Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters










Publication year range
1.
Biomolecules ; 13(4)2023 04 14.
Article in English | MEDLINE | ID: mdl-37189421

ABSTRACT

The noradrenaline system attracts attention for its role in mood disorders and neurodegenerative diseases but the lack of well-validated methods impairs our understanding when assessing its function and release in vivo. This study combines simultaneous positron emission tomography (PET) and microdialysis to explore if [11C]yohimbine, a selective antagonist radioligand of the α2 adrenoceptors, may be used to assess in vivo changes in synaptic noradrenaline during acute pharmacological challenges. Anesthetised Göttingen minipigs were positioned in a head holder in a PET/CT device. Microdialysis probes were placed in the thalamus, striatum and cortex and dialysis samples were collected every 10 min. Three 90 min [11C]yohimbine scans were acquired: at baseline and at two timepoints after the administration of amphetamine (1-10 mg/kg), a non-specific releaser of dopamine and noradrenaline, or nisoxetine (1 mg/kg), a specific noradrenaline transporter inhibitor. [11C]yohimbine volumes of distribution (VT) were obtained using the Logan kinetic model. Both challenges induced a significant decrease in yohimbine VT, with time courses reflecting their different mechanisms of action. Dialysis samples revealed a significant increase in noradrenaline extracellular concentrations after challenge and an inverse correlation with changes in yohimbine VT. These data suggest that [11C]yohimbine can be used to evaluate acute variations in synaptic noradrenaline concentrations after pharmacological challenges.


Subject(s)
Norepinephrine , Positron Emission Tomography Computed Tomography , Animals , Microdialysis , Norepinephrine/metabolism , Positron-Emission Tomography/methods , Renal Dialysis , Swine, Miniature , Yohimbine/metabolism
2.
Transl Psychiatry ; 10(1): 239, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32681022

ABSTRACT

The schizophrenia-associated gene, BRD1, encodes an epigenetic regulator in which chromatin interactome is enriched with genes implicated in mental health. Alterations in histone modifications and epigenetic regulation contribute to brain transcriptomic changes in affective disorders and preclinical data supports a role for BRD1 in psychopathology. However, the implication of BRD1 on affective pathology remains poorly understood. In this study, we assess affective behaviors and associated neurobiology in Brd1+/- mice along with their responses to Fluoxetine and Imipramine. This involves behavioral, neurostructural, and neurochemical characterizations along with regional cerebral gene expression profiling combined with integrative functional genomic analyses. We report behavioral changes in female Brd1+/- mice with translational value to depressive symptomatology that can be alleviated by the administration of antidepressant medications. Behavioral changes are accompanied by altered brain morphometry and imbalances in monoaminergic systems. In accordance, gene expression changes across brain tissues reveal altered neurotransmitter signaling and cluster in functional pathways associated with depression including 'Adrenergic-, GPCR-, cAMP-, and CREB/CREM-signaling'. Integrative gene expression analysis specifically links changes in amygdaloid intracellular signaling activity to the behavioral treatment response in Brd1+/- mice. Collectively, our study highlights the importance of BRD1 as a modulator of affective pathology and adds to our understanding of the molecular mechanisms underlying affective disorders and their treatment response.


Subject(s)
Histone Acetyltransferases , Schizophrenia , Animals , Depression/genetics , Epigenesis, Genetic , Female , Gene Expression , Mice , Schizophrenia/genetics
3.
Nat Commun ; 11(1): 1491, 2020 03 20.
Article in English | MEDLINE | ID: mdl-32198394

ABSTRACT

The serotonin transporter (SERT) terminates serotonin signaling by rapid presynaptic reuptake. SERT activity is modulated by antidepressants, e.g., S-citalopram and imipramine, to alleviate symptoms of depression and anxiety. SERT crystal structures reveal two S-citalopram binding pockets in the central binding (S1) site and the extracellular vestibule (S2 site). In this study, our combined in vitro and in silico analysis indicates that the bound S-citalopram or imipramine in S1 is allosterically coupled to the ligand binding to S2 through altering protein conformations. Remarkably, SERT inhibitor Lu AF60097, the first high-affinity S2-ligand reported and characterized here, allosterically couples the ligand binding to S1 through a similar mechanism. The SERT inhibition by Lu AF60097 is demonstrated by the potentiated imipramine binding and increased hippocampal serotonin level in rats. Together, we reveal a S1-S2 coupling mechanism that will facilitate rational design of high-affinity SERT allosteric inhibitors.


Subject(s)
Allosteric Site/drug effects , Citalopram/pharmacology , Imipramine/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin Plasma Membrane Transport Proteins/metabolism , Allosteric Regulation/drug effects , Allosteric Site/genetics , Animals , Antidepressive Agents/pharmacology , Citalopram/chemistry , Drug Development , Genetic Engineering , Imipramine/chemistry , Ligands , Molecular Docking Simulation , Mutagenesis, Site-Directed , Protein Conformation , Rats , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/genetics
4.
Neuropharmacology ; 128: 379-387, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29104073

ABSTRACT

The antidepressant vortioxetine exerts its effects via modulation of several serotonin (5-HT) receptors and inhibition of the 5-HT transporter (SERT). Additionally, vortioxetine has beneficial effects on aspects of cognitive dysfunction in depressed patients. However, a global examination of the drug effect on brain network connectivity is still missing. Here we compared the effects of vortioxetine and a serotonin norepinephrine reuptake inhibitor, duloxetine, on resting-state functional connectivity (RSFC) across the whole brain in awake rats using a combination of pharmacological and awake animal resting-state functional magnetic resonance imaging (rsfMRI) techniques. Our data showed that vortioxetine and duloxetine affected different inter-areal connections with limited overlap, indicating that in addition to different primary target profiles, these two antidepressants have distinct mechanisms of action at the systems level. Further, our data suggest that vortioxetine can affect specific brain areas with distinct 5-HT receptor expression profiles. Taken together, this study demonstrates that the awake animal fMRI approach provides a powerful tool to elucidate the effects of drugs on the brain with high spatial specificity and a global field of view. This capability is valuable to understand how different drugs affect the systems-level brain function, and provides important guidance to dissect specific brain regions and connections for further detailed mechanistic studies. This study also highlights the translational opportunity of the awake animal fMRI approach between preclinical results and human studies.


