Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Stem Cells ; 28(10): 1816-28, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20979137

ABSTRACT

Neuronal precursor cells (NPCs) are temporally regulated and have the ability to proliferate and differentiate into mature neurons, oligodendrocytes, and astrocytes in the presence of growth factors (GFs). In the present study, the role of the Jak pathway in brain development was investigated in NPCs derived from neurosphere cultures using Jak2 and Jak3 small interfering RNAs and specific inhibitors. Jak2 inhibition profoundly decreased NPC proliferation, preventing further differentiation into neurons and glial cells. However, Jak3 inhibition induced neuronal differentiation accompanied by neurite growth. This phenomenon was due to the Jak3 inhibition-mediated induction of neurogenin (Ngn)2 and NeuroD in NPCs. Jak3 inhibition induced NPCs to differentiate into scattered neurons and increased the expression of Tuj1, microtubule associated protein 2 (MAP2), Olig2, and neuroglial protein (NG)2, but decreased glial fibrillary acidic protein (GFAP) expression, with predominant neurogenesis/polydendrogenesis compared with astrogliogenesis. Therefore, Jak2 may be important for NPC proliferation and maintenance, whereas knocking-down of Jak3 signaling is essential for NPC differentiation into neurons and oligodendrocytes but does not lead to astrocyte differentiation. These results suggest that NPC proliferation and differentiation are differentially regulated by the Jak pathway.


Subject(s)
Cell Differentiation/drug effects , Janus Kinase 3/metabolism , Neurons/cytology , Neurons/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Blotting, Western , Brain/embryology , Brain/metabolism , Cell Differentiation/genetics , Cell Proliferation/drug effects , Cells, Cultured , Immunohistochemistry , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Janus Kinase 3/antagonists & inhibitors , Janus Kinase 3/genetics , Mice , Neurogenesis/drug effects , Neurogenesis/genetics , Neurons/drug effects , Quinazolines/pharmacology , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/drug effects , Tyrphostins/pharmacology
2.
Cardiovasc Res ; 87(1): 119-26, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20154065

ABSTRACT

AIMS: Because apoptotic death plays a critical role in cardiomyocyte loss during ischaemic heart injury, a detailed understanding of the mechanisms involved is likely to have a substantial impact on the optimization and development of treatment strategies. The goal of this study was to assess gene profiling during ischaemia/hypoxia and to evaluate the functions of ischaemia/hypoxia-responsive genes in in vivo and in vitro ischaemia/hypoxia-induced cardiomyocyte apoptosis models. METHODS AND RESULTS: DNA microarray analysis and real-time polymerase chain reaction were performed on hearts obtained from an in vivo rat transient ischaemia model and on neonatal rat cardiomyocytes from an in vitro hypoxia model. Three genes, namely Ddit4, Gadd45beta and Atf3, were found to be up-regulated in vivo and in vitro. Using loss-of-function and gain-of-function techniques, the functions of these ischaemia/hypoxia-responsive genes were evaluated. Ischaemia/hypoxia-induced cardiomyocyte apoptosis was remarkably attenuated by the small interfering RNA-mediated down-regulation of Gadd45beta in vivo and in vitro, whereas ectopic Gadd45beta expression significantly aggravated hypoxia-induced apoptosis in vitro. CONCLUSION: These results suggest that Gadd45beta is a key player in ischaemia/hypoxia-induced apoptotic cardiomyocyte death, and that strategies based on its inhibition might be of benefit in the treatment of acute ischaemic heart disease.


Subject(s)
Antigens, Differentiation/genetics , Apoptosis , Myocardial Ischemia/genetics , Myocytes, Cardiac/metabolism , Activating Transcription Factor 3/genetics , Animals , Animals, Newborn , Antigens, Differentiation/metabolism , Apoptosis/genetics , Cell Hypoxia , Cell Line , Disease Models, Animal , Gene Expression Profiling/methods , Gene Knockdown Techniques , Hemodynamics , Male , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Myocytes, Cardiac/pathology , Oligonucleotide Array Sequence Analysis , RNA Interference , Rats , Rats, Sprague-Dawley , Repressor Proteins/genetics , Time Factors , Transcription Factors , Transfection , Up-Regulation
3.
Neurosci Lett ; 468(3): 254-8, 2010 Jan 14.
Article in English | MEDLINE | ID: mdl-19900507

ABSTRACT

Blood-brain barrier (BBB) dysfunction contributes to the pathophysiology of cerebrovascular diseases such as stroke. In the present study, we investigated the role of PKC isoforms in aglycemic hypoxia-induced hyperpermeability using an in vitro model of the BBB consisting of mouse bEnd.3 cells. PKCbetaII and PKCdelta isoforms were activated during aglycemic hypoxia. CGP53353, a specific PKCbetaII inhibitor, significantly attenuated aglycemic hypoxia-induced BBB hyperpermeability and disruption of occludin and zonula occludens-1 (ZO-1), indicating a deleterious role of PKCbetaII in the regulation of BBB permeability during aglycemic hypoxia. Conversely, rottlerin, a specific PKCdelta inhibitor, exacerbated BBB hyperpermeability and tight junction (TJ) disruption during aglycemic hypoxia, indicating a protective role of PKCdelta against aglycemic hypoxia-induced BBB hyperpermeability. Furthermore, disruption of TJ proteins during aglycemic hypoxia was attenuated by PKCbetaII DN and PKCdelta WT overexpression, and aggravated by PKCbetaII WT and PKCdelta DN overexpression. These results suggest that PKCbetaII and PKCdelta counter-regulate BBB permeability during aglycemic hypoxia.


