced by SOD1(G93A). These effects of M3 were attenuated by treatment with a PI3K inhibitor (LY294002). Moreover, fasudil slowed disease progression, increased survival time and reduced motor neuron loss, in SOD1(G93A) mice. Fasudil also attenuated the increase in ROCK activity and PTEN, and the reduction in Akt in SOD1(G93A) mice. CONCLUSIONS AND IMPLICATIONS: These findings indicate that fasudil may be effective at suppressing motor neuron degeneration and symptom progression in ALS. Hence, fasudil may have potential as a therapeutic agent for ALS treatment.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Amyotrophic Lateral Sclerosis/drug therapy , Motor Neurons/drug effects , Protein Kinase Inhibitors/pharmacology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/administration & dosage , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Disease Models, Animal , Disease Progression , Dose-Response Relationship, Drug , Male , Mice, Transgenic , Motor Neurons/pathology , Neuroprotective Agents/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins c-akt/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Survival Rate , rho-Associated Kinases/antagonists & inhibitors
4.
Neuroscience ; 220: 302-12, 2012 Sep 18.
Article in English | MEDLINE | ID: mdl-22710066

ABSTRACT

Thrombolysis with tissue plasminogen activator (tPA) is the only FDA-approved therapy for acute ischemic stroke. However, hemorrhagic transformation, neurotoxicity, and a short treatment time window comprise major limitations for thrombolytic therapy. The purpose of the present study was to investigate whether fasudil, a Rho kinase (ROCK) inhibitor, would prevent tPA-associated hemorrhagic transformation and extend the reperfusion window in an experimental stroke model in mice. Mice subjected to 6-h middle cerebral artery occlusion were treated with delayed tPA alone, with combined tPA plus fasudil, or with a vehicle. We used histological and neurobehavioral measures to assess the effects of the treatment at 18 h and 7 days after the reperfusion. To investigate the mechanism of fasudil's beneficial effects further, we also performed an in vitro study with tPA and fasudil in human brain microvascular endothelial cells. Combination therapy with tPA plus fasudil prevented the development of hemorrhagic transformation, but did not reduce the infarct volumes. These changes significantly reduced mortality and increased locomotor activity at 7 days after the reperfusion. Furthermore, the administration of both drugs prevented injury to the human brain endothelial cells via the reduction of matrix metalloproteinase-9 (MMP-9) activity. These findings indicate that fasudil prevents the hemorrhagic transformation induced by focal cerebral ischemia in mice treated with tPA, at least in part, by inhibiting the increased activity of MMP-9 in endothelial cells.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Cerebral Hemorrhage/prevention & control , Fibrinolytic Agents/adverse effects , Matrix Metalloproteinase 9/metabolism , Protein Kinase Inhibitors/pharmacology , Tissue Plasminogen Activator/adverse effects , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Blotting, Western , Cells, Cultured , Cerebral Hemorrhage/chemically induced , Cerebral Hemorrhage/enzymology , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Humans , Male , Mice , Rats, Sprague-Dawley , Stroke/drug therapy
5.
Braz. j. med. biol. res ; 45(3): 212-215, Mar. 2012. ilus, tab
Article in English | LILACS | ID: lil-618043

ABSTRACT

Agmatine, an endogenous polyamine and putative neuromodulator, is known to have neuroprotective effects on various neurons in the central nervous system. We determined whether or not topically administered agmatine could reduce ischemic retinal injury. Transient ocular ischemia was achieved by intraluminal occlusion of the middle cerebral artery of ddY mice (30-35 g) for 2 h, which is known to also induce occlusion of the ophthalmic artery. In the agmatine group (N = 6), a 1.0 mM agmatine-containing ophthalmic solution was administered four times daily for 2 weeks before occlusion. In the control group (N = 6), a 0.1 percent hyaluronic acid ophthalmic solution was instilled at the same times. At 22 h after reperfusion, the eyeballs were enucleated and the retinal sections were stained by terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL). Transient ocular ischemia induced apoptosis of retinal cells in the entire retinal layer, and topically administered agmatine can significantly reduce this ischemic retinal injury. The proportion of apoptotic cells was definitely decreased (P < 0.001; Kruskal-Wallis test). Overall, we determined that topical agmatine application effectively decreases retinal damage in an in vivo ocular ischemic injury model. This implies that agmatine is a good candidate as a direct neuroprotective agent for eyes with ocular ischemic diseases.


