Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
Add more filters










Publication year range
1.
Psychopharmacology (Berl) ; 240(8): 1789-1804, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37394539

ABSTRACT

BACKGROUND: Major depressive disorder (MDD) is defined as a complex mental disorder which is characterized by a pervasive low mood and aversion to activity. Several types of neurotransmitter systems e.g. serotonergic, glutamatergic and noradrenergic systems have been suggested to play an important role in the origination of depression, but neurotrophins such as brain derived neurotrophic factor (BDNF) have also been implicated in the disease process. OBJECTIVES: The purpose of this study was to examine the effects of a newly developed class of molecules, characterized as positive allosteric modulators of neurotrophin/Trk receptor mediated signaling (Trk-PAM), on neurotransmitter release and depression-like behavior in vivo. METHODS: The effect of and possible interaction of neurotrophin/Trk signaling pathways with serotonergic and glutamatergic systems in the modulation of depression-related responses was studied using newly developed Trk-PAM compounds (ACD855, ACD856 and AC26845), as well as ketamine and fluoxetine in the forced swim test (FST) in rodents. Moreover, in vivo microdialysis in freely moving rats was used to assess changes in neurotransmitter levels in the rat. RESULTS: The results from the study show that several different compounds, which all potentiate Trk-receptor mediated signaling, display antidepressant-like activity in the FST. Moreover, the data also indicate that the effects of both fluoxetine and ketamine in the FST, both used in clinical practice, are mediated via BDNF/TrkB signaling, which could have implications for novel therapies in MDD. CONCLUSIONS: Trk-PAMs could provide an interesting avenue for the development of novel therapeutics in this area.


Subject(s)
Depressive Disorder, Major , Ketamine , Rats , Animals , Fluoxetine/pharmacology , Brain-Derived Neurotrophic Factor/metabolism , Depressive Disorder, Major/drug therapy , Ketamine/pharmacology , Antidepressive Agents/pharmacology , Receptor, trkB/metabolism
2.
Behav Brain Res ; 405: 113178, 2021 05 07.
Article in English | MEDLINE | ID: mdl-33607166

ABSTRACT

There is evidence that interaction between the neuropeptide galanin and the 5-HT1A receptor represents an integrative mechanism in the regulation of serotonergic neurotransmission. Thus, in rats intracerebroventricular (i.c.v.) galanin did not impair retention in the passive avoidance (PA) test 24 h after training, but attenuated the retention deficit caused by subcutaneous (s.c.) administration of the 5-HT1A receptor agonist 8-OH-DPAT. This impairment has been linked to postsynaptic 5-HT1A receptor activation. To confirm these results in mice, galanin was infused i.c.v. (1 nmol/mouse) in C57BL/6/Bkl mice 30 min prior to training followed by s.c. injection (0.3 mg/kg) of 8-OH-DPAT or saline 15 min before PA training. In line with previous results, i.c.v. galanin significantly attenuated the PA impairment caused by 5-HT1A receptor activation in mice. To study if the galanin 5-HT1A receptor interaction involved the dorsal hippocampus, galanin (1 nmol/mouse) was directly infused into this brain region alone or in combination with s.c. 8-OH-DPAT. However, unlike i.c.v. galanin, galanin infusion into the dorsal hippocampus alone impaired PA retention and failed to attenuate the 8-OH-DPAT-mediated PA impairment. These results indicate that the ability of i.c.v. galanin to modify 5-HT1A receptor activation is not directly mediated via receptor interactions in the dorsal hippocampus. Instead, the galanin-mediated PA impairment suggests an important inhibitory role of galanin receptors in the dorsal hippocampus for acquisition (encoding) and/or consolidation of emotional memory. In addition, the interaction between galanin and 5-HT1A receptors probably involves a wide serotonergic network that is important for the integration of emotional and cognitive behaviors.


Subject(s)
Emotions , Galanin/pharmacology , Hippocampus/drug effects , Memory Consolidation/drug effects , Memory/drug effects , Receptor, Serotonin, 5-HT1A/drug effects , Animals , Avoidance Learning/drug effects , Behavior, Animal/drug effects , Galanin/administration & dosage , Male , Mice, Inbred C57BL
3.
Biosci Rep ; 40(1)2020 01 31.
Article in English | MEDLINE | ID: mdl-31919522

ABSTRACT

Adult neurogenesis, the production of newborn neurons from neural stem cells (NSCs) has been suggested to be decreased in patients with schizophrenia. A similar finding was observed in an animal model of schizophrenia, as indicated by decreased bromodeoxyuridine (BrdU) labelling cells in response to a non-competitive N-methyl-d-aspartate (NMDA) receptor antagonist. The antipsychotic drug clozapine was shown to counteract the observed decrease in BrdU-labelled cells in hippocampal dentate gyrus (DG). However, phenotypic determination by immunohistochemistry analysis could not reveal whether BrdU-positive cells were indeed NSCs. Using a previously established cell model for analysing NSC protection in vitro, we investigated a protective effect of clozapine on NSCs. Primary NSCs were isolated from the mouse subventricular zone (SVZ), we show that clozapine had a NSC protective activity alone, as evident by employing an ATP cell viability assay. In contrast, haloperidol did not show any NSC protective properties. Subsequently, cells were exposed to the non-competitive NMDA-receptor antagonist ketamine. Clozapine, but not haloperidol, had a NSC protective/anti-apoptotic activity against ketamine-induced cytotoxicity. The observed NSC protective activity of clozapine was associated with increased expression of the anti-apoptotic marker Bcl-2, decreased expression of the pro-apoptotic cleaved form of caspase-3 and associated with decreased expression of the autophagosome marker 1A/1B-light chain 3 (LC3-II). Collectively, our findings suggest that clozapine may have a protective/anti-apoptotic effect on NSCs, supporting previous in vivo observations, indicating a neurogenesis-promoting activity for clozapine. If the data are further confirmed in vivo, the results may encourage an expanded use of clozapine to restore impaired neurogenesis in schizophrenia.