Subject(s)
Antidepressive Agents/pharmacology , Brain/drug effects , Duloxetine Hydrochloride/pharmacology , Piperazines/pharmacology , Rest , Sulfides/pharmacology , Wakefulness/drug effects , Animals , Brain/diagnostic imaging , Image Processing, Computer-Assisted , Magnetic Resonance Imaging , Male , Neural Pathways/diagnostic imaging , Neural Pathways/drug effects , Oxygen/blood , Rats , Rats, Long-Evans , Receptor, Serotonin, 5-HT1A/metabolism , Receptor, Serotonin, 5-HT1B , Vortioxetine
5.
Transl Psychiatry ; 7(11): 1261, 2017 11 30.
Article in English | MEDLINE | ID: mdl-29187755

ABSTRACT

1q21.1 hemizygous microdeletion is a copy number variant leading to eightfold increased risk of schizophrenia. In order to investigate biological alterations induced by this microdeletion, we generated a novel mouse model (Df(h1q21)/+) and characterized it in a broad test battery focusing on schizophrenia-related assays. Df(h1q21)/+ mice displayed increased hyperactivity in response to amphetamine challenge and increased sensitivity to the disruptive effects of amphetamine and phencyclidine hydrochloride (PCP) on prepulse inhibition. Probing of the direct dopamine (DA) pathway using the DA D1 receptor agonist SKF-81297 revealed no differences in induced locomotor activity compared to wild-type mice, but Df(h1q21)/+ mice showed increased sensitivity to the DA D2 receptor agonist quinpirole and the D1/D2 agonist apomorphine. Electrophysiological characterization of DA neuron firing in the ventral tegmental area revealed more spontaneously active DA neurons and increased firing variability in Df(h1q21)/+ mice, and decreased feedback reduction of DA neuron firing in response to amphetamine. In a range of other assays, Df(h1q21)/+ mice showed no difference from wild-type mice: gross brain morphology and basic functions such as reflexes, ASR, thermal pain sensitivity, and motor performance were unaltered. Similarly, anxiety related measures, baseline prepulse inhibition, and seizure threshold were unaltered. In addition to the central nervous system-related phenotypes, Df(h1q21)/+ mice exhibited reduced head-to tail length, which is reminiscent of the short stature reported in humans with 1q21.1 deletion. With aspects of both construct and face validity, the Df(h1q21)/+ model may be used to gain insight into schizophrenia-relevant alterations in dopaminergic transmission.


Subject(s)
Abnormalities, Multiple , Behavior, Animal , Chromosome Deletion , Dopamine Agonists/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Dopaminergic Neurons/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Megalencephaly , Nucleus Accumbens/metabolism , Prepulse Inhibition , Receptors, Dopamine/metabolism , Schizophrenia , Ventral Tegmental Area/metabolism , Abnormalities, Multiple/metabolism , Abnormalities, Multiple/pathology , Abnormalities, Multiple/physiopathology , Amphetamine/pharmacology , Animals , Apomorphine/pharmacology , Behavior, Animal/drug effects , Benzazepines/pharmacology , Chromosomes, Human, Pair 1/metabolism , Disease Models, Animal , Dopamine Agonists/administration & dosage , Dopamine Uptake Inhibitors/administration & dosage , Dopaminergic Neurons/drug effects , Excitatory Amino Acid Antagonists/administration & dosage , Megalencephaly/metabolism , Megalencephaly/pathology , Megalencephaly/physiopathology , Mice , Mice, Inbred C57BL , Nucleus Accumbens/drug effects , Phencyclidine/pharmacology , Phenotype , Prepulse Inhibition/drug effects , Quinpirole/pharmacology , Receptors, Dopamine/drug effects , Schizophrenia/metabolism , Schizophrenia/pathology , Schizophrenia/physiopathology , Ventral Tegmental Area/drug effects
6.
Neuropharmacology ; 125: 50-63, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28711518

