Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Int J Neuropsychopharmacol ; 18(3)2014 Oct 31.
Article in English | MEDLINE | ID: mdl-25522408

ABSTRACT

BACKGROUND: Substantial clinical data support the addition of low doses of atypical antipsychotic drugs to selective serotonin reuptake inhibitors (SSRIs) to rapidly enhance the antidepressant effect in treatment-resistant depression. Preclinical studies suggest that this effect is at least partly explained by an increased catecholamine outflow in the medial prefrontal cortex (mPFC). METHODS: In the present study we used in vivo microdialysis in freely moving rats and in vitro intracellular recordings of pyramidal cells of the rat mPFC to investigate the effects of adding the novel atypical antipsychotic drug asenapine to the SSRI escitalopram with regards to monoamine outflow in the mPFC and dopamine outflow in nucleus accumbens as well as glutamatergic transmission in the mPFC. RESULTS: The present study shows that addition of low doses (0.05 and 0.1 mg/kg) of asenapine to escitalopram (5 mg/kg) markedly enhances dopamine, noradrenaline, and serotonin release in the rat mPFC as well as dopamine release in the nucleus accumbens. Moreover, this drug combination facilitated both N-methyl-d-Aspartate (NMDA)- and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-induced currents as well as electrically evoked excitatory postsynaptic potentials in pyramidal cells of the rat mPFC. CONCLUSIONS: Our results support the notion that the augmentation of SSRIs by atypical antipsychotic drugs in treatment-resistant depression may, at least in part, be related to enhanced catecholamine output in the prefrontal cortex and that asenapine may be clinically used to achieve this end. In particular, the subsequent activation of the D1 receptor may be of importance for the augmented antidepressant effect, as this mechanism facilitated both NMDA and AMPA receptor-mediated transmission in the mPFC. Our novel observation that the drug combination, like ketamine, facilitates glutamatergic transmission in the mPFC may contribute to explain the rapid and potent antidepressant effect obtained when atypical antipsychotic drugs are added to SSRIs.


Subject(s)
Antipsychotic Agents/pharmacology , Biogenic Monoamines/metabolism , Citalopram/pharmacology , Glutamic Acid/metabolism , Heterocyclic Compounds, 4 or More Rings/pharmacology , Prefrontal Cortex/drug effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Synaptic Transmission/drug effects , Animals , Benzazepines/pharmacology , Bicuculline/pharmacology , Dibenzocycloheptenes , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Drug , Excitatory Amino Acid Agents/pharmacology , Excitatory Postsynaptic Potentials/drug effects , GABA-A Receptor Antagonists/pharmacology , Male , Neurons/drug effects , Prefrontal Cortex/cytology , Rats , Rats, Wistar , Time Factors
2.
Synapse ; 66(7): 650-60, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22362425

ABSTRACT

The psychotropic drug asenapine is approved for the treatment of schizophrenia and manic or mixed episodes associated with bipolar I disorder. Asenapine exhibits higher affinity for several 5-HT receptors and α(2)-adrenoceptors than for D(2) receptors. Noteworthy, blockage of both the 5-HT(2A) and α(2)-adrenergic receptors has been shown to enhance prefrontal dopamine release induced by D(2) receptor antagonists. Previous results show that asenapine, both systemically and locally, increases dopamine, noradrenaline, and serotonin release in the medial prefrontal cortex (mPFC), and that the increased dopamine release largely depends on an intracortical action. Using reverse microdialysis in freely moving rats, we here assessed the potency of low concentrations of asenapine to cause a pharmacologically significant blockage in vivo of 5-HT(2A) receptors and α(2)-adrenoceptors within the mPFC, and thus its ability to affect cortical monoamine release by these receptors. Intracortical administration of 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane hydrochloride (DOI), a 5-HT(2A/2C) receptor agonist, increased cortical monoamine release, effects that were antagonized both by asenapine and the selective 5-HT(2A) antagonist M100907. Application of clonidine, an α(2)-adrenoceptor agonist, significantly reduced monoamine release in the mPFC. The selective α(2)-adrenoceptor antagonist idazoxan blocked, whereas asenapine partially blocked clonidine-induced cortical dopamine and noradrenaline decrease. The effects of asenapine and idazoxan on clonidine-induced serotonin decrease were less pronounced. Our results propose that low concentrations of asenapine in the mPFC exhibit a pharmacologically significant 5-HT(2A) and α(2) receptor antagonistic activity, which may contribute to enhance prefrontal monoamine release in vivo and, secondarily, its clinical effects in schizophrenia and bipolar disorder.


Subject(s)
Antipsychotic Agents/pharmacology , Biogenic Monoamines/metabolism , Heterocyclic Compounds, 4 or More Rings/pharmacology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Receptor, Serotonin, 5-HT2A/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Amphetamines/pharmacology , Animals , Dibenzocycloheptenes , Fluorobenzenes/pharmacology , Male , Microdialysis , Norepinephrine/metabolism , Piperidines/pharmacology , Rats , Rats, Wistar , Serotonin/metabolism , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Up-Regulation
3.
Synapse ; 66(4): 277-90, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22121030