Subject(s)
Animals , Male , Mice , Agmatine/administration & dosage , Arterial Occlusive Diseases/complications , Ischemia/drug therapy , Neuroprotective Agents/administration & dosage , Ophthalmic Artery , Retinal Diseases/drug therapy , Disease Models, Animal , Ischemia/etiology , Retinal Diseases/etiology
6.
Braz J Med Biol Res ; 45(3): 212-5, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22331138

ABSTRACT

Agmatine, an endogenous polyamine and putative neuromodulator, is known to have neuroprotective effects on various neurons in the central nervous system. We determined whether or not topically administered agmatine could reduce ischemic retinal injury. Transient ocular ischemia was achieved by intraluminal occlusion of the middle cerebral artery of ddY mice (30-35 g) for 2 h, which is known to also induce occlusion of the ophthalmic artery. In the agmatine group (N = 6), a 1.0 mM agmatine-containing ophthalmic solution was administered four times daily for 2 weeks before occlusion. In the control group (N = 6), a 0.1% hyaluronic acid ophthalmic solution was instilled at the same times. At 22 h after reperfusion, the eyeballs were enucleated and the retinal sections were stained by terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL). Transient ocular ischemia induced apoptosis of retinal cells in the entire retinal layer, and topically administered agmatine can significantly reduce this ischemic retinal injury. The proportion of apoptotic cells was definitely decreased (P < 0.001; Kruskal-Wallis test). Overall, we determined that topical agmatine application effectively decreases retinal damage in an in vivo ocular ischemic injury model. This implies that agmatine is a good candidate as a direct neuroprotective agent for eyes with ocular ischemic diseases.


Subject(s)
Agmatine/administration & dosage , Arterial Occlusive Diseases/complications , Ischemia/drug therapy , Neuroprotective Agents/administration & dosage , Ophthalmic Artery , Retinal Diseases/drug therapy , Animals , Disease Models, Animal , Ischemia/etiology , Male , Mice , Retinal Diseases/etiology
7.
Neuroscience ; 205: 39-48, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22244977

ABSTRACT

Delayed activation of tissue plasminogen activator (tPA) can lead to the disruption of the blood-brain barrier (BBB), resulting in hemorrhagic complications. In the present study, we focused on tight junction proteins (TJPs), occludin, zona occludens (ZO)-1, and claudin-5, which are important structural components of the BBB, and investigated whether inhibition of matrix metalloproteinases (MMPs) provides a protective effect against hemorrhagic complications induced by tPA. We subjected mice to 6-h filamental middle cerebral artery occlusion (MCAO) with vehicle, delayed tPA alone, or combined tPA (10 mg/kg, i.v.) plus GM6001 (100 mg/kg, i.p.), a broad-spectrum MMP inhibitor. We evaluated brain hemoglobin and the expression of MMP-9 and TJPs by immunoblotting. GM6001 significantly reduced tPA-elevated brain hemoglobin, MMP-9, and inhibited the degradation of occludin and ZO-1 induced by tPA, but not claudin-5. Treatment with GM6001 also significantly prevented the decrease in the survival rate and the reduction in locomotor activity caused by tPA at 7 days after ischemia/reperfusion. Furthermore, GM6001 treatment also significantly prevented cell damage, determined by release of lactase dehydrogenase (LDH) activity, and the decrease in transendothelial electrical resistance (TEER) induced by tPA. These findings indicate that GM6001 prevented the hemorrhagic complications and improved the behavioral abnormalities induced by tPA, partly via protection of TJPs. This suggests that GM6001 may be a useful candidate for combination therapy against the hemorrhagic complications induced by tPA.


Subject(s)
Cerebral Hemorrhage/chemically induced , Cerebral Hemorrhage/prevention & control , Dipeptides/pharmacology , Fibrinolytic Agents/toxicity , Matrix Metalloproteinase 3/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Tissue Plasminogen Activator/antagonists & inhibitors , Tissue Plasminogen Activator/toxicity , Animals , Animals, Outbred Strains , Cerebral Hemorrhage/enzymology , Dipeptides/therapeutic use , Disease Models, Animal , Male , Matrix Metalloproteinase Inhibitors/therapeutic use , Mice
8.
Neuroscience ; 185: 116-24, 2011 Jun 30.
Article in English | MEDLINE | ID: mdl-21524692

ABSTRACT

Heparin-binding epidermal growth factor-like growth factor (HB-EGF) is a hypoxia-inducible neuroprotective protein that also stimulates proliferation of neuronal precursor cells. In this study, we investigated the possible role of HB-EGF in ischemia and reperfusion injury by measuring the changes in its mRNA expression following focal cerebral ischemia. We also examined neural damage after a middle cerebral artery occlusion (MCAO) and reperfusion in ventral forebrain specific HB-EGF knockout (KO) mice. The levels of HB-EGF mRNA in the cerebral cortex of wild-type (WT) mice were significantly increased 3-24 h after MCAO and reperfusion. Cerebral infraction in HB-EGF KO mice was aggravated at 1 day and 6 days after MCAO and reperfusion compared with WT mice. The number of terminal deoxynucleotidyl transferase (TdT)-mediated dNTP nick end labeling (TUNEL) and an oxidative stress marker, 8-hydroxy-2'-deoxyguanosine (8-OHdG) positive cells, were higher in HB-EGF KO mice than in WT mice. On the other hand, fewer bromodeoxyuridine (BrdU) positive cells were found in the subventricular zone in HB-EGF KO mice compared with WT mice. These results indicate that HB-EGF may play a pivotal role in ischemia and reperfusion injury and that endogenously synthesized HB-EGF is necessary for both the neuroprotective effect and for regulation of cell proliferation in the subventricular zone.