Subject(s)
Adult Stem Cells/drug effects , Apoptosis/drug effects , Autophagy/drug effects , Clozapine/pharmacology , Excitatory Amino Acid Antagonists/toxicity , Ketamine/toxicity , Lateral Ventricles/drug effects , Neural Stem Cells/drug effects , Neuroprotective Agents/pharmacology , Adult Stem Cells/metabolism , Adult Stem Cells/pathology , Animals , Caspase 3/metabolism , Cells, Cultured , Haloperidol/pharmacology , Lateral Ventricles/metabolism , Lateral Ventricles/pathology , Male , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neurogenesis/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction
4.
Eur Neuropsychopharmacol ; 29(5): 616-628, 2019 05.
Article in English | MEDLINE | ID: mdl-30910381

ABSTRACT

Schizophrenia is associated with cognitive impairments related to hypofunction in glutamatergic N-methyl-D-aspartate receptor (NMDAR) transmission. Phencyclidine (PCP), a non-competitive NMDAR antagonist, models schizophrenia-like behavioral symptoms including cognitive deficits in rodents. This study examined the effects of PCP on emotional memory function examined in the passive avoidance (PA) task in mice and the ability of typical and atypical antipsychotic drugs (APDs) to rectify the PCP-mediated impairment. Pre-training administration of PCP (0.5, 1, 2 or 3 mg/kg) dose-dependently interfered with memory consolidation in the PA task. In contrast, PCP was ineffective when administered after training, and immediately before the retention test indicating that NMDAR blockade interferes with memory encoding mechanisms. The typical APD haloperidol and the dopamine D2/3 receptor antagonist raclopride failed to block the PCP-induced PA impairment suggesting a negligible role of D2 receptors in the PCP impairment. In contrast, the memory impairment was blocked by the atypical APDs clozapine and olanzapine in a dose-dependent manner while risperidone was effective only at the highest dose tested (1 mg/kg). The PCP-induced impairment involves 5-HT1A receptor mechanisms since the antagonist NAD-299 blocked the memory impairment caused by PCP and the ability of clozapine to attenuate the impairment by PCP. These results indicate that atypical but not typical APDs can ameliorate NMDAR-mediated memory impairments and support the view that atypical APDs such as clozapine can modulate glutamatergic memory dysfunctions through 5-HT1A receptor mechanisms. These findings suggest that atypical APDs may improve cognitive impairments related to glutamatergic dysfunction relevant for emotional memories in schizophrenia.


Subject(s)
Antipsychotic Agents/therapeutic use , Clozapine/therapeutic use , Emotional Regulation/drug effects , Haloperidol/therapeutic use , Memory Disorders/drug therapy , Phencyclidine/toxicity , Animals , Antipsychotic Agents/pharmacology , Clozapine/pharmacology , Dose-Response Relationship, Drug , Emotional Regulation/physiology , Excitatory Amino Acid Antagonists/toxicity , Haloperidol/pharmacology , Male , Memory Disorders/chemically induced , Memory Disorders/psychology , Mice , Mice, Inbred C57BL , Serotonin 5-HT1 Receptor Antagonists/pharmacology , Serotonin 5-HT1 Receptor Antagonists/therapeutic use
5.
Front Neural Circuits ; 12: 106, 2018.
Article in English | MEDLINE | ID: mdl-30627087

ABSTRACT

Neuropeptides are auxiliary messenger molecules that always co-exist in nerve cells with one or more small molecule (classic) neurotransmitters. Neuropeptides act both as transmitters and trophic factors, and play a role particularly when the nervous system is challenged, as by injury, pain or stress. Here neuropeptides and coexistence in mammals are reviewed, but with special focus on the 29/30 amino acid galanin and its three receptors GalR1, -R2 and -R3. In particular, galanin's role as a co-transmitter in both rodent and human noradrenergic locus coeruleus (LC) neurons is addressed. Extensive experimental animal data strongly suggest a role for the galanin system in depression-like behavior. The translational potential of these results was tested by studying the galanin system in postmortem human brains, first in normal brains, and then in a comparison of five regions of brains obtained from depressed people who committed suicide, and from matched controls. The distribution of galanin and the four galanin system transcripts in the normal human brain was determined, and selective and parallel changes in levels of transcripts and DNA methylation for galanin and its three receptors were assessed in depressed patients who committed suicide: upregulation of transcripts, e.g., for galanin and GalR3 in LC, paralleled by a decrease in DNA methylation, suggesting involvement of epigenetic mechanisms. It is hypothesized that, when exposed to severe stress, the noradrenergic LC neurons fire in bursts and release galanin from their soma/dendrites. Galanin then acts on somato-dendritic, inhibitory galanin autoreceptors, opening potassium channels and inhibiting firing. The purpose of these autoreceptors is to act as a 'brake' to prevent overexcitation, a brake that is also part of resilience to stress that protects against depression. Depression then arises when the inhibition is too strong and long lasting - a maladaption, allostatic load, leading to depletion of NA levels in the forebrain. It is suggested that disinhibition by a galanin antagonist may have antidepressant activity by restoring forebrain NA levels. A role of galanin in depression is also supported by a recent candidate gene study, showing that variants in genes for galanin and its three receptors confer increased risk of depression and anxiety in people who experienced childhood adversity or recent negative life events. In summary, galanin, a neuropeptide coexisting in LC neurons, may participate in the mechanism underlying resilience against a serious and common disorder, MDD. Existing and further results may lead to an increased understanding of how this illness develops, which in turn could provide a basis for its treatment.