ABSTRACT

Antidepressant drugs are frequently used to treat affective symptoms in schizophrenia. We have recently shown that escitalopram, but not citalopram or R-citalopram, increases firing rate and burst firing of midbrain dopamine neurons, potentiates cortical N-methyl-D-aspartate (NMDA) receptor-mediated transmission and enhances cognition, effects that might influence the outcome of concomitant antipsychotic medication. Here, we studied, in rats, the behavioral and neurobiological effects of adding escitalopram, citalopram, or R-citalopram to the second-generation antipsychotic drug risperidone. We examined antipsychotic efficacy using the conditioned avoidance response (CAR) test, extrapyramidal side effect (EPS) liability using a catalepsy test, dopamine outflow in the medial prefrontal cortex (mPFC) and nucleus accumbens using in vivo microdialysis in freely moving animals, and NMDA receptor-mediated transmission in the mPFC using intracellular electrophysiological recording in vitro. Only escitalopram (5 mg/kg), but not citalopram (10 mg/kg), or R-citalopram (10 mg/kg), dramatically enhanced the antipsychotic-like effect of a low dose of risperidone (0.25 mg/kg), without increasing catalepsy. Given alone, escitalopram, but not citalopram or R-citalopram, markedly enhanced both cortical dopamine output and NMDA receptor-mediated transmission. Addition of escitalopram and to some extent R-citalopram, but not citalopram, significantly enhanced both cortical dopamine output and cortical NMDA receptor-mediated transmission induced by a suboptimal dose/concentration of risperidone. These results suggest that adjunct treatment with escitalopram, but not citalopram, may enhance the effect of a subtherapeutic dose of risperidone on positive, negative, cognitive, and depressive symptoms in schizophrenia, yet without increased EPS liability.


Subject(s)
Antipsychotic Agents/administration & dosage , Brain/drug effects , Citalopram/administration & dosage , Risperidone/administration & dosage , Selective Serotonin Reuptake Inhibitors/administration & dosage , Animals , Avoidance Learning/drug effects , Behavior, Animal/drug effects , Brain/metabolism , Chromatography, High Pressure Liquid , Dopamine/metabolism , Drug Synergism , Drug Therapy, Combination , Electrophysiological Phenomena/drug effects , Male , Microdialysis , Patch-Clamp Techniques , Rats , Rats, Wistar
4.
Synapse ; 65(5): 357-67, 2011 May.
Article in English | MEDLINE | ID: mdl-20730799

ABSTRACT

Escitalopram, the S-enantiomer of citalopram, possesses superior efficacy compared to other selective serotonin reuptake inhibitors (SSRIs) in the treatment of major depression. Escitalopram binds to an allosteric site on the serotonin transporter, which further enhances the blockade of serotonin reuptake, whereas R-citalopram antagonizes this positive allosteric modulation. Escitalopram's effects on neurotransmitters other than serotonin, for example, dopamine and glutamate, are not well studied. Therefore, we here studied the effects of escitalopram, citalopram, and R-citalopram on dopamine cell firing in the ventral tegmental area, using single-cell recording in vivo and on NMDA receptor-mediated currents in pyramidal neurons in the medial prefrontal cortex using in vitro electrophysiology in rats. The cognitive effects of escitalopram and citalopram were also compared using the novel object recognition test. Escitalopram (40-640 µg/kg i.v.) increased both firing rate and burst firing of dopaminergic neurons, whereas citalopram (80-1280 µg/kg) had no effect on firing rate and only increased burst firing at high dosage. R-citalopram (40-640 µg/kg) had no significant effects. R-citalopram (320 µg/kg) antagonized the effects of escitalopram (320 µg/kg). A very low concentration of escitalopram (5 nM), but not citalopram (10 nM) or R-citalopram (5 nM), potentiated NMDA-induced currents in pyramidal neurons. Escitalopram's effect was antagonized by R-citalopram and blocked by the dopamine D(1) receptor antagonist SCH23390. Escitalopram, but not citalopram, improved recognition memory. Our data suggest that the excitatory effect of escitalopram on dopaminergic and NMDA receptor-mediated neurotransmission may have bearing on its cognitive-enhancing effect and superior efficacy compared to other SSRIs in major depression.


Subject(s)
Action Potentials/drug effects , Citalopram/pharmacology , Cognition/physiology , Dopamine/metabolism , Neurons/drug effects , Prefrontal Cortex/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Ventral Tegmental Area/cytology , Analysis of Variance , Animals , Benzazepines/pharmacology , Dopamine Agents/pharmacology , Dose-Response Relationship, Drug , Male , Neuropsychological Tests , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Recognition, Psychology
5.
Sci Adv ; 7(16)2021 04.
Article in English | MEDLINE | ID: mdl-33863724

ABSTRACT

Several important drug targets, e.g., ion channels and G protein-coupled receptors, are extremely difficult to approach with current antibody technologies. To address these targets classes, we explored kinetically controlled proteases as structural dynamics-sensitive druggability probes in native-state and disease-relevant proteins. By using low-Reynolds number flows, such that a single or a few protease incisions are made, we could identify antibody binding sites (epitopes) that were translated into short-sequence antigens for antibody production. We obtained molecular-level information of the epitope-paratope region and could produce high-affinity antibodies with programmed pharmacological function against difficult-to-drug targets. We demonstrate the first stimulus-selective monoclonal antibodies targeting the transient receptor potential vanilloid 1 (TRPV1) channel, a clinically validated pain target widely considered undruggable with antibodies, and apoptosis-inducing antibodies selectively mediating cytotoxicity in KRAS-mutated cells. It is our hope that this platform will widen the scope of antibody therapeutics for the benefit of patients.


Subject(s)
Antibodies, Monoclonal , Antigens , Antibodies, Monoclonal/chemistry , Binding Sites, Antibody , Epitopes , Humans
6.
Int J Neuropsychopharmacol ; 13(7): 891-903, 2010 Aug.
Article in English | MEDLINE | ID: mdl-19835668

