Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Exp Anim ; 65(3): 303-10, 2016 Jul 29.
Article in English | MEDLINE | ID: mdl-27041456

ABSTRACT

Chlorine dioxide (ClO2) is a strong oxidant that possesses an antimicrobial activity. We demonstrated here that ClO2 gas is easily generated by mixing 3.35% sodium chlorite solution (Purogene) and 85% phosphoric acid at a 10:1 volume ratio without using an expensive machine. In a test room (87 m(3)), experiments were carried out using various amounts of sodium chlorite solution (0.25 ml/m(3) to 20.0 ml/m(3)). The gas concentration increased in a sodium chlorite volume-dependent manner and reached peak values of from 0.8 ppm to 40.8 ppm at 2 h-3 h, and then gradually decreased. No differences in gas concentrations were observed between 0.1 and 2.5 m above the floor, indicating that the gas was evenly distributed. Under high-humidity (approximately 80% relative humidity), colony formation of both Staphylococcus aureus and Escherichia coli was completely inhibited by ClO2 gas exposure at 1.0 ml/m(3) sodium chlorite solution (mean maximal concentration of 3.0 ppm). Exposure at 4.0 ml/m(3) sodium chlorite solution (mean maximal concentration of 10.6 ppm) achieved complete inactivation of Bacillus atrophaeus spores. In contrast, without humidification, the efficacy of ClO2 gas was apparently attenuated, suggesting that the atmospheric moisture is indispensable. Delicate electronic devices (computer, camera, etc.) operated normally, even after being subjected to more than 20 times of fumigation. Considering that our method for gas generation is simple, reproducible, and highly effective at decontaminating microbes, our approach is expected to serve as an inexpensive alternative method for cleaning and disinfecting animal facilities.


Subject(s)
Chlorine Compounds , Disinfection/methods , Fumigation/methods , Oxides , Animals , Bacillus/drug effects , Chlorides/chemistry , Chlorine Compounds/chemical synthesis , Chlorine Compounds/pharmacology , Dose-Response Relationship, Drug , Escherichia coli/drug effects , Gases , Housing, Animal , Humidity , Oxides/chemical synthesis , Oxides/pharmacology , Phosphoric Acids/chemistry , Solutions , Staphylococcus aureus/drug effects
2.
Thromb Res ; 133(4): 622-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23932349

ABSTRACT

INTRODUCTION: Intracerebral hemorrhage (ICH) is a major clinical concern with anticoagulation therapy. The effect of a new oral direct FXa inhibitor, edoxaban, was determined in a rat model of ICH and compared with a direct thrombin inhibitor, melagatran, and heparin. METHODS: To induce ICH, 0.1 U collagenase type VII was injected into the striatum of male Wistar rats under anesthesia with thiopental or halothane. Immediately after ICH induction, edoxaban, melagatran, or heparin were infused intravenously. Five hours after ICH induction, the brain was removed and ICH size was measured. To estimate the margin of safety, antithrombotic effects were evaluated in a rat venous thrombosis model. RESULTS: Edoxaban at 6mg/kg/h significantly increased ICH volume (1.8-fold) and prolonged prothrombin time (PT) 2.8-fold compared to the vehicle group. No deaths were observed with edoxaban. Melagatran at 1mg/kg/h increased ICH volume at 1mg/kg/h (2.8-fold) with 6.1-fold PT prolongation. At 3mg/kg/h, all rats died due to severe ICH (3.9-fold). Heparin at both 100 and 500U/kg/h significantly increased ICH. At 500U/kg/h, 5 out of 8 rats died. The doses required for 50% inhibition of thrombosis of edoxaban, melagatran, and heparin were 0.045mg/kg/h, 0.14mg/kg/h, and 55U/kg/h, respectively. The safety margins between antithrombotic and ICH exacerbation effects of these anticoagulants were 133, 7.1, and 1.8, respectively. CONCLUSION: The safety margin of edoxaban was wider than that of melagatran or heparin. These results suggest that edoxaban may be preferable from the perspective of ICH exacerbation risk.


Subject(s)
Anticoagulants/pharmacology , Antithrombins/pharmacology , Azetidines/pharmacology , Benzylamines/pharmacology , Cerebral Hemorrhage/drug therapy , Factor Xa Inhibitors/pharmacology , Heparin/pharmacology , Pyridines/pharmacology , Thiazoles/pharmacology , Animals , Cerebral Hemorrhage/blood , Collagenases , Disease Models, Animal , Male , Rats , Rats, Wistar
3.
J Med Primatol ; 42(3): 165-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23802316

ABSTRACT

BACKGROUND: Physiologic stress has been demonstrated to impair glucose tolerance. METHODS: Glucose tolerance tests were performed using six cynomolgus monkeys. RESULTS: Chair-restrained subjects elicited higher elevations of plasma glucose and cortisol compared with squeezing device-restrained subjects. CONCLUSIONS: The responses to a glucose challenge are altered by different restraint procedures.