Subject(s)
Neuropeptides/metabolism , Neurotransmitter Agents/metabolism , Animals , Humans , Locus Coeruleus/metabolism , Mental Disorders/metabolism , Receptors, Neurotransmitter/metabolism
6.
Eur Neuropsychopharmacol ; 27(12): 1298-1307, 2017 12.
Article in English | MEDLINE | ID: mdl-29102248

ABSTRACT

The endogenous neuropeptide nociceptin (N/OFQ), which mediates its actions via the nociceptin receptor (NOP), is implicated in multiple behavioural and physiological functions. This study examined the effects of the NOP agonists N/OFQ and the synthetic agonist Ro 64-6198, the antagonists NNN and NalBzoH, as well as deletion of the Pronociceptin gene on emotional memory in mice. The animals were tested in the passive avoidance (PA) task, dependent on hippocampal and amygdala functions. N/OFQ injected intraventricularly (i.c.v.) prior to training produced a biphasic effect on PA retention; facilitation at a low dose and impairment at higher doses. Ro 64-6198 also displayed a biphasic effect with memory facilitation at lower doses and impairment at a high dose. None of the agonists influenced PA training latencies. NNN did not significantly modulate retention in the PA task but antagonized the inhibitory effects of N/OFQ. NalBzoH facilitated memory retention in a dose-dependent manner and blocked the impairing effects of N/OFQ. However, neither NNN nor NalBzoH blocked the memory-impairing effects of Ro 64-6198. Finally, the Pnoc knockout mice exhibited enhanced PA retention latencies compared to the wild type mice. The biphasic effect of the natural ligand and Ro 64-6198 and the failure of the antagonists to block the action of Ro 64-6198 indicate complexity in ligand-receptor interaction. These results indicate that brain nociceptin and its NOP has a subtle role in regulation of mechanisms of relevance for treatment of disorders with processing disturbances of aversive events e.g. Alzheimer's disease, anxiety, depression and PTSD.


Subject(s)
Avoidance Learning/physiology , Opioid Peptides/metabolism , Receptors, Opioid/deficiency , Animals , Association Learning/drug effects , Avoidance Learning/drug effects , Dose-Response Relationship, Drug , Imidazoles/pharmacology , Injections, Intraventricular , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Naloxone/analogs & derivatives , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Opioid Peptides/genetics , Opioid Peptides/pharmacology , Peptide Fragments/pharmacology , Receptors, Opioid/agonists , Receptors, Opioid/genetics , Retention, Psychology/drug effects , Spiro Compounds/pharmacology , Nociceptin Receptor , Nociceptin
7.
PLoS One ; 12(1): e0170496, 2017.
Article in English | MEDLINE | ID: mdl-28125634

ABSTRACT

Ketamine administration is a well-established approach to mimic experimentally some aspects of schizophrenia. Adult neurogenesis dysregulation is associated with psychiatric disorders, including schizophrenia. The potential role of neurogenesis in the ketamine-induced phenotype is largely unknown. Recent results from human genetic studies have shown the pituitary adenylate cyclase-activating polypeptide (PACAP) gene is a risk factor for schizophrenia. Its potential role on the regulation of neurogenesis in experimental model of schizophrenia remains to be investigated. We aimed to determine whether ketamine affects the viability of adult neural stem cells (NSC). We also investigated whether the detrimental effect mediated by ketamine could be counteracted by PACAP. NSCs were isolated from the subventricular zone of the mouse and exposed to ketamine with/without PACAP. After 24 hours, cell viability, potential involvement of apoptosis, endoplasmic reticulum (ER) stress, mTOR and AMPA pathway activation were assessed by quantitative RT-PCR and Western blot analysis. We show that ketamine impairs NSC viability in correlation with increased apoptosis, ER stress and mTOR activation. The results also suggest that the effect of ketamine occurs via AMPA receptor activation. Finally, we show that PACAP counteracted the decreased NSC viability induced by ketamine via the specific activation of the PAC-1 receptor subtype. Our study shows that the NSC viability may be negatively affected by ketamine with putative importance for the development of a schizophrenia phenotype in the ketamine induced animal model of schizophrenia. The neuroprotective effect via PAC-1 activation suggests a potentially novel pharmacological target for the treatment of schizophrenia, via neurogenesis normalization.