Subject(s)
Blood-Brain Barrier/metabolism , Glucose/metabolism , Protein Kinase C-delta/physiology , Protein Kinase C/physiology , Animals , Capillary Permeability , Cell Hypoxia , Cell Line , Isoenzymes/antagonists & inhibitors , Isoenzymes/physiology , Mice , Protein Kinase C/antagonists & inhibitors , Protein Kinase C beta , Protein Kinase C-delta/antagonists & inhibitors , Rats , Tight Junctions/physiology
4.
J Pharmacol Sci ; 110(3): 405-9, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19609071

ABSTRACT

In this study, we examined the effect of Cd on the expression of vascular cell adhesion molecule-1 (VCAM-1) and its mechanisms in bEnd.3 cells. The treatment with Cd increased protein and mRNA expressions of VCAM-1 and increased the phosphorylations of p38, JNK, and ERK. The Cd-induced VCAM-1 expression was significantly suppressed by either a specific p38 mitogen-activated protein kinase (MAPK) inhibitor (SB202190) or a JNK inhibitor (SP600125), but not by an ERK inhibitor (U0126). These results suggest that Cd induces the expression of VCAM-1, at least in part, via p38 and JNK pathways in bEnd.3 cells.


Subject(s)
Cadmium/pharmacology , Endothelium, Vascular/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Brain/cytology , Humans , Mice
5.
J Neurosci ; 29(18): 5974-84, 2009 May 06.
Article in English | MEDLINE | ID: mdl-19420263

ABSTRACT

Neural-cadherin (N-cadherin), a member of the classical cadherin family of transmembrane glycoproteins, mediates cellular recognition and cell-cell adhesion through calcium-dependent homophilic interactions and plays important roles in the development and maintenance of the nervous system. Metalloproteinase is known to cleave N-cadherin, which is further cleaved by gamma-secretase. The intracellular domain of N-cadherin interacts with beta-catenin, and beta-catenin stability is critical for cell-cell adhesion and cell survival. In the present study, we showed that N-cadherin is cleaved specifically by calpain, resulting in the generation of a novel 110 kDa fragment. The cleavage occurred in ischemic brain lesions and in vitro neural cells in the presence of NMDA and ionomycin, and was restored by calpain inhibitors but not matrix metalloproteinase or gamma-secretase inhibitors. Calpain directly cleaved N-cadherin in in vitro calpain assays, and calpain inhibitors prevented its cleavage in a dose-dependent manner. Using N-cadherin deletion mutants, we found that calpain cleavage sites exist in at least four regions of the cytoplasmic domain. Treatment with NMDA induced neuronal death, and it suppressed the expression of surface N-cadherin and the N-cadherin/beta-catenin interaction, effects that were prevented by calpain inhibitor. Furthermore, calpain-mediated N-cadherin cleavage significantly affected cell-cell adhesion, AKT signaling, the N-cadherin/beta-catenin interaction and the Wnt target gene expressions through the accumulation of nuclear beta-catenin.


Subject(s)
Brain Injuries/metabolism , Cadherins/metabolism , Calpain/metabolism , Animals , Animals, Newborn , Biotinylation/methods , Brain Injuries/pathology , Cadherins/genetics , Calcium/metabolism , Calpain/pharmacology , Cells, Cultured , Disease Models, Animal , Dizocilpine Maleate/pharmacology , Embryo, Mammalian , Endocytosis/drug effects , Endocytosis/physiology , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/genetics , In Vitro Techniques , Ionomycin/pharmacology , Ionophores/pharmacology , Mice , Models, Biological , Mutation/genetics , N-Methylaspartate/pharmacology , Neurons/drug effects , Protein Structure, Tertiary/drug effects , Protein Structure, Tertiary/physiology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Subcellular Fractions/drug effects , Transfection/methods , Wnt Proteins/metabolism , beta Catenin/metabolism
6.
Eur J Nutr ; 48(4): 235-42, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19234663

