Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
J Pharmacol Toxicol Methods ; 104: 106896, 2020.
Article in English | MEDLINE | ID: mdl-32622755

ABSTRACT

INTRODUCTION: Clinical observations are routinely used to address drug-induced behavioural changes in dogs. To deal with the limitations of this methodology, we evaluated Actiwatch-Mini® as a tool to monitor continuously locomotor activity in dogs and to provide objective parameters that could be linked to behavioural changes after compound administration. METHODS: Eight Beagles were equipped with Actiwatch-Mini®. As a reference drug, a PDE2-inhibitor was administered daily for six days at doses known to cause minor or severe behavioural changes. RESULTS: While traditional observation showed no behavioural changes - except sedation - dogs receiving 0.5 mg/kg/day, showed significant increases in % immobile time (+15.8%) and mean length of immobile phases (+1.2 min) but no change in number of immobile phases (+2.2). At 1 mg/kg/day, light to severe changes in behaviour were present. Actiwatch-Mini® recorded an increase in mean length of immobile phases (+1.9 min) and a decrease in mean number of immobile phases (-7.4) in the first four hours after each dosing while total % immobile time was not significantly increased (+4.9%). DISCUSSION: Actiwatch-Mini® was able to detect changes in immobility parameters in dogs dosed with a PDE2-inhibitor while no or only minor behavioural changes were observed. The system can be used for continuously monitoring the activity of dogs, to provide objective scores for evaluation of activity. It provides an inexpensive and low labour-intensive method for detecting changes in activity and possible early indications of drug-induced behavioural changes.


Subject(s)
Behavior, Animal/drug effects , Monitoring, Physiologic/methods , Motor Activity/drug effects , Phosphodiesterase Inhibitors/toxicity , Animals , Cyclic Nucleotide Phosphodiesterases, Type 2/antagonists & inhibitors , Dogs , Female , Male , Monitoring, Physiologic/instrumentation , Phosphodiesterase Inhibitors/administration & dosage , Time Factors
2.
Circ Heart Fail ; 13(5): e006609, 2020 05.
Article in English | MEDLINE | ID: mdl-32418479

ABSTRACT

BACKGROUND: Low myocardial cGMP-PKG (cyclic guanosine monophosphate-protein kinase G) activity has been associated with increased cardiomyocyte diastolic stiffness in heart failure with preserved ejection fraction. Cyclic guanosine monophosphate is mainly hydrolyzed by PDE (phosphodiesterases) 5a and 9a. Importantly, PDE9a expression has been reported to be upregulated in human heart failure with preserved ejection fraction myocardium and chronic administration of a PDE9a inhibitor reverses preestablished cardiac hypertrophy and systolic dysfunction in mice subjected to transverse aortic constriction (TAC). We hypothesized that inhibiting PDE9a activity ameliorates diastolic dysfunction. METHODS: To examine the effect of chronic PDE9a inhibition, 2 diastolic dysfunction mouse models were studied: (1) TAC-deoxycorticosterone acetate and (2) Leprdb/db. PDE9a inhibitor (5 and 8 mg/kg per day) was administered to the mice via subcutaneously implanted osmotic minipumps for 28 days. The effect of acute PDE9a inhibition was investigated in intact cardiomyocytes isolated from TAC-deoxycorticosterone acetate mice. Atrial natriuretic peptide together with PDE9a inhibitor were administered to the isolated intact cardiomyocytes through the cell perfusate. RESULTS: For acute inhibition, no cellular stiffness reduction was found, whereas chronic PDE9a inhibition resulted in reduced left ventricular chamber stiffness in TAC-deoxycorticosterone acetate, but not in Leprdb/db mice. Passive cardiomyocyte stiffness was reduced by chronic PDE9a inhibition, with no differences in myocardial fibrosis or cardiac morphometry. PDE9a inhibition increased the ventricular-arterial coupling ratio, reflecting impaired systolic function. CONCLUSIONS: Chronic PDE9a inhibition lowers left ventricular chamber stiffness in TAC-deoxycorticosterone acetate mice. However, the usefulness of PDE9a inhibition to treat high-diastolic stiffness may be limited as the required PDE9a inhibitor dose also impairs systolic function, observed as a decline in ventricular-arterial coordination, in this model.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Myocytes, Cardiac/drug effects , Phosphodiesterase Inhibitors/pharmacology , Ventricular Dysfunction, Left/drug therapy , Ventricular Function, Left/drug effects , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Animals , Diastole , Disease Models, Animal , Male , Mice, Inbred C57BL , Myocytes, Cardiac/enzymology , Phosphodiesterase Inhibitors/toxicity , Ventricular Dysfunction, Left/enzymology , Ventricular Dysfunction, Left/physiopathology
3.
Thromb Haemost ; 117(9): 1722-1729, 2017 08 30.
Article in English | MEDLINE | ID: mdl-28837204