Subject(s)
Blood Glucose/metabolism , Macaca fascicularis/physiology , Restraint, Physical/methods , Animals , Female , Glucose Tolerance Test , Stress, Physiological
4.
J Am Assoc Lab Anim Sci ; 51(3): 345-51, 2012 May.
Article in English | MEDLINE | ID: mdl-22776193

ABSTRACT

A method for blood collection from the jugular vein of mice without anesthesia was compared with a tail-incision technique. Jugular vein blood collection allowed withdrawal of almost 15% of the circulating blood volume at a time in less than 1 min. Hemolysis, hematocrit, and plasma thrombin-antithrombin complexes (a marker of blood coagulation) were higher in samples collected from the tail vein than the jugular vein. Mice produced similar plasma corticosterone levels after serial blood collection by either method. Tail incision led to a slight but significant increase in C-reactive protein levels. Using the jugular venipuncture technique, we then performed a pharmacokinetic study and an oral glucose tolerance test. Plasma concentrations of levofloxacin, an antimicrobial agent, were dose-dependently elevated after oral administration, and linear increases in C(max) and AUC were observed. We also confirmed that overall glucose excursion is significantly decreased in mice treated with exendin 4, a glucagon-like peptide 1 agonist. These results indicate that the jugular venipuncture is a useful technique from the point of view of no requirement for anesthetics, serial blood collection at short intervals, large volume of blood collection, quality of sample and animal welfare. This technique is of particular interest for studies that examine time-dependent changes in blood variables.


Subject(s)
Animal Welfare , Jugular Veins , Mice , Phlebotomy/methods , Administration, Oral , Animals , Anti-Bacterial Agents/blood , Anti-Bacterial Agents/pharmacokinetics , Blood Chemical Analysis/veterinary , Blood Glucose/drug effects , Blood Glucose/metabolism , Exenatide , Glucagon-Like Peptide 1/agonists , Glucose Tolerance Test/veterinary , Hypoglycemic Agents/administration & dosage , Levofloxacin , Male , Mice, Inbred C57BL , Mice, Inbred ICR , Ofloxacin/blood , Ofloxacin/pharmacokinetics , Peptides/administration & dosage , Phlebotomy/veterinary , Venoms/administration & dosage
5.
PLoS One ; 6(9): e24724, 2011.
Article in English | MEDLINE | ID: mdl-21931829

ABSTRACT

Ca²âº/calmodulin-dependent protein kinase II δB (CaMKIIδB) is one of the predominant isoforms of CaMKII in the heart. The precise role of CaMKIIδB in the transcriptional cross-talk of Ca²âº-handling proteins during heart failure remains unclear. In this work, we aim to determine the mechanism of CaMKIIδB in modulating the expression of sarcolemmal Na⁺-Ca²âº exchange (NCX1). We also aim to address the potential effects of calmodulin antagonism on the imbalance of NCX1 and sarcoendoplasmic reticulum Ca²âº ATPase (SERCA) during heart failure. Eight weeks after transverse aortic constriction (TAC)-induced heart failure in mice, we found that the heart weight/tibia length (HW/TL) ratio and the lung weight/body weight (LW/BW) ratio increased by 59% and 133%, respectively. We further found that the left ventricle-shortening fraction decreased by 40% compared with the sham-operated controls. Immunoblotting revealed that the phosphorylation of CaMKIIδB significantly increased 8 weeks after TAC-induced heart failure. NCX1 protein levels were also elevated, whereas SERCA2 protein levels decreased in the same animal model. Moreover, transfection of active CaMKIIδB significantly increased NCX1 protein levels in adult mouse cardiomyocytes via class IIa histone deacetylase (HDAC)/myocyte enhancer factor-2 (MEF2)-dependent signaling. In addition, pharmacological inhibition of calmodulin/CaMKIIδB activity improved cardiac function in TAC mice, which partially normalized the imbalance between NCX1 and SERCA2. These data identify NCX1 as a cellular target for CaMKIIδB. We also suggest that the CaMKIIδB-induced imbalance between NCX1 and SERCA2 is partially responsible for the disturbance of intracellular Ca²âº homeostasis and the pathological process of heart failure.


Subject(s)
Aortic Diseases/complications , Aortic Diseases/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Heart Failure/etiology , Heart Failure/metabolism , Sodium-Calcium Exchanger/metabolism , Animals , Blotting, Western , Body Weight , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Heart/physiology , Immunohistochemistry , Lung/metabolism , Lung/physiology , Male , Mice , Organ Size/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases , Sodium-Calcium Exchanger/genetics
6.
Eur J Pharmacol ; 643(1): 48-57, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20599932