ABSTRACT

BACKGROUND: Although there is growing awareness of the beneficial potential of onion intake to lower the risk of cardiovascular disease, there is little information about the effect of onion on ischemic heart injury, one of the most common cardiovascular diseases. AIM OF THE STUDY: This study investigates the effect of the methanol-soluble extract of onion on ischemic injury in heart-derived H9c2 cells in vitro and in rat hearts in vivo. The underlying mechanism is also investigated. METHODS: To evaluate the effect of onion on ischemia-induced cell death, LDH release and TUNEL-positivity were assessed in H9c2 cells, and the infarct size was measured in a myocardial infarct model. To investigate the mechanism of the cardioprotection by onion, the reactive oxygen species (ROS) level and the mitochondrial membrane potential (DeltaPsi(m)) were measured using an imaging technique; the caspase-3 activity was assayed, and Western blotting was performed to examine cytochrome c release in H9c2 cells. RESULTS: The methanolic extract of onion had a preventive effect on ischemia/hypoxia-induced apoptotic death in H9c2 cells in vitro and in rat heart in vivo. The onion extract (0.05 g/ml) inhibited the elevation of the ROS, mitochondrial membrane depolarization, cytochrome c release and caspase-3 activation during hypoxia in H9c2 cells. In the in vivo rat myocardial infarction model, onion extract (10 g/kg) significantly reduced the infarct size, the apoptotic cell death of the heart and the plasma MDA level. CONCLUSION: In conclusion, the results of this study suggest that the methanolic extract of onion attenuates ischemia/hypoxia-induced apoptosis in heart-derived H9c2 cells in vitro and in rat hearts in vivo, through, at least in part, an antioxidant effect.


Subject(s)
Apoptosis/drug effects , Cell Hypoxia/drug effects , Ischemia/drug therapy , Onions/chemistry , Plant Extracts/pharmacology , Animals , Antioxidants/pharmacology , Blotting, Western , Caspase 3/metabolism , Coronary Vessels , Cytochromes c/metabolism , Humans , In Situ Nick-End Labeling , Malondialdehyde/blood , Membrane Potential, Mitochondrial/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Rats , Reactive Oxygen Species/metabolism
7.
Brain Res ; 1248: 22-30, 2009 Jan 12.
Article in English | MEDLINE | ID: mdl-19022230

ABSTRACT

We investigated the effects of a novel Na(+)/H(+) exchanger-1 (NHE-1) inhibitor KR-33028 on glutamate excitotoxicity in cultured neuron cells in vitro and cerebral infarct in vivo by comparing its potency with that of zoniporide, a well-known, highly potent NHE-1 inhibitor. KR-33028 inhibited NHE-1 activation in a concentration-dependent manner (IC(50)=2.2 nM), with 18-fold greater potency than that of zoniporide (IC(50)=40.7 nM). KR-33028 significantly attenuated glutamate-induced LDH release with approximately 100 times lower EC(25) than that of zoniporide in cortical neurons in vitro (EC(25) of 0.007 and 0.81 microM, respectively), suggesting its 100-fold greater potency than zoniporide in producing anti-necrotic effect. In addition, the EC(50) of KR-33028 for anti-apoptotic effect was 100 times lower than that of zoniporide shown by TUNEL positivity (0.005 and 0.62 microM, respectively) and caspase-3 activity (0.01 and 2.64 microM, respectively). Furthermore, the EC(50) value of KR-33028 against glutamate-induced intracellular Ca(2+) overload was also 100 times lower than that of zoniporide (EC(50) of 0.004 and 0.65 microM, respectively). In the in vivo cerebral infarct model (60 min middle cerebral artery occlusion followed by 24 h reperfusion), KR-33028 reduced infarct size in a dose-dependent manner. Its ED(25) value, however, was quite similar to that of zoniporide (ED(25) of 0.072 and 0.097 mg/kg, respectively). Hence these results suggest that the novel NHE-1 inhibitor, KR-33028, could be an efficient therapeutic tool to protect neuronal cells against ischemic injury.


Subject(s)
Cerebral Cortex/drug effects , Cerebral Infarction/pathology , Glutamic Acid/pharmacology , Guanidines/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Thiophenes/pharmacology , Animals , Apoptosis/drug effects , Brain Ischemia , Caspase 3/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Cerebral Infarction/drug therapy , Cerebral Infarction/physiopathology , Enzyme Activation/drug effects , In Situ Nick-End Labeling , Mice , Microscopy, Confocal , Microscopy, Fluorescence , Necrosis , Neurons/chemistry , Neurons/physiology , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley
8.
Brain Res ; 1193: 153-61, 2008 Feb 08.
Article in English | MEDLINE | ID: mdl-18178179

ABSTRACT

COX-2 and prostaglandins (PGs) might play important roles in epilepsy. In kainic acid-induced seizures, the brain largely increases PGD(2), first from COX-1 and later COX-2-induced PGF(2alpha). Pre-treatment with COX-2 inhibitors such as indomethacin, nimesulide, and celecoxib is known to aggravate kainic acid (KA)-induced seizure activity. However it is not known whether the proconvulsant effect of those non-steroidal anti-inflammatory drugs (NSAIDs) is due to changes in endogenous prostaglandins (PGs), or what types of PGs are involved. The purpose of this study was to determine the effect of intracisternally administered PGs on KA-induced seizures aggravated by pre- or post-treatment with COX-2 inhibitors. Systemic KA injection (10 mg/kg i.p.) in mice evoked mild seizure activity within 15 min. PGs were administrated intracisternally 20 min prior to KA administration. COX inhibitors (indomethacin, nimesulide, and ketoprofen, 10 mg/kg i.p.) were injected 1 h before or 15 min after KA. An additional COX-2 inhibitor, celecoxib, was administered orally. Intracisternally administered PGF(2alpha) (700 ng), but not PGD(2) (700 ng) or PGE(2) (700 ng) completely alleviated KA-induced seizures potentiated by COX-2 inhibitors, and also reduced KA-induced hippocampal neuronal death aggravated by indomethacin. PGF(2alpha) alone did not affect KA-induced seizures. However, an FP receptor antagonist, AL 8810 (10 or 50 ng) which is an 11beta-fluoro analogue of PGF(2alpha) potentiated KA-induced seizure activity dose-dependently. In summary, pre- or post-treatment with COX-2 inhibitors aggravates KA-induced seizures, which suggests to change the endogenous PGF(2alpha). Seizure-induced PGF(2alpha) might act as an endogenous anticonvulsant through FP receptors.