Subject(s)
Gene Expression Regulation/genetics , Infarction, Middle Cerebral Artery/pathology , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/metabolism , Prosencephalon/metabolism , Reperfusion Injury/pathology , 8-Hydroxy-2'-Deoxyguanosine , Adult Stem Cells/physiology , Analysis of Variance , Animals , Brain Infarction/etiology , Brain Infarction/pathology , Bromodeoxyuridine/metabolism , Cerebral Ventricles/physiology , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Disease Models, Animal , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Heparin-binding EGF-like Growth Factor , In Situ Nick-End Labeling/methods , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , RNA, Messenger/metabolism , Reperfusion Injury/complications , Transforming Growth Factor alpha/genetics , Transforming Growth Factor alpha/metabolism
9.
Neuroscience ; 171(1): 258-67, 2010 Nov 24.
Article in English | MEDLINE | ID: mdl-20804821

ABSTRACT

Toll-like receptors (TLRs) are signaling receptors in the innate immune system that is a specific immunologic response to systemic bacterial infection. We investigated whether cerebral ischemia induced by the middle cerebral artery occlusion (MCAO) for 2 h differed in mice that lack a functional TLR3, TLR4, or TLR9 signaling pathway. TLR4, but not TLR3 or TLR9, knock-out (KO) mice had significantly smaller infarct area and volume at 24 h after ischemia-reperfusion (I/R) compared with wild-type mice. In addition, TLR4 KO mice improved in neurological deficits after I/R compared with wild-type mice. Moreover, we investigated the expression of TLR4 in the ischemic brain with immunohistochemistry. The number of TLR4-positive cells gradually increased from 1 h after MCAO to 22 h after I/R. We also examined the localization of TLR4 in the ischemic area. TLR4 was localized with CD11b-positive microglial cells in the ischemic striatum and the number of CD11b-positive microglial cells was smaller in TLR4 KO mice than in wild-type mice. In addition, we investigated the translocation of NF-κB among TLR3, 4, and 9 KO mice after I/R injury using western blotting. NF-κB's p65 subunit was decreased in TLR4 KO mice compared to wild-type mice, but not TLR3 or 9 KO mice. These data suggest that TLR4 knockout, but not TLR3 or TLR9 knockout, may play a neuroprotective role in ischemic brain injury induced by MCAO in mice.


Subject(s)
Brain Ischemia/complications , Cerebral Infarction/etiology , Cerebral Infarction/prevention & control , Gene Expression Regulation/genetics , Nervous System Diseases/etiology , Nervous System Diseases/prevention & control , Toll-Like Receptor 4/deficiency , Animals , Brain Ischemia/genetics , CD11b Antigen/metabolism , Cell Count/methods , Cerebral Infarction/pathology , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , Nervous System Diseases/pathology , Neuroglia/metabolism , Neuroglia/pathology , Neurons/metabolism , Neurons/pathology , Phosphopyruvate Hydratase/metabolism , Protein Serine-Threonine Kinases/metabolism , Statistics, Nonparametric , Time Factors , Toll-Like Receptor 3/deficiency , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 9/deficiency , Toll-Like Receptor 9/metabolism , NF-kappaB-Inducing Kinase
10.
Neuroscience ; 159(2): 760-9, 2009 Mar 17.
Article in English | MEDLINE | ID: mdl-19166916

ABSTRACT

We aimed to examine whether thalidomide might inhibit the neuronal damage resulting from focal cerebral ischemia, and if so to explore the neuroprotective mechanism. Focal cerebral ischemia was induced by permanent middle cerebral artery occlusion (MCAO) in mice, and thalidomide was intraperitoneally administered a total of three times (at 10 min before, just before, and 1 h after MCAO). Thalidomide significantly reduced (a) the infarct area and volume at 24 and 72 h after MCAO and (b) the neurological score at 72 h after MCAO. Brains were also histochemically assessed for apoptosis and lipid peroxidation using terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL) staining and an antibody recognizing 8-hydroxy-2'-deoxyguanosine (8-OHdG), respectively. Thalidomide reduced both the number of TUNEL-positive cells and the oxidative damage. However, post-treatment of thalidomide [20 mg/kg, three times (at just after, 1 h after, 3 h after MCAO)] did not reduce the infarct volume. In an in vitro study, we examined the effects of thalidomide on lipid peroxidation in mouse brain homogenates and on the production of various radical species. Thalidomide inhibited both the lipid peroxidation and the production of H(2)O(2) and O(2).(-) (but not HO(-)) radicals. We also measured the brain concentration of TNF-alpha by ELISA. The TNF-alpha level in the brain was significantly increased at 9-24 h after MCAO. However, thalidomide did not reduce the elevated TNF-alpha level at either 12 or 24 h after MCAO. These findings indicate that thalidomide has neuroprotective effects against ischemic neuronal damage in mice, and that an inhibitory action of thalidomide against oxidative stress may be partly responsible for these neuroprotective effects.