ABSTRACT

Antagonism of the 5-HT6 receptor is a promising approach for the symptomatic treatment of Alzheimer's disease (AD). There is compelling preclinical evidence for the procognitive potential of 5-HT6 receptor antagonists and several compounds are in clinical development, as adjunct therapy to acetylcholinesterase inhibitors (AChEIs). This manuscript summarizes the scientific rationale for the use of 5-HT6 receptor antagonists as AD treatment, with some focus on the selective and high-affinity 5-HT6 receptor antagonist idalopirdine (Lu AE58054). The 5-HT6 receptor is enriched in brain regions that mediate cognition, where expression predominates on glutamatergic and GABAergic neurons and subsets of GABAergic interneurons. It is proposed that 5-HT6 receptor antagonism modulates the balance between neuronal excitation (glutamate) and inhibition (GABA), which may have widespread implications for neurotransmission and neuronal activity. This is supported by preclinical studies showing that 5-HT6 receptor antagonists increase concentrations of multiple neurotransmitters, and strengthened by recent evidence that idalopirdine facilitates neuronal oscillations and contributes to the recruitment of several neuronal networks relevant in cognition. Some of these effects are observed with idalopirdine monotherapy, whereas others require concomitant treatment with an AChEI. Several hypotheses for the mechanism underlying the synergistic actions between 5-HT6 receptor antagonists and AChEIs are discussed. Collectively, the current evidence suggests that 5-HT6 receptor antagonism adds a unique, complementary mechanism of action to that of AChEIs. The facilitation of multiple neurotransmitters and neuronal activity in brain regions that mediate cognition, and the synergy with AChEIs, are proposed to mediate the procognitive effects of 5-HT6 receptor antagonists in AD patients.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Psychotropic Drugs/pharmacology , Receptors, Serotonin/metabolism , Serotonin Antagonists/pharmacology , Animals , Brain/drug effects , Brain/metabolism , Cognition/drug effects , Cognition/physiology , Humans
7.
Eur J Pharmacol ; 799: 1-6, 2017 Mar 15.
Article in English | MEDLINE | ID: mdl-28188762

ABSTRACT

Idalopirdine (Lu AE58054) is a high affinity and selective antagonist for the human serotonin 5-HT6 receptor (Ki 0.83nM) in phase III development for mild-to-moderate Alzheimer's disease as an adjunct therapy to acetylcholinesterase inhibitors (AChEIs). We have studied the effects of idalopirdine on extracellular levels of monoamines, glutamate and acetylcholine in the medial prefrontal cortex (mPFC) of freely-moving rats using microdialysis. Idalopirdine (10mg/kg p.o.) increased extracellular levels of dopamine, noradrenaline and glutamate in the mPFC and showed a trend to increase serotonin levels. No effect was observed on acetylcholine levels. The AChEI donepezil (1.3mg/kg s.c.) significantly increased the levels of acetylcholine. Pretreatment with idalopirdine 2h prior to donepezil administration potentiated the effect of donepezil on extracellular acetylcholine levels. The idalopirdine potentiation of donepezil-induced increase in acetylcholine levels was also observed during local infusion of idalopirdine (6µg/ml) into the mPFC by reverse dialysis. The data from the current study may provide a mechanistic model for the pro-cognitive effects observed with administration of idalopirdine in donepezil-treated patients with Alzheimer's disease observed in the phase 2 studies (Wilkinson et al. 2014).


Subject(s)
Acetylcholine/metabolism , Benzylamines/pharmacology , Biogenic Monoamines/metabolism , Extracellular Space/drug effects , Glutamic Acid/metabolism , Indoles/pharmacology , Prefrontal Cortex/cytology , Receptors, Serotonin/metabolism , Animals , Extracellular Space/metabolism , Male , Rats , Rats, Sprague-Dawley , Serotonin Antagonists/pharmacology
8.
Pharmacol Biochem Behav ; 153: 141-146, 2017 02.
Article in English | MEDLINE | ID: mdl-28057524

ABSTRACT

Previous studies have shown that partial and full 5-HT1A receptor agonists reduce antipsychotic-induced catalepsy. Consequently, some antipsychotics combining balanced efficacy between dopamine (DA) D2 antagonism or partial agonism and 5-HT1A receptor agonism have a low propensity to induce extrapyramidal side effects (EPS), as reflected by low cataleptogenic activity in rodents. In the present experiments, we attempted to explore the importance of pre- and postsynaptic 5-HT1A agonistic properties of brexpiprazole and aripiprazole in the context of neurological side-effect liabilities. Additional measures of prefrontal cortical serotonin (5-HT) and DA levels using microdialysis were used to support that brexpiprazole has a preferential agonist effect on presynaptic 5-HT1A receptors. Brexpiprazole (3.0 and 10mg/kg, p.o.) as well as aripiprazole (8.0 and 30mg/kg, p.o.) failed to induce catalepsy in rats. Brexpiprazole (10mg/kg, p.o.) significantly reduced the cataleptic response induced by haloperidol (0.63mg/kg, s.c.), while aripiprazole (1.0-100mg/kg, p.o.) failed to reverse the effect of haloperidol and only showed a numeric decrease at 10mg/kg, (p.o.). When 5-HT1A receptors were blocked by the selective antagonist, WAY100635 (1.0mg/kg, s.c.), cataleptogenic properties of brexpiprazole (10mg/kg; p.o), but not aripiprazole (8.0 and 30mg/kg, p.o.) were unmasked. The ("biased") 5-HT1A receptor agonists F15599 (postsynaptic preference) and F13714 (presynaptic preference) had differential effects on haloperidol-induced catalepsy: F13714 (0.16mg/kg, s.c.) counteracted catalepsy, whereas F15599 (0.040mg/kg, s.c.) had no significant effect at regionally-selective doses. These data support a role of presynaptic 5-HT1A receptors in the anticataleptic effect of brexpiprazole. The selective 5-HT2A antagonist M100907 (0.10mg/kg, s.c.) had no effect on haloperidol-induced catalepsy, arguing against a major role of 5-HT2A receptors in the cataleptogenic profile of brexpiprazole. The findings with brexpiprazole were supported using microdialysis studies: Brexpiprazole (3.0 and 10mg/kg, p.o.) decreased extracellular 5-HT levels in the medial prefrontal cortex (mPFC), while it failed to affect extracellular DA in the same samples, suggesting that the 5-HT1A agonist properties of brexpiprazole may be preferentially presynaptic. In conclusion, these results confirm that brexpiprazole and aripiprazole have low propensities to induce EPS. However, the low EPS risk of brexpiprazole is more likely dependent on its agonist properties on presynaptic 5-HT1A receptors, while that of aripiprazole is less sensitive to 5-HT1A receptor antagonism.


