Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Nucl Med ; 49(7): 1183-8, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18552135

ABSTRACT

UNLABELLED: The purpose of the present study was to measure adenosine A(2A) receptor (A(2A)R) occupancy in the brain by a novel adenosine A(1)/A(2A) antagonist, 5-[5-amino-3-(4fluorophenyl)pyrazin-2-yl]-1-isopropylpyridine-2(1H)-one (ASP5854), and to determine the degree of receptor occupancy necessary to inhibit haloperidol-induced catalepsy in rhesus monkeys. METHODS: A(2A)R occupancy by ASP5854 (0.001-0.1 mg/kg) was examined in the striatum using an A(2A)R-specific radiotracer, (11)C-SCH442416, and PET in conscious rhesus monkeys. A(2A)R occupancy was monitored after a single intravenous administration of ASP5854 in 3 animals, and a dynamic PET scan was performed at 1, 4, and 8 h after an intravenous bolus injection of the tracer for approximately 740 MBq. Catalepsy was induced by haloperidol (0.03 mg/kg, intramuscularly) and examined for incidence and duration. RESULTS: ASP5854 dose-dependently increased A(2A)R occupancy in the striatum and showed long-lasting occupancy even after the reduction of plasma concentration. Haloperidol induced severe catalepsy at 40 min after intramuscular injection. The incidence and duration of cataleptic posture were dose-dependently reduced by ASP5854 at 1 h after oral administration, and the minimum ED(50) value was 0.1 mg/kg. Administration of a dose of 0.1 mg/kg yielded a plasma concentration of 97 +/- 16.3 ng/mL, which corresponded to 85%-90% of A(2A)R occupancy. CONCLUSION: These results showed that ASP5854 antagonized A(2A)R in the striatum, and the dissociation from A(2A)R was relatively slow. In addition, more than 85% A(2A)R occupancy by ASP5854 resulted in an inhibition of haloperidol-induced catalepsy. Thus, such a pharmacodynamic study directly demonstrates both the kinetics of a drug in the brain and the relationship between dose-dependent receptor occupancy and plasma level.


Subject(s)
Adenosine A1 Receptor Antagonists , Aminopyridines/metabolism , Corpus Striatum/metabolism , Piperazines/metabolism , Receptor, Adenosine A2A/metabolism , Adenosine A2 Receptor Antagonists , Aminopyridines/therapeutic use , Animals , Carbon Radioisotopes , Catalepsy/chemically induced , Catalepsy/drug therapy , Corpus Striatum/diagnostic imaging , Haloperidol , Macaca mulatta , Male , Piperazines/therapeutic use , Positron-Emission Tomography , Pyrazoles/metabolism , Pyrimidines/metabolism
2.
Bioorg Med Chem Lett ; 17(20): 5577-81, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17804225

ABSTRACT

A successful design of conformationally restricted novel quinazolinone derivatives linked via a cyclopentene moiety as potent poly(ADP-ribose)polymerase-1 (PARP-1) inhibitors has been developed. One selected member of the new series, 8-chloro-2-[(3S)-3-(4-phenylpiperidin-1-yl)cyclopent-1-en-1-yl]quinazolin-4(3H)-one (S-16d), was found to be highly potent with IC(50)=8.7 nM and good brain penetration.


Subject(s)
Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/metabolism , Quinazolinones/chemistry , Quinazolinones/pharmacology , Animals , Cattle , Cross-Linking Reagents/chemistry , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Molecular Conformation , Molecular Structure , Poly(ADP-ribose) Polymerases/chemistry , Quinazolinones/chemical synthesis , Quinazolinones/pharmacokinetics , Structure-Activity Relationship
3.
J Pharmacol Exp Ther ; 323(2): 708-19, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17684118

