Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Cereb Blood Flow Metab ; 25(4): 451-9, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15674237

ABSTRACT

Repinotan is a highly potent 5-HT1A receptor agonist with strong neuroprotective efficacy in animal models of middle cerebral artery occlusion and traumatic brain injury. In this study, we characterized the time window for neuroprotective effects of repinotan in animal models. In the permanent middle cerebral artery occlusion model, repinotan showed neuroprotective efficacy when administered as a triple bolus injection (0.3-100 microg/kg) or an intravenous infusion (0.3-100 microg/kg per hour). A 73% reduction in infarct volume was observed with a 3 microg/kg intravenous bolus, and a 65% reduction was observed with a 3 and 10 microg/kg per hour intravenous infusion. When delayed until 5 hours after occlusion, repinotan (10 microg/kg per hour) reduced infarct volume by 43%. In the transient middle cerebral artery occlusion model, repinotan (10 microg/kg per hour) administered immediately after occlusion reduced infarct volume by 97%, and a delay to 5 hours reduced infarct volume by 81%. In the acute subdural hematoma model, repinotan (3 and 10 microg/kg per hour) reduced infarct volume by 65%. In this model, repinotan (3 microg/kg per hour) administered 5 hours after occlusion reduced infarct volume by 54%. The favorable neuroprotective efficacy, broad dose-response curve, and prolonged therapeutic window observed in all models strongly suggest that repinotan is a promising candidate for treating acute ischemic stroke in humans.


Subject(s)
Benzopyrans/pharmacology , Brain Injuries/drug therapy , Neuroprotective Agents , Receptor, Serotonin, 5-HT1A/drug effects , Serotonin Receptor Agonists/pharmacology , Stroke/drug therapy , Thiazoles/pharmacology , Animals , Biomarkers , Brain/pathology , Brain Injuries/pathology , Hematoma, Subdural, Acute/pathology , Infarction, Middle Cerebral Artery/pathology , Male , Rats , Rats, Long-Evans , Stroke/pathology , Succinate Dehydrogenase/metabolism
2.
Bioorg Med Chem Lett ; 15(2): 401-4, 2005 Jan 17.
Article in English | MEDLINE | ID: mdl-15603962

ABSTRACT

Novel cyclohexadienes have been identified as potent and specific IK(Ca)-channel blockers. In this communication we describe their synthesis as well as their chemical and biological properties. A selected derivative is being enriched in rat brain and reduces the infarct volume, intracranial pressure as well as the water content in a rat subdural hematoma model of traumatic brain injury after iv administration.


Subject(s)
Body Water/drug effects , Cyclohexanes/pharmacology , Intracranial Pressure/drug effects , Potassium Channel Blockers/pharmacology , Potassium Channels/metabolism , Animals , Body Water/metabolism , Brain Infarction/drug therapy , Brain Injuries/drug therapy , Cyclohexanes/chemical synthesis , Cyclohexenes , Disease Models, Animal , Hematoma, Subdural/drug therapy , Intermediate-Conductance Calcium-Activated Potassium Channels , Potassium Channel Blockers/chemical synthesis , Rats , Structure-Activity Relationship
3.
Eur J Neurosci ; 20(7): 1761-8, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15379997

ABSTRACT

Early deterioration and death after brain injury is often the result of oedema in the injured and peri-lesional tissue. So far, no pharmacotherapy is available that exhibits significant brain oedema-reducing efficacy in patients. We selected two low molecular weight compounds from different chemical classes, a triazole (1-[(2-chlorophenyl)diphenylmethyl]-1,2,3-triazole) and a cyclohexadiene (methyl 4-[4-chloro-3-(trifluoromethyl)phenyl]-6-methyl-3-oxo-1,4,7-tetrahydroisobenzofuran-5-carboxylate) to characterize their pharmacological properties on KCNN4 channels (intermediate/small conductance calcium-activated potassium channel, subfamily N, member 4) in vitro as well as in vivo. In vitro we replaced potassium by rubidium (Rb+) and determined Rb+ fluxes evoked by 10 micro m of the calcium ionophore A23187 on C6BU1 rat glioma cells. Compared with known KCNN4 blockers, such as clotrimazole (IC50=360 +/- 12 nm) and charybdotoxin (IC50=3.3 +/- 1.9 nm), the triazole and cyclohexadiene were considerably more potent than clotrimazole and displayed similar potencies (IC50=12.1 +/- 8.8 and 13.3 +/- 4.7 nm, respectively). In the rat acute subdural haematoma model, both the triazole and cyclohexadiene displayed reduction of brain water content (-26% at 0.3 mg/kg and -24% at 0.01 mg/kg) and reduction of the intracranial pressure (-46% at 0.1 mg/kg and -60% at 0.003 mg/kg) after 24 h when administered as a 4-h infusion immediately after brain injury. When infarct volumes were determined after 7 days, the triazole as well as the cyclohexadiene displayed strong neuroprotective efficacy (-52% infarct volume reduction at 1.2 mg/kg and -43% at 0.04 mg/kg, respectively). It is concluded that blockade of KCNN4 channels is a new pharmacological approach to attenuate acute brain damage caused by traumatic brain injury.