Subject(s)
Basal Ganglia Diseases/chemically induced , Quinolones/toxicity , Receptor, Serotonin, 5-HT1A/physiology , Thiophenes/toxicity , Animals , Aripiprazole/toxicity , Catalepsy/chemically induced , Dopamine/analysis , Haloperidol/pharmacology , Male , Piperazines/pharmacology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Serotonin/analysis
9.
Biol Psychiatry ; 82(1): 62-76, 2017 07 01.
Article in English | MEDLINE | ID: mdl-27837920

ABSTRACT

BACKGROUND: The schizophrenia-associated BRD1 gene encodes a transcriptional regulator whose comprehensive chromatin interactome is enriched with schizophrenia risk genes. However, the biology underlying the disease association of BRD1 remains speculative. METHODS: This study assessed the transcriptional drive of a schizophrenia-associated BRD1 risk variant in vitro. Accordingly, to examine the effects of reduced Brd1 expression, we generated a genetically modified Brd1+/- mouse and subjected it to behavioral, electrophysiological, molecular, and integrative genomic analyses with focus on schizophrenia-relevant parameters. RESULTS: Brd1+/- mice displayed cerebral histone H3K14 hypoacetylation and a broad range of behavioral changes with translational relevance to schizophrenia. These behaviors were accompanied by striatal dopamine/serotonin abnormalities and cortical excitation-inhibition imbalances involving loss of parvalbumin immunoreactive interneurons. RNA-sequencing analyses of cortical and striatal micropunches from Brd1+/- and wild-type mice revealed differential expression of genes enriched for schizophrenia risk, including several schizophrenia genome-wide association study risk genes (e.g., calcium channel subunits [Cacna1c and Cacnb2], cholinergic muscarinic receptor 4 [Chrm4)], dopamine receptor D2 [Drd2], and transcription factor 4 [Tcf4]). Integrative analyses further found differentially expressed genes to cluster in functional networks and canonical pathways associated with mental illness and molecular signaling processes (e.g., glutamatergic, monoaminergic, calcium, cyclic adenosine monophosphate [cAMP], dopamine- and cAMP-regulated neuronal phosphoprotein 32 kDa [DARPP-32], and cAMP responsive element binding protein signaling [CREB]). CONCLUSIONS: Our study bridges the gap between genetic association and pathogenic effects and yields novel insights into the unfolding molecular changes in the brain of a new schizophrenia model that incorporates genetic risk at three levels: allelic, chromatin interactomic, and brain transcriptomic.


Subject(s)
Behavior, Animal/physiology , Gene Expression/genetics , Histone Acetyltransferases/physiology , Schizophrenia/genetics , Synaptic Transmission/genetics , Acetylation , Animals , Animals, Genetically Modified/genetics , Corpus Striatum/metabolism , Dopamine/metabolism , Histone Acetyltransferases/genetics , Histones/metabolism , Interneurons/physiology , Mice , Serotonin/metabolism
10.
Pharmacol Biochem Behav ; 150-151: 147-152, 2016.
Article in English | MEDLINE | ID: mdl-27984094

ABSTRACT

The goal of the present work was to characterise the effects of selegiline on the rat sleep pattern. Furthermore, for comparative purposes, the pharmacokinetics of selegiline and its metabolites in brain and plasma were investigated, and microdialysis experiments were performed to examine the resulting effect on dopamine, noradrenaline and serotonin levels. Selegiline (1, 5, 10 and 30mg/kg) was found to dose-dependently increase the time spent awake following acute dosing. The pharmacokinetic assessment of selegiline showed that, following an oral dose of 5mg/kg, low circulating levels of the parent compound were found relative to those of biotransformed l-methamphetamine and l-amphetamine. The time course of selegiline-induced wakefulness was shown to follow the time course of l-methamphetamine and l-amphetamine in brain, suggesting that these metabolites are responsible for the modulation of sleep architecture. Furthermore, selegiline (5mg/kg) caused a significant increase of extracellular levels of DA (250%) and NA (200%), but not of 5-HT, in the rat prefrontal cortex. In summary, an integrated experimental approach was undertaken here to evaluate selegiline's effect on sleep architecture in rats in relation to its pharmacokinetics and changes in monoaminergic neurotransmitter levels in the brain. The effect of selegiline on sleep was likely mediated by an increase of dopamine and noradrenaline levels in the brain caused by the formed metabolites.