ABSTRACT

Central adenosine A(2A) receptor is a promising target for drugs to treat Parkinson's disease (PD), and the central blockade of adenosine A(1) receptor improves cognitive function. In the present study, we investigated the effect of a novel adenosine A(1) and A(2A) dual antagonist, 5-[5-amino-3-(4-fluorophenyl) pyrazin-2-yl]-1-isopropylpyridine-2(1H)-one (ASP5854), in animal models of PD and cognition. The binding affinities of ASP5854 for human A(1) and A(2A) receptors were 9.03 and 1.76 nM, respectively, with higher specificity and no species differences. ASP5854 also showed antagonistic action on A(1) and A(2A) agonist-induced increases of intracellular Ca(2+) concentration. ASP5854 ameliorated A(2A) agonist 2-[p-(2-carboxyethyl) phenethylamino]-5'-N-ethylcarboxamidoadenosine (CGS21680)- and haloperidol-induced catalepsy in mice, with the minimum effective doses of 0.32 and 0.1 mg/kg, respectively, and it also improved haloperidol-induced catalepsy in rats at doses higher than 0.1 mg/kg. In unilateral 6-hydroxydopamine-lesioned rats, ASP5854 significantly potentiated l-dihydroxyphenylalanine (L-DOPA)-induced rotational behavior at doses higher than 0.032 mg/kg. ASP5854 also significantly restored the striatal dopamine content reduced by 1-metyl-4-phenyl-1,2,3,6-tetrahydropyridine treatment in mice at doses higher than 0.1 mg/kg. Furthermore, in the rat passive avoidance test, ASP5854 significantly reversed the scopolamine-induced memory deficits, whereas the specific adenosine A(2A) antagonist 8-((E)-2-(3,4-dimethoxyphenyl)ethenyl)-1,3-diethyl-7-methyl-3,7-dihydro-1H-purine-2,6-dione (KW-6002; istradefylline) did not. Scopolamine- or 5H-dibenzo[a,d]cyclohepten-5,10-imine (dizocilpine maleate) (MK-801)-induced impairment of spontaneous alternation in the mouse Y-maze test was ameliorated by ASP5854, whereas KW-6002 did not exert improvement at therapeutically relevant dosages. These results demonstrate that the novel, selective, and orally active dual adenosine A(1) and A(2A) receptors antagonist ASP5854 improves motor impairments, is neuroprotective via A(2A) antagonism, and also enhances cognitive function through A(1) antagonism.


Subject(s)
Adenosine A1 Receptor Antagonists , Adenosine A2 Receptor Antagonists , Aminopyridines/therapeutic use , Cognition Disorders/drug therapy , MPTP Poisoning/drug therapy , Piperazines/therapeutic use , Adenosine/analogs & derivatives , Adenosine/pharmacology , Aminopyridines/pharmacology , Animals , CHO Cells , Cricetinae , Cricetulus , Dopamine/analysis , Dopamine/metabolism , Haloperidol/pharmacology , Humans , Male , Memory Disorders/drug therapy , Mice , Mice, Inbred C57BL , Phenethylamines/pharmacology , Piperazines/pharmacology , Rats , Rats, Inbred F344 , Rats, Wistar , Receptor, Adenosine A1/physiology , Receptor, Adenosine A2A/physiology , Scopolamine/pharmacology
4.
J Pharmacol Exp Ther ; 320(3): 1087-96, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17167170

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear receptor superfamily and function as ligand-modulated transcription factors that regulate gene expression in many important biological processes. The PPARdelta subtype has the highest expression in the brain and is postulated to play a major role in neuronal cell function; however, the precise physiological roles of this receptor remain to be elucidated. Herein, we show that the high-affinity PPARdelta agonists L-165041 [4-[3-(4-acetyl-3-hydroxy-2-propylphenoxy)-propoxyl]phenoxy]-acetic acid] and GW501516 [2-methyl4-((4-methyl-2-(4-trifluoromethylphenyl)-1,3-triazol-5-yl)-methylsulfanyl)phenoxy acetic acid] protect against cytotoxin-induced SH-SY5Y cell injury in vitro and both ischemic brain injury and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) neurotoxicity in vivo. In the SH-SY5Y studies, treatment with L-165041 or GW501516 significantly and concentration-dependently attenuated cell death following thapsigargin, 1-methyl-4-phenylpyridinium, or staurosporine exposure, with the extent of damage correlated with the level of caspase-3 inhibition. In the transient (90 min) middle cerebral artery occlusion model of ischemic brain injury in rats, i.c.v. infusion of L-165041 or GW501516 significantly attenuated the ischemic brain damage measured 24 h after reperfusion. Moreover, the PPARdelta agonists also significantly attenuated MPTP-induced depletion of striatal dopamine and related metabolite contents in mouse brain. These results demonstrate that subtype-selective PPARdelta agonists possess antiapoptotic properties in vitro, which may underlie their potential neuroprotective potential in in vivo experimental models of cerebral ischemia and Parkinson's disease (PD). These findings suggest that PPARdelta agonists could be useful tools for understanding the role of PPARdelta in other neurodegenerative disorders, as well as attractive therapeutic candidates for stroke and neurodegenerative diseases such as PD.