ABSTRACT

Compared to both first- and second-generation antipsychotic drugs (APDs), clozapine shows superior efficacy in treatment-resistant schizophrenia. In contrast to most APDs clozapine possesses high affinity for alpha2-adrenoceptors, and clinical and preclinical studies provide evidence that the alpha2-adrenoceptor antagonist idazoxan enhances the antipsychotic efficacy of typical D2 receptor antagonists as well as olanzapine. Risperidone has lower affinity for alpha2-adrenoceptors than clozapine but higher than most other APDs. Here we examined, in rats, the effects of adding idazoxan to risperidone on antipsychotic effect using the conditioned avoidance response (CAR) test, extrapyramidal side-effect (EPS) liability using the catalepsy test, brain dopamine efflux using in-vivo microdialysis in freely moving animals, cortical N-methyl-D-aspartate (NMDA) receptor-mediated transmission using intracellular electrophysiological recording in vitro, and ex-vivo autoradiography to assess the in-vivo alpha2A- and alpha2C-adrenoceptor occupancies by risperidone. The dose of risperidone needed for antipsychotic effect in the CAR test was approximately 0.4 mg/kg, which produced 11% and 17% in-vivo receptor occupancy at alpha2A- and alpha2C-adrenoceptors, respectively. Addition of idazoxan (1.5 mg/kg) to a low dose of risperidone (0.25 mg/kg) enhanced the suppression of CAR, but did not enhance catalepsy. Both cortical dopamine release and NMDA receptor-mediated responses were enhanced. These data propose that the therapeutic effect of risperidone in schizophrenia can be enhanced and its EPS liability reduced by adjunctive treatment with an alpha2-adrenoceptor antagonist, and generally support the notion that the potent alpha2-adrenoceptor antagonistic action of clozapine may be highly important for its unique efficacy in schizophrenia.


Subject(s)
Adrenergic alpha-2 Receptor Antagonists/pharmacology , Antipsychotic Agents/pharmacology , Cerebral Cortex/drug effects , Idazoxan/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism , Risperidone/pharmacology , Synaptic Transmission/drug effects , Adrenergic alpha-2 Receptor Antagonists/administration & dosage , Animals , Antipsychotic Agents/administration & dosage , Antipsychotic Agents/metabolism , Avoidance Learning/drug effects , Catalepsy/drug therapy , Catalepsy/metabolism , Cerebral Cortex/metabolism , Dopamine Agonists/administration & dosage , Dopamine Agonists/metabolism , Dopamine Agonists/pharmacology , Electrophysiological Phenomena/drug effects , Excitatory Amino Acid Agents/administration & dosage , Excitatory Amino Acid Agents/metabolism , Excitatory Amino Acid Agents/pharmacology , Idazoxan/administration & dosage , Idazoxan/metabolism , Male , Microdialysis , Rats , Rats, Sprague-Dawley , Rats, Wistar , Risperidone/administration & dosage , Risperidone/metabolism
7.
Synapse ; 64(11): 870-4, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20842721

ABSTRACT

Asenapine is a novel psychopharmacologic agent being developed for schizophrenia and bipolar disorder. Like clozapine, asenapine facilitates cortical dopaminergic and N-methyl-D-aspartate (NMDA) receptor-mediated transmission in rats. The facilitation of NMDA-induced currents in cortical pyramidal cells by clozapine is dependent on dopamine and D(1) receptor activation. Moreover, previous results show that clozapine prevents and reverses the blockade of NMDA-induced currents and firing activity in the pyramidal cells by the noncompetitive NMDA receptor antagonist phencyclidine (PCP). Here, we investigated the effects of asenapine in these regards using intracellular electrophysiological recording in vitro. Asenapine (5 nM) significantly facilitated NMDA-induced currents (162 ± 15% of control) in pyramidal cells of the medial prefrontal cortex (mPFC). The asenapine-induced facilitation was blocked by the D(1) receptor antagonist SCH23390 (1 µM). Furthermore, the PCP-induced blockade of cortical NMDA-induced currents was effectively reversed by 5 nM asenapine. Our results demonstrate a clozapine-like facilitation of cortical NMDA-induced currents by asenapine that involves prefrontal dopamine and activation of D(1) receptors. Asenapine and clozapine also share the ability to reverse functional PCP-induced hypoactivity of cortical NMDA receptors. The ability of asenapine to increase both cortical dopaminergic and NMDA receptor-mediated glutamatergic transmission suggests that this drug may have an advantageous effect not only on positive symptoms in patients with schizophrenia, but also on negative and cognitive symptoms.


Subject(s)
Antipsychotic Agents/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Neurons/drug effects , Prefrontal Cortex/cytology , Receptors, Dopamine D1/metabolism , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Benzazepines/pharmacology , Dibenzocycloheptenes , Drug Interactions , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , In Vitro Techniques , Male , Membrane Potentials/drug effects , N-Methylaspartate/pharmacology , Patch-Clamp Techniques/methods , Phencyclidine/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D1/antagonists & inhibitors
8.
Synapse ; 63(10): 913-20, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19582782

ABSTRACT

Treatment with topiramate may improve negative symptoms in schizophrenia when added to typical antipsychotic drugs (APDs) but not to clozapine. Both dopaminergic and glutamatergic transmissions in the medial prefrontal cortex (mPFC) are facilitated by atypical, but not typical, APDs, which is thought to improve negative symptoms and cognitive dysfunction in schizophrenia. Our previous results show that topiramate increases prefrontal dopamine (DA) outflow when added to the D(2/3) receptorantagonist raclopride. Here, using intracellular recording in vitro, we investigated the effects of topiramate on glutamatergic neurotransmission in the rat mPFC, both when given alone and in combination with raclopride or clozapine. Neither topiramate nor raclopride alone had any effect on N-methyl-D-aspartate (NMDA)-induced currents in pyramidal cells of the mPFC. However, the combination of topiramate and raclopride facilitated the NMDA-induced currents, and this effect was blocked by the D1 receptor antagonist SCH23390. Topiramate also facilitated the effect of a submaximal, but inhibited the effect of a maximal, concentration of clozapine on these currents. The effect of combined topiramate and a submaximal concentration of clozapine could be blocked by SCH23390. In addition, combined topiramate and raclopride facilitated excitatory postsynaptic potentials. In contrast, topiramate inhibited clozapine's facilitating effect on these potentials. These data may help explain the improvement of negative symptoms when topiramate is used as adjunctive therapy in schizophrenic patients receiving typical APDs, but they may also shed light on the observed deterioration of symptoms when topiramate is added to full dose clozapine.