ABSTRACT

Bromocriptine, a dopamine D(2) receptor agonist, has widely been used for patients with Parkinson's disease. The aim of the present study was to investigate the effect of bromocriptine on glutamate transporter. Since the astroglial glutamate transporter GLT-1 (EAAT2) is the predominant isoform in the forebrain, we generated EAAT2-expressing human embryonic kidney cells and immortalized mouse astrocytes. In the present studies, we observed a GLT-1-immunoreactive band and significant Na(+)-dependent d-[(3)H] aspartate uptake. Furthermore, the glutamate transporter inhibitors, dl-threo-beta-benzyloxyaspartic acid (TBOA) and dihydrokainate (DHK), displayed a dose-dependent reduction of d-[(3)H] aspartate uptake in both types of cells. In contrast, cells exposed to either chemical anoxia or high KCl elicited a marked release of d-[(3)H] aspartate, and the release was inhibited by TBOA and DHK, implying the contribution of glutamate transporter reversal. Interestingly, we found that bromocriptine dose-dependently inhibits d-[(3)H] aspartate release elicited by chemical anoxia or high KCl, while no changes occurred in the uptake. The inhibitory action of bromocriptine was not affected by sulpiride, a dopamine D(2) receptor antagonist. On the other hand, bromocriptine had no effect on swelling-induced d-[(3)H] aspartate release, which is mediated by volume-regulated anion channels. In vivo studies revealed that bromocriptine suppresses the excessive elevation of glutamate levels in gerbils subjected to transient forebrain ischemia in a manner similar to DHK. Taken together, these results provide evidence that bromocriptine inhibits excitatory amino acid release via reversed operation of GLT-1 without altering forward transport.


Subject(s)
Astrocytes/drug effects , Bromocriptine/pharmacology , Dopamine Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Amino Acid Transporter 2/metabolism , Excitatory Amino Acids/antagonists & inhibitors , Animals , Astrocytes/metabolism , Blotting, Western , Cell Line , Dose-Response Relationship, Drug , Glutamic Acid/metabolism , Humans , Mice , Transfection
7.
J Pharmacol Exp Ther ; 332(2): 421-8, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19889795

ABSTRACT

Using a heart ischemia/reperfusion model in rats, we recently demonstrated that 3-[2-[4-(3-chloro-2-methylphenyl)-1-piperazinyl]ethyl]-5,6-dimethoxy-1-(4-imidazolylmethyl)-1H-indazole dihydrochloride 3.5 hydrate (DY-9760e), a calmodulin inhibitor, is a cardioprotective drug. Here, we examined cardioprotective mechanisms of DY-9760e in hypertrophy and heart failure using a mouse transverse aortic constriction (TAC) model. Mice were subjected to TAC and 2 weeks later they were administered DY-9760e for another 6 weeks (at 10 or 20 mg/kg/day p.o.). Chronic administration inhibited TAC-induced increased heart-to-body weight ratio dose-dependently. Consistent with inhibition of hypertrophy, fraction shortening, an indicator of heart contractile function, assessed by echocardiography was completely restored by DY-9760e (20 mg/kg/day) administration. Inhibition of TAC-induced atrial natriuretic peptide (ANP) up-regulation further confirmed an antihypertrophic effect of DY-9760e. It is noteworthy that we found that breakdown of dystrophin and spectrin by calpain was associated with heart failure in TAC mice. Caveolin-3 breakdown was closely associated with endothelial nitric-oxide synthase (eNOS) dissociation from the plasma membrane and its subsequent uncoupling. Uncoupled monomeric eNOS formation was associated with increased protein tyrosine nitration, suggesting peroxynitrite production and NO and superoxide formation. It is important to note that 6 weeks of DY-9760e treatment significantly blocked hypertrophic responses, such as increased heart weight and ANP induction. Overall, we show that inhibition of both dystrophin/spectrin breakdown and uncoupling of eNOS probably underlies the cardioprotective mechanisms of DY-9760e. The observed protection of sarcolemmal proteins and eNOS by DY-9760e during pressure overload suggests a novel therapeutic strategy to rescue the heart from hypertrophy-induced failure.


Subject(s)
Cardiotonic Agents/pharmacology , Dystrophin/metabolism , Heart/drug effects , Hypertrophy, Left Ventricular/drug therapy , Indazoles/pharmacology , Nitric Oxide Synthase Type III/metabolism , Animals , Atrial Natriuretic Factor/metabolism , Cardiotonic Agents/administration & dosage , Caveolin 3/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Glycoproteins/pharmacology , Heart Failure/prevention & control , Indazoles/administration & dosage , Male , Mice , Mice, Inbred Strains , Myocardium/metabolism , Myocardium/pathology , Spectrin/metabolism
8.
Cardiovasc Ther ; 27(1): 17-27, 2009.
Article in English | MEDLINE | ID: mdl-19207476