Subject(s)
Brain Ischemia/prevention & control , Brain/drug effects , Neuroprotective Agents/pharmacology , PPAR delta/agonists , Parkinson Disease/prevention & control , Acetates/pharmacokinetics , Acetates/pharmacology , Animals , Brain/metabolism , Brain Ischemia/metabolism , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Dopamine/metabolism , Genes, Reporter , Humans , Ligands , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacokinetics , PPAR delta/genetics , Parkinson Disease/metabolism , Phenols/pharmacokinetics , Phenols/pharmacology , Phenoxyacetates , Rats , Rats, Wistar , Substrate Specificity , Thiazoles/pharmacokinetics , Thiazoles/pharmacology
5.
Bioorg Med Chem ; 14(5): 1378-90, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16288880

ABSTRACT

We disclose herein our efforts aimed at discovery of selective PARP-1 and PARP-2 inhibitors. We have recently discovered several novel classes of quinazolinones, quinazolidinones, and quinoxalines as potent PARP-1 inhibitors, which may represent attractive therapeutic candidates. In PARP enzyme assays using recombinant PARP-1 and PARP-2, the quinazolinone derivatives displayed relatively high selectivity for PARP-1 and quinoxaline derivatives showed superior selectivity for PARP-2, and the quinazolidinone derivatives did not have selectivity for PARP-1/2. Structure-based drug design analysis via a combination of X-ray structural study utilizing the complexes of inhibitors and human PARP-1 catalytic domain, and homology modeling using murine PARP-2 suggested distinct interactions of inhibitors with PARP-1 and PARP-2. These findings provide a new structural framework for the design of selective inhibitors for PARP-1 and PARP-2.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Poly(ADP-ribose) Polymerase Inhibitors , Quinazolines/chemical synthesis , Quinoxalines/chemical synthesis , Binding Sites , Catalytic Domain , Enzyme Inhibitors/pharmacology , Humans , Models, Chemical , Models, Molecular , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases , Quinazolines/pharmacology , Quinoxalines/pharmacology , Structure-Activity Relationship
6.
J Neurotrauma ; 22(11): 1362-73, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16305324

ABSTRACT

Intracerebral hemorrhage (ICH) is the most serious side effect of antithrombotic agents, especially in cases of cerebrovascular disease. In the present study, we compared the exacerbation of ICH and prolongation of bleeding time (BT) in guinea pigs with recombinant tissue plasminogen activator (rt-PA), heparin, aspirin, and FK419, a novel nonpeptide platelet glycoprotein (GP) IIb/IIIa receptor antagonist. ICH was induced by injection of bacterial collagenase into the caudate nucleus; BT was measured with a Simplate R device. Neither heparin nor aspirin prolonged BT. In contrast, rt-PA at the highest dose used in the study did prolong BT, and FK419 caused a dose-dependent prolongation of BT. Moreover, rt-PA and heparin increased the degree of ICH in a dose-dependent manner, leading to death in more than half of the animals treated with higher doses of these drugs. These findings show that the prohemorrhagic mechanisms underlying the prolongation of BT differ from those in collagenase-induced ICH, and that the risk of an agent with antithrombotic effects potentiating hemorrhage in the collagenase-induced model of ICH more closely parallels that in stroke patients than does the effect of the agent on BT. The findings also suggest that antiplatelet agents, including FK419, may be safer than thrombolytic or anticoagulant agents for use in patients at risk for ICH, such as those with stroke or cerebral aneurysm.