Subject(s)
Clozapine/pharmacology , Dopamine Antagonists/pharmacology , Fructose/analogs & derivatives , Glutamic Acid/metabolism , Neuroprotective Agents/pharmacology , Prefrontal Cortex/drug effects , Raclopride/pharmacology , Serotonin Antagonists/pharmacology , Synaptic Transmission/drug effects , Animals , Biophysical Phenomena/drug effects , Dose-Response Relationship, Drug , Drug Interactions , Electric Stimulation , Excitatory Postsynaptic Potentials/drug effects , Fructose/pharmacology , In Vitro Techniques , Male , Membrane Potentials/drug effects , N-Methylaspartate/pharmacology , Patch-Clamp Techniques/methods , Prefrontal Cortex/cytology , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Rats , Rats, Sprague-Dawley , Topiramate
9.
Eur Neuropsychopharmacol ; 29(11): 1282-1287, 2019 11.
Article in English | MEDLINE | ID: mdl-31537475

ABSTRACT

Recently, a single injection of the nitric oxide donor sodium nitroprusside (SNP) was found to induce a rapid and sustained antipsychotic effect in treatment-resistant schizophrenia (TRS). Moreover, a single i.p. injection of SNP in rats was found to generate both rapid and persisting changes in brain synaptic plasticity, including enhanced excitatory postsynaptic current responses and spine morphology in layer V pyramidal cells in the medial prefrontal cortex (mPFC) brain slices. Here we used the conditioned avoidance response (CAR) test in rats to investigate the antipsychotic-like efficacy of SNP in combination with low-dose risperidone. In addition, we performed microdialysis experiments in freely moving rats to measure neurotransmitter efflux in the mPFC and the nucleus accumbens (NAc). Risperidone caused only 20% suppression of CAR, which is far below the degree of CAR suppression required to predict a significant clinical antipsychotic effect. Addition of a low dose of SNP to risperidone dramatically enhanced the antipsychotic-like effect to a clinically relevant level. SNP significantly enhanced the risperidone-induced dopamine output in the mPFC but not in the NAc. The increased prefrontal dopamine release induced by the drug combination may also improve cognition as indicated by previous preclinical and clinical studies and, furthermore, via enhanced synaptic spine function and morphology in mPFC generate a both rapid and prolonged antipsychotic and pro-cognitive effect. Our results delineate SNP as a promising new treatment option for schizophrenia, including TRS, when added to currently available antipsychotic medication in order to improve efficacy at maintained or even reduced dosage.


Subject(s)
Avoidance Learning/drug effects , Nitroprusside/pharmacology , Risperidone/pharmacology , Animals , Dopamine/metabolism , Drug Synergism , Male , Microdialysis , Nucleus Accumbens/metabolism , Prefrontal Cortex/metabolism , Rats
10.
Psychopharmacology (Berl) ; 196(3): 417-29, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17940749

ABSTRACT

RATIONALE: Asenapine is a novel psychopharmacologic agent being developed for the treatment of schizophrenia and bipolar disorder. MATERIALS AND METHODS: The present study was undertaken to investigate the effects of asenapine using animal models predictive of antipsychotic efficacy (conditioned avoidance response [CAR]) and extrapyramidal side effects (EPS; catalepsy). In parallel, the effects of asenapine on regional dopamine output using in vivo microdialysis in freely moving rats, dopamine output in the core and shell subregions of nucleus accumbens (NAc) using in vivo voltammetry in anesthetized rats, and N-methyl-D: -aspartate (NMDA)-induced currents in pyramidal neurons of the medial prefrontal cortex (mPFC) using the electrophysiological technique intracellular recording in vitro were assessed. RESULTS: Asenapine (0.05-0.2 mg/kg, subcutaneous [s.c.]) induced a dose-dependent suppression of CAR (no escape failures recorded) and did not induce catalepsy. Asenapine (0.05-0.2 mg/kg, s.c.) increased dopamine efflux in both the mPFC and the NAc. Low-dose asenapine (0.01 mg/kg, intravenous [i.v.]) increased dopamine efflux preferentially in the shell compared to the core of NAc, whereas at a higher dose (0.05 mg/kg, i.v.), the difference disappeared. Finally, like clozapine (100 nM), but at a considerably lower concentration (5 nM), asenapine significantly potentiated the NMDA-induced responses in pyramidal cells of the mPFC. CONCLUSIONS: These preclinical data suggest that asenapine may exhibit highly potent antipsychotic activity with very low EPS liability. Its ability to increase both dopaminergic and glutamatergic activity in rat mPFC suggests that asenapine may possess an advantageous effect not only on positive symptoms in patients with schizophrenia, but also on negative and cognitive symptoms.


Subject(s)
Antipsychotic Agents/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Schizophrenia/drug therapy , Animals , Antipsychotic Agents/administration & dosage , Antipsychotic Agents/adverse effects , Avoidance Learning , Catalepsy/chemically induced , Conditioning, Classical , Dibenzocycloheptenes , Disease Models, Animal , Dopamine/metabolism , Dose-Response Relationship, Drug , Electrophysiology , Heterocyclic Compounds, 4 or More Rings/administration & dosage , Heterocyclic Compounds, 4 or More Rings/adverse effects , Injections, Subcutaneous , Male , Microdialysis , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Rats, Wistar
11.
Eur Neuropsychopharmacol ; 28(5): 620-629, 2018 05.
Article in English | MEDLINE | ID: mdl-29571967