Subject(s)
Cerebral Infarction/etiology , Cerebral Infarction/prevention & control , Infarction, Middle Cerebral Artery/complications , Neuroprotective Agents/therapeutic use , Thalidomide/therapeutic use , 8-Hydroxy-2'-Deoxyguanosine , Analysis of Variance , Animals , Blood Pressure/drug effects , Cell Death/drug effects , Cells, Cultured , Cerebral Infarction/metabolism , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Free Radical Scavengers/metabolism , Heart Rate/radiation effects , In Situ Nick-End Labeling/methods , In Vitro Techniques , Lipid Peroxidation/drug effects , Male , Mice , Nervous System Diseases/etiology , Nervous System Diseases/prevention & control , Retinal Ganglion Cells/drug effects , Time Factors , Tumor Necrosis Factor-alpha/metabolism
11.
J Neurosci Res ; 87(4): 906-17, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18855935

ABSTRACT

We investigated the expression of Na(+)/Ca(2+) exchanger (NCX) and the functional role of NCX in retinal damage by using NCX1-heterozygous deficient mice (NCX1(+/-)) and SEA0400 (2-[4-[(2,5-difluorophenyl)methoxy] phenoxy]-5-ethoxyaniline), a selective NCX inhibitor in vivo. We also examined the role of NCX in oxygen-glucose deprivation (OGD) stress with a retinal ganglion cell line (RGC-5) cell culture in vitro. The expression of NCX1 was confirmed and entirely localized in retina by immunoblotting and immunohistochemistry, respectively. NCX1(+/-) mice possessed significant protection against retinal damage induced by intravitreal injection of N-methyl-D-aspartate (NMDA). SEA0400 at 3 and 10 mg/kg significantly reduced NMDA- or high intraocular pressure-induced retinal cell damage in mice. Furthermore, SEA0400 reduced the number of TUNEL (terminal deoxynucleotidyl transferase dUTP nick-end labeling)-positive cells and the expression of phosphorylated mitogen-activated protein kinases (ERK1/2, JNK, p38) induced by NMDA injection. In RGC-5, SEA0400 at 0.3 and 1 microM significantly inhibited OGD-induced cell damage. OGD-induced cell damage was aggravated by ouabain (a Na(+),K(+)-ATPase inhibitor) at 100 microM, and this increased damage was significantly reduced by SEA0400 at 1 microM. In conclusion, these results suggest that NCX1 may play a role in retinal cell death induced by NMDA and ischemia-reperfusion.


Subject(s)
N-Methylaspartate/toxicity , Reperfusion Injury/physiopathology , Retina/cytology , Retinal Ganglion Cells/physiology , Retinal Neurons/physiology , Sodium-Calcium Exchanger/metabolism , Aniline Compounds/pharmacology , Animals , Cell Death/drug effects , Cell Line , Enzyme Inhibitors/pharmacology , In Situ Nick-End Labeling , Intraocular Pressure/physiology , Ionomycin/toxicity , Male , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinases/metabolism , Neuroprotective Agents/pharmacology , Neurotoxins/pharmacology , Ouabain/pharmacology , Phenyl Ethers/pharmacology , Rats , Retina/drug effects , Sodium-Calcium Exchanger/antagonists & inhibitors , Sodium-Calcium Exchanger/genetics , Stress, Physiological , Thapsigargin/toxicity
12.
Neuroscience ; 157(2): 309-18, 2008 Nov 19.
Article in English | MEDLINE | ID: mdl-18835333

ABSTRACT

Cerebral ischemia induces Ca(2+) influx into neuronal cells, and activates several proteases including calpains. Since calpains play important roles in neuronal cell death, calpain inhibitors may have potential as drugs for cerebral infarction. ((1S)-1((((1S)-1-Benzyl-3- cyclopropylamino-2,3-di-oxopropyl)amino)carbonyl)-3-methylbutyl) carbamic acid 5-methoxy-3-oxapentyl ester (SNJ-1945) is a novel calpain inhibitor that has good membrane permeability and water solubility. We evaluated the effect of SNJ-1945 on the focal brain ischemia induced by middle cerebral artery occlusion (MCAO) in mice. Brain damage was evaluated by assessing neurological deficits at 24 h or 72 h after MCAO and also by examining 2,3,5-triphenyltetrazolium chloride (TTC) staining and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) staining of brain sections. When injected at 1 h after MCAO, SNJ-1945 at 30 and 100 mg/kg, i.p. decreased the infarction volume and improved the neurological deficits each assessed at 24 h. SNJ-1945 at 100 mg/kg, i.p. also showed neuroprotective effects at 72 h and reduced the number of TUNEL-positive cells at 24 h. SNJ-1945 was able to prevent neuronal cell death even when it was injected at up to 6 h, but not at 8 h, after MCAO. In addition, SNJ-1945 decreased cleaved alpha-spectrin at 6 h and 12 h, and active caspase-3 at 12 h and 24 h in ischemic brain hemisphere. These findings indicate that SNJ-1945 inhibits the activation of calpain, and offers neuroprotection against the effects of acute cerebral ischemia in mice even when given up to 6 h after MCAO. SNJ-1945 may therefore be a potential drug for stroke.