Subject(s)
Aspirin/pharmacology , Fibrinolytic Agents/pharmacology , Heparin/pharmacology , Intracranial Hemorrhages/chemically induced , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Tissue Plasminogen Activator/pharmacology , Animals , Bleeding Time , Collagenases/pharmacology , Dose-Response Relationship, Drug , Guinea Pigs , Intracranial Hemorrhages/mortality , Male , Piperidines/pharmacology , Platelet Aggregation/drug effects , Propionates/pharmacology , Recombinant Proteins/pharmacology , Whole Blood Coagulation Time
7.
Bioorg Med Chem Lett ; 15(19): 4221-5, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16098744

ABSTRACT

We have shown that a 4-phenyl-1,2,3,6-tetrahydropyridine fragment plays an important role in improving inhibitory potency against poly(ADP-ribose) polymerase-1 (PARP-1). Various benzamide analogues linked with this fragment via alkyl spacers have been prepared and evaluated. As a result, some of them have been found to be highly potent PARP-1 inhibitors.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/analogs & derivatives , Poly(ADP-ribose) Polymerase Inhibitors , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/chemical synthesis , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Antipyrine , Benzamides , Humans , Inhibitory Concentration 50 , Poly (ADP-Ribose) Polymerase-1 , Structure-Activity Relationship
8.
Bioorg Med Chem ; 13(13): 4343-52, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15927840

ABSTRACT

The discovery of the non-peptide antiplatelet injectable agent FK419 is reported. Based on the beta-turn structure of RGD peptide sequences in the alpha chain of fibrinogen, which binds the glycoprotein IIb/IIIa (GPIIb/IIIa) on the surface of platelets to induce platelet aggregation, the prototype 2 was designed. After further substituent effects were investigated at the alpha-position of the carboxylic acid in 2, we enhanced platelet aggregation inhibition, and discovered the useful feature of reduced prolongation of bleeding time. Finally, the potent platelet aggregation inhibitor FK419 (3) could be discovered. FK419 shows a safe feature of reduced prolongation of bleeding time, as well as potent inhibition of platelet aggregation.


Subject(s)
Piperidines/chemical synthesis , Piperidines/pharmacology , Platelet Aggregation Inhibitors/therapeutic use , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Propionates/chemical synthesis , Propionates/pharmacology , Blood Coagulation/drug effects , Blood Platelets/chemistry , Blood Platelets/metabolism , Humans , Male , Molecular Structure , Oligopeptides/chemistry , Piperidines/chemistry , Propionates/chemistry , Structure-Activity Relationship , von Willebrand Factor/metabolism
9.
J Cereb Blood Flow Metab ; 25(1): 75-86, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15678114

ABSTRACT

The antithrombotic efficacy of FK419, a novel nonpeptide platelet glycoprotein IIb/IIIa antagonist, was compared with tirofiban in guinea-pigs. FK419 and tirofiban similarly inhibited platelet aggregation in vitro (IC50 values: 0.43+/-0.076 and 0.41+/-0.053 micromol/L) and dispersed aggregated platelets (EC50 values: 2.3+/-0.88 and 2.0+/-0.81 micromol/L). FK419 inhibited retention of platelets and neutrophils in a collagen-coated bead column with greater potency than tirofiban (IC50 values of 0.90+/-0.133 and 2.4+/-0.21 micromol/L for platelet retention and 0.32+/-0.078 and 0.57+/-0.180 micromol/L for neutrophil retention). When FK419 or tirofiban were administered after photochemically induced middle cerebral artery (MCA) occlusion in guinea-pigs, they dose-dependently improved MCA patency. FK419 reduced neurological deficits and ischemic brain damage in a dose-dependent fashion, whereas tirofiban did not. Reduced regional cerebral blood flow in the striatum gradually returned to the preoccluded level with FK419 treatment; however, no restoration was observed with tirofiban even though the MCA was recanalized. These results indicate that FK419 ameliorates ischemic brain damage by not only lysing the obstructive thrombus in MCA but also preventing or restoring microcirculation deficits after occlusion/reperfusion, suggesting that FK419 would be an attractive intervention for the treatment of ischemic stroke patients.


Subject(s)
Cerebrovascular Circulation/drug effects , Infarction, Middle Cerebral Artery/drug therapy , Piperidines/administration & dosage , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Propionates/administration & dosage , Tyrosine/analogs & derivatives , Tyrosine/administration & dosage , Animals , Blood Platelets/metabolism , Blood Platelets/pathology , Brain/blood supply , Brain/metabolism , Brain/pathology , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Brain Ischemia/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Guinea Pigs , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Intracranial Thrombosis/metabolism , Intracranial Thrombosis/pathology , Male , Microcirculation/drug effects , Microcirculation/metabolism , Neutrophils/metabolism , Neutrophils/pathology , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Recovery of Function/drug effects , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Tirofiban
10.
J Cereb Blood Flow Metab ; 25(1): 108-18, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15678117