ABSTRACT

Antithrombotic medications target coagulation factors. Their use is associated with an increased bleeding risk. Safer drugs are needed. The heat shock protein 70 (Hsp70) exhibits antithrombotic properties that do not influence bleeding. By using murine models, we aimed to test the hypothesis that overexpressing Hsp70 with CM-695, a first in class dual inhibitor of HDAC6 and phosphodiesterase 9, protects against thrombosis while leaves bleeding tendency unaltered. CM-695 was used to induce Hsp70 overexpression. Hsp70 overexpressing mice were submitted to three thrombosis-triggering procedures. The ferric chloride carotid artery model was used to compare the antithrombotic role of CM-695 and rivaroxaban, a direct oral anticoagulant. The mouse tail transection model was used to compare the bleeding tendency upon CM-695 or rivaroxaban administration. Intraperitoneal (i. p.) 20 mg/kg CM-695 increased Hsp70 expression markedly in the murine aortic tissue. This treatment delayed thrombosis in the collagen/epinephrine [p=0.04 (Log-Rank test), n=10], Rose Bengal/laser [median vessel occlusion time (OT): 58.6 vs 39.0 minutes (min) in the control group (CG), p=0.008, n≥10] and ferric chloride (OT: 14.7 vs 9.2 min in the CG, p=0.032, n≥10) models. I.p. 80 mg/kg CM-695 (n≥9) and intravenous 3 mg/kg rivaroxaban (n≥8) significantly delayed thrombosis. CM-695 did not induce bleeding [median bleeding time (BT): 8.5 vs 7.5 min in the CG, n≥10]. However, BT was dramatically increased by rivaroxaban (30.0 vs 13.7 min in the CG, p=0.001, n=10). In conclusion, CM-695 is a new antithrombotic small molecule devoid of bleeding risk that may be envisioned as a useful clinical tool.


Subject(s)
Blood Coagulation/drug effects , Fibrinolytic Agents/pharmacology , HSP70 Heat-Shock Proteins/metabolism , Histone Deacetylase Inhibitors/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Rivaroxaban/pharmacology , Thromboembolism/prevention & control , Thrombosis/prevention & control , 3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Animals , Bleeding Time , Female , Fibrinolytic Agents/toxicity , HSP70 Heat-Shock Proteins/deficiency , HSP70 Heat-Shock Proteins/genetics , Hemorrhage/chemically induced , Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase 6/metabolism , Histone Deacetylase Inhibitors/toxicity , Mice , Mice, Knockout , Nervous System Autoimmune Disease, Experimental , Phosphodiesterase Inhibitors/toxicity , Risk Assessment , Rivaroxaban/toxicity , Thromboembolism/blood , Thromboembolism/genetics , Thrombosis/blood , Thrombosis/genetics , Time Factors , Up-Regulation
4.
Toxicol Sci ; 148(2): 544-54, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26377646

ABSTRACT

Atrazine (ATR) is a broad-spectrum triazine herbicide that disrupts steroidogenesis resulting in reproductive and developmental toxicity at high doses. Mouse BLTK1 Leydig cells were used as a steroidogenic model to investigate the effects of ATR on testosterone (T) biosynthesis. Induction of steroidogenesis by 3 ng/ml recombinant human chorionic gonadotropin (rhCG) induced intracellular 3',5' cyclic adenosine monophosphate (cAMP) approximately 20-fold and T approximately 3-fold at 4 h. Co-treatment with 300 µM ATR super-induced cAMP levels 100-fold yet antagonized rhCG-mediated induction of T approximately 20% at 4 h. ATR inhibited cAMP-specific phosphodiesterase (cPDE) with an IC50 of ≥98 µM, suggesting cPDE inhibition contributes to the super-induction of cAMP. However, concentrations of up to 3 mM db-cAMP did not antagonize rhCG induction of T levels, suggesting cAMP super-induction alone does not decrease T biosynthesis. Western analysis of cAMP-activated protein kinase A (PKA) target proteins identified ATR-mediated concentration-dependent alterations in phosphorylation including phospho-CREB. These results suggest the cPDE inhibition by ATR and super-induction of cAMP are independent of effects on T levels, and that altered phosphorylation of key steroidogenic regulatory proteins may underlie ATR-mediated disruption of steroidogenesis.


Subject(s)
Atrazine/toxicity , Endocrine Disruptors/toxicity , Herbicides/toxicity , Leydig Cells/drug effects , Testosterone/biosynthesis , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Animals , Cell Line, Tumor , Chorionic Gonadotropin/pharmacology , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Dose-Response Relationship, Drug , Leydig Cells/metabolism , Male , Mice , Phosphodiesterase Inhibitors/toxicity , Phosphorylation , Second Messenger Systems/drug effects , Time Factors
5.
Chem Res Toxicol ; 28(10): 1914-25, 2015 Oct 19.
Article in English | MEDLINE | ID: mdl-26313431

ABSTRACT

During drug discovery and development, the early identification of adverse effects is expected to reduce costly late-stage failures of candidate drugs. As risk/safety assessment takes place rather late during the development process and due to the limited ability of animal models to predict the human situation, modern unbiased high-dimensional biology readouts are sought, such as molecular signatures predictive for in vivo response using high-throughput cell-based assays. In this theoretical proof of concept, we provide findings of an in-depth exploration of a single chemical core structure. Via transcriptional profiling, we identified a subset of close analogues that commonly downregulate multiple tubulin genes across cellular contexts, suggesting possible spindle poison effects. Confirmation via a qualified toxicity assay (in vitro micronucleus test) and the identification of a characteristic aggregate-formation phenotype via exploratory high-content imaging validated the initial findings. SAR analysis triggered the synthesis of a new set of compounds and allowed us to extend the series showing the genotoxic effect. We demonstrate the potential to flag toxicity issues by utilizing data from exploratory experiments that are typically generated for target evaluation purposes during early drug discovery. We share our thoughts on how this approach may be incorporated into drug development strategies.