Subject(s)
Carbamates/therapeutic use , Cerebral Infarction/pathology , Cerebral Infarction/prevention & control , Docosahexaenoic Acids/therapeutic use , Analysis of Variance , Animals , Brain Ischemia/complications , Caspase 3/metabolism , Cell Death/drug effects , Cerebral Infarction/etiology , Disease Models, Animal , Dose-Response Relationship, Drug , In Situ Nick-End Labeling/methods , Male , Mice , Neurologic Examination , Spectrin/metabolism , Tetrazolium Salts , Time Factors
13.
Brain Res ; 1214: 169-76, 2008 Jun 12.
Article in English | MEDLINE | ID: mdl-18457816

ABSTRACT

SUN N8075 is a novel antioxidant with neuroprotective properties. This study was designed to elucidate its neuroprotective effects against 6-hydroxy dopamine (6-OHDA)-induced cell death and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced neurotoxicity (known as in vitro and in vivo models of Parkinson's disease, respectively). In the in vitro study, on human neuroblastoma SH-SY5Y cells, SUN N8075 decreased the hydrogen peroxide (H2O2)-induced production of reactive oxygen species and protected against 6-OHDA-induced cell death. In the in vivo study, SUN N8075, when injected intraperitoneally (i.p.) twice with a 5-h interval, inhibited lipid peroxidation (viz. the production of thiobarbituric acid reactive substance) in the mouse forebrain at 1 h after the second injection. Mice were injected i.p. with MPTP (10 mg/kg) four times at 1-h intervals, and brains were analyzed 7 days later. SUN N8075 at 30 mg/kg (i.p., twice) exhibited a protective effect against the MPTP-induced decrease in tyrosine hydroxylase (TH)-positive fibers in the striatum. Moreover, SUN N8075 at 10 and 30 mg/kg (i.p., twice) had a similar protective effect against the MPTP-induced decrease in TH-positive cells in the substantia nigra. Further, SUN N8075 30 mg/kg (i.p. twice) markedly suppressed the MPTP-induced accumulation of 8-hydroxy-deoxyguanosine (8-OHdG) in the striatum. These findings indicate that SUN N8075 exerts protective effects, at least in part via an anti-oxidation mechanism, in these in vitro and in vivo models of Parkinson's disease.


Subject(s)
Aniline Compounds/therapeutic use , Antioxidants/therapeutic use , Parkinsonian Disorders/prevention & control , Piperazines/therapeutic use , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Adrenergic Agents/toxicity , Analysis of Variance , Animals , Body Weight/drug effects , Body Weight/physiology , Cell Death/drug effects , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Humans , Hydrogen Peroxide/pharmacology , Lipid Peroxidation/drug effects , Male , Mice , Mice, Inbred C57BL , Neuroblastoma/pathology , Oxidopamine/toxicity , Parkinsonian Disorders/chemically induced , Reactive Oxygen Species/metabolism
14.
Brain Res ; 1208: 217-24, 2008 May 07.
Article in English | MEDLINE | ID: mdl-18395193

ABSTRACT

Endoplasmic reticulum (ER) stress, which is caused by the accumulation of unfolded proteins in the ER lumen, is associated with stroke and neurodegenerative diseases such as Parkinson's and Alzheimer's diseases. We evaluated the effect of a selective inducer of immunoglobulin heavy chain binding protein (BiP) (BiP inducer X; BIX) against both tunicamycin-induced cell death (in SH-SY5Y cells) and the effects of global transient forebrain ischemia (in gerbils). BIX significantly induced BiP expression both in vitro and in vivo. Pretreatment with BIX at 2 or 5 microM reduced the cell death induced by tunicamycin in SH-SY5Y cells. In gerbils subjected to forebrain ischemia, prior treatment with BIX (intracerebroventricular injection at 10 or 40 microg) protected against cell death and decreased TUNEL-positive cells in the hippocampal CA1 subfield. These findings indicate that this selective inducer of BiP could be used to prevent the neuronal damage both in vitro and in vivo.