Subject(s)
Adult Stem Cells/drug effects , Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Ketamine/pharmacology , Neural Stem Cells/drug effects , Pituitary Adenylate Cyclase-Activating Polypeptide/pharmacology , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Adult Stem Cells/metabolism , Animals , Cell Survival , Male , Mice , Neural Stem Cells/metabolism , Neuroprotective Agents/pharmacology , Neurotoxins/pharmacology
8.
Eur Neuropsychopharmacol ; 26(3): 467-76, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26811292

ABSTRACT

Newer, "atypical" antipsychotics carry a lower risk of motor side-effects than older, "typical" compounds. It has been proposed that a ~100-fold faster dissociation from the dopamine D2 receptor (D2R) distinguishes atypical from typical antipsychotics. Furthermore, differing antipsychotic D2R affinities have been suggested to reflect differences in dissociation rate constants (koff), while association rate constants (kon) were assumed to be similar. However, it was recently demonstrated that lipophilic accumulation of ligand in the cell interior and/or membrane can cause underestimation of koff, and as high-affinity D2R antagonists are frequently lipophilic, this may have been a confounding factor in previous studies. In the present work, a functional electrophysiology assay was used to measure the recovery of dopamine-mediated D2R responsivity from antipsychotic antagonism, using elevated concentrations of dopamine to prevent the potential bias of re-binding of lipophilic ligands. The variability of antipsychotic kon was also reexamined, capitalizing on the temporal resolution of the assay. kon was estimated from the experimental recordings using a simple mathematical model assumed to describe the binding process. The time course of recovery from haloperidol (typical antipsychotic) was only 6.4- to 2.5-fold slower than that of the atypical antipsychotics, amisulpride, clozapine, and quetiapine, while antipsychotic kons were found to vary more widely than previously suggested. Finally, affinities calculated using our kon and koff estimates correlated well with functional potency and with affinities reported from radioligand binding studies. In light of these findings, it appears unlikely that typical and atypical antipsychotics are primarily distinguished by their D2R binding kinetics.


Subject(s)
Antipsychotic Agents/pharmacology , Dopamine D2 Receptor Antagonists/pharmacology , Receptors, Dopamine D2/metabolism , Animals , Dopamine/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Humans , Kinetics , Oocytes , Receptors, Dopamine D2/genetics , Statistics as Topic
9.
Colloids Surf B Biointerfaces ; 140: 342-352, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26774571

ABSTRACT

Clozapine is an effective atypical antipsychotic drug that unfortunately exhibits poor oral bioavailability. Moreover, the clinical use of the compound is limited because of its numerous unfavorable and unsafe side effects. Therefore, the aim of the present study was the development of a new nanocarrier for a more effective clozapine delivery. Here, clozapine was encapsulated into polymeric nanocapsules (NCs). Polyelectrolyte multilayer shells were constructed by the technique of sequential adsorption of polyelectrolytes (LbL) using biocompatible polyanion PGA (Poly-L-glutamic acid, sodium salt) and polycation PLL (poly-L-lysine) on clozapine-loaded nanoemulsion cores. Pegylated external layers were prepared using PGA-g-PEG (PGA grafted by PEG (polyethylene glycol)). Clozapine was successfully loaded into the PLL-PGA nanocarriers (CLO-NCs) with an average size of 100 nm. In vitro analysis of the interactions of the CLO-NCs with the cells of the mononuclear phagocytic system (MPS) was conducted. Cell biocompatibility, phagocytosis potential, and cellular uptake were studied. Additionally, the biodistribution and behavioral effects of the encapsulated clozapine were also studied. The results indicate that surface modified (by PEG grafting) polymeric PLL-PGA CLO-NCs are very promising nanovehicles for improving clozapine delivery.


Subject(s)
Clozapine/chemistry , Nanocapsules/chemistry , Polyglutamic Acid/chemistry , Polylysine/chemistry , Animals , Antipsychotic Agents/chemistry , Antipsychotic Agents/pharmacokinetics , Antipsychotic Agents/pharmacology , Cell Line , Cell Survival/drug effects , Clozapine/pharmacokinetics , Clozapine/pharmacology , Drug Compounding , Flow Cytometry , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred C57BL , Microscopy, Confocal , Motor Activity/drug effects , Tissue Distribution
10.
Int J Neuropsychopharmacol ; 19(6)2016 06 01.
Article in English | MEDLINE | ID: mdl-26657176

ABSTRACT

BACKGROUND: The psychotomimetic phencyclidine (PCP) produces behavioral symptoms similar to those observed in schizophrenia, accompanied by increased motor activity. The dopamine and adenosine 3',5'-cyclic monophosphate-regulated phosphoprotein of 32kDa (DARPP-32) is enriched in the medium spiny neurons (MSNs) of the striatum and has been implicated in the actions of PCP. We examined the effects of deletion of DARPP-32 in distinct populations of striatal MSNs, on the ability of PCP to induce motor activation and memory deficit. METHODS: The effects of PCP were examined in mice with conditional knockout of DARPP-32 in the MSNs of the direct, or indirect pathway. DARPP-32 phosphorylation was determined by Western blotting. The motor stimulant effects of PCP were determined by measuring locomotion following acute and chronic administration. Memory deficit was evaluated using the passive avoidance test. RESULTS: Loss of DARPP-32 in direct MSNs prevents PCP-induced phosphorylation and abolishes the motor stimulation effects of PCP. In contrast, lack of DARPP-32 in indirect MSNs does not affect the ability of PCP to promote DARPP-32 phosphorylation and to increase motor activity. The impairment in passive avoidance induced by PCP is independent of the expression of DARPP-32 in direct or indirect MSNs. CONCLUSIONS: The increase in DARPP-32 phosphorylation induced by PCP occurs selectively in the MSNs of the direct pathway, which are also specifically involved in the motor stimulant effects of this drug. The memory deficit induced by PCP is not linked to the expression of DARPP-32 in striatal MSNs.