Subject(s)
Cyclooxygenase Inhibitors/administration & dosage , Dinoprost/metabolism , Dinoprost/therapeutic use , Receptors, Prostaglandin/metabolism , Seizures/metabolism , Seizures/prevention & control , Analysis of Variance , Animals , Cell Survival/drug effects , Dinoprost/administration & dosage , Dinoprost/analogs & derivatives , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Electroencephalography/methods , Kainic Acid , Male , Mice , Mice, Inbred ICR , Receptors, Prostaglandin/antagonists & inhibitors , Seizures/chemically induced , Seizures/pathology
9.
Eur J Pharmacol ; 578(1): 11-8, 2008 Jan 06.
Article in English | MEDLINE | ID: mdl-17961543

ABSTRACT

Cadmium (Cd), an ubiquitous heavy metal, is known to be accumulated outside of the blood-brain barrier. In this study, we investigated whether Cd has cytotoxicity in mouse brain microvascular endothelial cells (bEnd.3). Results from the cell viability assay showed that Cd caused a remarkable decrease in cell viability in a dose-dependent manner. The cell death induced by Cd appeared to involve apoptosis, based on our results from annexin V staining, electron microscopy and TUNEL staining. And the cell death induced by Cd was inhibited by caspase inhibitor ZVAD-fmk. To further investigate the mechanism of the Cd-induced cell death, we examined the effects of selective inhibitors for mitogen activated protein kinase (MAPK) pathways on the cell death. The Cd-induced cell death was significantly inhibited by p38 MAPK inhibitor SB202190, but not by either, c-Jun N-terminal kinase (JNK) inhibitor SP600125 or extracellular signal-regulated kinase (ERK) inhibitor U0126. Phosphorylations of p38 MAPK, JNK and ERK were stimulated by treatment with CdCl(2). In summary, our results suggest that Cd can induce apoptotic cell death, at least in part, through the p38 MAPK pathway in brain microvascular endothelial cells.


Subject(s)
Apoptosis/drug effects , Cadmium Chloride/toxicity , Endothelium, Vascular/drug effects , p38 Mitogen-Activated Protein Kinases/drug effects , Animals , Annexin A5/metabolism , Blood-Brain Barrier/metabolism , Brain/blood supply , Cadmium Chloride/administration & dosage , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , In Situ Nick-End Labeling , Mice , Microcirculation/cytology , Microcirculation/drug effects , Microcirculation/metabolism , Microscopy, Electron , Phosphorylation/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Arch Pharm Res ; 30(4): 493-501, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17489367

ABSTRACT

The present study examined the effect of the methanol extract of Isaria sinclairii, a kind of Donchunghacho (Tochukaso), on blood pressure in spontaneously hypertensive rats (SHR). Blood pressure and heart rate were measured after treatment with the methanol extract of I. sinclairii by the indirect tail-cuff method and the direct in vivo model. Starting at 12 weeks of age, male SHR were treated with the extracts for 2 or 4 weeks. We found that, when compared to untreated control SHR, oral treatment with I. sinclairii methanol extract (30 mg/kg/day) remarkably decreased systolic blood pressure from 200 to 112 mmHg and decreased diastolic blood pressure from 114 to 88 mmHg. Furthermore, efficacy of methanol extract of I. sinclairii was superior to captopril (30 mg/kg/mL, positive control), an angiotensin-converting enzyme inhibitor, with a lowering effect that dropped systolic blood pressure from 201 to 130 mmHg and diastolic blood pressure from 102 to 92 mmHg. However, in normal Wistar Kyoto rats, I. sinclairii methanol extract did not significantly change the normal blood pressure, suggesting that this type of Dongchunghacho has a selective effect against hypertension. Therefore, methanol extract of I. sinclairii may be used as an anti-hypertensive food/agent. Furthermore, this extract also has multiple actions such as No production in endothelial cells, inhibiting thrombin-induced blood coagulation by thrombin and mildly decreasing in prostaglandin E2 levels in cultured macrophage cells, all of which might contribute to protection against atherogenesis and thrombus formation. HPLC and MS analysis of methanol extract of I. sinclairii revealed the presence of adenosine.