ABSTRACT

Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) and extracellular signal-regulated kinase (ERK) have pivotal roles in endothelin-1 (ET-1)-induced cardiomyocyte hypertrophy. We here tested whether a novel CaM antagonist, DY-9760e inhibits ET-1-induced hypertrophy through inhibition of CaMKII and ERK activities. We first confirmed that Ca(2+) oscillation induced by ET-1 treatment elicits transient activation of CaMKII and ERK in cultured cardiomyocytes. DY-9760e treatment with 3 microM totally and partially inhibited the ET-1-induced CaMKII and ERK activation, respectively. The ET-1-induced ERK activation was also partially blocked by a CaMKII inhibitor, KN93. To confirm involvement of CaMKII activity in the ERK activation by ET-1 and A23187, cultured cardiomyocytes were transfected with a constitutively active CaMKII. The transfection with the active CaMKII elicited ERK activation in cultured cardiomyocytes and cotransfection with dominant negative CaMKII eliminated its ERK activation. Consistent with inhibitory actions of DY-9760e on the ET-1-induced CaMKII and ERK activation, induction of hypertrophy-related genes including atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) was significantly inhibited by DY-9760e treatment. Combination treatment with DY-9760e and U0126, a MEK inhibitor, totally blocked the ET-1-induced ANP and BNP expression. DY-9760e treatment (3 microM) significantly inhibited the ET-1-induced hypertrophy and combination treatment with DY-9760e and U0126 totally blocked the ET-1-induced hypertrophy in cultured cardiomyocytes. These results suggest that DY-9760e elicits antihypertrophic action on ET-1-induced cardiac hypertrophy through inhibition of CaMKII and ERK activation and that CaMKII activity in part mediates ET-1-induced ERK activation.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Cardiomegaly/prevention & control , Endothelin-1/metabolism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Indazoles/pharmacology , Myocytes, Cardiac/drug effects , Protein Kinase Inhibitors/pharmacology , Animals , Animals, Newborn , Atrial Natriuretic Factor/metabolism , Benzylamines/pharmacology , Butadienes/pharmacology , Calcimycin/pharmacology , Calcium Signaling/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cardiomegaly/enzymology , Cardiomegaly/pathology , Cell Proliferation/drug effects , Cell Size/drug effects , Cells, Cultured , DNA Replication/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Ionophores/pharmacology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Natriuretic Peptide, Brain/metabolism , Nitriles/pharmacology , Phosphorylation , RNA, Messenger/metabolism , Rats , Rats, Wistar , Sulfonamides/pharmacology , Time Factors , Transfection
9.
Mol Pharmacol ; 75(1): 101-12, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18952768

ABSTRACT

The pathophysiological relevance of endothelial nitric-oxide synthase (eNOS)-induced superoxide production in cardiomyocyte injury after prolonged phenylephrine (PE) exposure remains unclear. The aims of this study were to define the mechanism of O2(*) production by uncoupled eNOS and evaluate the therapeutic potential of a novel calmodulin antagonist 3-[2-[4-(3-chloro-2-methylphenyl)-1-piperazinyl]ethyl]-5,6-dimethoxyindazole (DY-9836) to rescue hypertrophied cardiomyocytes from PE-induced injury. In cultured rat cardiomyocytes, prolonged exposure for 96 h to PE led to translocation from membrane to cytosol of eNOS and breakdown of caveolin-3 and dystrophin. When NO and O2(*) production were monitored in PE-treated cells by 4-amino-5-methylamino-2',7'-difluorofluorescein and dihydroethidium, respectively, Ca(2+)-induced NO production elevated by 5.7-fold (p < 0.01) after 48-h PE treatment, and the basal NO concentration markedly elevated (16-fold; p < 0.01) after 96-h PE treatment. On the other hand, the O2(*) generation at 96 h was closely associated with an increased uncoupled eNOS level. Coincubation with DY-9836 (3 microM) during the last 48 h inhibited the aberrant O2(*) generation nearly completely and NO production by 72% (p < 0.01) after 96 h of PE treatment and inhibited the breakdown of caveolin-3/dystrophin in cardiomyocytes. PE-induced apoptosis assessed by TdT-mediated dUTP nick-end labeling staining was also attenuated by DY-9836 treatment. These results suggest that O2(*) generation by uncoupled eNOS probably triggers PE-induced cardiomyocyte injury. Inhibition of abnormal O2(*) and NO generation by DY-9836 treatment represents an attractive therapeutic strategy for PE/hypertrophy-induced cardiomyocyte injury.


Subject(s)
Calmodulin/antagonists & inhibitors , Cardiotonic Agents/pharmacology , Indazoles/pharmacology , Myocytes, Cardiac/drug effects , Piperazines/pharmacology , Superoxides/metabolism , Animals , Animals, Newborn , Caveolin 3/metabolism , Cells, Cultured , Dystrophin/metabolism , Fluorescent Antibody Technique, Indirect , Fluorescent Dyes/metabolism , Heart Ventricles/cytology , Immunohistochemistry , Myocytes, Cardiac/enzymology , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type III/metabolism , Phalloidine/metabolism , Phenylephrine/pharmacology , Rats , Rats, Wistar , Rhodamines/metabolism , Superoxides/analysis , Time Factors
10.
Biochem Pharmacol ; 74(12): 1727-37, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-17888407

ABSTRACT

Cardiac hypertrophy impairs Ca(2+) handling in the sarcoplasmic reticulum, thereby impairing cardiac contraction. To identify the mechanisms underlying impaired Ca(2+) release from the sarcoplasmic reticulum in hypertrophic cardiomyocytes, we assessed Ca(2+)-dependent signaling and the phosphorylation of phospholamban, which regulates Ca(2+) uptake during myocardial relaxation and is in turn regulated by Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) and calcineurin. In cultured rat cardiomyocytes, treatment with endothelin-1, angiotensin II, and phenylephrine-induced hypertrophy and increased CaMKII autophosphorylation and calcineurin expression. The calcineurin level reached its maximum at 72h and remained elevated for at least 96h after endothelin-1 or angiotensin II treatment. By contrast, CaMKII autophosphorylation, phospholamban phosphorylation, and caffeine-induced Ca(2+) mobilization all peaked 48h after these treatments. By 96h after treatment, CaMKII autophosphorylation and phospholamban phosphorylation had returned to baseline, and caffeine-induced Ca(2+) mobilization was impaired relative to baseline. A similar biphasic change was observed in dystrophin levels in endothelin-1-induced hypertrophic cardiomyocytes, and treatment with the novel CaM antagonists DY-9760e and DY-9836 significantly inhibited the hypertrophy-induced dystrophin breakdown. Taken together, the abnormal Ca(2+) regulation in cardiomyocytes following hypertrophy is in part mediated by an imbalance in calcineurin and CaMKII activities, which leads to abnormal phospholamban activity.