11.
Front Pharmacol ; 6: 162, 2015.
Article in English | MEDLINE | ID: mdl-26300776

ABSTRACT

Serotonin [5-hydroxytryptamine (5-HT)] is a multifunctional neurotransmitter innervating cortical and limbic areas involved in cognition and emotional regulation. Dysregulation of serotonergic transmission is associated with emotional and cognitive deficits in psychiatric patients and animal models. Drugs targeting the 5-HT system are widely used to treat mood disorders and anxiety-like behaviors. Among the fourteen 5-HT receptor (5-HTR) subtypes, the 5-HT1AR and 5-HT7R are associated with the development of anxiety, depression and cognitive function linked to mechanisms of emotional learning and memory. In rodents fear conditioning and passive avoidance (PA) are associative learning paradigms to study emotional memory. This review assesses the role of 5-HT1AR and 5-HT7R as well as their interplay at the molecular, neurochemical and behavioral level. Activation of postsynaptic 5-HT1ARs impairs emotional memory through attenuation of neuronal activity, whereas presynaptic 5-HT1AR activation reduces 5-HT release and exerts pro-cognitive effects on PA retention. Antagonism of the 5-HT1AR facilitates memory retention possibly via 5-HT7R activation and evidence is provided that 5HT7R can facilitate emotional memory upon reduced 5-HT1AR transmission. These findings highlight the differential role of these 5-HTRs in cognitive/emotional domains of behavior. Moreover, the results indicate that tonic and phasic 5-HT release can exert different and potentially opposing effects on emotional memory, depending on the states of 5-HT1ARs and 5-HT7Rs and their interaction. Consequently, individual differences due to genetic and/or epigenetic mechanisms play an essential role for the responsiveness to drug treatment, e.g., by SSRIs which increase intrasynaptic 5-HT levels thereby activating multiple pre- and postsynaptic 5-HTR subtypes.

12.
Neural Plast ; 2015: 370432, 2015.
Article in English | MEDLINE | ID: mdl-26257960

ABSTRACT

Stimulating regeneration in the brain has the potential to rescue neuronal networks and counteract progressive pathological changes in Alzheimer's disease (AD). This study investigated whether drugs with different mechanisms of action could enhance neurogenesis and improve cognition in mice receiving human neural stem cell (hNSC) transplants. Six- to nine-month-old AD Tg2576 mice were treated for five weeks with the amyloid-modulatory and neurotrophic drug (+)-phenserine or with the partial α7 nicotinic receptor (nAChR) agonist JN403, combined with bilateral intrahippocampal hNSC transplantation. We observed improved spatial memory in hNSC-transplanted non-drug-treated Tg2576 mice but not in those receiving drugs, and this was accompanied by an increased number of Doublecortin- (DCX-) positive cells in the dentate gyrus, a surrogate marker for newly generated neurons. Treatment with (+)-phenserine did however improve graft survival in the hippocampus. An accumulation of α7 nAChR-expressing astrocytes was observed around the injection site, suggesting their involvement in repair and scarring processes. Interestingly, JN403 treatment decreased the number of α7 nAChR-expressing astrocytes, correlating with a reduction in the number of DCX-positive cells in the dentate gyrus. We conclude that transplanting hNSCs enhances endogenous neurogenesis and prevents further cognitive deterioration in Tg2576 mice, while simultaneous treatments with (+)-phenserine or JN403 result in countertherapeutic effects.


Subject(s)
Alzheimer Disease/psychology , Alzheimer Disease/therapy , Amyloid beta-Peptides/antagonists & inhibitors , Cognition , Neural Stem Cells/transplantation , Neurogenesis , Stem Cell Transplantation , alpha7 Nicotinic Acetylcholine Receptor/agonists , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Astrocytes/drug effects , Carbamates/pharmacology , Cell Proliferation , Doublecortin Protein , Female , Hippocampus/surgery , Humans , Male , Maze Learning/drug effects , Memory/drug effects , Mice , Nerve Regeneration , Nicotinic Agonists/pharmacology , Physostigmine/analogs & derivatives , Physostigmine/pharmacology , Quinuclidines/pharmacology
13.
Proc Natl Acad Sci U S A ; 112(31): E4326-35, 2015 Aug 04.
Article in English | MEDLINE | ID: mdl-26195742

ABSTRACT

Synaptosomal-associated protein of 25 kDa (SNAP-25) is a key molecule in the soluble N-ethylmaleimide-sensitive factor attachment protein (SNARE) complex mediating fast Ca(2+)-triggered release of hormones and neurotransmitters, and both splice variants, SNAP-25a and SNAP-25b, can participate in this process. Here we explore the hypothesis that minor alterations in the machinery mediating regulated membrane fusion can increase the susceptibility for metabolic disease and precede obesity and type 2 diabetes. Thus, we used a mouse mutant engineered to express normal levels of SNAP-25 but only SNAP-25a. These SNAP-25b-deficient mice were exposed to either a control or a high-fat/high-sucrose diet. Monitoring of food intake, body weight, hypothalamic function, and lipid and glucose homeostases showed that SNAP-25b-deficient mice fed with control diet developed hyperglycemia, liver steatosis, and adipocyte hypertrophy, conditions dramatically exacerbated when combined with the high-fat/high-sucrose diet. Thus, modified SNARE function regulating stimulus-dependent exocytosis can increase the vulnerability to and even provoke metabolic disease. When combined with a high-fat/high-sucrose diet, this vulnerability resulted in diabesity. Our SNAP-25b-deficient mouse may represent a diabesity model.