Subject(s)
Drug Discovery , Gene Expression Profiling , Animals , Cell Line, Tumor , HEK293 Cells , Humans , Microscopy, Confocal , Phosphodiesterase Inhibitors/chemistry , Phosphodiesterase Inhibitors/metabolism , Phosphodiesterase Inhibitors/toxicity , Phosphoric Diester Hydrolases/chemistry , Phosphoric Diester Hydrolases/metabolism , Pyrrolidines/chemistry , Pyrrolidines/metabolism , Pyrrolidines/toxicity , Structure-Activity Relationship , Transcriptome/drug effects , Tubulin/metabolism
6.
Eur J Pharmacol ; 761: 79-85, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-25941078

ABSTRACT

Here, we present a preliminary pharmacological characterisation of Lu AF33241, a novel, brain penetrant phosphodiesterase inhibitor of (PDE) 2A and 10A tool compound, in in vitro/in vivo assays indicative of PDE2A and/or PDE10A inhibition, and in vivo models/assays relevant to cognitive processing and antipsychotic-like activity. An assay was also included to investigate potential effects on motor activity. The in vitro selectivity of Lu AF33241 was determined against a panel of PDE enzymes. Lu AF33241 potently inhibited both full-length recombinant hPDE2A (Ki=4.2nM) and hPDE10A (Ki=42nM). The compound moderately inhibited both hPDE1C (Ki=1200nM), hPDE7B (Ki=890nM), and hPDE11A (Ki=1800nM). Lu AF33241 displayed a Ki above 5000nM against all other tested members of the PDE family. Albeit within a narrow dose range, Lu AF33241 attenuated sub-chronic phencyclidine-induced deficits in novel object recognition (3 and 10mg/kg), displayed antipsychotic-like activity in the conditioned avoidance response paradigm (10mg/kg), and did not induce catalepsy within a dose-range of 2-6mg/kg. Further catalepsy studies are needed to investigate a predictive safety window. Lu AF33241 represents a novel PDE2A/PDE10A inhibitor tool compound that may serve to further the understanding of the roles played by these enzymes in various CNS disorders.


Subject(s)
Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Blood-Brain Barrier/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/antagonists & inhibitors , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/metabolism , Quinoxalines/pharmacology , Triazoles/pharmacology , Animals , Antipsychotic Agents/metabolism , Antipsychotic Agents/toxicity , Avoidance Learning/drug effects , Capillary Permeability , Catalepsy/chemically induced , Cognition/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Exploratory Behavior/drug effects , Humans , Male , Motor Activity/drug effects , Phosphodiesterase Inhibitors/metabolism , Phosphodiesterase Inhibitors/toxicity , Quinoxalines/metabolism , Rats, Sprague-Dawley , Rats, Wistar , Recognition, Psychology/drug effects , Triazoles/metabolism
7.
Behav Brain Res ; 278: 129-36, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25281278

ABSTRACT

In this work, we report the effect of post-training intraperitoneal administration of zaprinast on rat memory retention in the Morris water maze task that revealed a significant memory impairment at the intermediate dose of 10mg/kg. Zaprinast is capable of inhibiting both striatal and hippocampal PDE activity but to a different extent which is probably due to the different PDE isoforms expressed in these areas. To assess the possible involvement of cyclic nucleotides in rat memory impairment, we compared the effects obtained 30 min after the zaprinast injection with respect to 24h after injection by measuring both cyclic nucleotide levels and PDE activity. As expected, 30 min after the zaprinast administration, we observed an increase of cyclic nucleotides, which returned to a basal level within 24h, with the exception of the hippocampal cGMP which was significantly decreased at the dose of 10mg/kg of zaprinast. This increase in the hippocampal region is the result of a cGMP-specific PDE5 induction, confirmed by sildenafil inhibition, in agreement with literature data that demonstrate transcriptional regulation of PDE5 by cAMP/cGMP intracellular levels. Our results highlight the possible rebound effect of PDE inhibitors.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Hippocampus/drug effects , Memory Disorders/chemically induced , Memory Disorders/pathology , Phosphodiesterase Inhibitors/toxicity , Purinones/toxicity , Analysis of Variance , Animals , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Dose-Response Relationship, Drug , Escape Reaction/drug effects , Hippocampus/metabolism , In Vitro Techniques , Male , Maze Learning/drug effects , Motor Activity/drug effects , Rats , Rats, Wistar , Reaction Time/drug effects , Time Factors
8.
J Neuroinflammation ; 11: 172, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-25301432