Subject(s)
Heat-Shock Proteins/metabolism , Ischemic Attack, Transient , Molecular Chaperones/metabolism , Prosencephalon/pathology , Analysis of Variance , Animals , Antiviral Agents/pharmacology , Cell Death/drug effects , Cell Death/physiology , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Endoplasmic Reticulum Chaperone BiP , Gene Expression Regulation/drug effects , Gerbillinae , Heat-Shock Proteins/genetics , Humans , In Situ Nick-End Labeling/methods , Ischemic Attack, Transient/complications , Ischemic Attack, Transient/metabolism , Ischemic Attack, Transient/pathology , Male , Molecular Chaperones/genetics , Phosphopyruvate Hydratase/metabolism , Thiocyanates/pharmacology , Tunicamycin/pharmacology
15.
Neuroscience ; 151(1): 111-9, 2008 Jan 02.
Article in English | MEDLINE | ID: mdl-18082969

ABSTRACT

Endoplasmic reticulum (ER) stress, which is caused by an accumulation of unfolded proteins in the ER lumen, is associated with stroke and with neurodegenerative diseases such as Parkinson's and Alzheimer diseases. We assessed the expression patterns of immunoglobulin heavy chain binding protein (BiP)/glucose-regulated protein (GRP) 78 (an ER-resident molecular chaperone whose expression serves as a good marker of ER-stress), activating transcription factor (ATF)-4, and C/EBP homology protein (CHOP) by immunohistochemistry and/or Western blotting after transient forebrain ischemia in gerbils. Double-fluorescent staining involving CHOP immunohistochemistry and the terminal deoxynucleotidyl transferase-mediated DNA nick-end labeling (TUNEL) method was performed to clarify the involvement of CHOP in cell death. Immunohistochemical and Western blot analyses of the hippocampal Cornet d'Ammon (CA)1 subfield showed that BiP expression was increased at 12 h, peaked at 3 days, then decreased (versus the control group). A transient increase was detected in CA3 at 1 day after ischemia, but BiP expression was unchanged in dentate gyrus and cortex. Signals for ATF-4 and CHOP were increased at 1 day and 3 days in CA1, and at 12 h in CA3. Co-localization of CHOP immunoreactivity and DNA fragmentation was detected by the TUNEL method at 3 days after ischemia in CA1, but not at 12 h in CA3. These findings are consistent with ER stress playing a pivotal role in post-ischemic neuronal death in the gerbil hippocampal CA1 subfield.


Subject(s)
Cell Death/physiology , Endoplasmic Reticulum/pathology , Ischemic Attack, Transient/pathology , Neurons/pathology , Prosencephalon/pathology , Stress, Physiological/pathology , Activating Transcription Factor 4/metabolism , Animals , Apoptosis/physiology , Biomarkers , Blotting, Western , DNA Fragmentation , Fluorescent Dyes , Gerbillinae , Glial Fibrillary Acidic Protein/metabolism , Immunoglobulin Heavy Chains/metabolism , Immunohistochemistry , In Situ Nick-End Labeling , Male , Reverse Transcriptase Polymerase Chain Reaction
16.
Neuroscience ; 149(4): 779-88, 2007 Nov 23.
Article in English | MEDLINE | ID: mdl-17945433

ABSTRACT

(2S)-1-(4-Amino-2,3,5-trimethylphenoxy)-3-{4-[4-(4-fluorobenzyl) phenyl]-1-piperazinyl}-2-propanol dimethanesulfonate (SUN N8075) is a novel antioxidant with neuroprotective properties. We examined whether SUN N8075 inhibited the neuronal damage resulting from permanent focal cerebral ischemia, and examined its neuroprotective properties in vivo and in vitro mechanism. Focal cerebral ischemia was induced by permanent middle cerebral artery occlusion in mice, and the resulting infarction, brain swelling, and neurological deficits were evaluated after 24 h or 72 h. Brain damage was assessed histochemically using terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL) staining and antibody recognizing 4-hydroxynonenal histidine adduct (4-HNE). In the in vitro study, we examined the effects of SUN N8075 on 1) lipid peroxidation in mouse brain homogenates and 2) cell viability and caspase-3 protease activity under a hypoxic insult or FeSO(4) in rat cultured cerebrocortical neurons. SUN N8075 administered either 10 min before or at 1 h after the occlusion reduced both infarction size and neurological deficits. SUN N8075 reduced brain swelling when administered 10 min before, 1 h, or 3 h after occlusion. Furthermore, only pretreatment (administered 10 min before) decreased infarct volume and brain swelling at 72 h after middle cerebral artery occlusion. SUN N8075 reduced the number of TUNEL-positive cells and decreased the level of oxidative damage, as assessed by immunopositive staining to 4-HNE. SUN N8075 inhibited lipid peroxidation, leakage of lactate dehydrogenase, caspase-3 activation induced by in vitro hypoxia, and the neuronal damage induced by in vitro FeSO(4) exposure. These findings indicate that SUN N8075 has neuroprotective effects against acute ischemic neuronal damage in mice and may prove promising as a therapeutic drug for stroke.