Subject(s)
Metabolic Diseases/metabolism , Synaptosomal-Associated Protein 25/metabolism , Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Adiposity , Animals , Blood Glucose/metabolism , Body Weight , Dyslipidemias/pathology , Energy Intake , Energy Metabolism , Feeding Behavior , Female , Homeostasis , Hypertrophy , Hypothalamus/metabolism , Insulin/metabolism , Insulin Secretion , Leptin/blood , Liver/metabolism , Liver/pathology , Male , Metabolic Diseases/blood , Mice, Obese , Phenotype , Receptors, Leptin/metabolism , Synaptosomal-Associated Protein 25/deficiency
14.
Eur Neuropsychopharmacol ; 25(8): 1314-23, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25963581

ABSTRACT

In light of the involvement of serotonergic 5-HT1A receptors in the mediation of the memory of aversive events, the potent and selective 5-HT1A receptor antagonists, MC18 fumarate and VP08/34 fumarate, were tested in the passive avoidance task (PA), a rodent model of instrumental conditioning. Either alone or in combination with the prototypical agonist 8-OH-DPAT, MC18 fumarate at doses (0.1, 0.3 and 1mg/kg given 15min prior to training) exerted a dose-dependent facilitation of PA memory retention. When administered 15min prior to 8-OH-DPAT (0.3 and 1mg/kg), MC18 fumarate at a dose of 0.3mg/kg, enhanced significantly the impairment of PA retention caused by 8-OH-DPAT (1mg/kg). However, VP08/34 fumarate given at the same doses exerted no statistically effect on PA retention memory. Furthermore, VP08/34 fumarate given 15min prior to 8-OH-DPAT (0.3 and 1mg/kg) only slightly enhanced the PA impairment induced by 8-OH-DPAT. In conclusion, the profile of MC18 fumarate is intriguing since it behaves in a manner which differs from both full receptor antagonists such as NAD-299 or partial receptor agonists. The results also illustrate the importance of subtle receptor interaction and probably ligand efficacy in determining the actions of two almost identical 5-HT1A receptor ligands in cognitive function such as instrumental learning.


Subject(s)
Memory/physiology , Receptor, Serotonin, 5-HT1A/metabolism , 8-Hydroxy-2-(di-n-propylamino)tetralin/chemistry , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Animals , Avoidance Learning/drug effects , Avoidance Learning/physiology , Benzopyrans/chemistry , Benzopyrans/pharmacology , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Dose-Response Relationship, Drug , Fumarates/chemistry , Fumarates/pharmacology , Male , Memory/drug effects , Mice, Inbred C57BL , Molecular Structure , Serotonin 5-HT1 Receptor Agonists/chemistry , Serotonin 5-HT1 Receptor Agonists/pharmacology , Serotonin 5-HT1 Receptor Antagonists/chemistry , Serotonin 5-HT1 Receptor Antagonists/pharmacology
15.
Eur Neuropsychopharmacol ; 25(5): 591-8, 2015 May.
Article in English | MEDLINE | ID: mdl-25799919

ABSTRACT

Pharmacotherapy is effective in helping many patients suffering from psychiatric and neurological disorders, and both psychotherapeutic and stimulation-based techniques likewise have important roles to play in their treatment. However, therapeutic progress has recently been slow. Future success for improving the control and prevention of brain disorders will depend upon deeper insights into their causes and pathophysiological substrates. It will also necessitate new and more rigorous methods for identifying, validating, developing and clinically deploying new treatments. A field of Research and Development (R and D) that remains critical to this endeavour is Neuropsychopharmacology which transformed the lives of patients by introducing pharmacological treatments for psychiatric disorder some 60 years ago. For about half of this time, the European College of Neuropsychopharmacology (ECNP) has fostered efforts to enhance our understanding of the brain, and to improve the management of psychiatric disorders. Further, together with partners in academia and industry, and in discussions with regulators and patients, the ECNP is implicated in new initiatives to achieve this goal. This is then an opportune moment to survey the field, to analyse what we have learned from the achievements and failures of the past, and to identify major challenges for the future. It is also important to highlight strategies that are being put in place in the quest for more effective treatment of brain disorders: from experimental research and drug discovery to clinical development and collaborative ventures for reinforcing "R and D". The present article sets the scene, then introduces and interlinks the eight articles that comprise this Special Volume of European Neuropsychopharmacology. A broad-based suite of themes is covered embracing: the past, present and future of "R and D" for psychiatric disorders; complementary contributions of genetics and epigenetics; efforts to improve the treatment of depression, neurodevelopmental and neurodegenerative disorders; and advances in the analysis and neuroimaging of cellular and cerebral circuits.


Subject(s)
Brain Diseases/drug therapy , Brain/drug effects , Central Nervous System Agents/pharmacology , Mental Disorders/drug therapy , Animals , Brain/growth & development , Brain/metabolism , Brain/pathology , Brain Diseases/genetics , Brain Diseases/metabolism , Brain Diseases/pathology , Epigenesis, Genetic/drug effects , Humans , Mental Disorders/genetics , Mental Disorders/metabolism , Mental Disorders/pathology , Models, Neurological
16.
Int J Neuropsychopharmacol ; 17(1): 149-55, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24074141

ABSTRACT

It has been suggested that the favorable side-effect profiles of atypical antipsychotics (e.g. clozapine and amisulpride) are related to their ∼100-fold faster dissociation from dopamine D2 receptors (D2R) compared with typical antipsychotics (e.g. haloperidol and chlorpromazine). Fast dissociation would entail rapidly reversible antagonism; however, this has not been thoroughly studied using functional assays. We compared the reversibilities of D2R antagonism by 17 compounds using an electrophysiological method to measure dopamine-evoked potassium channel activation via D2R. Varying rates and amplitudes of D2R response recovery were observed following antagonist removal. Whereas recovery rates differed 15-fold between atypical drugs, recovery from clozapine and amisulpride antagonism was, unexpectedly, less than twofold faster than from chlorpromazine. The recovery amplitude correlated with calculated water solubility and lipid/water distribution coefficients, suggesting variable drug partitioning into cell membranes. Our data do not support the notion that the rate of reversibility of D2R antagonism is what distinguishes atypical from typical antipsychotics.