Subject(s)
Antihypertensive Agents/pharmacology , Paecilomyces/chemistry , Adenosine/analysis , Animals , Blood Pressure/drug effects , Dinoprostone/biosynthesis , Dose-Response Relationship, Drug , Male , Nitric Oxide/biosynthesis , Rats , Rats, Inbred SHR , Tandem Mass Spectrometry
11.
Toxicol Lett ; 165(3): 212-20, 2006 Sep 10.
Article in English | MEDLINE | ID: mdl-16777358

ABSTRACT

In order to get insight into the mechanism of cadmium (Cd)-induced brain injury, we investigated the effects of Cd on the induction of COX-2 and ICAM-1 in bEnd.3 mouse brain endothelial cells (EC). Cd stimulated PGE(2) release in a time and dose dependent manner, which was accompanied by increase of COX-2 expression. The thiol-reducing antioxidant N-acetylcyteine attenuated Cd-induced PGE(2) production and COX-2 expression. Cd increased phosphorylation of p38 MAPK, but not of JNK and ERK1/2. A blockade of p38 MAPK pathway abrogated Cd-induced COX-2 expression and PGE(2) production. Cd-induced ICAM-1 expression and leukocyte-EC adhesion were diminished by non-steroidal anti-inflammatory drugs such as indomethacin and NS-398, which was reversed by addition of PGE(2). Together, these data suggest that Cd induces COX-2 expression through the activation of p38 MAPK, an oxidative stress-sensitive cellular signaling molecule, and induction of COX-2 is associated with ICAM-1 expression in brain endothelial cells following Cd exposure.


Subject(s)
Cadmium/toxicity , Cyclooxygenase 2/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Intercellular Adhesion Molecule-1/metabolism , Animals , Brain/blood supply , Cell Adhesion/physiology , Dinoprostone/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation , Leukocytes/metabolism , Mice , Up-Regulation , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Life Sci ; 78(10): 1091-7, 2006 Feb 02.
Article in English | MEDLINE | ID: mdl-16153663

ABSTRACT

In the present study, the mechanism of antiplatelet activity of DK-002, a synthesized (6aS,cis)-9,10-Dimethoxy-7,11b-dihydro-indeno[2,1-c]chromene-3,6a-diol, was investigated. DK-002 inhibited the thrombin, collagen, and ADP-induced rat platelet aggregation in a concentration-dependent manner, with IC50 values of 120, 27, and 47 microM, respectively. DK-002 also inhibited thrombin-induced dense granule secretion, thromboxane A2 synthesis, and [Ca2+]i elevation in platelets. DK-002 did not show any significant effect on ADP-induced inhibition of cyclic AMP elevation by prostaglandin E1, but DK-002 was confirmed to inhibit ADP-induced [Ca2+]i elevation and shape change. DK-002 inhibited 4-bromo-A23187-induced [Ca2+]i elevation in the presence of creatine phosphate/creatine phosphokinase (CP/CPK, a ADP scavenging system) and indomethacin (a specific inhibitor of cyclooxygenase). DK-002 also inhibited Ca2+ mobilization in thrombin- or 4-bromo-A23187-stimulated platelets through its inhibitory effects on both Ca2+ release from intracellular stores and Ca2+ influx, in the presence of CP/CPK and indomethacin. Taken together, the present study shows that DK-002 has inhibitory effects on stimulation of platelets, and suggests that its antiplatelet activity might be related to the inhibitory mechanism on Ca2+ mobilization in stimulated platelets.


Subject(s)
Benzopyrans/pharmacology , Blood Platelets/drug effects , Indenes/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Adenosine Diphosphate/pharmacology , Animals , Benzopyrans/chemistry , Blood Platelets/ultrastructure , Calcium/blood , Creatine Kinase/metabolism , Cyclic AMP/metabolism , Cyclooxygenase Inhibitors/pharmacology , Cytoplasmic Granules/drug effects , In Vitro Techniques , Indenes/chemistry , Indomethacin/pharmacology , Ionophores/pharmacology , Rats , Rats, Sprague-Dawley , Serotonin/blood , Thrombin/pharmacology , Thromboxane B2/blood
13.
Eur J Pharmacol ; 525(1-3): 1-7, 2005 Nov 21.
Article in English | MEDLINE | ID: mdl-16289528

ABSTRACT

A novel Na+/H+ exchanger-1 (NHE-1) inhibitor [5-(2-methoxy-5-chloro-5-phenyl)furan-2-ylcarbonyl]guanidine (KR-32570) has been previously demonstrated to elicit cardioprotective effect against ischemic injury in rat heart. In the present study, we examined the effects of KR-32570 on cell death induced by hypoxic insult in heart-derived H9c2 cells. Treatment with KR-32570 (1-10 microM) significantly reduced hypoxia-induced necrotic cell death (lactate dehydrogenase release) and apoptotic cell death (TUNEL-positivity, caspase-3 activity). KR-32570 also decreased the cytosolic and mitochondrial Ca2+ overload induced by hypoxia. Inhibition of mitochondrial Ca2+ overload by ruthenium red mimicked the anti-apoptotic effect of KR-32570. In addition, KR-32570 significantly recovered the large reduction in mitochondrial membrane potential (delta psi(m)) and cytochrome c release induced by hypoxia. Taken together, our results suggest that a new NHE-1 inhibitor KR-32570 elicits potent cardioprotective effects in H9c2 cells, and its effects may be mediated by inhibition of intracellular Ca2+ overload and mitochondrial death pathway during hypoxia.