ABSTRACT

The binding of platelet glycoprotein (GP) IIb/IIIa to fibrinogen is the final common pathway in platelet aggregation, a process known to play a key role in the pathogenesis of ischemic brain damage. We compared the effects of FK419, a novel nonpeptide GPIIb/IIIa antagonist, with recombinant tissue plasminogen activator (rt-PA) on middle cerebral artery (MCA) patency and ischemic brain damage in a thrombotic stroke model in squirrel monkeys. FK419 not only inhibited in vitro platelet aggregation (IC50: 88 nmol/L), but also showed disaggregatory activity to aggregated platelet (EC50: 286 nmol/L). FK419 dose-dependently reduced the time to first reperfusion and total occlusion time of MCA blood flow when administered immediately after the termination of photoirradiation. FK419 reduced cerebral infarction and ameliorated neurologic deficits with similar dose-dependency. Although rt-PA reduced the time to first reperfusion, total occlusion time, and cerebral infarction, it did not significantly ameliorate neurologic deficits and induced petechial intracerebral hemorrhages. These results indicate: (1) FK419 restored cerebral blood flow after thrombotic occlusion of MCA, (2) FK419 reduced ischemic brain injury by its thrombolytic actions in a non-human primate stroke model, and (3) FK419 has superior antithrombotic efficacy and is safer than rt-PA.


Subject(s)
Brain Ischemia/drug therapy , Fibrinolytic Agents/administration & dosage , Infarction, Middle Cerebral Artery/drug therapy , Piperidines/administration & dosage , Platelet Aggregation Inhibitors/administration & dosage , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Propionates/administration & dosage , Tissue Plasminogen Activator/administration & dosage , Animals , Blood Platelets/metabolism , Blood Platelets/pathology , Brain/blood supply , Brain/metabolism , Brain/pathology , Brain Ischemia/metabolism , Brain Ischemia/pathology , Cerebral Hemorrhage/drug therapy , Cerebral Hemorrhage/metabolism , Cerebral Hemorrhage/pathology , Cerebrovascular Circulation/drug effects , Dose-Response Relationship, Drug , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Male , Microcirculation/drug effects , Microcirculation/metabolism , Middle Cerebral Artery/metabolism , Middle Cerebral Artery/pathology , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Recovery of Function/drug effects , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Saimiri
11.
Ann Nucl Med ; 18(5): 427-31, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15462405

ABSTRACT

OBJECTIVE: Recent animal PET research has led to the development of PET scanners for small animals. A planar positron imaging system (PPIS) was newly developed to study physiological function in small animals and plants in recent years. To examine the usefulness of PPIS for functional study in small animals, we examined dopaminergic images of mouse striata in MPTP-induced parkinsonism. METHODS: Male C57BL/6NCrj mice were treated with MPTP 7 days before the PPIS study. Scans were performed to measure dopamine D1 receptor binding and dopamine transporter availability with [11C]SCH23390 (about 2 MBq) and [11C]beta-CFT (about 2 MBq), respectively. After the PPIS study, dopamine content in the striatum was measured by HPLC. RESULTS: The MPTP treatment significantly reduced dopamine content in the striatum 7 days after treatment. In the MPTP-treated group, [11C]beta-CFT binding in the striatum was significantly decreased compared with the control group, while striatal [11C]SCH23390 binding was not affected. Dopamine content in the striatum was significantly correlated with the striatal binding of [11C]beta-CFT. CONCLUSION: The present results suggest that PPIS is able to determine brain function in a small animal. Using PPIS, high throughput imaging of small animal brain functions could be achieved.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Brain/diagnostic imaging , Parkinsonian Disorders/diagnostic imaging , Positron-Emission Tomography/methods , Animals , Benzazepines/pharmacokinetics , Disease Models, Animal , Dopamine Antagonists/pharmacokinetics , Mice , Mice, Inbred C57BL , Parkinsonian Disorders/chemically induced , Receptors, Dopamine D1/drug effects , Receptors, Dopamine D1/physiology
12.
Eur J Pharmacol ; 498(1-3): 179-88, 2004 Sep 13.
Article in English | MEDLINE | ID: mdl-15363993