Subject(s)
Antipsychotic Agents/classification , Antipsychotic Agents/pharmacology , Dopamine D2 Receptor Antagonists , Animals , Dose-Response Relationship, Drug , Hydrophobic and Hydrophilic Interactions , Membrane Potentials/drug effects , Membrane Potentials/physiology , Oocytes , Potassium Channels/metabolism , Solubility , Xenopus
17.
PLoS One ; 8(11): e80497, 2013.
Article in English | MEDLINE | ID: mdl-24244693

ABSTRACT

Pregnant women with epilepsy have to balance maternal and fetal risks associated with uncontrolled seizures against the potential teratogenic effects from antiepileptic drugs (AEDs). Carbamazepine (CBZ) is among the four most commonly used AEDs for treatment of pregnant epileptic women. We previously reported that new-born children had a decreased head circumference after in utero CBZ exposure. This study investigates how prenatal exposure of CBZ influences the number of neurons in new-born and young mouse hippocampus, amygdala and cortex cerebri. Clinical studies describe inconclusive results on if prenatal CBZ treatment influences cognition. Here we investigate this issue in mice using two well characterized cognitive tasks, the passive avoidance test and the Morris water maze test. Prenatal exposure of CBZ reduced the number of neurons (NeuN-immunoreactive cells) in the new-born mouse hippocampus with 50% compared to non-exposed mice. A reduction of neurons (20%) in hippocampus was still observed when the animals were 5 weeks old. These mice also displayed a 25% reduction of neurons in cortex cerebri. Prenatal CBZ treatment did not significantly impair learning and memory measured in the passive avoidance test and in the Morris water maze. However, these mice displayed a higher degree of thigmotaxic behaviour than the control mice. The body weight of prenatally CBZ exposed five-week old mice were lower compared to control mice not exposed to CBZ (p = 0.001). In conclusion, prenatal exposure to CBZ reduces the number of neurons dramatically in areas important for cognition such as hippocampus and cortex, without severe impairments on learning and memory. These results are in line with some clinical studies, reporting that CBZ has minor negative effects on cognition. The challenge for future studies are to segment out what possible effects a reduction of neurons could have on different types of cognition, like intellectual ability and social interaction.


Subject(s)
Carbamazepine/adverse effects , Hippocampus/cytology , Learning/drug effects , Memory/drug effects , Neurons/drug effects , Amygdala/cytology , Animals , Cerebral Cortex/cytology , Female , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Pregnancy , Prenatal Exposure Delayed Effects
18.
Br J Pharmacol ; 170(4): 859-70, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23902597

ABSTRACT

BACKGROUND AND PURPOSE: The beat-by-beat fluctuation (dynamics) of heart rate (HR) depends on centrally mediated control of the autonomic nervous system (ANS) reflecting the physiological state of an organism. 5-HT1A receptors are implicated in affective disorders,associated with ANS dysregulation which increases cardiac risk but their role in autonomic HR regulation under physiological conditions is insufficiently characterized. EXPERIMENTAL APPROACH: The effects of subcutaneously administered 5-HT1A receptor ligands on HR dynamics were investigated in C57BL/6 mice during stress-free conditions and emotional challenge (recall of fear conditioned to an auditory stimulus and novelty exposure) using time domain and non-linear HR analyses. KEY RESULTS: Pre-training treatment with of 8-OH-DPAT (0.5 mg·kg(-1) , s.c.) prevented conditioned tachycardia in the retention test indicating impaired fear memory. Pretest 5-HT1A receptor activation by 8-OH-DPAT (0.5 but not 0.1 and 0.02 mg·kg(-1) ) caused bradycardia and increased HR variability. 8-OH-DPAT (0.5 mg·kg(-1) ) lowered the unconditioned and conditioned tachycardia from ∼750 to ∼550 bpm, without changing the conditioned HR response to the sound. 8-OH-DPAT induced profound QT prolongation and bradyarrhythmic episodes. Non-linear analysis indicated a pathological state of HR dynamics after 8-OH-DPAT (0.5 mg·kg(-1) ) with ANS hyperactivation impairing HR adaptability. The 5-HT1A receptor antagonist WAY-100635 (0.03 mg·kg(-1) ) blocked these effects of 8-OH-DPAT. CONCLUSIONS AND IMPLICATIONS: Pre-training 5-HT1A receptor activation by 8-OH-DPAT (0.5 mg·kg(-1) ) impaired memory of conditioned auditory fear based on an attenuated HR increase, whereas pretest administration did not prevent the fear-conditioned HR increase but induced pathological HR dynamics through central ANS dysregulation with cardiac effects similar to acute SSRI overdose.