Subject(s)
Calcineurin/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium/metabolism , Cardiomegaly/metabolism , Angiotensin II/pharmacology , Animals , Calcium-Binding Proteins/metabolism , Cardiomegaly/chemically induced , Cardiomegaly/enzymology , Cells, Cultured , Endothelin-1/pharmacology , Immunohistochemistry , Indazoles/pharmacology , Phenylephrine/pharmacology , Phosphorylation , Rats , Rats, Wistar
11.
J Neurochem ; 99(1): 97-106, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16987238

ABSTRACT

Microsphere embolism (ME)-induced up-regulation of endothelial nitric oxide synthase (eNOS) in endothelial cells of brain microvessels was observed 2-48 h after ischemia. eNOS induction preceded disruption of the blood-brain barrier (BBB) observed 6-72 h after ischemia. In vascular endothelial cells, ME-induced eNOS expression was closely associated with protein tyrosine nitration, which is a marker of generation of peroxynitrite. Leakage of rabbit IgG from microvessels was also evident around protein tyrosine nitration-immunoreactive microvessels. To determine whether eNOS expression and protein tyrosine nitration in vascular endothelial cells mediates BBB disruption in the ME brain, we tested the effect of a novel calmodulin-dependent NOS inhibitor, 3-[2-[4-(3-chloro-2-methylphenyl)-1-piperazinyl]ethyl]-5,6-dimethoxy-1-(4-imidazolylmethyl)-1H-indazole dihydrochloride 3.5 hydrate (DY-9760e), which inhibits eNOS activity and, in turn, protein tyrosine nitration. Concomitant with inhibition of protein tyrosine nitration in vascular endothelial cells, DY-9760e significantly inhibited BBB disruption as assessed by Evans blue (EB) excretion. DY-9760e also inhibited cleavage of poly (ADP-ribose) polymerase as a marker of the apoptotic pathway in vascular endothelial cells. Taken together with previous evidence in which DY-9760e inhibited brain edema, ME-induced eNOS expression in vascular endothelial cells likely mediates BBB disruption and, in turn, brain edema.


Subject(s)
Blood-Brain Barrier/physiology , Brain/enzymology , Embolism/physiopathology , Nitric Oxide Synthase Type III/biosynthesis , Animals , Brain Ischemia/enzymology , Brain Ischemia/physiopathology , Calmodulin/antagonists & inhibitors , Cerebrovascular Circulation , Embolism/etiology , Endothelium, Vascular/enzymology , Enzyme Induction , Immunohistochemistry , Indazoles/pharmacology , Microspheres , Rats
12.
Cardiovasc Drug Rev ; 24(2): 88-100, 2006.
Article in English | MEDLINE | ID: mdl-16961723

ABSTRACT

DY-9760e (3-[2-[4-(3-chloro-2-methylphenyl)-1-piperazinyl]ethyl]-5,6-dimethoxy-1-(4-imidazolylmethyl)-1H-indazole dihydrochloride-3.5 hydrate) inhibits Ca(2+)/CaM-dependent nitric oxide synthase (NOS), thereby inhibiting nitric oxide (NO) production. In cardiomyocytes from ischemic rat heart NO and superoxide levels are increased causing protein tyrosine nitration. In hearts subjected to ischemia/reperfusion DY-9760e totally abolishes protein tyrosine nitration. Notably, DY-9760e also inhibits calpain and cas-pase-3 activation that occurs prior to apoptosis in cardiomyocytes. In ischemic hearts fodrin is the substrate for calpain. DY-9760e inhibits fodrin breakdown in the peri-infarct area rather than in the infarct core. In the ischemic rat brain DY-9760e inhibits caspase-3-induced proteolysis of calpastatin, an endogenous calpain inhibitor, suggesting that crosstalk between calpain and caspase-3 is mediated by calpastatin breakdown. Thus, DY-9760e rescues neurons and cardiomyocytes from ischemic injury by inhibiting crosstalk between calpain and caspase-3 as well as protein tyrosine nitration.


Subject(s)
Calmodulin/antagonists & inhibitors , Cardiotonic Agents/therapeutic use , Indazoles/therapeutic use , Myocardial Ischemia/drug therapy , Animals , Cardiotonic Agents/chemistry , Cardiotonic Agents/pharmacology , Disease Models, Animal , Humans , Indazoles/chemistry , Indazoles/pharmacology , Models, Cardiovascular , Molecular Structure , Myocardial Ischemia/physiopathology , Myocardial Reperfusion Injury/physiopathology , Myocardial Reperfusion Injury/prevention & control
13.
J Neurochem ; 98(1): 310-20, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16805817

ABSTRACT

Calpain, a Ca(2+)-dependent cysteine protease, in vitro converts calcineurin (CaN) to constitutively active forms of 45 kDa and 48 kDa by cleaving the autoinhibitory domain of the 60 kDa subunit. In a mouse middle cerebral artery occlusion (MCAO) model, calpain converted the CaN A subunit to the constitutively active form with 48 kDa in vivo. We also confirmed increased Ca(2+)/CaM-independent CaN activity in brain extracts. The generation of constitutively active and Ca(2+)/CaM-independent activity of CaN peaked 2 h after reperfusion in brain extracts. Increased constitutively active CaN activity was associated with dephosphorylation of dopamine-regulated phosphoprotein-32 in the brain. Generation of constitutively active CaN was accompanied by translocation of nuclear factor of activated T-cells (NFAT) into nuclei of hippocampal CA1 pyramidal neurons. In addition, a novel calmodulin antagonist, DY-9760e, blocked the generation of constitutively active CaN by calpain, thereby inhibiting NFAT nuclear translocation. Together with previous studies indicating that NFAT plays a critical role in apoptosis, we propose that calpain-induced CaN activation in part mediates delayed neuronal death in brain ischemia.


Subject(s)
Brain Ischemia/pathology , Calcineurin/metabolism , Calpain/pharmacology , Neurons/drug effects , Analysis of Variance , Animals , Brain Ischemia/metabolism , Carrier Proteins/metabolism , Caseins/pharmacokinetics , Cell Death/drug effects , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Enzyme Activation/drug effects , Immunohistochemistry/methods , Immunoprecipitation/methods , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins/metabolism , Molecular Weight , Nerve Tissue Proteins/metabolism , Phosphorus Isotopes/pharmacokinetics , Protein Subunits/metabolism , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism
14.
Brain Res ; 1083(1): 189-95, 2006 Apr 14.
Article in English | MEDLINE | ID: mdl-16545345

ABSTRACT

A large body of evidence indicates that disturbances of Ca(2+) homeostasis may be a causative factor in the neurotoxicity following cerebral ischemia. However, the mechanisms by which Ca(2+) overload leads to neuronal cell death have not been fully elucidated. Calmodulin, a major intracellular Ca(2+)-binding protein found mainly in the central nervous system, mediates many physiological functions in response to changes in the intracellular Ca(2+) concentration, whereas Ca(2+) overload in neurons after excitotoxic insult may induce excessive activation of calmodulin signaling pathways, leading to neuronal cell death. To determine the role of calmodulin in the induction of neuronal cell death, we generated primary rat cortical neurons that express a mutant calmodulin with a defect in Ca(2+)-binding affinity. Neurons expressing the mutant had low responses of calmodulin-dependent signaling to membrane depolarization by high KCl and became resistant to glutamate-triggered excitotoxic neuronal cell death compared with the vector or wild-type calmodulin-transfected cells, indicating that blocking calmodulin function is protective against excitotoxic insult. These results suggest that calmodulin plays a crucial role in the processes of Ca(2+)-induced neuronal cell death and the possibility that the blockage of calmodulin attenuates brain injury after cerebral ischemia.


Subject(s)
Brain Ischemia/metabolism , Calcium Signaling/genetics , Calcium/metabolism , Calmodulin/metabolism , Cerebral Infarction/metabolism , Nerve Degeneration/metabolism , Animals , Binding Sites/genetics , Brain/drug effects , Brain/metabolism , Brain/physiopathology , Brain Ischemia/drug therapy , Brain Ischemia/physiopathology , Calcium Signaling/drug effects , Calmodulin/antagonists & inhibitors , Calmodulin/genetics , Cell Death/drug effects , Cell Death/genetics , Cells, Cultured , Cerebral Infarction/physiopathology , Cerebral Infarction/prevention & control , Cytoprotection/drug effects , Cytoprotection/genetics , Glutamic Acid/metabolism , Glutamic Acid/pharmacology , Humans , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mutation/genetics , Nerve Degeneration/physiopathology , Neurotoxins/metabolism , Neurotoxins/pharmacology , Potassium Chloride/pharmacology , Rats , Rats, Wistar , Transfection
15.
J Pharmacol Exp Ther ; 317(2): 529-36, 2006 May.
Article in English | MEDLINE | ID: mdl-16467455

ABSTRACT

Microsphere embolism (ME)-induced cerebral ischemia can elicit various pathological events leading to neuronal death. Western blotting and immunohistochemical studies revealed that expression of calpastatin, an endogenous calpain inhibitor, decreased after ME induction. Calpain activation after ME was apparently due to, in part, a decrease in calpastatin in a late phase of neuronal injury. The time course of that decrease also paralleled caspase-3 activation. In vitro studies demonstrated that calpastatin was degraded by caspase-3 in a Ca(2+)/calmodulin (CaM)-dependent manner. Because CaM binds directly to calpastatin, we asked whether a novel CaM antagonist, 3-[2-[4-(3-chloro-2-methylphenylmethyl)-1-piperazinyl]ethyl]-5,6-dimethoxy-1-(4-imidazolylmethyl)-1H-indazole dihydro-chloride 3.5 hydrate (DY-9760e), inhibits caspase-3-induced calpastatin degradation during ME-induced neuronal damage. We also tested the effect of DY-9760e on degradation of fodrin, a calpain substrate. Consistent with our hypothesis, DY-9760e (25 or 50 mg/kg i.p.) treatment inhibited degradation of calpastatin and fodrin in a dose-dependent manner. Because DY-9760e showed powerful neuroprotective activity with concomitant inhibition of calpastatin degradation, cross-talk between calpain and caspase-3 through calpastatin possibly accounts for ME-induced neuronal injury. Taken together, both inhibition of caspase-3-induced calpastatin degradation and calpain-induced fodrin breakdown by DY-9760e in part mediate its neuroprotective action.