Subject(s)
Brain Edema/therapy , Brain Injuries/therapy , Clotrimazole/therapeutic use , Hematoma, Subdural/therapy , Potassium Channel Blockers/therapeutic use , Animals , Brain Chemistry , Calcimycin/pharmacology , Cell Line, Tumor , Cerebral Infarction/pathology , Charybdotoxin/therapeutic use , DNA Primers , Erythrocytes/physiology , Glioma/genetics , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels , Potassium Channels, Calcium-Activated , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Rubidium/blood , Water/analysis
4.
CNS Drug Rev ; 9(4): 343-58, 2003.
Article in English | MEDLINE | ID: mdl-14647528

ABSTRACT

Traumatic brain injury (TBI) is the most common cause of mortality and morbidity in adults under 40 years of age in industrialized countries. Worldwide the incidence is increasing, about 9.5 million people are hospitalized per year due to TBI, and the death rate is estimated to be more than one million people per year. Recently BAY 38-7271 has been characterized as a structurally novel, selective and highly potent cannabinoid CB1/CB2 receptor agonist in vitro and in vivo with pronounced neuroprotective efficacy in a rat traumatic brain injury model, showing a therapeutic window of at least 5 h. Furthermore, neuroprotective efficacy was also found in models of transient and permanent occlusion of the middle cerebral artery and brain edema models as well. In this article we review the in vitro and in vivo pharmacology of BAY 38-7271, the results from acute and subacute toxicity studies, pharmacokinetics and drug metabolism in animals and healthy male volunteers. In phase I studies BAY 38-7271 was safe and well tolerated when administered by i.v. infusion for either 1 or 24 h. As the doses of BAY 38-7271 in animals needed for maximal neuroprotective efficacy were significantly lower than those inducing typical cannabinoid-like side effects, it is to be expected that the compound will offer a novel therapeutic approach with a favorable therapeutic window for the treatment of TBI or cerebral ischemia.


Subject(s)
Brain Injuries/drug therapy , Cannabinoid Receptor Agonists , Indans/therapeutic use , Neuroprotective Agents/therapeutic use , Sulfonic Acids/therapeutic use , Animals , Area Under Curve , Half-Life , Humans , Indans/metabolism , Indans/pharmacokinetics , Metabolic Clearance Rate , Sulfonic Acids/metabolism , Sulfonic Acids/pharmacokinetics
5.
Brain Res ; 989(1): 99-111, 2003 Oct 31.
Article in English | MEDLINE | ID: mdl-14519516

ABSTRACT

BAY 38-7271 is a new high-affinity cannabinoid receptor agonist with strong neuroprotective efficacy in a rat model of traumatic brain injury (acute subdural hematoma, SDH). In the present study we investigated CB1 receptor signal transduction by [35S]GTPgammaS binding in situ and in vitro to assess changes in receptor functionality after SDH. Further, we continued to investigate the neuroprotective properties of BAY 38-7271 in the rat SDH and transient middle cerebral artery occlusion (tMCA-O) model as well as the efficacy with respect to SDH-induced brain edema. [35S]GTPgammaS binding revealed minor attenuation of CB1 receptor functionality on brain membranes from injured hemispheres when compared to non-injured hemispheres or controls. In the rat SDH model, BAY 38-7271 displayed strong neuroprotective efficacy when administered immediately after SDH either as a 1 h (65% infarct volume reduction at 0.1 microg/kg) or short-duration (15 min) infusion (53% at 10 microg/kg). When administered as a 4 h infusion with a 5 h delay after injury, significant neuroprotection was observed (49% at 1.0 microg/kg/h). This was also observed when BAY 38-7271 was administered as a 5 h delayed 15 min short-duration infusion (64% at 3 microg/kg). In addition, the neuroprotective potential of BAY 38-7271 was demonstrated in the rat tMCA-O model, displaying pronounced neuroprotective efficacy in the cerebral cortex (91% at 1 ng/kg/h) and striatum (53% at 10 ng/kg/h). BAY 38-7271 also reduced intracranial pressure (28% at 250 ng/kg/h) and brain water content (20% at 250 ng/kg/h) when determined 24 h post-SDH. Based on these data it is concluded that the neuroprotective efficacy of BAY 38-7271 is mediated by multiple mechanisms triggered by cannabinoid receptors.


Subject(s)
Brain Edema/drug therapy , Hematoma, Subdural/drug therapy , Indans/therapeutic use , Neuroprotective Agents/therapeutic use , Receptor, Cannabinoid, CB1/agonists , Sulfonic Acids/therapeutic use , Animals , Brain Edema/metabolism , Dose-Response Relationship, Drug , Hematoma, Subdural/metabolism , Indans/pharmacology , Male , Neuroprotective Agents/pharmacology , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/metabolism , Sulfonic Acids/pharmacology
7.
J Pharmacol Exp Ther ; 302(1): 359-68, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12065738

ABSTRACT

(-)-(R)-3-(2-Hydroxymethylindanyl-4-oxy)phenyl-4,4,4-trifluoro-1-sulfonate (BAY 38-7271) is a new high-affinity cannabinoid receptor subtype 1 (CB1 receptor) ligand (K(i) = 0.46-1.85 nM; rat brain, human cortex, or recombinant human CB1 receptor), structurally unrelated to any cannabinoid receptor ligand known so far. BAY 38-7271 was characterized as a CB1 receptor agonist in 5-[gamma(35)S]-thiophosphate triethylammonium salt binding assays using rat or human CB1 receptors. In the rat hypothermia assay, BAY 38-7271 induced a dose-dependent reduction in body temperature (minimal effective dose = 6 microg/kg, i.v.); whereas in rats trained to discriminate the CB1/CB2 receptor agonist (-)-cis-3-[2-hydroxy-4(1,1-dimethyl-heptyl)phenyl]-trans-4-(3-hydroxypropyl) cyclohexanol (CP 55,940; 0.03 mg/kg, i.p.) from vehicle, BAY 38-7271 induced complete generalization (3 microg/kg, i.v.). In both in vivo models, a specific CB1 receptor-mediated mechanism was confirmed by demonstrating that the effects of CP 55,940 and BAY 38-7271 were blocked by pretreatment with the selective CB1 receptor antagonist N-(piperidin-1-yl)-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamidehydrochloride. In the rat traumatic brain injury model, BAY 38-7271 demonstrated highly potent and efficient neuroprotective properties when administered as a 4-h infusion immediately after induction of subdural hematoma (70% infarct volume reduction at 100 ng/kg/h). Even when applied with a 3-h delay, a significant neuroprotective efficacy could be observed (59% infarct volume reduction at 300 ng/kg/h). The neuroprotective potential of BAY 38-7271 was confirmed in a rat model of focal cerebral ischemia induced by permanent occlusion of the middle cerebral artery. It is concluded that the CB1/CB2 receptor agonist BAY 38-7271 shows pronounced neuroprotective properties that do not result from drug-induced hypothermia and that occur in a dose range devoid of typical cannabinoid-like side effects.


Subject(s)
Neuroprotective Agents/pharmacology , Receptor, Cannabinoid, CB2 , Receptors, Drug/agonists , Animals , Binding, Competitive/drug effects , Body Temperature/drug effects , Brain Injuries/drug therapy , Brain Injuries/pathology , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Cyclohexanols/pharmacology , Discrimination, Psychological/drug effects , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Hematoma, Subdural/drug therapy , Indans/pharmacology , Indans/therapeutic use , Male , Middle Cerebral Artery/physiology , Neuroprotective Agents/therapeutic use , Rats , Rats, Long-Evans , Rats, Wistar , Receptors, Cannabinoid , Signal Transduction/drug effects , Succinate Dehydrogenase/metabolism , Sulfonic Acids/pharmacology , Sulfonic Acids/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...