Subject(s)
Autonomic Nervous System/metabolism , Behavior, Animal , Brain/metabolism , Conditioning, Psychological , Fear , Heart Rate , Heart/innervation , Receptor, Serotonin, 5-HT1A/metabolism , Acoustic Stimulation , Animals , Autonomic Nervous System/drug effects , Behavior, Animal/drug effects , Brain/drug effects , Conditioning, Psychological/drug effects , Dose-Response Relationship, Drug , Electrocardiography , Emotions , Fear/drug effects , Heart Rate/drug effects , Ligands , Male , Mental Recall/drug effects , Mice , Mice, Inbred C57BL , Nonlinear Dynamics , Receptor, Serotonin, 5-HT1A/drug effects , Serotonin 5-HT1 Receptor Agonists/pharmacology , Serotonin 5-HT1 Receptor Antagonists/pharmacology , Signal Processing, Computer-Assisted , Time Factors
19.
Behav Brain Res ; 238: 160-9, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23098796

ABSTRACT

Local infusion of the GABA(A) receptor agonist muscimol is used for reversible inactivation of septohippocampal brain structures associated with cognitive functions. However, information on the effective duration, affected processes and site(s) of action of muscimol in the hippocampus is lacking. Therefore, the dose- and time-dependent effects of bilateral dorsohippocampal infusion of muscimol (0.01-2.0 µg/mouse) below the CA1 area were examined on processing of fear memory in male C57BL/6J mice. Infusion of muscimol 15 min-6 h but not 9 h or 24 h before training impaired conditioned context-dependent fear tested 24 h or 48 h after training. Post-training infusion of muscimol also impaired context-dependent fear when applied either 4 h or 6 h after training, although with lower efficacy. Muscimol was ineffective when administered immediately, 1 h or 24 h after training. Infusion of muscimol 15 min before training impaired context-dependent fear 4-6 h after training indicating preserved short-term but impaired long-term memory. Regardless of infusion time and dose, muscimol had no effect on tone-dependent (cued) fear memory. The impairment by the fluorescently-labeled muscimol-bodipy (5.3 µg/mouse) were similar to those of an equimolar dose of muscimol (1 µg/mouse). The distribution profile after local infusion indicated that muscimol-bodipy (5.3 µg/mouse) was confined to the CA1 area of the dorsal hippocampus. These results demonstrated that GABA(A) receptor activation in the CA1 area of the dorsal hippocampus causes a long-term memory impairment of conditioned context-dependent fear mediated by a long-lasting (≥6 h) muscimol action most likely affecting consolidation processes.


Subject(s)
CA1 Region, Hippocampal/drug effects , Conditioning, Classical/drug effects , Fear/drug effects , GABA-A Receptor Agonists/pharmacology , Memory, Long-Term/drug effects , Muscimol/pharmacology , Acoustic Stimulation , Animals , Association Learning/drug effects , Cues , Male , Mice , Mice, Inbred C57BL
20.
Neuropharmacology ; 63(6): 1150-60, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22801295

ABSTRACT

This study utilized pharmacological manipulations to analyze the role of direct and indirect activation of 5-HT(7) receptors (5-HT(7)Rs) in passive avoidance learning by assessing emotional memory in male C57BL/6J mice. Additionally, 5-HT(7)R binding affinity and 5-HT(7)R-mediated protein phosphorylation of downstream signaling targets were determined. Elevation of 5-HT by the selective serotonin reuptake inhibitor (SSRI) fluoxetine had no effect by itself, but facilitated emotional memory performance when combined with the 5-HT(1A)R antagonist NAD-299. This facilitation was blocked by the selective 5-HT(7)R antagonist SB269970, revealing excitatory effects of the SSRI via 5-HT(7)Rs. The enhanced memory retention by NAD-299 was blocked by SB269970, indicating that reduced activation of 5-HT(1A)Rs results in enhanced 5-HT stimulation of 5-HT(7)Rs. The putative 5-HT(7)R agonists LP-44 when administered systemically and AS19 when administered both systemically and into the dorsal hippocampus failed to facilitate memory. This finding is consistent with the low efficacy of LP-44 and AS19 to stimulate protein phosphorylation of 5-HT(7)R-activated signaling cascades. In contrast, increasing doses of the dual 5-HT(1A)R/5-HT(7)R agonist 8-OH-DPAT impaired memory, while co-administration with NAD-299 facilitated of emotional memory in a dose-dependent manner. This facilitation was blocked by SB269970 indicating 5-HT(7)R activation by 8-OH-DPAT. Dorsohippocampal infusion of 8-OH-DPAT impaired passive avoidance retention through hippocampal 5-HT(1A)R activation, while 5-HT(7)Rs appear to facilitate memory processes in a broader cortico-limbic network and not the hippocampus alone.


Subject(s)
Basigin/physiology , Emotions/physiology , Memory/physiology , Receptor Cross-Talk/physiology , Receptor, Serotonin, 5-HT1A/physiology , Selective Serotonin Reuptake Inhibitors/pharmacology , Animals , Avoidance Learning/drug effects , Basigin/drug effects , Blotting, Western , Emotions/drug effects , Fear/drug effects , Fear/physiology , Fluoxetine/pharmacology , HeLa Cells , Heart Rate/drug effects , Hippocampus/metabolism , Hippocampus/physiology , Humans , Male , Memory/drug effects , Mice , Mice, Inbred C57BL , Phosphorylation , Radioligand Assay , Receptor Cross-Talk/drug effects , Receptor, Serotonin, 5-HT1A/drug effects , Serotonin/metabolism , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacology , Stereotaxic Techniques , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...