Subject(s)
Monoamine Oxidase Inhibitors/pharmacology , Selegiline/pharmacology , Wakefulness/drug effects , Animals , Biotransformation , Dopamine/analysis , Dose-Response Relationship, Drug , Male , Microdialysis , Norepinephrine/analysis , Prefrontal Cortex/chemistry , Rats , Rats, Sprague-Dawley , Selegiline/metabolism , Sleep/drug effects
11.
J Pharmacol Exp Ther ; 358(3): 472-82, 2016 09.
Article in English | MEDLINE | ID: mdl-27402279

ABSTRACT

Major depressive disorder (MDD) is a common psychiatric disorder that often features impairments in cognitive function, and these cognitive symptoms can be important determinants of functional ability. Vortioxetine is a multimodal antidepressant that may improve some aspects of cognitive function in patients with MDD, including attention, processing speed, executive function, and memory. However, the cause of these effects is unclear, and there are several competing theories on the underlying mechanism, notably including regionally-selective downstream enhancement of glutamate neurotransmission and increased acetylcholine (ACh) neurotransmission. The current work sought to evaluate the ACh hypothesis by examining vortioxetine's ability to reverse scopolamine-induced impairments in rodent tests of memory and attention. Additionally, vortioxetine's effects on hippocampal extracellular ACh levels were examined alongside studies of vortioxetine's pharmacokinetic profile. We found that acute vortioxetine reversed scopolamine-induced impairments in social and object recognition memory, but did not alter scopolamine-induced impairments in attention. Acute vortioxetine also induced a modest and short-lived increase in hippocampal ACh levels. However, this short-term effect is at variance with vortioxetine's moderately long brain half life (5.1 hours). Interestingly, subchronic vortioxetine treatment failed to reverse scopolamine-induced social recognition memory deficits and had no effects on basal hippocampal ACh levels. These data suggest that vortioxetine has some effects on memory that could be mediated through cholinergic neurotransmission, however these effects are modest and only seen under acute dosing conditions. These limitations may argue against cholinergic mechanisms being the primary mediator of vortioxetine's cognitive effects, which are observed under chronic dosing conditions in patients with MDD.


Subject(s)
Acetylcholine/metabolism , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/drug therapy , Extracellular Space/drug effects , Hippocampus/pathology , Piperazines/pharmacology , Scopolamine/pharmacology , Sulfides/pharmacology , Animals , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/physiopathology , Extracellular Space/metabolism , Hippocampus/drug effects , Hippocampus/physiopathology , Male , Piperazines/therapeutic use , Rats , Rats, Wistar , Recognition, Psychology/drug effects , Social Behavior , Sulfides/therapeutic use , Synaptic Transmission/drug effects , Vortioxetine
12.
Neuropharmacology ; 107: 351-363, 2016 08.
Article in English | MEDLINE | ID: mdl-27039041

ABSTRACT

The 5-HT6 receptor has emerged as a promising target for cognitive disorders and combining a 5-HT6 receptor antagonist with an acetylcholinesterase inhibitor (AChEI) represents a novel approach for the symptomatic treatment of Alzheimer's disease (AD). A recent phase 2 trial showed that the selective 5-HT6 receptor antagonist idalopirdine (Lu AE58054) improved cognition in patients with moderate AD on stable treatment with the AChEI donepezil. Here we investigated the effects of idalopirdine in combination with donepezil on hippocampal function using in vivo electrophysiology and microdialysis. Network oscillations in the hippocampus were recorded during electrical stimulation of the brainstem nucleus pontis oralis (nPO) in the anesthetized rat and hippocampal acetylcholine (ACh) levels were measured in the freely-moving rat. In addition, potential pharmacokinetic interactions between idalopirdine and donepezil were assessed. Idalopirdine alone did not affect hippocampal network oscillations or ACh levels. Donepezil (0.3 and 1.0 mg/kg i.v.) dose-dependently increased hippocampal theta and gamma power during nPO stimulation. Idalopirdine (2 mg/kg i.v.), administered 1 h prior to donepezil, potentiated the theta and gamma response to 0.3 mg/kg donepezil and prolonged the gamma response to 1 mg/kg donepezil. Donepezil (1.3 mg/kg s.c.) increased extracellular ACh levels in the hippocampus and this was further augmented by administration of idalopirdine (10 mg/kg p.o.) 2 h prior to donepezil. These effects could not be attributed to a pharmacokinetic interaction between the compounds. This study demonstrates that idalopirdine potentiates the effects of donepezil on two pharmacodynamic biomarkers associated with cognition, i.e. neuronal oscillations and extracellular ACh levels in the hippocampus. Such potentiation could contribute to the procognitive effects of idalopirdine observed in donepezil-treated AD patients.


Subject(s)
Acetylcholine/metabolism , Benzylamines/administration & dosage , Brain Waves/physiology , Cholinesterase Inhibitors/administration & dosage , Hippocampus/metabolism , Indoles/administration & dosage , Receptors, Serotonin/metabolism , Animals , Biological Clocks/drug effects , Biological Clocks/physiology , Brain Waves/drug effects , Extracellular Fluid/drug effects , Extracellular Fluid/metabolism , Hippocampus/drug effects , Male , Nerve Net/drug effects , Nerve Net/metabolism , Rats , Rats, Sprague-Dawley , Serotonin Antagonists/pharmacology
13.
Pharm Res ; 33(5): 1133-43, 2016 May.
Article in English | MEDLINE | ID: mdl-26780452

ABSTRACT

PURPOSE: Gabapentin displays non-linear drug disposition, which complicates dosing for optimal therapeutic effect. Thus, the current study was performed to elucidate the pharmacokinetic/pharmacodynamic (PKPD) relationship of gabapentin's effect on mechanical hypersensitivity in a rat model of CFA-induced inflammatory hyperalgesia. METHODS: A semi-mechanistic population-based PKPD model was developed using nonlinear mixed-effects modelling, based on gabapentin plasma and brain extracellular fluid (ECF) time-concentration data and measurements of CFA-evoked mechanical hyperalgesia following administration of a range of gabapentin doses (oral and intravenous). RESULTS: The plasma/brain ECF concentration-time profiles of gabapentin were adequately described with a two-compartment plasma model with saturable intestinal absorption rate (K m = 44.1 mg/kg, V max = 41.9 mg/h∙kg) and dose-dependent oral bioavailability linked to brain ECF concentration through a transit compartment. Brain ECF concentration was directly linked to a sigmoid E max function describing reversal of hyperalgesia (EC 50, plasma = 16.7 µg/mL, EC 50, brain = 3.3 µg/mL). CONCLUSIONS: The proposed semi-mechanistic population-based PKPD model provides further knowledge into the understanding of gabapentin's non-linear pharmacokinetics and the link between plasma/brain disposition and anti-hyperalgesic effects. The model suggests that intestinal absorption is the primary source of non-linearity and that the investigated rat model provides reasonable predictions of clinically effective plasma concentrations for gabapentin.