ABSTRACT

We compared the antithrombotic efficacy of FK419 [(S)-2-acetylamino-3-[(R)-[1-[3-(piperidin-4-yl)propionyl]piperidin-3-ylcarbonyl]amino] propionic acid trihydrate], a novel nonpeptide glycoprotein IIb/IIIa antagonist, with recombinant tissue plasminogen activator (rt-PA) and other antithrombotic agents (aspirin, ozagrel, argatroban and heparin). FK419 not only inhibited ADP- and collagen-induced guinea pig platelet aggregation, but also induced disaggregation for ADP-induced aggregated platelets in vitro. In the photochemically induced middle cerebral artery thrombosis model in guinea pigs, FK419 dose-dependently shortened the time to first reperfusion and the total middle cerebral artery occlusion time and reduced ischemic brain damage and ameliorated neurological deficits measured 24 h after middle cerebral artery occlusion. Rt-PA similarly improved the middle cerebral artery patency, brain damage and neurological deficits. Neither aspirin, ozagrel, argatroban nor heparin restored the middle cerebral artery blood flow and improved the brain damage or neurological deficits. These results demonstrated that novel glycoprotein IIb/IIIa antagonist FK419 could disperse thrombus and ameliorated ischemic brain damage, suggesting that FK419 would be an attractive intervention for stroke patients.


Subject(s)
Fibrinolytic Agents/pharmacology , Middle Cerebral Artery/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Thrombosis/drug therapy , Animals , Blood Coagulation/drug effects , Blood Flow Velocity/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Fibrinolysis/drug effects , Fibrinolytic Agents/therapeutic use , Guinea Pigs , Male , Middle Cerebral Artery/pathology , Middle Cerebral Artery/physiopathology , Platelet Aggregation/drug effects , Quinolines/pharmacology , Time Factors
13.
J Med Chem ; 47(17): 4151-4, 2004 Aug 12.
Article in English | MEDLINE | ID: mdl-15293985

ABSTRACT

A novel class of quinazolinone derivatives as potent poly(ADP-ribose)polymerase-1 (PARP-1) inhibitors has been discovered. Key to success was application of a rational discovery strategy involving structure-based design, combinatorial chemistry, and classical SAR for improvement of potency and bioavailability. The new inhibitors were shown to bind to the nicotinamide-ribose binding site (NI site) and the adenosine-ribose binding site (AD site) of NAD+.


Subject(s)
Blood-Brain Barrier/metabolism , Poly(ADP-ribose) Polymerase Inhibitors , Quinazolines/chemical synthesis , Administration, Oral , Animals , Biological Availability , Brain/metabolism , Catalytic Domain , Combinatorial Chemistry Techniques , Dogs , Humans , In Vitro Techniques , Mice , Mice, Inbred C57BL , Microsomes, Liver/metabolism , Models, Molecular , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/pathology , Poly(ADP-ribose) Polymerases/chemistry , Quinazolines/chemistry , Quinazolines/pharmacokinetics , Rats , Structure-Activity Relationship
14.
J Pharmacol Exp Ther ; 310(3): 1114-24, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15113847

ABSTRACT

Methamphetamine (METH) administration in mice, results in a chronic dopamine (DA) depletion associated with nerve terminal damage, with DA oxidation and generation of reactive oxygen species (ROS) primarily mediating this neurotoxicity. The oxidative stress induced by METH putatively activates nuclear enzyme poly(ADP-ribose) polymerase (PARP), with excessive PARP activation eventually leading to cell death. In this study, we show that prevention of PARP activation by treatment with FR261529 [2-(4-chlorophenyl)-5-quinoxalinecarboxamide], the compound that was recently identified as a novel PARP inhibitor (IC50 for PARP-1 = 33 nM, IC50 for PARP-2 = 7 nM), protects against both ROS-induced cells injury in vitro and METH-induced dopaminergic neuronal damage in an in vivo Parkinson's disease (PD) model. In PC12 cells, exposure of hydrogen peroxide or METH markedly induced PARP activation, and treatment with FR261529 (1 microM) significantly reduced PARP activation and attenuated cell death. In the mouse METH model, METH (15 mg/kg x 2 i.p., 2 h apart) intoxication accelerated DA metabolism and oxidation in the striatum, with subsequent cell damage in nigrostriatal dopaminergic neurons after 4 days. Oral administration of FR261529 (10 or 32 mg/kg) attenuated the damage of dopaminergic neurons via marked reduction of PARP activity and not via changes in dopamine metabolism or body temperature. These findings indicate that the neuroprotective effects of a novel PARP inhibitor, FR261529, were accompanied by inhibition of METH-induced PARP activation, suggesting that METH induces nigrostriatal dopaminergic neurodegeneration involving PARP activation and also orally active and brain-penetrable PARP inhibitor FR261529 could be a novel attractive therapeutic candidate for neurodegenerative disorders such as PD.


Subject(s)
Enzyme Inhibitors/pharmacology , Methamphetamine/toxicity , Poly(ADP-ribose) Polymerase Inhibitors , Quinoxalines/pharmacology , Animals , Benzamides/pharmacology , Body Temperature/drug effects , Brain/drug effects , Brain/enzymology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine/metabolism , Enzyme Inhibitors/pharmacokinetics , Lipid Peroxidation/drug effects , Methamphetamine/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/pharmacology , PC12 Cells , Rats , Substantia Nigra/drug effects , Substantia Nigra/metabolism
15.
J Pharmacol Exp Ther ; 309(3): 1067-78, 2004 Jun.
Article in English | MEDLINE | ID: mdl-14985416

ABSTRACT

The massive activation of poly(ADP-ribose) polymerase-1 (PARP-1) by DNA-damaging stimuli, such as exposure to reactive oxygen species (ROS), can lead to cell injury via severe, irreversible depletion of the NAD and ATP pool, and PARP-1 inhibitors have been expected to rescue neurons from degeneration in a number of disease models. We have recently identified 2-[3-[4-(4-chlorophenyl)-1-piperazinyl] propyl]-4(3H)-quinazolinone (FR255595) as a novel and potent PARP-1 inhibitor through structure-based drug design and high-throughput screening. This compound potently inhibited PARP activity with an IC(50) value of 11 nM and was orally active and highly brain penetrable. Here, we show that prevention of PARP activation by FR255595 protects against both ROS-induced cells injury in vitro and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced nigrostriatal dopaminergic damage in an in vivo Parkinson's disease (PD) model. In cell death models in vitro, exposure of hydrogen peroxide induced cell death with PARP overactivation in PC12 cells and SH-SY5Y cells, and pre- and post-treatment with FR255595 (10(-9)-10(-5) M) significantly reduced PARP activation and cell death. In mouse MPTP model, MPTP (20 mg/kg i.p.) intoxication lead to PARP activation and cell damage in the nigrostriatal dopaminergic pathway, which was significantly ameliorated by oral administration of FR255595 (10-32 mg/kg), both in the substantia nigra and in the striatum via marked reduction of PARP activation, even with delayed treatment. These findings clearly indicate that the novel PARP-1 inhibitor FR255595 exerts neuroprotective effect through its potent PARP-1 inhibitory actions in PD model, suggesting that the drug could be an attractive candidate for several neurodegenerative disorders, including PD.


Subject(s)
Cell Death/drug effects , MPTP Poisoning/prevention & control , Piperazines/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors , Quinazolines/therapeutic use , Animals , Cells, Cultured , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , PC12 Cells , Piperazines/pharmacokinetics , Piperazines/pharmacology , Poly(ADP-ribose) Polymerases , Quinazolines/pharmacokinetics , Quinazolines/pharmacology , Quinazolinones , Rats
16.
J Pharmacol Exp Ther ; 308(3): 1094-101, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14634048