Subject(s)
Aniline Compounds/therapeutic use , Brain Infarction/prevention & control , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Piperazines/therapeutic use , Aldehydes/metabolism , Analysis of Variance , Animals , Brain Infarction/etiology , Brain Ischemia/complications , Caspase 3/metabolism , Cell Count/methods , Cell Death/drug effects , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , In Situ Nick-End Labeling/methods , In Vitro Techniques , Iron/pharmacology , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology , Male , Mice , Neurons/physiology , Time Factors
17.
Neuroscience ; 148(1): 105-14, 2007 Aug 10.
Article in English | MEDLINE | ID: mdl-17624681

ABSTRACT

Metallothioneins (MTs) are small cysteine-rich proteins found widely throughout the mammalian body, including the CNS. MT-1 and -2 protect against reactive oxygen species and free radicals. We investigated the role of MT-1 and -2 using MT-1,-2 knockout (KO) mice. MT-1,-2 KO mice exhibited greater neuronal damage after permanent middle cerebral artery occlusion (MCAO) than wild-type mice. MT-2 mRNA was significantly increased at 6, 12, and 24 h after MCAO in the wild-type mouse brain [as detected by real-time reverse-transcription polymerase chain reaction (RT-PCR)], while MT-1 and MT-3 were decreased at 12 and 24 h. In an immunohistochemical study, MT expression displayed colocalization with glial fibrillary acidic protein (GFAP)-positive cells (astrocytes) in the penumbra area in wild-type mice. Since erythropoietin (EPO) has been reported to induce MT-1 and -2 gene expression in vitro, we examined its effect after permanent MCAO, and explored the possible underlying mechanism by examining MT-1 and -2 induction in vivo. In wild-type mice, EPO significantly reduced both infarct area and volume at 24 h after the ischemic insult. However, in MT-1,-2 KO mice EPO-treatment did not alter infarct volume (vs. vehicle-treatment). In wild-type mice at 6 h after EPO administration, real-time RT-PCR revealed increased MT-1 and -2 mRNA expression in the cerebral cortex (without MCAO). Further, MT-1 and -2 immunoreactivity was increased in the cortex of EPO-treated mice. These findings indicate that MTs are induced, and may be neuroprotective against neuronal damage, after MCAO. Furthermore, EPO is neuroprotective in vivo during permanent MCAO, and this may be at least partly mediated by MTs.


Subject(s)
Brain Infarction/drug therapy , Brain Ischemia/drug therapy , Brain/drug effects , Erythropoietin/pharmacology , Metallothionein/genetics , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Brain/metabolism , Brain/physiopathology , Brain Infarction/metabolism , Brain Infarction/physiopathology , Brain Ischemia/metabolism , Brain Ischemia/physiopathology , Cytoprotection/drug effects , Cytoprotection/physiology , Erythropoietin/therapeutic use , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Glial Fibrillary Acidic Protein/metabolism , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/physiopathology , Male , Metallothionein/metabolism , Metallothionein 3 , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Degeneration/drug therapy , Nerve Degeneration/metabolism , Nerve Degeneration/physiopathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Up-Regulation/genetics
18.
Neuroscience ; 147(4): 957-67, 2007 Jul 29.
Article in English | MEDLINE | ID: mdl-17590517

ABSTRACT

The endoplasmic reticulum (ER) plays an important role in ischemic neuronal cell death. ER stress-related markers [immunoglobulin binding protein (BiP)/glucose-regulated protein (GRP) 78, activating transcription factor-4 (ATF-4), and C/EBP-homologous protein (CHOP)] in the striatum and the cortex were investigated after permanent middle cerebral artery occlusion (MCAO) in mice. Using endoplasmic reticulum stress-activated indicator (ERAI) transgenic mice, which show splicing of X-box protein 1 (XBP-1) mRNA as green fluorescence, we monitored the regional changes in fluorescence after MCAO. BiP mRNA (by reverse-transcription polymerase chain reaction [RT-PCR] analysis) was increased in the cortex at 6 h. In immunohistochemical and/or Western blot analysis, the expressions of ER stress-related markers (BiP, ATF-4, and CHOP) were increased in the infarct region, more strongly in the cortex than in the striatum. ERAI fluorescence was observed in the ischemic area starting from 6 and 12 h, respectively, after MCAO, with the peaks at 1 day and the fluorescence co-localized with the 2,3,5-triphenyltetrazolium chloride (TTC)-visible extension of brain infarction. These findings suggest that permanent MCAO induces expression of ER-stress related genes mainly in the periphery of the MCA territory.