Subject(s)
Amines/pharmacology , Amines/pharmacokinetics , Analgesics/pharmacology , Analgesics/pharmacokinetics , Cyclohexanecarboxylic Acids/pharmacology , Cyclohexanecarboxylic Acids/pharmacokinetics , Hyperalgesia/drug therapy , gamma-Aminobutyric Acid/pharmacology , gamma-Aminobutyric Acid/pharmacokinetics , Amines/administration & dosage , Amines/therapeutic use , Analgesics/administration & dosage , Analgesics/therapeutic use , Animals , Biological Availability , Brain/metabolism , Computer Simulation , Cyclohexanecarboxylic Acids/administration & dosage , Cyclohexanecarboxylic Acids/therapeutic use , Gabapentin , Hyperalgesia/metabolism , Male , Models, Biological , Rats , Rats, Sprague-Dawley , gamma-Aminobutyric Acid/administration & dosage , gamma-Aminobutyric Acid/therapeutic use
14.
Neuropsychopharmacology ; 41(2): 638-45, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26129678

ABSTRACT

Sensory gating is the brain's ability to filter out irrelevant information before it reaches high levels of conscious processing. In the current study we aimed to investigate the involvement of the noradrenergic and dopaminergic neurotransmitter systems in sensory gating. Furthermore, we investigated cross-species reliability by comparing effects in both healthy humans and rats, while keeping all experimental conditions as similar as possible between the species. The design of the human experiment (n=21) was a double-blind, placebo-controlled, cross-over study where sensory gating was assessed following a dose of either reboxetine (8 mg), haloperidol (2 mg), their combination or placebo at four separate visits. Similarly in the animal experiment sensory gating was assessed in rats, (n=22) following a dose of reboxetine (2 mg/kg), haloperidol (0.08 mg/kg), their combination or placebo. The sensory gating paradigms in both experiments were identical. In humans, we found significantly reduced P50 suppression following separate administration of reboxetine or haloperidol, while their combined administration did not reach statistical significance compared with placebo. In the rats, we found a similar significant reduction of sensory gating (N40) following treatment with haloperidol and the combination of haloperidol and reboxetine, but not with separate reboxetine treatment, compared with placebo. Our study indicates that even when experimental conditions are kept as similar as possible, direct human to rat cross-species translation of pharmacological effects on sensory gating is challenging, which calls for more focussed research in this important translational area.


Subject(s)
Adrenergic Uptake Inhibitors/administration & dosage , Dopamine Antagonists/administration & dosage , Haloperidol/administration & dosage , Morpholines/administration & dosage , Sensory Gating/drug effects , Sensory Gating/physiology , Administration, Oral , Adolescent , Adult , Animals , Auditory Perception/drug effects , Auditory Perception/physiology , CA3 Region, Hippocampal/drug effects , CA3 Region, Hippocampal/physiology , Cross-Over Studies , Double-Blind Method , Evoked Potentials, Auditory/drug effects , Evoked Potentials, Auditory/physiology , Humans , Male , Rats , Reboxetine , Species Specificity , Young Adult
15.
J Alzheimers Dis ; 42(4): 1179-89, 2014.
Article in English | MEDLINE | ID: mdl-25024305

ABSTRACT

P300 (P3) event-related potentials (ERPs) have been suggested to be an endogenous marker of cognitive function and auditory oddball paradigms are frequently used to evaluate P3 ERPs in clinical settings. Deficits in P3 amplitude and latency reflect some of the neurological dysfunctions related to several psychiatric and neurological diseases, e.g., Alzheimer's disease (AD). However, only a very limited number of rodent studies have addressed the back-translational validity of the P3-like ERPs as suitable markers of cognition. Thus, the potential of rodent P3-like ERPs to predict pro-cognitive effects in humans remains to be fully validated. The current study characterizes P3-like ERPs in the 192-IgG-SAP (SAP) rat model of the cholinergic degeneration associated with AD. Following training in a combined auditory oddball and lever-press setup, rats were subjected to bilateral intracerebroventricular infusion of 1.25 µg SAP or PBS (sham lesion) and recording electrodes were implanted in hippocampal CA1. Relative to sham-lesioned rats, SAP-lesioned rats had significantly reduced amplitude of P3-like ERPs. P3 amplitude was significantly increased in SAP-treated rats following pre-treatment with 1 mg/kg donepezil. Infusion of SAP reduced the hippocampal choline acetyltransferase activity by 75%. Behaviorally defined cognitive performance was comparable between treatment groups. The present study suggests that AD-like deficits in P3-like ERPs may be mimicked by the basal forebrain cholinergic degeneration induced by SAP. SAP-lesioned rats may constitute a suitable model to test the efficacy of pro-cognitive substances in an applied experimental setup.