ABSTRACT

BACKGROUND: cGMP-degrading phosphodiesterase 6 (PDE6) mutations cause around 4 to 5% of retinitis pigmentosa (RP), a rare form of retinal dystrophy. Growing evidence suggests that inflammation is involved in the progression of RP. The aims of this study were to corroborate the presence of high TNFα concentration in the eyes of RP patients and to evaluate whether the blockade of TNFα with Infliximab, a monoclonal anti-TNFα antibody, prevented retinal degeneration induced by PDE6 inhibition in cultures of porcine retina. METHODS: Aqueous humor from 30 patients with RP and 13 healthy controls were used to quantify the inflammatory mediators IL-6, TNFα, IL-1ß, IL-10 by a multiplex enzyme-linked immunosorbent assay (ELISA) system. Retinal explants from pig were exposed to Zaprinast, a PDE6 inhibitor, for 24 hours in the absence or the presence of Infliximab. Cell death was evaluated by TUNEL assay. The number and distribution of caspase-3 positive cells, indirect poly(ADP)ribose polymerase (PARP) activation and glial fibrillary acidic protein (GFAP) content were visualized by immunolabeling. Antioxidant total capacity, nitrites and thiobarbituric acid reactive substances (TBARS) formation were determined to evaluate antioxidant-oxidant status. RESULTS: IL-6 and TNFα concentrations were higher in the aqueous humor of RP patients than in controls. Infliximab prevented retinal degeneration, as judging by the reduced presence of TUNEL-positive cells, the reduction of caspase-3 activation and also reduction of glial activation, in an ex vivo model of porcine retina. Additionally, Infliximab partially reduced oxidative stress in retinal explants exposed to Zaprinast. CONCLUSIONS: Inflammatory mediators IL-6 and TNFα were elevated in the aqueous humor of RP patients corroborating previous studies suggesting sustained chronic inflammation. Our study suggests that TNFα is playing an important role in cell death in an ex vivo model of retinal degeneration by activating different cell pathways at different cell layers of the retina that should be further studied.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antibodies, Monoclonal/pharmacology , Retinitis Pigmentosa/immunology , Animals , Aqueous Humor/immunology , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Female , Humans , In Situ Nick-End Labeling , Infliximab , Interleukin-6/immunology , Male , Middle Aged , Phosphodiesterase Inhibitors/toxicity , Purinones/toxicity , Retina/drug effects , Swine , Tumor Necrosis Factor-alpha/immunology
9.
Pharmacol Biochem Behav ; 118: 36-45, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24211858

ABSTRACT

Akathisia is a subset of the larger antipsychotic side effect profile known as extrapyramidal syndrome (EPS). It is associated with antipsychotic treatment and is characterized as a feeling of inner restlessness that results in a compulsion to move. There are currently no primate models available to assess drug-induced akathisia; the present research was designed to address this shortcoming. We developed a novel rating scale based on both the Barnes Akathisia Rating Scale (BARS) and the Hillside Akathisia Scale (HAS) to measure the objective, observable incidence of antipsychotic-induced akathisia-like behavior in Cebus apella non-human primates (NHPs). To induce akathisia, we administered the atypical antipsychotic aripiprazole (1 mg/kg) or the selective phosphodiesterase 10A (PDE10A) inhibitor MP-10 (1-3 mg/kg). Treatment with both compounds produced significantly greater akathisia scores on the rating scale than vehicle treatment. Characteristic behaviors observed included vocalizations, stereotypies, teeth grinding, restless limb movements, and hyperlocomotion. Adenosine A2A receptor antagonists have previously been shown to be effective in blocking antipsychotic-induced EPS in primates. The selective A2A receptor antagonist, SCH 412348 (10-30 mg/kg), effectively reduced or reversed akathisia-like behavior induced by both aripiprazole and MP-10. This work represents the first NHP measurement scale of akathisia and demonstrates that NHPs are responsive to akathisia-inducing agents. As such, it provides a useful tool for the preclinical assessment of putative antipsychotics. In addition, these results provide further evidence of the utility of A2A receptor antagonists for the treatment of antipsychotic-induced movement disorders.


Subject(s)
Adenosine A2 Receptor Antagonists/pharmacology , Akathisia, Drug-Induced/drug therapy , Akathisia, Drug-Induced/physiopathology , Akathisia, Drug-Induced/psychology , Animals , Antipsychotic Agents/administration & dosage , Antipsychotic Agents/antagonists & inhibitors , Antipsychotic Agents/toxicity , Aripiprazole , Behavior, Animal/drug effects , Cebus , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Haloperidol/administration & dosage , Haloperidol/antagonists & inhibitors , Haloperidol/toxicity , Humans , Male , Motor Activity/drug effects , Phosphodiesterase Inhibitors/administration & dosage , Phosphodiesterase Inhibitors/toxicity , Piperazines/administration & dosage , Piperazines/antagonists & inhibitors , Piperazines/toxicity , Pyrazoles/administration & dosage , Pyrazoles/antagonists & inhibitors , Pyrazoles/toxicity , Pyrimidines/pharmacology , Quinolines/administration & dosage , Quinolines/antagonists & inhibitors , Quinolines/toxicity , Quinolones/administration & dosage , Quinolones/antagonists & inhibitors , Quinolones/toxicity , Triazoles/pharmacology
10.
Adv Exp Med Biol ; 756: 341-7, 2013.
Article in English | MEDLINE | ID: mdl-22836652