Subject(s)
Brain Ischemia/prevention & control , Calcium-Binding Proteins/biosynthesis , Calpain/antagonists & inhibitors , Caspases/metabolism , Indazoles/therapeutic use , Neuroprotective Agents/therapeutic use , Animals , Brain Ischemia/enzymology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Calmodulin/antagonists & inhibitors , Calmodulin/metabolism , Caspase 3 , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Activation , Indazoles/pharmacology , Intracranial Embolism/complications , Male , Microspheres , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/pharmacology , Protein Binding , Rats , Rats, Wistar
16.
Biol Pharm Bull ; 28(9): 1658-61, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16141535

ABSTRACT

Microsphere embolism (ME)-induced ischemia model in rat resembles to multiple brain embolism in human with several clinical features. We here tested whether nitric oxide (NO) production contributes to the neuronal injury in the ME model. A novel calmodulin antagonist, DY-9760e, having a potent inhibitory effect on neuronal nitric oxide synthase (nNOS), reduced brain infarct size in the ME-induced brain ischemia. Consistent with our previous observation with gerbil ischemia/reperfusion model, DY-9760e completely inhibited NO production immediately after and 24 or 48 h after ME. Unlike the gerbil ischemia/reperfusion model, protein tyrosine nitration markedly increased 6-48 h after ME. DY-9760e treatment completely inhibited the marked increase in the protein tyrosine nitration at 24 h after ME. These results suggest that the inhibition of NO production and protein tyrosine nitration by DY-9760e contribute to its neuroprotective action in the ME-induced brain damage.


Subject(s)
Calmodulin/antagonists & inhibitors , Indazoles/pharmacology , Intracranial Embolism/drug therapy , Intracranial Embolism/metabolism , Neuroprotective Agents , Nitrates/metabolism , Nitric Oxide/biosynthesis , Tyrosine/metabolism , Animals , Blotting, Western , Brain/pathology , Cerebral Infarction/pathology , Injections, Intraperitoneal , Intracranial Embolism/pathology , Microdialysis , Microspheres , Neostriatum/drug effects , Neostriatum/metabolism , Rats
17.
J Pharmacol Sci ; 98(2): 142-50, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15937403

ABSTRACT

We here assessed the effects of 3-[2-[4-(3-chloro-2-methylphenyl)-1-piperazinyl]ethyl]-5,6-dimethoxy-1-(4-imidazolylmethyl)-1H-indazole dihydrochloride 3.5 hydrate (DY-9760e), a novel calmodulin antagonist, on infarct size in the rat heart subjected to ischemia/reperfusion. Rats were subjected to a 30-min coronary occlusion followed by a 24-h reperfusion. DY-9760e was intravenously infused for 20 min, starting at 20 min after coronary occlusion. Treatment with DY-9760e (10 mg/kg) significantly reduced the infarct size in the risk area assessed by Evans Blue/TTC (triphenyltetrazolium chloride) staining. DY-9760e treatment also ameliorated contractile dysfunction of the left ventricle 72 h after reperfusion. DY-9760e significantly inhibited fodrin breakdown and caspase-3 activation. The inhibitory effect of DY-9760e on the fodrin breakdown was prominent in the rim rather than in the center of the risk area. DY-9760e also blocked protein tyrosine nitration associated with infarction. These results suggest that the cardioprotective effect of DY-9760e involved inhibition of calpain/caspase activation and protein tyrosine nitration.


Subject(s)
Carrier Proteins/metabolism , Indazoles/pharmacology , Microfilament Proteins/metabolism , Myocardial Reperfusion Injury/prevention & control , Proteins/metabolism , Tyrosine/metabolism , Animals , Blotting, Western , Caspase 3 , Caspase Inhibitors , Caspases/metabolism , Enzyme Activation , Male , Proteins/chemistry , Rats , Rats, Sprague-Dawley , Ventricular Function, Left/drug effects
18.
Biol Pharm Bull ; 28(4): 629-33, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15802800

ABSTRACT

DY-9760e (3-[2-[4-(3-chloro-2-methylphenyl)-1-piperazinyl]ethyl]-5,6-dimethoxy-1-(4-imidazolylmethyl)-1H-indazole dihydrochloride 3.5 hydrate), a calmodulin antagonist, provides protection against Ca(2+) overload-associated cytotoxicity and brain injury after cerebral ischemia in rats. In this study, we assessed the effect of DY-9760e on ischemic infarct volume in cats subjected to permanent focal cerebral ischemia. DY-9760e was infused for 6 h, beginning 5 min after occlusion of the middle cerebral artery. The infarct volume was measured at the end of drug infusion. DY-9760e, at the dose of 0.25 but not 0.1 mg/kg/h, significantly reduced cerebral infarct volume without affecting any physiological parameters, and its protective effect was mainly evident in the cerebral cortex, where the penumbra, a salvageable zone, exists. The present study demonstrates that DY-9760e protects against brain injury after focal ischemia in a gyrencephalic animal as well as in the rodents reported previously and suggests its therapeutic value for the treatment of acute stroke.