Subject(s)
DNA-Binding Proteins/metabolism , Endoplasmic Reticulum/metabolism , Gene Expression Regulation/physiology , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Nuclear Proteins/metabolism , Stress, Physiological/physiopathology , Activating Transcription Factor 4/metabolism , Analysis of Variance , Animals , Cerebral Cortex/metabolism , Corpus Striatum/metabolism , DNA-Binding Proteins/genetics , Endoplasmic Reticulum Chaperone BiP , Functional Laterality , Glial Fibrillary Acidic Protein/metabolism , Heat-Shock Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Chaperones/metabolism , Nuclear Proteins/genetics , Regulatory Factor X Transcription Factors , Stress, Physiological/metabolism , Stress, Physiological/pathology , Time Factors , Transcription Factor CHOP/metabolism , Transcription Factors , X-Box Binding Protein 1
19.
Brain Res ; 1116(1): 187-93, 2006 Oct 20.
Article in English | MEDLINE | ID: mdl-16952340

ABSTRACT

The neuroprotective effect of cilostazol, an antiplatelet drug, was examined after 24 h permanent middle cerebral artery (MCA) occlusion in mice, and explored the possible underlying mechanism by examining metallothionein (MT)-1 and -2 induction in vivo. Cilostazol (30 mg/kg) was intraperitoneally administered at 12 h before, 1 h before, and just after MCA occlusion. Mice were euthanized at 24 h after the occlusion, and the neuronal damage was evaluated using 2,3,5-triphenyltetrazolium chloride (TTC) staining. Cilostazol significantly reduced the infarct area and volume, especially in the cortex. Real-time RT-PCR revealed increased mRNA expressions for MT-1 and -2 in the cortex of normal brains at 6 h after cilostazol treatment without MCA occlusion. MT-1 and -2 immunoreactivity was also increased in the cortex of such mice, and this immunoreactivity was observed in the ischemic hemisphere at 24 h after MCA occlusion (without cilostazol treatment). The strongest MT-1 and -2 immunoreactivity was detected in MCA-occlused mice treated with cilostazol [in the peri-infarct zone of the cortex (penumbral zone)]. These findings indicate that cilostazol has neuroprotective effects in vivo against permanent focal cerebral ischemia, especially in the penumbral zone in the cortex, and that MT-1 and -2 may be partly responsible for these neuroprotective effects.


Subject(s)
Brain Ischemia/pathology , Brain Ischemia/prevention & control , Metallothionein/biosynthesis , Neuroprotective Agents/pharmacology , Tetrazoles/pharmacology , Animals , Cerebral Cortex/pathology , Cilostazol , Enzyme Induction/drug effects , Immunohistochemistry , Infarction, Anterior Cerebral Artery/pathology , Male , Mice , Mice, Inbred C57BL , Middle Cerebral Artery/physiology , Reverse Transcriptase Polymerase Chain Reaction
20.
Brain Res ; 1082(1): 196-204, 2006 Apr 12.
Article in English | MEDLINE | ID: mdl-16515773

ABSTRACT

Rifampicin, an antibacterial drug, is highly effective in the treatment of tuberculosis and leprosy. Recently, it has been reported to have neuroprotective effects in in vitro and in vivo models. This study was designed to elucidate its neuroprotective effects against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced neurotoxicity (known as an in vivo mouse model of Parkinson's disease). Mice were injected intraperitoneally (i.p.) with MPTP (10 mg/kg) four times at 1-h intervals, and brains were analyzed 3 or 7 days later. Rifampicin at 20 mg/kg (i.p., twice) had protective effects against MPTP-induced neuronal damage (immunohistochemical changes in tyrosine hydroxylase) in both the substantia nigra and striatum. Rifampicin also protected against the MPTP-induced depletions of dopamine, 3,4-dihydroxyphenylacetic acid (DOPAC), and homovanillic acid (HVA) in the striatum. The maximal concentrations of rifampicin between 30 and 240 min after a single rifampicin injection (20 mg/kg, i.p.) were 2.6 microM (at 30 min) in plasma and 0.77 microM (at 60 min) in striatum. Next, the effects of rifampicin on oxidative stress [lipid peroxidation in mouse brain homogenates and free radical-scavenging activity against diphenyl-p-picrylhydrazyl (DPPH)] were evaluated to clarify the underlying mechanism. At 1 microM or more, rifampicin significantly inhibited both lipid peroxidation in the striatum and free radical production. These findings suggest that in mice, rifampicin can reach brain tissues at concentrations sufficient to attenuate MPTP-induced neurodegeneration in the nigrostriatal dopaminergic neuronal pathway, and that an inhibitory effect against oxidative stress may be partly responsible for its observed neuroprotective effects.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Brain/drug effects , Brain/pathology , Enzyme Inhibitors/therapeutic use , Parkinsonian Disorders/drug therapy , Rifampin/therapeutic use , 3,4-Dihydroxyphenylacetic Acid/metabolism , Analysis of Variance , Animals , Body Weight/drug effects , Brain Chemistry/drug effects , Cell Count/methods , Dopamine/metabolism , Dose-Response Relationship, Drug , Drug Interactions , Free Radicals/metabolism , Homovanillic Acid/metabolism , Immunohistochemistry/methods , Lipid Peroxidation/drug effects , Male , Mice , Mice, Inbred C57BL , Parkinsonian Disorders/chemically induced , Rifampin/blood , Tyrosine 3-Monooxygenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...