ABSTRACT

The 5-HT5A receptor is arguably the least understood 5-HT receptor. Despite widespread expression in human and rodent brains it lacks specific ligands. Our previous results suggest that 5-HT5A receptor antagonists may be effective against cognitive impairment in schizophrenia. In this study, using behavioral, immunohistochemical, electrophysiological and microdialysis techniques, we examined the mechanism by which ASP5736, a novel and selective 5-HT5A receptor antagonist, exerts a positive effect in animal models of cognitive impairment. We first confirmed the effect of ASP5736 on cognitive deficits in rats treated subchronically with phencyclidine hydrochloride (PCP) using an attentional set shifting task. Subsequently, we identified 5-HT5A receptors in dopaminergic (DAergic) neurons and parvalbumin (PV)-positive interneurons in the ventral tegmental area (VTA) and in PV-positive interneurons in the medial prefrontal cortex (mPFC). Burst firing of the DAergic cells in the parabrachial pigmental nucleus (PBP) in the VTA, which predominantly project to the mPFC, was significantly enhanced by treatment with ASP5736. In contrast, ASP5736 exerted no significant effect on either the firing rate or burst firing in the DA cells in the paranigral nucleus (PN), that project to the nucleus accumbens (N. Acc.). ASP5736 increased the release of DA and gamma-aminobutyric acid (GABA) in the mPFC of subchronically PCP-treated rats. These results support our hypothesis that ASP5736 might block the inhibitory 5-HT5A receptors on DAergic neurons in the VTA that project to the mPFC, and interneurons in the mPFC, and thereby improve cognitive impairment by preferentially enhancing DAergic and GABAergic neurons in the mPFC.


Subject(s)
Cognitive Dysfunction/complications , Cognitive Dysfunction/drug therapy , Guanidines/pharmacology , Isoquinolines/pharmacology , Receptors, Serotonin/drug effects , Receptors, Serotonin/metabolism , Schizophrenia/complications , Schizophrenia/drug therapy , Action Potentials/physiology , Animals , Cognitive Dysfunction/chemically induced , Discrimination, Psychological/drug effects , Dopamine/metabolism , Dopaminergic Neurons/physiology , Interneurons/physiology , Male , Phencyclidine , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiology , Rats , Schizophrenic Psychology , Serotonin Antagonists/pharmacology , Ventral Tegmental Area/physiology , gamma-Aminobutyric Acid/metabolism
12.
Eur J Pharmacol ; 573(1-3): 148-60, 2007 Nov 14.
Article in English | MEDLINE | ID: mdl-17689529

ABSTRACT

The present study describes the pharmacological profile of the putative antipsychotic drug Lu 35-138 ((+)-(S)-3-{1-[2-(1-acetyl-2,3-dihydro-1H-indol-3-yl)ethyl]-3,6-dihydro-2H-pyridin-4-yl}-6-chloro-1H-indole). The in vitro receptor profile of Lu 35-138 revealed high affinity (K(i)=5 nM) and competitive antagonism (K(b)=8 nM) at dopamine D(4) receptors combined with potent 5-HT uptake inhibition (IC(50)=3.2 nM) and moderate alpha(1)-adrenoceptor affinity (K(i)=45 nM). In vivo, Lu 35-138 selectively counteracted hyperlocomotion induced by d-amphetamine (0.5 mg/kg; ED(50)=4.0 mg/kg, s.c.) in rats and phencyclidine (PCP; 2.5 mg/kg; ED(50)=13 mg/kg, s.c.) in mice. Lu 35-138 was unable to affect hyperlocomotion induced by a high dose of d-amphetamine (2.0 mg/kg), which indicates a preferential action on limbic versus striatal structures. A similar limbic selectivity of Lu 35-138 was indicated in voltammetric measure of dopamine output in the core and shell subdivisions of the nucleus accumbens in rats. Furthermore, a relatively large dose of Lu 35-138 (18 mg/kg, s.c.) counteracted d-amphetamine-induced disruption of pre-pulse inhibition in rats and repeated administration of Lu 35-138 (0.31 or 1.25 mg/kg, p.o. once daily for 3 weeks) reduced the number of spontaneously active dopamine neurones in the ventral tegmental area, underlining its antipsychotic-like profile. Lu 35-138 failed to induce catalepsy in rats or dystonia in Cebus apella monkeys and did not deteriorate spatial memory in rats as assessed by water maze performance. Collectively, these results suggest that Lu 35-138 possesses antipsychotic activity combined with a low extrapyramidal and cognitive side effect liability.


Subject(s)
Dihydropyridines/pharmacology , Indoles/pharmacology , Motor Activity/drug effects , Receptors, Dopamine D4/antagonists & inhibitors , Selective Serotonin Reuptake Inhibitors/pharmacology , Adrenergic alpha-1 Receptor Antagonists , Animals , Animals, Outbred Strains , Benzodiazepines/pharmacology , Cebus , Citalopram/pharmacology , Clozapine/pharmacology , Cognition/drug effects , Dihydropyridines/chemistry , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Female , Haloperidol/pharmacology , Haplorhini , Humans , Indoles/chemistry , Male , Mice , Molecular Structure , Olanzapine , Piperazines/chemistry , Piperazines/pharmacology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Risperidone/pharmacology , Selective Serotonin Reuptake Inhibitors/chemistry , Sulfonamides/pharmacology
13.
Eur Neuropsychopharmacol ; 27(4): 411-417, 2017 04.
Article in English | MEDLINE | ID: mdl-28190661

ABSTRACT

Brexpiprazole (Rexulti®), a novel D2/3 receptor (R) partial agonist, was recently approved as monotherapy for schizophrenia, demonstrating effectiveness against both positive and negative symptoms, and also approved as add-on treatment to antidepressant drugs, inducing a potent antidepressant effect with a faster onset compared to an antidepressant given alone. Moreover, brexpiprazole has demonstrated pro-cognitive effects in preclinical studies. To explore whether the observed effects may be mediated via modulation of prefrontal glutamatergic transmission, we investigated the effect of brexpiprazole, alone and in combination with the SSRI escitalopram, on prefrontal glutamatergic transmission using in vitro electrophysiological intracellular recordings of deep layer pyramidal cells of the rat medial prefrontal cortex (mPFC). Nanomolar concentrations of brexpiprazole potentiated NMDAR-induced currents and electrically evoked EPSPs via activation of dopamine D1Rs, in similarity with the effect of the atypical antipsychotic drug clozapine. The effect of an ineffective concentration of brexpiprazole was significantly potentiated by the addition of escitalopram. When combined with escitalopram, brexpiprazole also potentiated AMPAR-mediated transmission, in similarity with the clinically rapid acting antidepressant drug ketamine. The effect on the AMPAR-mediated currents was also D1R dependent. In conclusion, our data propose that brexpiprazole exerts a clozapine-like potentiation of NMDAR-mediated currents in the mPFC, which can explain its efficacy on negative symptoms of schizophrenia and the pro-cognitive effects observed preclinically. Moreover, add-on brexpiprazole to escitalopram also potentiated AMPAR-mediated transmission, which may provide a neurobiological explanation to the faster antidepressant effect of add-on brexpiprazole in major depression.