ABSTRACT

As inflammation plays an important role in the pathogenesis of neonatal meconium aspiration syndrome (MAS), anti-inflammatory agents including inhibitors of phosphodiesterases (PDE) are increasingly used in the treatment. To evaluate side effects of PDE inhibitors, this study analyzed changes in blood pressure, heart rate (HR) and heart rate variability (HRV) during and after intravenous aminophylline in the animal model of MAS. Oxygen-ventilated rabbits were given meconium intratracheally (25 mg/ml, 4 ml/kg) or saline. Thirty minutes later, the animals were treated by intravenous aminophylline (Syntophyllin, 2 mg/kg) or saline (sham-treated controls). A second dose of the treatment was given 2 h later. During (5 min) and immediately after (5 min) the treatment, and during 5 h after the treatment, mean blood pressure in the femoral artery (MAP), HR and HRV were evaluated. In meconium-instilled animals, increases in MABP, HR, and HRV were observed already 5 min after aminophylline administration, while in saline-instilled animals aminophylline increased HR and caused inconsistant changes in HRV parameters compared to sham-treated animals. Within 5 h after the treatment administration, MAP, HR, and HRV parameters gradually returned to the initial values. Concluding, intravenous aminophylline may lead to acute cardiovascular changes. Thus, if aminophylline is used for treatment of MAS, its possible cardiovascular effects should be considered, particularly in patients with cardiovascular instability.


Subject(s)
Aminophylline/toxicity , Bronchodilator Agents/pharmacology , Bronchodilator Agents/toxicity , Cardiovascular System/drug effects , Meconium Aspiration Syndrome/drug therapy , Aminophylline/therapeutic use , Animals , Blood Pressure/drug effects , Bronchodilator Agents/therapeutic use , Disease Models, Animal , Heart Rate/drug effects , Humans , Infant, Newborn , Lung/drug effects , Meconium , Phosphodiesterase Inhibitors/therapeutic use , Phosphodiesterase Inhibitors/toxicity , Rabbits
11.
Br J Pharmacol ; 160(4): 860-77, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20590585

ABSTRACT

BACKGROUND AND PURPOSE: Caffeine exacerbates the hyperthermia associated with an acute exposure to 3,4 methylenedioxymethamphetamine (MDMA, 'Ecstasy') in rats. The present study investigated the mechanisms mediating this interaction. EXPERIMENTAL APPROACH: Adult male Sprague-Dawley rats were treated with caffeine (10 mg x kg(-1); i.p.) and MDMA (15 mg x kg(-1); i.p.) alone and in combination. Core body temperatures were monitored before and after drug administration. KEY RESULTS: Central catecholamine depletion blocked MDMA-induced hyperthermia and its exacerbation by caffeine. Caffeine provoked a hyperthermic response when the catecholamine releaser d-amphetamine (1 mg x kg(-1)) was combined with the 5-HT releaser D-fenfluramine (5 mg x kg(-1)) or the non-selective dopamine receptor agonist apomorphine (1 mg x kg(-1)) was combined with the 5-HT(2) receptor agonist DOI (2 mg x kg(-1)) but not following either agents alone. Pretreatment with the dopamine D(1) receptor antagonist Schering (SCH) 23390 (1 mg x kg(-1)), the 5-HT(2) receptor antagonist ketanserin (5 mg x kg(-1)) or alpha(1)-adreno- receptor antagonist prazosin (0.2 mg x kg(-1)) blocked MDMA-induced hyperthermia and its exacerbation by caffeine. Co-administration of a combination of MDMA with the PDE-4 inhibitor rolipram (0.025 mg x kg(-1)) and the adenosine A(1/2) receptor antagonist 9-chloro-2-(2-furanyl)-[1,2,4]triazolo[1,5-C]quinazolin-5-amine 15943 (10 mg x kg(-1)) or the A(2A) receptor antagonist SCH 58261 (2 mg x kg(-1)) but not the A(1) receptor antagonist DPCPX (10 mg x kg(-1)) exacerbated MDMA-induced hyperthermia. CONCLUSIONS AND IMPLICATIONS: A mechanism comprising 5-HT and catecholamines is proposed to mediate MDMA-induced hyperthermia. A combination of adenosine A(2A) receptor antagonism and PDE inhibition can account for the exacerbation of MDMA-induced hyperthermia by caffeine.


Subject(s)
Adrenergic Uptake Inhibitors/toxicity , Caffeine/toxicity , Fever/chemically induced , N-Methyl-3,4-methylenedioxyamphetamine/toxicity , Phosphodiesterase Inhibitors/toxicity , Adenosine A1 Receptor Antagonists , Adenosine A2 Receptor Antagonists , Adrenergic Uptake Inhibitors/antagonists & inhibitors , Adrenergic Uptake Inhibitors/metabolism , Adrenergic Uptake Inhibitors/pharmacokinetics , Adrenergic alpha-Antagonists/pharmacology , Animals , Body Temperature Regulation/drug effects , Caffeine/antagonists & inhibitors , Catecholamines/antagonists & inhibitors , Catecholamines/physiology , Dopamine Agents/pharmacology , Drug Interactions , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , Male , N-Methyl-3,4-methylenedioxyamphetamine/antagonists & inhibitors , N-Methyl-3,4-methylenedioxyamphetamine/metabolism , N-Methyl-3,4-methylenedioxyamphetamine/pharmacokinetics , Neurons/drug effects , Neurons/metabolism , Phosphodiesterase 4 Inhibitors , Rats , Rats, Sprague-Dawley , Serotonin/physiology , Serotonin Agents/pharmacology , Time Factors
12.
Pharmacol Rep ; 61(5): 930-4, 2009.
Article in English | MEDLINE | ID: mdl-19904018

ABSTRACT

The convulsant action of cocaine and the proconvulsant effects of sildenafil, a drug which is widely used in the treatment of erectile dysfunction, have been documented both in humans and mice. Since it was reported that sildenafil alone, and in conjunction with cocaine, is used recreationally, the present study was performed to examine the influence of sildenafil on cocaine-induced seizures in mice.We showed that administration of sildenafil (5-20 mg/kg, ip) did not affect latency to clonic seizures induced by ip administration of cocaine at a dose of 85 mg/kg, nor did it influence seizure incidence and mortality.We conclude that sildenafil does not significantly increase the risk of seizures when co-administered with cocaine.


Subject(s)
Cocaine/toxicity , Phosphodiesterase Inhibitors/toxicity , Piperazines/toxicity , Seizures/chemically induced , Sulfones/toxicity , Animals , Dose-Response Relationship, Drug , Drug Interactions , Male , Mice , Phosphodiesterase Inhibitors/administration & dosage , Piperazines/administration & dosage , Purines/administration & dosage , Purines/toxicity , Sildenafil Citrate , Sulfones/administration & dosage
13.
J Med Toxicol ; 5(1): 3-7, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19191208

ABSTRACT

OBJECTIVE: Calcium channel blocker (CCB) toxicity, in particular that induced by verapamil and diltiazem, presents clinical challenges with no true antidote. Levosimendan, a calcium sensitizer, improves cardiac contractility in patients with heart failure. We tested the hypothesis that calcium channel sensitization will prolong survival in a rat model of severe verapamil poisoning. METHODS: This was a blinded, randomized, controlled animal study. Wistar rats (mean weight, 371 +/- 50 g) were used. Verapamil (2.5 mg/ml) was infused at a rate of 37.5 mg/kg per hour. Bolus doses of levosimendan (5 microg/mL) were given at 0 min (12 microg/kg) and 5 min (18 microg/kg); saline control was of equal volume. The rats were intubated and maintained under general anesthesia with isoflurane. Electrocardiographic activity and core temperature were monitored during the poisoning and treatment phases. Each rat underwent femoral vein cannulation and was then randomized, in blinded fashion, to receive either levosimendan or an equal volume of saline at 0 and 5 minutes. Death, defined as 1 minute of asystole, was used as the primary endpoint. RESULTS: Rats treated with levosimendan died before the control group (7.37 +/- 0.7 min [n=7] vs. 16.4 +/- 4.2 [n=7] [ p=.053]). All animals experienced bradycardia prior to asystole. DISCUSSION: Although levosimendan has the ability to sensitize and enhance binding of troponin C to Ca2+, this study did not show an improvement in survival time in the setting of verapamil toxicity. This may be attributed to levosimendan's inhibition of phosphodiesterase, which possibly exacerbated the CCB-induced hypotension. CONCLUSION: In this rat model, levosimendan as a solitary antidotal treatment for verapamil toxicity was not beneficial.


Subject(s)
Antidotes/pharmacology , Bradycardia/drug therapy , Heart Arrest/drug therapy , Hydrazones/pharmacology , Hypotension/drug therapy , Phosphodiesterase Inhibitors/pharmacology , Pyridazines/pharmacology , Animals , Antidotes/toxicity , Body Temperature/drug effects , Bradycardia/chemically induced , Bradycardia/physiopathology , Calcium Channel Blockers , Drug Interactions , Electrocardiography , Heart Arrest/chemically induced , Heart Arrest/physiopathology , Heart Rate/drug effects , Hydrazones/toxicity , Hypotension/chemically induced , Hypotension/physiopathology , Male , Models, Animal , Phosphodiesterase Inhibitors/toxicity , Pyridazines/toxicity , Rats , Rats, Wistar , Simendan , Time Factors , Verapamil
14.
Pharmacol Biochem Behav ; 91(4): 537-41, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18835293

ABSTRACT

Rolipram, a phosphodiesterase-4 (PDE4) inhibitor, is of current interest as a cognitive enhancer and as a treatment for inflammatory diseases. Originally developed as an anti-depressant, rolipram's efficacy was limited due to its side effects of nausea and vomiting. The experiments reported here evaluated the potential of rolipram to produce conditioned gaping (a selective measure of nausea in rats) to a flavor in the taste reactivity test (Experiment 1) and to a context (Experiment 2). In Experiment 1, rats were intra-orally infused with 17% sucrose solution prior to being injected with rolipram (Vehicle, 0.03, 0.1 or 0.3 mg/kg). Following 3 conditioning trials, rats conditioned with 0.3 mg/kg rolipram displayed conditioned gaping reactions during the infusion of sucrose. In Experiment 2, rats received 4 conditioning trials in which they were injected with 0.3 mg/kg rolipram and placed into a distinctive chamber. At test, when returned to the chamber rats displayed conditioned gaping. These results demonstrate the ability of the conditioned gaping model to detect the nauseating properties of a rolipram-paired flavor (Experiment 1) and rolipram-paired context (Experiment 2), further validating the potential use of the conditioned gaping model as a pre-clinical screening tool to evaluate the side effect of nausea produced by newly developed drugs.