Subject(s)
Alzheimer Disease/physiopathology , CA1 Region, Hippocampal/physiopathology , Cholinergic Neurons/physiology , Evoked Potentials, Auditory/physiology , Nerve Degeneration/physiopathology , Alzheimer Disease/drug therapy , Animals , Antibodies, Monoclonal , Auditory Perception/drug effects , Auditory Perception/physiology , CA1 Region, Hippocampal/drug effects , Choline O-Acetyltransferase/metabolism , Cholinergic Neurons/drug effects , Disease Models, Animal , Donepezil , Electrodes, Implanted , Evoked Potentials, Auditory/drug effects , Indans/pharmacology , Male , Nerve Degeneration/drug therapy , Neuroprotective Agents/pharmacology , Neuropsychological Tests , Piperidines/pharmacology , Psychomotor Performance/drug effects , Psychomotor Performance/physiology , Rats, Sprague-Dawley , Ribosome Inactivating Proteins, Type 1 , Saporins
16.
J Pharmacol Exp Ther ; 350(3): 589-604, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24947465

ABSTRACT

Brexpiprazole (OPC-34712, 7-{4-[4-(1-benzothiophen-4-yl)piperazin-1-yl]butoxy}quinolin-2(1H)-one) is a novel drug candidate in clinical development for psychiatric disorders with high affinity for serotonin, dopamine, and noradrenaline receptors. In particular, it bound with high affinity (Ki < 1 nM) to human serotonin 1A (h5-HT1A)-, h5-HT2A-, long form of human D2 (hD2L)-, hα1B-, and hα2C-adrenergic receptors. It displayed partial agonism at h5-HT1A and hD2 receptors in cloned receptor systems and potent antagonism of h5-HT2A receptors and hα1B/2C-adrenoceptors. Brexpiprazole also had affinity (Ki < 5 nM) for hD3-, h5-HT2B-, h5-HT7-, hα1A-, and hα1D-adrenergic receptors, moderate affinity for hH1 (Ki = 19 nM), and low affinity for hM1 receptors (Ki > 1000 nM). Brexpiprazole potently bound to rat 5-HT2A and D2 receptors in vivo, and ex vivo binding studies further confirmed high 5-HT1A receptor binding potency. Brexpiprazole inhibited DOI (2,5-dimethoxy-4-iodoamphetamine)-induced head twitches in rats, suggestive of 5-HT2A antagonism. Furthermore, in vivo D2 partial agonist activity of brexpiprazole was confirmed by its inhibitory effect on reserpine-induced DOPA accumulation in rats. In rat microdialysis studies, brexpiprazole slightly reduced extracellular dopamine in nucleus accumbens but not in prefrontal cortex, whereas moderate increases of the dopamine metabolites, homovanillic acid and DOPAC (3,4-dihydroxy-phenyl-acetic acid), in these areas also suggested in vivo D2 partial agonist activity. In particular, based on a lower intrinsic activity at D2 receptors and higher binding affinities for 5-HT1A/2A receptors than aripiprazole, brexpiprazole would have a favorable antipsychotic potential without D2 receptor agonist- and antagonist-related adverse effects. In conclusion, brexpiprazole is a serotonin-dopamine activity modulator with a unique pharmacology, which may offer novel treatment options across a broad spectrum of central nervous system disorders.


Subject(s)
Dopamine Agents/chemistry , Dopamine Agents/metabolism , Dopamine/metabolism , Quinolones/chemistry , Quinolones/metabolism , Serotonin Agents/chemistry , Serotonin Agents/metabolism , Serotonin/metabolism , Thiophenes/chemistry , Thiophenes/metabolism , Animals , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Dopamine D2 Receptor Antagonists , Dose-Response Relationship, Drug , Humans , Male , Protein Binding/physiology , Quinolones/pharmacology , Rats , Rats, Wistar , Receptor, Serotonin, 5-HT1A/metabolism , Receptors, Dopamine D2/agonists , Receptors, Dopamine D2/metabolism , Thiophenes/pharmacology
17.
Behav Brain Res ; 266: 85-93, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24613239

ABSTRACT

Patients with schizophrenia exhibit disturbances in information processing. These disturbances can be investigated with different paradigms of auditory event related potentials (ERP), such as sensory gating in a double click paradigm (P50 suppression) and the mismatch negativity (MMN) component in an auditory oddball paradigm. The aim of the current study was to test if rats subjected to social isolation, which is believed to induce some changes that mimic features of schizophrenia, displays alterations in sensory gating and MMN-like response. Male Lister-Hooded rats were separated into two groups; one group socially isolated (SI) for 8 weeks and one group housed (GH). Both groups were then tested in a double click sensory gating paradigm and an auditory oddball paradigm (MMN-like) paradigm. It was observed that the SI animals showed reduced sensory gating of the cortical N1 amplitude. Furthermore, the SI animals showed significant reduction in cortical MMN-like response compared with the GH animals. No deficits in sensory gating or MMN-like response were observed in the hippocampus (CA3) of the SI animals compared with GH animals. In conclusion, the change in sensory gating of the N1 amplitude supports previous findings in SI rats and the reduced MMN-like response is similar to the deficits of MMN seen in patients with schizophrenia. Since reduced auditory MMN amplitude is believed to be more selectively associated with schizophrenia than other measures of sensory gating deficits, the current study supports the face validity of the SI reared rat model for schizophrenia.