Subject(s)
Cardiotonic Agents/pharmacology , Cell Death/drug effects , Guanidines/pharmacology , Hypoxia , Myocytes, Cardiac/drug effects , Animals , Calcium/metabolism , Caspase 3 , Caspases/metabolism , Cell Line , L-Lactate Dehydrogenase/metabolism , Membrane Potentials/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/physiology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Rats , Sodium-Hydrogen Exchangers/antagonists & inhibitors
14.
Brain Res ; 1061(1): 67-71, 2005 Nov 02.
Article in English | MEDLINE | ID: mdl-16225853

ABSTRACT

We investigated the effects of an Na(+)/H(+) exchanger inhibitor, sabiporide, on excitotoxicity in cultured neuronal cells and in vivo. Sabiporide attenuated glutamate- or NMDA (N-methyl-d-aspartic acid)-induced neuronal cell death. Sabiporide also reduced glutamate or NMDA-induced increase in [Ca(2+)](i). In in vivo brain ischemia model, sabiporide produced protective effects, decreasing the infarct size and edema volume. Our results suggest that sabiporide elicits neuroprotective effect both in vitro and in vivo.


Subject(s)
Brain Ischemia/pathology , Guanidines/pharmacology , Neurons/drug effects , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Animals , Brain Infarction/etiology , Brain Infarction/pathology , Brain Infarction/prevention & control , Brain Ischemia/chemically induced , Brain Ischemia/drug therapy , Calcium/metabolism , Cell Death/drug effects , Cells, Cultured , Cerebral Cortex/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Extracellular Space/drug effects , Glutamic Acid/adverse effects , Guanidines/therapeutic use , Mice , N-Methylaspartate/adverse effects , Neurons/pathology , Reperfusion/methods , Tetrazolium Salts , Thiazoles
15.
J Neurosci Res ; 81(1): 73-84, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15931672

ABSTRACT

Arachidonic acid (AA) is released from membrane phospholipids during normal and pathologic processes such as neurodegeneration. AA is metabolized via lipoxygenase (LOX)-, cyclooxygenase (COX)-, and cytochrome P450 (CYP450)-catalyzed pathways. We investigated the relative contributions of these pathways in AA-induced neuronal death. Exposure of cultured cortical neurons to AA (50 microM) yielded significantly apoptotic neuronal death, which was attenuated greatly by LOX inhibitors (nordihydroguaiaretic acid, AA861, and baicalein), or CYP450 inhibitors (SKF525A and metyrapone), rather than COX inhibitors (indomethacin and NS398). AA (10 microM)-induced neurotoxicity was prevented by all kinds of inhibitors. Compared, the neurotoxic effects of three pathway metabolites, 12-hydroxyeicosatetraenoic acid (12-HETE), a major LOX metabolite, induced a significant neurotoxicity. AA also produced reactive oxygen species within 30 min, which was reduced by all inhibitors tested, including COX inhibitors, and AA neurotoxicity was abolished by the antioxidant Trolox. AA treatment also depleted glutathione levels; this depletion was reduced by the LOX or CYP450 inhibitors rather than by the COX inhibitors. Taken together, our data suggested that the LOX pathway likely plays a major role in AA-induced neuronal death with the modification of intracellular free radical levels.


Subject(s)
Apoptosis/physiology , Arachidonic Acid/metabolism , Cytochrome P-450 Enzyme System/metabolism , Lipoxygenase/metabolism , Neurons/enzymology , Analysis of Variance , Animals , Arachidonic Acid/administration & dosage , Cerebral Cortex/cytology , Cerebral Cortex/enzymology , Coculture Techniques , Dose-Response Relationship, Drug , Mice , Neuroglia/physiology , Prostaglandin-Endoperoxide Synthases/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , Statistics, Nonparametric
16.
Neuroreport ; 16(7): 741-4, 2005 May 12.
Article in English | MEDLINE | ID: mdl-15858417

ABSTRACT

We investigated the role of individual protein kinase C (PKC) isoforms during kainate toxicity in cortical neurons. Treatment with 50 microM kainate induced isoform-specific activation of PKC-delta according to the translocation from the soluble to the particulate fraction, while it caused remarkable decreases in PKC alpha, beta, epsilon and zeta in both fractions. Kainate-induced neuronal death was significantly increased by pharmacological inhibition of PKC-delta with rottlerin, suggesting a protective role of PKC-delta against kainate toxicity. A PKC activator phorbol 12-myristate 13-acetate remarkably attenuated the kainate-induced neuronal death. Although phorbol 12-myristate 13-acetate activates PKC-epsilon and PKC-delta, the protective effect of phorbol 12-myristate 13-acetate was almost completely abolished by rottlerin, but not by epsilonV1-2. These results suggest that activation of PKC-delta attenuates the kainate-induced cell death of cortical neurons.


Subject(s)
Cell Death/physiology , Neurons/cytology , Neurons/enzymology , Protein Kinase C/metabolism , Animals , Carcinogens/pharmacology , Cell Death/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Excitatory Amino Acid Agonists/toxicity , Intracellular Signaling Peptides and Proteins/pharmacology , Kainic Acid/toxicity , Mice , Protein Kinase C/antagonists & inhibitors , Protein Kinase C-delta , Tetradecanoylphorbol Acetate/pharmacology
17.
Thromb Res ; 115(4): 309-18, 2005.
Article in English | MEDLINE | ID: mdl-15668191

ABSTRACT

Brazilin (7,11b-dihydrobenz[b]indeno[1,2-d]pyran-3,6a,9,10 (6H)-tetrol), the major component of Caesalpinia sappan L., was reported to show antiplatelet activity through the inhibition of phospholipase A2 (PLA2) activity and the increase in intracellular free Ca2+ concentration ([Ca2+]i). To search more potential antiplatelet agent, brazilin derivatives were synthesized and examined for their effects on the platelet aggregation. Among those compounds, BRX-018, (6aS,cis)-Malonic acid 3-acetoxy-6a9-bis-(2-methoxycarbonyl-acetoxy)-6,6a,7,11b-tetrahydro-indeno[2,1-c]chromen-10-yl ester methylester, was confirmed as one of the potential antiplatelet agents. In the present study, we investigated the antiplatelet mechanism of BRX-018. BRX-018 inhibited the thrombin-, collagen-, and ADP-induced rat platelet aggregation in a concentration-dependent manner, with IC50 values of 35, 15, and 25 microM, respectively. BRX-018 also inhibited thrombin-induced dense granule secretion, thromboxane A2 (TXA2) synthesis, and [Ca2+]i elevation in platelets. BRX-018 was also found to inhibit A23187-induced [Ca2+]i and aggregation in the presence of apyrase (ADP scavenger) but not in the presence of both apyrase and indomethacin (a specific inhibitor of cyclooxygenase, COX). Although BRX-018 significantly inhibited arachidonic acid (AA)-induced aggregation and TXA2 synthesis, it had no significant inhibitory effect on cyclooxygenase activity in vitro. In contrast, BRX-018 inhibited the activity of purified PLA2. Dixon plot showed that this inhibition was mixed type with an inhibition constant of Ki=23 microM. Taken together, the present study suggests that BRX-018 may be a promising antiplatelet agent and that its antiplatelet activity may be based on the inhibitory mechanisms on TXA2 synthesis in stimulated platelets.


Subject(s)
Benzopyrans/pharmacology , Malonates/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Adenosine Diphosphate/pharmacology , Animals , Arachidonic Acid/pharmacology , Benzopyrans/chemical synthesis , Benzopyrans/chemistry , Blood Platelets/drug effects , Blood Platelets/physiology , Calcimycin/pharmacology , Calcium/blood , Cattle , Cell Degranulation/drug effects , Collagen/pharmacology , In Vitro Techniques , Ionophores/pharmacology , Male , Malonates/chemical synthesis , Malonates/chemistry , Phospholipases A/antagonists & inhibitors , Phospholipases A2 , Platelet Aggregation Inhibitors/chemical synthesis , Platelet Aggregation Inhibitors/chemistry , Prostaglandin-Endoperoxide Synthases/blood , Rats , Rats, Sprague-Dawley , Swine , Thrombin/pharmacology , Thromboxane B2/blood
18.
Arch Pharm Res ; 28(12): 1358-64, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16392669

ABSTRACT

In this study, we investigated whether a novel anti-ischemic KATP opener KR-31378 [(2S,3S,4R)-N"-cyano-N-(6-amino-3,4-dihydro-3-hydroxy-2-methly-2-dimethoxymethly-2H-benzopyran-4-yl)-N'-benzylguanidine] has protective effect against oxidative stress-induced death in heart-derived H9c2 cells. Cell death was induced by BSO, butionine sulfoximine, which inhibits GSH synthesis and subsequently increases reactive oxygen species (ROS) level. Cell death was quantitatively determined by measuring lactate dehydrogenase (LDH) activity and stained by Hoechst 33258. BSO-induced ROS production and mitochondrial membrane potential (MMP) were measured using 2',7'-dichlorofluorescein diacetate oxidation and rhodamine 123, respectively. Both the LDH release and the ROS elevation induced by treatment of H9c2 cells with 10 mM BSO, were significantly decreased by KR-31378. These protective effect and antioxidant effect of KR-31378 appeared to be independent on KATP channel opening. Cells exposed to BSO showed an early reduction in MMP, and this reduction in MMP was significantly reversed by treatment with KR-31378. Caspase-3 activity in BSO treated H9c2 cells was remarkably increased, and this increased caspase-3 activity was significantly reversed by KR-31378. In conclusion, our results suggest that KR-31378 can produce cardioprotective effect against oxidative stress-induced cell death through antioxidant mechanism.


Subject(s)
Guanidines/pharmacology , Myocytes, Cardiac/drug effects , Oxidative Stress/drug effects , Pyrans/pharmacology , Animals , Apoptosis/drug effects , Benzimidazoles/chemistry , Buthionine Sulfoximine/antagonists & inhibitors , Buthionine Sulfoximine/toxicity , Caspase 3 , Caspase Inhibitors , Caspases/metabolism , Cell Line , Decanoic Acids/pharmacology , Dose-Response Relationship, Drug , Fluoresceins/chemistry , Hydroxy Acids/pharmacology , L-Lactate Dehydrogenase/antagonists & inhibitors , L-Lactate Dehydrogenase/metabolism , Membrane Potentials/drug effects , Microscopy, Fluorescence , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/physiology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Potassium Channel Blockers/pharmacology , Reactive Oxygen Species/metabolism , Sulfonamides/pharmacology , Thiourea/analogs & derivatives , Thiourea/pharmacology , Time Factors
19.
Eur J Pharmacol ; 506(1): 27-35, 2004 Dec 03.
Article in English | MEDLINE | ID: mdl-15588621

ABSTRACT

A novel compound KR-31378 [(2S,3S,4R)-N''-cyano-N-(6-amino-3,4-dihydro-3-hydroxy-2-methly-2-dimethoxy-methly-2H-benzo-pyran-4-yl)-N-benzylguanidine] has been demonstrated as an anti-ischemic agent in rat heart and brain. Here, we report the effects of this compound on hypoxia-induced cell death and possible signaling pathways in heart-derived H9c2 cells. Treatment with KR-31378 (3-30 microM) 1 h before and during hypoxia significantly reduced hypoxia-induced cell death in a concentration-dependent manner. In addition, increase in hypoxia-induced transferase UTP nick end labeling (TUNEL)-positive cells was reduced by KR-31378, suggesting its antiapoptotic potential in H9c2 cells. The protective effect conferred by KR-31378 (10 microM) was abolished by cotreatment with 5-hydroxydecanoate (5HD), a specific blocker of the mitochondrial KATP (mtKATP) channel, but not by HMR-1883 (1-[[5-[2-(5-chloro-o-anisamido)ethyl]-methoxyphenyl]sulfonyl]-3-methylthiourea), a specific blocker of the sarcolemmal KATP channel. We observed that the treatment with KR-31378 could increase the expression of protein kinase C (PKC)-epsilon protein, but not other PKC isotypes (-alpha, -beta, -delta, -zeta), in the particulate fraction. This increased level of PKC-epsilon was sustained during the hypoxic period up to 8 h. In addition, our results showed that treatment with KR-31378 induced the expression of PKC-epsilon mRNA as early as 15 min after the treatment. A specific inhibitor for PKC-epsilon isoform, epsilonV1-2, completely blocked the protective effect of KR-31378 against hypoxia-induced cell death. In conclusion, our results suggest that KR-31378 can protect cultured H9c2 cells from hypoxia-induced death via the mtKATP channel and PKC-epsilon.


Subject(s)
Apoptosis/drug effects , Guanidines/pharmacology , Membrane Proteins/metabolism , Myocytes, Cardiac/drug effects , Protein Kinase C/metabolism , Pyrans/pharmacology , Thiourea/analogs & derivatives , Animals , Blotting, Western , Cell Hypoxia , Cell Line , Decanoic Acids/pharmacology , Dose-Response Relationship, Drug , Hydroxy Acids/pharmacology , In Situ Nick-End Labeling , Membrane Proteins/antagonists & inhibitors , Mitochondria, Heart/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Potassium Channels/metabolism , Protein Kinase C-epsilon , Sulfonamides/pharmacology , Thiourea/pharmacology , Time Factors
20.
Brain Res ; 1026(2): 295-301, 2004 Nov 12.
Article in English | MEDLINE | ID: mdl-15488492

ABSTRACT

Fructose-1,6-bisphosphate (FBP) is a glucose metabolism intermediate that shows a neuroprotective action in animal models of ischemia and other injuries. The intracellular mechanism of FBP on neuroprotection has not been previously defined. Here, we examined whether FBP has a neuroprotective effect against excitotoxicity, and whether it affects the production of reactive oxygen species (ROS), which are involved in the MAPK pathway in cortical neurons. FBP prevented neuronal death in a dose-dependent manner following 24 h of treatment with the excitotoxin, NMDA. After 8 h of NMDA treatment, we observed FBP-induced inhibition of the production of intracellular ROS, and at the earlier time FBP suppressed NMDA-induced p-p38 and p-ERK expression. In addition, MAPK inhibitors reduced NMDA-induced excitotoxicity and also ROS production. Taken together, our results suggest that the neuroprotective effects of FBP could be explained by down-regulation of free radical production through the p38MAPK/ERK pathway.


Subject(s)
Fructosediphosphates/pharmacology , MAP Kinase Signaling System/physiology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Blotting, Western/methods , Brain/cytology , Cell Count/methods , Cell Death/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Immunohistochemistry/methods , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , L-Lactate Dehydrogenase/metabolism , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred ICR , N-Methylaspartate/toxicity , Phosphopyruvate Hydratase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...