Subject(s)
Dopamine Agonists/pharmacology , Glutamic Acid/metabolism , Prefrontal Cortex/drug effects , Quinolones/pharmacology , Receptors, Dopamine D1/metabolism , Synaptic Transmission/drug effects , Thiophenes/pharmacology , Animals , Citalopram/pharmacology , Dose-Response Relationship, Drug , Drug Combinations , Electric Stimulation , Excitatory Postsynaptic Potentials/drug effects , In Vitro Techniques , Male , Neurotransmitter Agents/pharmacology , Patch-Clamp Techniques , Prefrontal Cortex/metabolism , Rats , Rats, Sprague-Dawley , Selective Serotonin Reuptake Inhibitors/pharmacology
14.
Eur Neuropsychopharmacol ; 26(9): 1401-1411, 2016 09.
Article in English | MEDLINE | ID: mdl-27474687

ABSTRACT

Nicotine has been found to improve cognition and reduce negative symptoms in schizophrenia and a genetic and pathophysiological link between the α7 nicotinic acetylcholine receptors (nAChRs) and schizophrenia has been demonstrated. Therefore, there has been a large interest in developing drugs affecting the α7 nAChRs for schizophrenia. In the present study we investigated, in rats, the effects of a selective α7 agonist (PNU282987) and a α7 positive allosteric modulator (PAM; NS1738) alone and in combination with the atypical antipsychotic drug risperidone for their utility as adjunct treatment in schizophrenia. Moreover we also investigated their utility as adjunct treatment in depression in combination with the SSRI citalopram. We found that NS1738 and to some extent also PNU282987, potentiated a subeffective dose of risperidone in the conditioned avoidance response test. Both drugs also potentiated the effect of a sub-effective concentration of risperidone on NMDA-induced currents in pyramidal cells of the medial prefrontal cortex. Moreover, NS1738 and PNU282987 enhanced recognition memory in the novel object recognition test, when given separately. Both drugs also potentiated accumbal but not prefrontal risperidone-induced dopamine release. Finally, PNU282987 reduced immobility in the forced swim test, indicating an antidepressant-like effect. Taken together, our data support the utility of drugs targeting the α7 nAChRs, perhaps especially α7 PAMs, to potentiate the effect of atypical antipsychotic drugs. Moreover, our data suggest that α7 agonists and PAMs can be used to ameliorate cognitive symptoms in schizophrenia and depression.


Subject(s)
Antidepressive Agents/pharmacology , Antipsychotic Agents/pharmacology , Benzamides/pharmacology , Bridged Bicyclo Compounds/pharmacology , Phenylurea Compounds/pharmacology , Schizophrenia/drug therapy , alpha7 Nicotinic Acetylcholine Receptor/agonists , Animals , Citalopram/pharmacology , Depression/drug therapy , Depression/metabolism , Disease Models, Animal , Dopamine/metabolism , Excitatory Amino Acid Agonists/pharmacology , Male , N-Methylaspartate/metabolism , N-Methylaspartate/pharmacology , Nicotinic Agonists/pharmacology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Rats, Sprague-Dawley , Rats, Wistar , Risperidone/pharmacology , Schizophrenia/metabolism , Schizophrenic Psychology , Selective Serotonin Reuptake Inhibitors/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/metabolism
15.
Eur Neuropsychopharmacol ; 23(7): 709-20, 2013 Jul.
Article in English | MEDLINE | ID: mdl-22732518

ABSTRACT

Quetiapine alleviates both positive and negative symptoms as well as certain cognitive impairments in schizophrenia despite a low D2 receptor occupancy and may also be used as monotherapy in bipolar and major depressive disorder. The mechanisms underlying the broad clinical utility of quetiapine remain to be clarified, but may be related to the potent inhibition of the norepinephrine transporter (NET) by norquetiapine, the major metabolite of quetiapine in humans. Since norquetiapine is not formed in rodents we here investigated in rats whether NET-inhibition may, in principle, contribute to the clinical effectiveness of quetiapine and allow for its low D2 receptor occupancy, by combining quetiapine with the selective NET-inhibitor reboxetine. Antipsychotic-like activity was assessed using the conditioned avoidance response (CAR) test, dopamine output in the medial prefrontal cortex (mPFC) and the nucleus accumbens was measured using in vivo microdialysis, and NMDA receptor-mediated transmission was measured using intracellular electrophysiological recordings in pyramidal cells of the mPFC in vitro. Adjunct reboxetine potentiated the suppression of CAR by quetiapine. Moreover, concomitant administration of quetiapine and reboxetine resulted in a synergistic increase in cortical, but not accumbal, dopamine output. The combination of low, clinically relevant concentrations of quetiapine (60 nM) and reboxetine (20 nM) markedly facilitated cortical NMDA receptor-mediated transmission in contrast to either drug alone, an effect that could be inhibited by the D1 receptor antagonist SCH23390. We conclude that concomitant NET-inhibition by norquetiapine may contribute to the overall antipsychotic effectiveness of quetiapine in spite of its relatively low level of D2 occupancy.