Subject(s)
Contingent Negative Variation/physiology , Evoked Potentials, Auditory/physiology , Schizophrenia/physiopathology , Sensory Gating/physiology , Social Isolation/psychology , Acoustic Stimulation , Analysis of Variance , Animals , Disease Models, Animal , Electroencephalography , Male , Motor Activity , Rats , Schizophrenic Psychology
18.
Curr Top Behav Neurosci ; 18: 123-59, 2014.
Article in English | MEDLINE | ID: mdl-24633891

ABSTRACT

Exposure to stressful situations may induce or deteriorate an already existing depression. Stress-related depression can be elicited at an adolescent/adult age but evidence also shows that early adverse experiences even at the fetal stage may predispose the offspring for later development of depression. The hypothalamus-pituitary-adrenal axis (HPA-axis) plays a key role in regulating the stress response and dysregulation in the system has been linked to depression both in humans and in animal models. This chapter critically reviews clinical and preclinical findings that may explain how stress can cause depression, including HPA-axis changes and alterations beyond the HPA-axis. As stress does not elicit depression in the majority of the population, this motivated research to focus on understanding the biology underlying resilient versus sensitive subjects. Animal models of depression have contributed to a deeper understanding of these mechanisms. Findings from these models will be presented.

19.
Basic Clin Pharmacol Toxicol ; 114(6): 451-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24314270

ABSTRACT

DOV 216,303, an inhibitor of serotonin, noradrenaline and dopamine reuptake, belongs to a new line of drugs called 'triple reuptake inhibitors' that have been proposed for treatment of depression. The addictive drug cocaine has similar mechanism of action and exerts rewarding effects by blocking reuptake of dopamine, leading to increased extracellular concentrations of dopamine in the nucleus accumbens. Thus, DOV 216,303 and other triple reuptake inhibitors might be speculated to exhibit abuse potential, limiting their future therapeutic use. To further elucidate potential addictive properties of DOV 216,303, we conducted a comparative study of addiction-related effects of DOV 216,303 and cocaine in mice using acute self-administration, conditioned place preference (CPP) and drug-induced hyperlocomotion. Effects on accumbal extracellular dopamine levels were determined using microdialysis, and we measured monoamine receptor occupancy as well as brain and plasma exposure. DOV 216,303 was self-administered acutely in the same dose range as cocaine. However, in the CPP model, DOV 216,303 did not induce place preference at doses where cocaine caused place preference. Higher doses of DOV 216,303 than cocaine were needed to induce hyperlocomotion and increase extracellular accumbal dopamine with effective doses being higher than effective doses used in depression models. Moreover, DOV 216,303 displayed a pharmacokinetic profile with lower potential for addiction than cocaine. Thus, high levels of DAT occupancy were reached slower and decayed more slowly after DOV 216,303 than cocaine administration. The present study shows that acute administration of DOV 216,303 displays some addictive-like properties in mice, but these were less pronounced than cocaine, most likely due to different pharmacokinetic profiles.


Subject(s)
Aza Compounds/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cocaine/pharmacology , Substance-Related Disorders/etiology , Animals , Conditioning, Psychological , Dopamine/analysis , Dopamine Plasma Membrane Transport Proteins/metabolism , Male , Mice , Motor Activity/drug effects , Nucleus Accumbens/metabolism , Self Administration , Serotonin Plasma Membrane Transport Proteins/metabolism
20.
Brain Res ; 1543: 253-62, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24231553

ABSTRACT

The Alzheimer's disease (AD) mouse model Tg2576 overexpresses an AD associated mutant variant of human APP and accumulates amyloid beta (Aß) in an age-dependent manner. Using the selective cholinergic immunotoxin mu p75-saporin (SAP), we induced a partial basal forebrain cholinergic degeneration (BFCD) in 3 months old male Tg2576 mice to co-express cholinergic degeneration with Aß overexpression as these characteristics constitutes key hallmarks of AD. At 9 months, SAP lesioned Tg2576 mice were cognitively impaired in two spatial paradigms addressing working memory and mid to long-term memory. Conversely, there was no deterioration of cognitive functioning in sham lesioned Tg2576 mice or wild type littermates (wt) receiving the immunotoxin. At 10 months of age, release of acetylcholine (ACh) was addressed by microdialysis in conscious mice. Scopolamine-induced increases in hippocampal ACh efflux was significantly reduced in SAP lesioned Tg2576 mice compared to sham lesioned Tg2576 mice. Intriguingly, there was no significant difference in ACh efflux between wt treatment groups. Following SAP treatment, choline acetyltransferase activity was reduced in the hippocampus and frontal cortex and the reduction was comparable between groups. Our results suggest that partial BFCD acts collectively with increased levels of Aß to induce cognitive decline and to compromise cholinergic release. Tg2576 mice with BFCD may constitute a new and suitable AD mouse model to study the interrelations between cholinergic deficits and amsyloid deposition.


Subject(s)
Acetylcholine/metabolism , Cholinergic Neurons/pathology , Hippocampus/metabolism , Memory Disorders/pathology , Nerve Degeneration/etiology , Prosencephalon/pathology , Alzheimer Disease/complications , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Choline O-Acetyltransferase/metabolism , Disease Models, Animal , Hippocampus/drug effects , Humans , Immunotoxins/pharmacology , Maze Learning , Memory Disorders/etiology , Mice , Mice, Transgenic , Presenilin-1/genetics , Recognition, Psychology/physiology , Ribosome Inactivating Proteins, Type 1/pharmacology , Saporins
SELECTION OF CITATIONS
SEARCH DETAIL
...