Subject(s)
Conditioning, Operant/drug effects , Nausea/chemically induced , Nausea/psychology , Phosphodiesterase 4 Inhibitors , Phosphodiesterase Inhibitors/toxicity , Rolipram/toxicity , Animals , Male , Motor Activity/drug effects , Rats , Rats, Sprague-Dawley
15.
Biol Pharm Bull ; 31(10): 1981-4, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18827368

ABSTRACT

Sildenafil is widely administered for the treatment of erectile dysfunction. Recently, sensorineural hearing loss following the ingestion of sildenafil was reported in one male patient. We examined hearing in mice that were administered high doses of sildenafil for up to 105 d. To assess hearing impairment, we evaluated auditory brainstem responses, auditory middle latency responses, and otoacoustic emissions. At high doses, sildenafil increased the hearing threshold shift of auditory brainstem responses. High-dose sildenafil treatment also resulted in delayed latency of both auditory brainstem responses and auditory middle responses. Otoacoustic emissions differed between control and high-dose sildenafil groups with long-term treatment. Collectively, these data demonstrate that high-dose and long-term sildenafil administration can induce hearing impairment in mice.


Subject(s)
Hearing Loss, Sensorineural/chemically induced , Phosphodiesterase Inhibitors/toxicity , Piperazines/toxicity , Sulfones/toxicity , Animals , Cochlea/physiology , Cochlear Nerve/pathology , Dose-Response Relationship, Drug , Evoked Potentials, Auditory, Brain Stem/drug effects , Hearing Loss, Sensorineural/pathology , Male , Mice , Mice, Inbred ICR , Otoacoustic Emissions, Spontaneous/drug effects , Purines/toxicity , Sildenafil Citrate
16.
Toxicol Pathol ; 36(6): 827-39, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18776163

ABSTRACT

Histopathological and immunohistochemical studies were conducted to characterize vascular injuries in rats treated with phosphodiesterase (PDE) IV inhibitors SCH 351591 or SCH 534385. Sprague-Dawley rats were administered PDE IV inhibitors by gavage at a range of doses and times. The two PDE IV inhibitors induced comparable levels of vascular injury, primarily in the mesentery and to a lesser extent in the pancreas, kidney, liver, small intestine, and stomach. Mesenteric vascular changes occurred as early as one hour, progressively developed over twenty-four to forty-eight hours, peaked at seventy-two hours, and gradually subsided from seven to nine days. The typical morphology of the vascular toxicity consisted of hemorrhage and necrosis of arterioles and arteries, microvascular injury, fibrin deposition, and perivascular inflammation of a variety of blood vessels. The incidence and severity of mesenteric vascular injury increased in a time- and dose-dependent manner in SCH 351591- or SCH 534385-treated rats. Mesenteric vascular injury was frequently associated with activation of mast cells (MC), endothelial cells (EC), and macrophages (MØ). Immunohistochemical studies showed increases in CD63 immunoreactivity of mesenteric MC and in nitrotyrosine immunoreactivity of mesenteric EC and MØ. The present study also provides a morphological and cellular basis for evaluating candidate biomarkers of drug-induced vascular injury.


Subject(s)
Blood Vessels/drug effects , Cyclic N-Oxides/toxicity , Phosphodiesterase 4 Inhibitors , Phosphodiesterase Inhibitors/toxicity , Quinolines/toxicity , Vascular Diseases/chemically induced , Vascular Diseases/pathology , Animals , Immunohistochemistry , Intestine, Small/blood supply , Intestine, Small/pathology , Kidney/blood supply , Kidney/pathology , Mesenteric Arteries/pathology , Pancreas/blood supply , Pancreas/pathology , Rats , Rats, Sprague-Dawley , Statistics, Nonparametric , Stomach/blood supply , Stomach/pathology
17.
Toxicol Pathol ; 36(6): 840-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18776166

ABSTRACT

Drug-associated vascular injury can be caused by phosphodiesterase (PDE) IV inhibitors and drugs from several other classes. The pathogenesis is poorly understood, but it appears to include vascular and innate immunological components. This research was undertaken to identify changes in peripheral blood associated with vascular injury caused by PDE IV inhibitors. We evaluated twelve proteins, serum nitrite, and leukocyte populations in peripheral blood of rats treated with experimental PDE IV inhibitors. We found that these compounds produced histological microvascular injury in a dose- and time-dependent manner. Measurement of these serum proteins showed changes in eight of the twelve examined. Changes were seen in the levels of: tissue inhibitor of metalloproteinase-1, alpha1-acid glycoprotein, GRO/CINC-1, vascular endothelial growth factor, C-reactive protein, haptoglobin, thrombomodulin, and interleukin-6. No changes were seen in levels of tumor necrosis factor-alpha, hepatocyte growth factor, nerve growth factor, and granulocyte-monocyte colony stimulating factor. Serum levels of nitrite were also increased. Circulating granulocyte numbers were increased, and lymphocyte numbers were decreased. The changes in these parameters showed both a dose- and time-dependent association with histopathologic changes. These biomarkers could provide an additional tool for the nonclinical and clinical evaluation of investigational compounds.


Subject(s)
Biomarkers/blood , Blood Vessels/drug effects , Cyclic N-Oxides/toxicity , Phosphodiesterase 4 Inhibitors , Phosphodiesterase Inhibitors/toxicity , Quinolines/toxicity , Vascular Diseases/chemically induced , Animals , Clinical Chemistry Tests , Dose-Response Relationship, Drug , Immunohistochemistry , Leukocyte Count , Mesenteric Arteries/pathology , Nitrates/blood , Nitrites/blood , Rats , Rats, Sprague-Dawley , Vascular Diseases/blood , Vascular Diseases/pathology
18.
Bioorg Med Chem ; 16(16): 7599-606, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18656371

ABSTRACT

A series of drug-like compounds derived from Sildenafil, Vardenafil and Tadalafil analogues were modelled through the MIA-QSAR (multivariate image analysis applied to quantitative structure-activity relationships) ligand-based approach. A highly predictive model was achieved and novel compounds, miscellany of substructures of these three representative phosphodiesterase type-5 (PDE-5) inhibitors were predicted using the calibration parameters obtained through partial least squares (PLS) regression. The high bioactivities of eight promising compounds were corroborated by docking evaluation. Calculated ADME-Tox (absorption, distribution, metabolism, excretion and toxicity) profiles for such compounds suggest advantages of some of them over the currently available, most common drugs used for the treatment of erectile dysfunction.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 5/chemistry , Phosphodiesterase 5 Inhibitors , Phosphodiesterase Inhibitors/chemistry , Phosphodiesterase Inhibitors/pharmacology , Animals , Carbolines/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Imidazoles/chemistry , Mice , Models, Molecular , Phosphodiesterase Inhibitors/metabolism , Phosphodiesterase Inhibitors/toxicity , Piperazines/chemistry , Purines/chemistry , Quantitative Structure-Activity Relationship , Rats , Sildenafil Citrate , Sulfones/chemistry , Tadalafil , Triazines/chemistry , Vardenafil Dihydrochloride
19.
Neurotoxicol Teratol ; 30(5): 440-7, 2008.
Article in English | MEDLINE | ID: mdl-18508234

ABSTRACT

Though caffeine is broadly distributed in many plants and foods, little is known about the teratogenic effects of caffeine during early embryonic development. Here, we used zebrafish as a model to test toxicity and teratogenicity since they have transparent eggs, making the organogenesis of zebrafish embryos easier to observe. When the exposure doses of caffeine were less than 150 ppm (17.5, 35, 50, 100 and 150 ppm), the zebrafish embryos exhibited no significant differences in survival rates after comparison with vehicle-control (0 ppm) group. As the exposure dosages increased, the survival rates decreased. No embryos survived after treatment with 300 ppm caffeine or higher dosages. The most evident change in embryos treated with caffeine was a shorter body length (vehicle-control: 3.26+/-0.01 mm, n=49; vs 150 ppm of caffeine: 2.67+/-0.03 mm, n=50). In addition, caffeine-treated embryos exhibited significantly reduced tactile sensitivity frequencies of touch-induced movement (vehicle-control: 9.93+/-0.77 vs 17.5-150 ppm caffeine: 5.37+/-0.52-0.10+/-0.06). Subtle changes are easily observed by staining with specific monoclonal antibodies F59, Znp1 and Zn5 to detect morphological changes in muscle fibers, primary motor axons and secondary motor axon projections, respectively. Our data show that the treatment of caffeine leads to misalignment of muscle fibers and motor neuron defects, especially secondary motor neuron axonal growth defects.


Subject(s)
Caffeine/toxicity , Dyskinesia, Drug-Induced/physiopathology , Embryo, Nonmammalian/drug effects , Motor Neurons/drug effects , Muscle, Skeletal/drug effects , Spinal Cord/drug effects , Animals , Antibodies, Monoclonal , Disease Models, Animal , Dose-Response Relationship, Drug , Dyskinesia, Drug-Induced/pathology , Embryo, Nonmammalian/abnormalities , Embryonic Development/drug effects , Embryonic Development/physiology , Motor Neurons/pathology , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/abnormalities , Muscle, Skeletal/innervation , Neuromuscular Junction/abnormalities , Neuromuscular Junction/drug effects , Neuromuscular Junction/physiopathology , Phosphodiesterase Inhibitors/toxicity , Sensation Disorders/chemically induced , Sensation Disorders/pathology , Sensation Disorders/physiopathology , Spinal Cord/abnormalities , Survival Rate , Time Factors , Toxicity Tests , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...