Subject(s)
Antipsychotic Agents/pharmacology , Avoidance Learning/drug effects , Dibenzothiazepines/pharmacology , Drug Synergism , Norepinephrine Plasma Membrane Transport Proteins/antagonists & inhibitors , 3,4-Dihydroxyphenylacetic Acid/metabolism , Adrenergic Uptake Inhibitors/pharmacology , Animals , Benzazepines/pharmacology , Dibenzothiazepines/antagonists & inhibitors , Dopamine/metabolism , Dopamine Antagonists/pharmacology , Dose-Response Relationship, Drug , Male , Membrane Potentials/drug effects , Morpholines/antagonists & inhibitors , Morpholines/pharmacology , N-Methylaspartate/antagonists & inhibitors , N-Methylaspartate/pharmacology , Nucleus Accumbens/metabolism , Prefrontal Cortex/metabolism , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Quetiapine Fumarate , Raclopride/pharmacology , Rats , Reboxetine
16.
Prog Neuropsychopharmacol Biol Psychiatry ; 39(1): 143-8, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22691715

ABSTRACT

A substantial number of patients with major depressive disorder (MDD) do not respond adequately to current antidepressant pharmacological treatments, which are all more or less based on a gradually increased enhancement of monoaminergic neurotransmission. Although a functional deficiency in monoaminergic neurotransmission may contribute to MDD, the etiology and pathophysiology are far from clarified. Recent studies suggest that inflammatory processes may contribute, since increased levels of pro-inflammatory cytokines and prostaglandin E(2) (PGE(2)) have repeatedly been observed in a subset of patients suffering from MDD. Interestingly, adjunct treatment with the anti-inflammatory drug celecoxib, a cyclo-oxygenase-2 (COX-2) inhibitor which blocks the PGE(2)-production, has shown to enhance the efficacy of both reboxetine, a selective noradrenaline reuptake inhibitor, as well as fluoxetine, a selective serotonin reuptake inhibitor, in treatment-resistant depression. To examine the neurobiological underpinnings to the clinical observations, we here studied the acute effects of a combined treatment with celecoxib and reboxetine on noradrenaline and dopamine output, as well as celecoxib and fluoxetine on 5-HT output in the medial prefrontal cortex, using in vivo microdialysis in awake freely moving rats. Celecoxib significantly potentiated the effects of reboxetine and fluoxetine on cortical noradrenaline and 5-HT output, respectively, but not the reboxetine-induced dopamine output. Moreover, celecoxib, when given alone, enhanced 5-HT output. These findings provide, in principle, novel experimental support for the clinical utility of combined treatment with antidepressant and anti-inflammatory drugs, such as COX-2 inhibitors, in MDD.


Subject(s)
Fluoxetine/pharmacology , Morpholines/pharmacology , Norepinephrine/metabolism , Prefrontal Cortex/drug effects , Pyrazoles/pharmacology , Serotonin/metabolism , Sulfonamides/pharmacology , Adrenergic Uptake Inhibitors/pharmacology , Animals , Celecoxib , Cyclooxygenase 2 Inhibitors/pharmacology , Dopamine/metabolism , Drug Synergism , Male , Microdialysis/methods , Prefrontal Cortex/metabolism , Rats , Rats, Wistar , Reboxetine , Selective Serotonin Reuptake Inhibitors/pharmacology
17.
Psychopharmacology (Berl) ; 221(1): 115-31, 2012 May.
Article in English | MEDLINE | ID: mdl-22068461

ABSTRACT

RATIONALE: The α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptor positive allosteric modulators (AMPA-PAMs), Org 24448 and Org 26576, and the glycine transporter-1 (GlyT-1) inhibitor Org 25935 are developed for treatment of schizophrenia. OBJECTIVES: Here we examined experimentally the ability of co-administration of these AMPA-PAMs or the GlyT-1 inhibitor to augment the antipsychotic activity and effect on cortical N-methyl-D: -aspartate (NMDA) receptor-mediated transmission of risperidone, olanzapine, or haloperidol. METHODS: We examined antipsychotic efficacy using the conditioned avoidance response (CAR) test, extrapyramidal side effect liability using a catalepsy test, and cortical NMDA receptor-mediated glutamatergic transmission using intracellular electrophysiological recording technique in vitro. RESULTS: Both AMPA-PAMs enhanced the suppression of CAR induced by risperidone or olanzapine, and Org 24448 also enhanced the effect of haloperidol. In contrast, the GlyT-1 inhibitor did not cause any behaviorally significant effect in the CAR test. However, the GlyT-1 inhibitor, but not the AMPA-PAMs, produced a large facilitation of NMDA-induced currents. All three drugs potentiated the effect of risperidone but not haloperidol on these currents. The GlyT-1 inhibitor also facilitated the effect of olanzapine. All drugs potentiated the effect of risperidone on electrically stimulated excitatory postsynaptic potentials (EPSP) in cortical pyramidal cells, whereas only the GlyT inhibitor facilitated the effect of olanzapine. CONCLUSIONS: Our results suggest that the AMPA-PAMs, when compared to the GlyT-1 inhibitor, show differential effects in terms of augmentation of antipsychotic efficacy, particularly when combined with risperidone or olanzapine. Both AMPA-PAMs and the GlyT-1 inhibitor may also improve negative symptoms and cognitive impairments in schizophrenia, in particular when combined with risperidone.