ABSTRACT

Platelet activation and subsequent aggregation play a key role in the pathogenesis of ischemic brain damage. Recent studies revealed that enhanced platelet activation is also observed after ischemia, suggesting that secondary thrombus formation might participate in the development of cerebral infarction. The binding of platelet glycoprotein GPIIb/IIIa (integrin alpha(IIb)beta3) to fibrinogen is the final common pathway in platelet aggregation. Therefore, GPIIb/IIIa antagonists might be useful in acute ischemic stroke as well as in the secondary prevention of ischemic stroke. In the present study, we evaluated the effect of three compounds, FK419 ((S)-2-acetylamino-3-[(R)-[1-[3-(piperidin-4-yl) propionyl] piperidin-3-ylcarbonyl] amino] propionic acid trihydrate), a novel nonpeptide GPIIb/IIIa antagonist, ozagrel, a selective thromboxane A(2) synthase inhibitor, and argatroban, a thrombin inhibitor, on middle cerebral artery (MCA) patency and ischemic brain damage using photochemically induced MCA thrombosis model in guinea pigs. FK419, ozagrel, or argatroban was administered 5 min after the termination of photoirradiation. FK419 dose-dependently improved MCA patency by decreasing the total occlusion time, time to continuous reperfusion, and the number of cyclic flow reductions, at doses that inhibited ADP-induced platelet aggregation ex vivo. In contrast, ozagrel only improved total occlusion time, and argatroban showed no improvement in MCA patency. FK419 also reduced ischemic brain damage in a dose-dependent fashion, whereas ozagrel and argatroban did not. Finally, FK419 ameliorated neurological deficits, whereas ozagrel and argatroban did not. These results indicate that FK419, a GPIIb/IIIa antagonist, ameliorates ischemic brain damage by improving MCA patency after occlusion and that FK419 is a promising candidate for the treatment of acute ischemic stroke.


Subject(s)
Antithrombins/therapeutic use , Infarction, Middle Cerebral Artery/drug therapy , Platelet Aggregation Inhibitors/therapeutic use , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Animals , Arginine/analogs & derivatives , Blood Coagulation/drug effects , Blood Platelets/drug effects , Blood Platelets/physiology , Brain Ischemia/complications , Brain Ischemia/drug therapy , Disease Models, Animal , Guinea Pigs , Male , Methacrylates/therapeutic use , Pipecolic Acids/therapeutic use , Reperfusion Injury/prevention & control , Sulfonamides , Thromboxane B2/metabolism
17.
Eur J Pharmacol ; 473(2-3): 163-9, 2003 Jul 25.
Article in English | MEDLINE | ID: mdl-12892834

ABSTRACT

The antiplatelet and antithrombotic effects of FR171113, 3-(4-chlorophenyl)-2-(2,4-dichlorobenzoylimino)-5-(methoxycarbonyl methylene)-1,3-thiazolidin-4-one, a non-peptide protease-activated receptor 1 (PAR1) antagonist, were evaluated in guinea pigs. FR171113 inhibited Ser-Phe-Leu-Leu-Arg-Asn-NH2 (a synthetic PAR1 agonist peptide)-induced and thrombin-induced aggregation of guinea pig platelets in a concentration-dependent manner in vitro (IC50=1.5 and 0.35 microM, respectively). Subcutaneous administration of FR171113 (0.1-3.2 mg/kg) produced a dose-dependent inhibition of platelet aggregation ex vivo. The ED50 value of FR171113 for platelet aggregation was 0.49 mg/kg s.c. However, FR171113 did not have an inhibitory effect on ADP- or collagen-induced platelet aggregation in vitro and ex vivo. One hour after FR171113 treatment at 1.0 mg/kg s.c., significant inhibition of arterial thrombosis without a prolongation of thrombin time or coagulation time was seen in the FeCl3-induced carotid artery thrombosis model in guinea pigs. Furthermore, FR171113 did not prolong bleeding time even at 32 mg/kg s.c., which is a much higher dose than that required in the thrombosis model. These observations indicate that FR171113 has desirable antiplatelet effects both in vitro and in vivo and that its in vivo antithrombotic activity is efficacious without causing a prolongation of bleeding time.


Subject(s)
Benzamides/pharmacology , Carotid Artery Thrombosis/drug therapy , Platelet Aggregation Inhibitors/pharmacology , Receptor, PAR-1/antagonists & inhibitors , Thiazoles/pharmacology , Animals , Arginine/analogs & derivatives , Benzamides/administration & dosage , Bleeding Time , Blood Coagulation/drug effects , Carotid Artery Thrombosis/blood , Carotid Artery Thrombosis/chemically induced , Chlorides , Dose-Response Relationship, Drug , Ferric Compounds , Guinea Pigs , Heparin/pharmacology , Injections, Subcutaneous , Male , Pipecolic Acids/pharmacology , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/administration & dosage , Receptors, Thrombin/antagonists & inhibitors , Sulfonamides , Thiazoles/administration & dosage , Thiazolidines
SELECTION OF CITATIONS
SEARCH DETAIL
...