Subject(s)
Brain Ischemia/drug therapy , Calmodulin/antagonists & inhibitors , Hypoxia, Brain/drug therapy , Indazoles/pharmacology , Animals , Brain Ischemia/physiopathology , Cats , Dose-Response Relationship, Drug , Hypoxia, Brain/physiopathology , Male , Telencephalon/pathology
19.
Biol Pharm Bull ; 27(11): 1788-91, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15516724

ABSTRACT

An excessive elevation of intracellular Ca(2+) levels is known to play a key role in the pathological events following cerebral ischemia. DY-9760e, 3-[2-[4-(3-chloro-2-methylphenylmethyl)-1-piperazinyl]ethyl]-5,6-dimethoxy-1-(4-imidazolylmethyl)-1H-indazole dihydrochloride 3.5 hydrate, is a potent calmodulin antagonist that attenuates brain damage in focal ischemia models. In the present study, we investigated the effect of DY-9760e on neuronal cell death induced by a variety of cell-toxic stimuli that increase intracellular Ca(2+). Cell death was induced by the exposure of primary cultured neurons to excitotoxic agents such as glutamate and N-methyl-D-aspartate, membrane-depolarizing agents such as veratridine and high KCl, or thapsigargin an endoplasmic reticulum Ca(2+)-ATPase inhibitor. Treatment with DY-9760e resulted in a dose-dependent prevention of neuronal cell death elicited by excitotoxicity, voltage-gated channel opening, and inhibition of endoplasmic reticulum Ca(2+)-ATPase. These results indicate that DY-9760e can rescue neurons from various types of cell-toxic stimuli, which may contribute to attenuation of brain injury after cerebral ischemia.


Subject(s)
Calmodulin/antagonists & inhibitors , Indazoles/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Calcium/metabolism , Calcium Channels/drug effects , Calcium Channels/physiology , Calcium-Transporting ATPases/antagonists & inhibitors , Cell Death/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Endoplasmic Reticulum/enzymology , Glutamic Acid/toxicity , Indazoles/administration & dosage , Ion Channel Gating , N-Methylaspartate/toxicity , Neurons/cytology , Neuroprotective Agents/administration & dosage , Patch-Clamp Techniques , Potassium Chloride/toxicity , Rats , Sodium Channels/drug effects , Sodium Channels/physiology , Thapsigargin/toxicity , Veratridine/toxicity
20.
J Pharmacol Sci ; 96(1): 65-72, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15359085

ABSTRACT

The novel calmodulin (CaM) antagonist DY-9760e (3-[2-[4-(3-chloro-2-methylphenyl)-1-piperazinyl]ethyl]-5,6-dimethoxy-1-(4-imidazolylmethyl)-1H-indazole dihydrochloride 3.5 hydrate) with an apparent neuroprotective effect in vivo preferentially inhibits neuronal nitric oxide synthase (nNOS), Ca2+/CaM-dependent protein kinase IIalpha (CaMKIIalpha), and calcineurin in vitro. In the present study, we investigated the molecular mechanism underlying its neuroprotective effect with the gerbil transient forebrain ischemia model, by focusing on its inhibition of these Ca2+/CaM-dependent enzymes. Post-ischemic DY-9760e treatment (5 mg/kg, i.p.) immediately after 5-min ischemia significantly reduced the delayed neuronal death in the hippocampal CA1 region. CaMKIIalpha was transiently autophosphorylated immediately after reperfusion with concomitant sustained decrease in its total amounts in the Triton X-100-soluble fractions. Calcineurin activity, accessed by the phosphorylation state of dopamine- and cAMP-regulated phosphoprotein of Mr 32,000 (DARPP-32) at Thr34, was elevated at 6 h after reperfusion. Post-treatment of DY-9760e had no effects on both CaMKIIalpha and DARPP-32 phosphorylation at 6 h after reperfusion. However, DY-9760e significantly inhibited nitrotyrosine formation, as a biomarker of NO, and in turn, peroxynitrite (ONOO-) production. These results suggest that DY-9760e primarily inhibits Ca2+/CaM-dependent neuronal NOS, without any effects on CaMKII and calcineurin, and the inhibition of NO production possibly accounts for its neuroprotective action in brain ischemic injury.


Subject(s)
Brain Ischemia/drug therapy , Calmodulin/antagonists & inhibitors , Hippocampus/drug effects , Indazoles/administration & dosage , Neurons/drug effects , Animals , Brain Ischemia/metabolism , Brain Ischemia/pathology , Calcium-Calmodulin-Dependent Protein Kinase Kinase , Calmodulin/metabolism , Cell Death/drug effects , Cell Death/physiology , Gerbillinae , Hippocampus/metabolism , Hippocampus/pathology , Male , Neurons/metabolism , Neurons/pathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...