Subject(s)
Antipsychotic Agents/agonists , Avoidance Learning/physiology , Glutamic Acid/metabolism , Oxadiazoles/pharmacology , Piperidines/pharmacology , Prefrontal Cortex/physiology , Receptors, AMPA/agonists , Risperidone/pharmacology , Synaptic Transmission/physiology , Animals , Antipsychotic Agents/pharmacology , Avoidance Learning/drug effects , Benzodiazepines/agonists , Benzodiazepines/pharmacology , Catalepsy/physiopathology , Drug Synergism , Glycine Plasma Membrane Transport Proteins/antagonists & inhibitors , Haloperidol/agonists , Haloperidol/pharmacology , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Olanzapine , Oxadiazoles/administration & dosage , Piperidines/administration & dosage , Prefrontal Cortex/drug effects , Rats , Rats, Wistar , Risperidone/agonists , Synaptic Transmission/drug effects , Tetrahydronaphthalenes/pharmacology , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/administration & dosage , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/analogs & derivatives , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
18.
Neuropsychopharmacology ; 35(9): 1952-61, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20463659

ABSTRACT

Preclinical data have shown that addition of the selective norepinephrine transporter (NET) inhibitor reboxetine increases the antipsychotic-like effect of the D(2/3) antagonist raclopride and, in parallel, enhances cortical dopamine output. Subsequent clinical results suggested that adding reboxetine to stable treatments with various antipsychotic drugs (APDs) may improve positive, negative and depressive symptoms in schizophrenia. In this study, we investigated in rats the effects of adding reboxetine to the second-generation APD olanzapine on: (i) antipsychotic efficacy, using the conditioned avoidance response (CAR) test, (ii) extrapyramidal side effect (EPS) liability, using a catalepsy test, (iii) dopamine efflux in the medial prefrontal cortex and the nucleus accumbens, using in vivo microdialysis in freely moving animals and (iv) cortical N-methyl-D-aspartate (NMDA) receptor-mediated transmission, using intracellular electrophysiological recording in vitro. Reboxetine (6 mg/kg) enhanced the suppression of CAR induced by a suboptimal dose (1.25 mg/kg), but not an optimal (2.5 mg/kg) dose of olanzapine without any concomitant catalepsy. Addition of reboxetine to the low dose of olanzapine also markedly increased cortical dopamine outflow and facilitated prefrontal NMDA receptor-mediated transmission. Our data suggest that adjunctive treatment with a NET inhibitor may enhance the therapeutic effect of low-dose olanzapine in schizophrenia without increasing EPS liability and add an antidepressant action, thus in principle allowing for a dose reduction of olanzapine with a concomitant reduction of dose-related side effects, such as EPS and weight gain.


Subject(s)
Antipsychotic Agents/pharmacology , Avoidance Learning/drug effects , Benzodiazepines/pharmacology , Cerebral Cortex/drug effects , Dopamine/metabolism , Morpholines/pharmacology , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Behavior, Animal/drug effects , Cerebral Cortex/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Excitatory Amino Acid Agonists/pharmacology , Freezing Reaction, Cataleptic/drug effects , In Vitro Techniques , Male , Membrane Potentials/drug effects , Microdialysis/methods , N-Methylaspartate/pharmacology , Olanzapine , Rats , Rats, Sprague-Dawley , Rats, Wistar , Reboxetine
19.
Vaccine ; 28(10): 2161-2168, 2010 Mar 02.
Article in English | MEDLINE | ID: mdl-20060511

ABSTRACT

The aim of the present study was to synthesise and screen a set of novel nicotine hapten immunogens used for the treatment of nicotine dependence. In the screening process we studied the amount of antibodies generated and their selectivity, using ELISA techniques, and their effects on nicotine-induced dopamine release in the NAC(shell) of the rat, assessed by in vivo voltammetry. We conclude that even small changes such as the linker attachment on the nicotine molecule as well as the structure of the linker may greatly influence the selectivity of the antibodies and the central neurobiological effects of nicotine that are considered critical for its dependence producing properties.


Subject(s)
Drug Evaluation, Preclinical/methods , Haptens/immunology , Nicotine/immunology , Substance-Related Disorders/therapy , Vaccines/therapeutic use , Animals , Antibodies/blood , Dopamine/metabolism , Enzyme-Linked Immunosorbent Assay , Haptens/chemistry , Male , Molecular Structure , Nicotine/chemistry , Rats , Rats, Wistar
20.
Psychopharmacology (Berl) ; 204(2): 251-64, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19198810

ABSTRACT

RATIONALE: Asenapine, a psychopharmacologic agent developed for schizophrenia and bipolar disorder, has higher affinity for 5-HT(2A/C,6,7) and alpha(2) adrenergic receptors than for D(2) receptors. Asenapine exhibits potent antipsychotic-like effects without inducing catalepsy, increases cortical and subcortical dopamine release, and facilitates cortical glutamatergic transmission in rats. In this study, we further analyzed the effects of asenapine on dopaminergic, noradrenergic, and serotonergic systems in the rat brain. MATERIALS AND METHODS: We studied the effects of asenapine on (1) dopaminergic neurons in the ventral tegmental area (VTA) and noradrenergic neurons in the locus coeruleus using in vivo single cell recording, (2) release of dopamine and noradrenaline (medial prefrontal cortex), serotonin (frontal cortex), and dopamine (nucleus accumbens), using in vivo microdialysis. RESULTS: Systemic asenapine increased dopaminergic (0.001-0.2 mg/kg, i.v.) and noradrenergic (0.025-0.05 mg/kg i.v.) neuronal firing, and asenapine (0.1-0.2 mg/kg, s.c) increased cortical noradrenaline and serotonin output. Local asenapine administration increased all three monoamines in the cortex but did not affect accumbal dopamine output. Intra-VTA tetrodotoxin perfusion blocked asenapine-induced accumbal but not cortical dopamine outflow. CONCLUSION: Asenapine at doses associated with antipsychotic activity enhanced cortical monoamine efflux. Whereas the asenapine-induced dopamine increase in nucleus accumbens is dependent on activation of dopaminergic neurons in the VTA, the increase of cortical dopamine outflow involves largely a local action at nerve terminals. Our data provide further insight on the pharmacologic characteristics of asenapine that may have bearing on its clinical efficacy in the treatment of schizophrenia and bipolar disorder.


Subject(s)
Cerebral Cortex/metabolism , Dopamine/metabolism , Dopamine/physiology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Norepinephrine/metabolism , Serotonin/metabolism , Animals , Brain/drug effects , Brain/physiology , Cerebral Cortex/drug effects , Dibenzocycloheptenes , Electrophysiology , Male , Microdialysis , Neurons/drug effects , Neurons/physiology , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Rats , Rats, Sprague-Dawley , Tetrodotoxin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL