Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 491
1.
Mol Cell Neurosci ; 120: 103719, 2022 05.
Article En | MEDLINE | ID: mdl-35283305

Pattern separation is a hippocampal process in which highly similar stimuli are recognized as separate representations, and deficits could lead to memory impairments in neuropsychiatric disorders such as schizophrenia. The 5-HT1A receptor (5-HT1AR) is believed to be involved in these hippocampal pattern separation processes. However, in the dorsal raphe nucleus (DRN), the 5-HT1AR is expressed as a somatodendritic autoreceptor, negatively regulates serotonergic signaling, and could thereby counteract the effects of hippocampal postsynaptic 5-HT1A receptors. Therefore, this study aims to identify how pre- and post-synaptic 5-HT1AR activity affects pattern separation. Object pattern separation (OPS) performance was measured in male Wistar rats after both acute and chronic treatment (i.p.) with 5-HT1AR biased agonists F13714 (0.0025 mg/kg acutely, 0.02 mg/kg/day chronically) or NLX-101 (0.08 mg/kg acutely, 0.32 mg/kg/day chronically), which preferentially activate autoreceptors or postsynaptic receptors respectively, for 14 days. Body temperature - a functional correlate of hypothalamic 5-HT1AR stimulation - was measured daily. Additionally, 5-HT1AR density (DRN) and plasticity markers (hippocampus) were assessed. Acute treatment with F13714 impaired OPS performance, whereas chronic treatment normalized this, and a drop in body temperature was found from day 4 onwards. NLX-101 enhanced OPS performance acutely and chronically, and caused an acute drop in body temperature. Chronic NLX-101 treatment increased doublecortin positive neurons in the dorsal hippocampus, while chronic treatment with F13714 resulted in a downregulation of 5-HT1A autoreceptors, which likely reversed the acute impairment in OPS performance. Chronic treatment with NLX-101 appears to have therapeutic potential to improve brain plasticity and OPS performance.


Aminopyridines , Autoreceptors , Hippocampus , Neuronal Plasticity , Pattern Recognition, Physiological , Piperidines , Pyrimidines , Receptor, Serotonin, 5-HT1A , Recognition, Psychology , Serotonin 5-HT1 Receptor Agonists , Aminopyridines/pharmacology , Animals , Autoreceptors/physiology , Hippocampus/drug effects , Hippocampus/physiology , Male , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Pattern Recognition, Physiological/drug effects , Pattern Recognition, Physiological/physiology , Piperidines/pharmacology , Pyrimidines/pharmacology , Rats , Rats, Wistar , Receptor, Serotonin, 5-HT1A/physiology , Recognition, Psychology/drug effects , Recognition, Psychology/physiology , Serotonin 5-HT1 Receptor Agonists/pharmacology , Serotonin 5-HT1 Receptor Agonists/therapeutic use
2.
Respir Physiol Neurobiol ; 296: 103810, 2022 02.
Article En | MEDLINE | ID: mdl-34728431

Systemic 8-OH-DPAT (a 5-HT1A receptor agonist) challenge evokes hyperventilation independent of peripheral 5-HT1A receptors. Though the pre-Botzinger Complex (PBC) is critical in generating respiratory rhythm and activation of local 5-HT1A receptors induces tachypnea via disinhibition of local GABAA neurons, its role in the respiratory response to systemic 8-OH-DPAT challenge is still unclear. In anesthetized rats, 8-OH-DPAT (100 µg/kg, iv) was injected twice to confirm the reproducibility of the evoked responses. The same challenges were performed after bilateral microinjections of (S)-WAY-100135 (a 5-HT1A receptor antagonist) or gabazine (a GABAA receptor antagonist) into the PBC. Our results showed that: 1) 8-OH-DPAT caused reproducible hyperventilation associated with hypotension and bradycardia; 2) microinjections of (S)-WAY-100135 into the PBC attenuated the hyperventilation by ˜60 % without effect on the evoked hypotension and bradycardia; and 3) the same hyperventilatory attenuation was also observed after microinjections of gabazine into the PBC. Our data suggest that PBC 5-HT1A receptors play a key role in the respiratory response to systemic 8-OH-DPAT challenge likely via disinhibiting local GABAergic neurons.


8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , GABA Antagonists/pharmacology , Hyperventilation/chemically induced , Hyperventilation/drug therapy , Medulla Oblongata/metabolism , Receptor, Serotonin, 5-HT1A/physiology , Respiratory Center/metabolism , Serotonin 5-HT1 Receptor Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacology , 8-Hydroxy-2-(di-n-propylamino)tetralin/administration & dosage , Animals , Disease Models, Animal , Male , Medulla Oblongata/drug effects , Piperazines/pharmacology , Pyridazines/pharmacology , Rats , Receptor, Serotonin, 5-HT1A/drug effects , Respiratory Center/drug effects
3.
Neuropharmacology ; 203: 108881, 2022 02 01.
Article En | MEDLINE | ID: mdl-34785162

Although serotonin 1A (5-HT1A) receptor agonists are widely used as the additive compound to reduce l-dopa-induced dyskinesia in Parkinson's disease (PD), few studies focused on the effect and mechanism of 5-HT1A receptor agonist on the motor symptoms of PD. Unilateral 6-hydroxydopamine (6-OHDA)-lesioned rats were used and implantation of electrodes was performed in the motor cortex of these rats. So the effect of 5-HT1A receptor agonist 8-OH-DPAT on motor behaviors and oscillatory activities were evaluated. In addition, 8-OH-DPAT combined with D2 receptor antagonist raclopride, NMDA receptor antagonist MK-801, or its agonist d-cycloserine (DCS) were co-administrated. 8-OH-DPAT administration significantly improved spontaneous locomotor activity and asymmetric forepaw function in 6-OHDA-lesioned rats. Meanwhile, 8-OH-DPAT identified selective modulation of the abnormal high beta oscillations (25-40 Hz) in the motor cortex of 6-OHDA-lesioned rats, without inducing pathological finely tuned gamma around 80 Hz. Different from 8-OH-DPAT, l-dopa treatment produced a prolonged improvement on motor performances and differential regulation of high beta and gamma oscillations. However, dopamine D2 receptor antagonist had no influence on the 8-OH-DPAT-mediated-motor behaviors and beta oscillations in 6-OHDA-lesioned rats. In contrast, subthreshold NMDA receptor antagonist MK-801 obviously elevated the 8-OH-DPAT-mediated-motor behaviors, while NMDA receptor agonist DCS partially impaired the 8-OH-DPAT-mediated symptoms in 6-OHDA-lesioned rats. This study suggests that 5-HT1A receptor agonist 8-OH-DPAT improves motor activity and modulates the oscillations in the motor cortex of parkinsonian rats. Different from l-dopa, 8-OH-DPAT administration ameliorates motor symptoms of PD through glutamatergic rather than the dopaminergic pathway.


Beta Rhythm/physiology , Motor Cortex/physiology , Parkinsonian Disorders/physiopathology , Receptor, Serotonin, 5-HT1A/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Serotonin 5-HT1 Receptor Agonists/pharmacology , 8-Hydroxy-2-(di-n-propylamino)tetralin/toxicity , Animals , Beta Rhythm/drug effects , Locomotion/drug effects , Locomotion/physiology , Male , Motor Cortex/drug effects , Motor Disorders/chemically induced , Motor Disorders/drug therapy , Motor Disorders/physiopathology , Oxidopamine/toxicity , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/drug therapy , Rats , Rats, Sprague-Dawley , Serotonin 5-HT1 Receptor Agonists/therapeutic use
4.
Front Neural Circuits ; 15: 718348, 2021.
Article En | MEDLINE | ID: mdl-34512276

Neuromodulatory systems may provide information on social context to auditory brain regions, but relatively few studies have assessed the effects of neuromodulation on auditory responses to acoustic social signals. To address this issue, we measured the influence of the serotonergic system on the responses of neurons in a mouse auditory midbrain nucleus, the inferior colliculus (IC), to vocal signals. Broadband vocalizations (BBVs) are human-audible signals produced by mice in distress as well as by female mice in opposite-sex interactions. The production of BBVs is context-dependent in that they are produced both at early stages of interactions as females physically reject males and at later stages as males mount females. Serotonin in the IC of males corresponds to these events, and is elevated more in males that experience less female rejection. We measured the responses of single IC neurons to five recorded examples of BBVs in anesthetized mice. We then locally activated the 5-HT1A receptor through iontophoretic application of 8-OH-DPAT. IC neurons showed little selectivity for different BBVs, but spike trains were characterized by local regions of high spike probability, which we called "response features." Response features varied across neurons and also across calls for individual neurons, ranging from 1 to 7 response features for responses of single neurons to single calls. 8-OH-DPAT suppressed spikes and also reduced the numbers of response features. The weakest response features were the most likely to disappear, suggestive of an "iceberg"-like effect in which activation of the 5-HT1A receptor suppressed weakly suprathreshold response features below the spiking threshold. Because serotonin in the IC is more likely to be elevated for mounting-associated BBVs than for rejection-associated BBVs, these effects of the 5-HT1A receptor could contribute to the differential auditory processing of BBVs in different behavioral subcontexts.


Inferior Colliculi , Receptor, Serotonin, 5-HT1A/physiology , Vocalization, Animal , Acoustic Stimulation , Animals , Auditory Perception , Female , Inferior Colliculi/physiology , Male , Mesencephalon , Mice , Serotonin
5.
Psychopharmacology (Berl) ; 238(8): 2249-2260, 2021 Aug.
Article En | MEDLINE | ID: mdl-33973045

RATIONALE: The prevalence of depression is ever-increasing throughout the population. However, available treatments are ineffective in around one-third of patients and there is a need for more effective and safer drugs. OBJECTIVES: The antidepressant-like and procognitive effects of the "biased agonists" F15599 (also known as NLX-101) which preferentially targets postsynaptic 5-HT1A receptors and F13714, which targets 5-HT1A autoreceptors, were investigated in mice. METHODS: Antidepressant-like properties of the compounds and their effect on cognitive functions were assessed using the forced swim test (FST) and the novel object recognition (NOR), respectively. Next, we induced a depressive-like state by an unpredictable chronic mild stress (UCMS) procedure to test the compounds' activity in the depression model, followed by measures of sucrose preference, FST, and locomotor activity. Levels of phosphorylated cyclic AMP response element-binding protein (p-CREB) and phosphorylated extracellular signal-regulated kinase (p-ERK1/2) were also determined. RESULTS: F15599 reduced immobility time in the FST over a wider dose-range (2 to 16 mg/kg po) than F13714 (2 and 4 mg/kg po), suggesting accentuated antidepressant-like properties in mice. F15599 did not disrupt long-term memory consolidation in the NOR at any dose tested, while F13714 impaired memory formation, notably at higher doses (4-16 mg/kg). In UCMS mice, a single administration of F15599 and F13714 was sufficient to robustly normalize depressive-like behavior in the FST but did not rescue disrupted sucrose preference. Both F15599 and F13714 rescued cortical and hippocampal deficits in p-ERK1/2 levels of UCMS mice but did not influence the p-CREB levels. CONCLUSIONS: Our studies showed that 5-HT1A receptor biased agonists such as F13714 and especially F15599, due to its less pronounced side effects, might have potential as fast-acting antidepressants.


Aminopyridines/administration & dosage , Antidepressive Agents/administration & dosage , Piperidines/administration & dosage , Pyrimidines/administration & dosage , Receptor, Serotonin, 5-HT1A , Serotonin 5-HT1 Receptor Agonists/administration & dosage , Stress, Psychological/drug therapy , Animals , Chronic Disease , Disease Models, Animal , Dose-Response Relationship, Drug , Locomotion , Male , Mice , Receptor, Serotonin, 5-HT1A/physiology , Single-Blind Method , Stress, Psychological/psychology
6.
Behav Brain Res ; 408: 113296, 2021 06 25.
Article En | MEDLINE | ID: mdl-33862061

Changes in 5-HT1A receptor (5-HT1AR)-mediated neurotransmission in the hippocampus have been associated with anxiety, depression and in the mode of action of antidepressant drugs. It has been commonly accepted that whereas the dorsal pole of the hippocampus (DH) is involved in cognitive processing, the ventral pole (VH) is associated with emotional regulation. However, to date, only a few studies have directly addressed the role played by VH 5-HT1ARs in anxiety and panic processing, and their results are conflicting. Here we report that intra-VH administration of the 5-HT1A receptor agonist 8-OH-DPAT, the endogenous agonist serotonin (5-HT), or the standard anxiolytic benzodiazepine midazolam impaired the acquisition of inhibitory avoidance in the elevated T-maze (ETM) of male Wistar rats, indicating an anxiolytic effect. Conversely, local injection of the 5-HT1AR antagonist WAY-100635 caused the opposite effect. These results were equally found in the Vogel conflict test. None of these drugs interfered with locomotor activity in the open-field test, nor did they alter the expression of the escape response in the ETM, a defensive behavior associated with panic. Pre-injection of a sub-effective dose of WAY-100635 in the VH blocked the anxiolytic effect of 5-HT or 8-OH-DPAT in the Vogel test, confirming the involvement of 5-HT1AR for this behavioral effect. The effect in this test was anxiety-selective as none of the drugs affected water consumption or nociception. In conclusion, our results suggest that 5-HT1ARs in the VH play a tonic inhibitory role in anxiety processing. These receptors, however, are not involved in the regulation of panic-related escape behavior.


Anxiety , Behavior, Animal/physiology , Hippocampus , Panic/physiology , Receptor, Serotonin, 5-HT1A/physiology , Animals , Anxiety/metabolism , Anxiety/physiopathology , Behavior, Animal/drug effects , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/physiopathology , Male , Panic/drug effects , Rats , Rats, Wistar , Serotonin Antagonists/pharmacology
7.
Behav Brain Res ; 404: 113161, 2021 04 23.
Article En | MEDLINE | ID: mdl-33571570

Serotonin (5-HT) neurotransmission has been associated with reward-related behaviour. Moreover, the serotonergic system modulates the basolateral amygdala (BLA), a structure involved in reward encoding, and reward prediction error. However, the role played by 5-HT on BLA during a reward-driven task has not been fully elucidated. In this paper, we investigated whether serotonergic modulation of the BLA is involved in reward-driven learning. To this end, we trained Long Evans rats in an operant conditioning task, and examined the effects of fluoxetine treatment (a selective serotonin reuptake inhibitor, 10 mg/kg) in combination with BLA lesions with NMDA (20 mg/mL) on extinction learning. We also investigated whether intra-BLA injection of the serotonergic 5-HT1A receptor agonist 8-OH DPAT, or antagonist WAY-100635, alters extinction performance. We found that fluoxetine treatment strongly accelerated extinction learning, while BLA lesions partially reverted this effect and slightly impaired consolidation of extinction. Stimulation and inhibition of 5-HT1A receptors in BLA induced opposite effects to those of fluoxetine, impairing or accelerating extinction performance, respectively. Our findings suggest that 5-HT modulates reward-driven learning, and 5-HT1A receptors located in the BLA are relevant for extinction.


Basolateral Nuclear Complex/drug effects , Conditioning, Operant/drug effects , Extinction, Psychological/drug effects , Receptor, Serotonin, 5-HT1A/drug effects , Serotonin/pharmacology , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Animals , Basolateral Nuclear Complex/metabolism , Basolateral Nuclear Complex/physiology , Biological Availability , Conditioning, Operant/physiology , Extinction, Psychological/physiology , Fluoxetine/pharmacology , Male , Piperazines/pharmacology , Pyridines/pharmacology , Rats , Rats, Long-Evans , Receptor, Serotonin, 5-HT1A/metabolism , Receptor, Serotonin, 5-HT1A/physiology , Reward , Serotonin/pharmacokinetics , Serotonin 5-HT1 Receptor Agonists/pharmacology , Serotonin 5-HT1 Receptor Antagonists/pharmacology
8.
Behav Brain Res ; 404: 113159, 2021 04 23.
Article En | MEDLINE | ID: mdl-33571572

Antidepressant drugs are first-line treatment for panic disorder. Facilitation of 5-HT1A receptor-mediated neurotransmission in the dorsal periaqueductal gray (dPAG), a key panic-associated area, has been implicated in the panicolytic effect of the selective serotonin reuptake inhibitor fluoxetine. However, it is still unknown whether this mechanism accounts for the antipanic effect of other classes of antidepressants drugs (ADs) and whether the 5-HT interaction with 5-HT2C receptors in this midbrain area (which increases anxiety) is implicated in the anxiogenic effect caused by short-term treatment with ADs. The results showed that previous injection of the 5-HT1A receptor antagonist WAY-100635 in the dPAG blocked the panicolytic-like effect caused by chronic systemic administration of the tricyclic AD imipramine in male Wistar rats tested in the elevated T-maze. Neither chronic treatment with imipramine nor fluoxetine changed the expression of 5-HT1A receptors in the dPAG. Treatment with these ADs also failed to significantly change ERK1/2 (extracellular-signal regulated kinase) phosphorylation level in this midbrain area. Blockade of 5-HT2C receptors in the dPAG with the 5-HT2C receptor antagonist SB-242084 did not change the anxiogenic effect caused by a single acute injection of fluoxetine or imipramine in the Vogel conflict test. These results reinforce the view that the facilitation of 5-HT1A receptor-mediated neurotransmission in the dPAG is a common mechanism involved in the panicolytic effect caused by chronic administration of ADs. On the other hand, the anxiogenic effect observed after short-term treatment with these drugs does not depend on 5-HT2C receptors located in the dPAG.


Antidepressive Agents/pharmacology , Anxiety/drug therapy , Panic/drug effects , Periaqueductal Gray/drug effects , Receptor, Serotonin, 5-HT1A/physiology , Receptor, Serotonin, 5-HT2C/physiology , Aminopyridines/pharmacology , Animals , Blotting, Western , Elevated Plus Maze Test , Fluoxetine/pharmacology , Imipramine/pharmacology , Indoles/pharmacology , Male , Open Field Test/drug effects , Periaqueductal Gray/metabolism , Periaqueductal Gray/physiology , Piperazines/pharmacology , Pyridines/pharmacology , Rats , Rats, Wistar , Receptor, Serotonin, 5-HT1A/drug effects , Receptor, Serotonin, 5-HT1A/metabolism , Receptor, Serotonin, 5-HT2C/drug effects , Receptor, Serotonin, 5-HT2C/metabolism , Serotonin 5-HT1 Receptor Antagonists/pharmacology
9.
Sci Rep ; 10(1): 16622, 2020 10 06.
Article En | MEDLINE | ID: mdl-33024202

Rats show mutual-reward preferences, i.e., they prefer options that result in a reward for both themselves and a conspecific partner to options that result in a reward for themselves, but not for the partner. In a previous study, we have shown that lesions of the basolateral amygdala (BLA) reduced choices for mutual rewards. Here, we aimed to explore the role of 5-HT1A receptors within the BLA in mutual-reward choices. Rats received daily injections of either 50 or 25 ng of the 5-HT1A receptor agonist 8-OH-DPAT or a vehicle solution into the BLA and mutual-reward choices were measured in a rodent prosocial choice task. Compared to vehicle injections, 8-OH-DPAT significantly increased mutual-reward choices when a conspecific was present. By contrast, mutual-reward choices were significantly reduced by 8-OH-DPAT injections in the presence of a toy rat. The effect of 8-OH-DPAT injections was statistically significant during the expression, but not during learning of mutual-reward behavior, although an influence of 8-OH-DPAT injections on learning could not be excluded. There were no differences between 8-OH-DPAT-treated and vehicle-treated rats in general reward learning, behavioral flexibility, locomotion or anxiety. In this study, we have shown that repeated injections of the 5-HT1A receptor agonist 8-OH-DPAT have the potential to increase mutual-reward choices in a social setting without affecting other behavioral parameters.


8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Amygdala/drug effects , Amygdala/physiology , Behavior, Animal/drug effects , Choice Behavior/drug effects , Learning/drug effects , Receptor, Serotonin, 5-HT1A/physiology , Reward , Serotonin 5-HT1 Receptor Agonists/pharmacology , 8-Hydroxy-2-(di-n-propylamino)tetralin/administration & dosage , Animals , Anxiety , Injections , Locomotion/drug effects , Rats , Serotonin 5-HT1 Receptor Agonists/administration & dosage , Social Behavior
10.
J Neurosci ; 40(23): 4551-4564, 2020 06 03.
Article En | MEDLINE | ID: mdl-32350040

Forming effective responses to threatening stimuli requires the adequate and coordinated emergence of stress-related internal states. Such ability depends on early-life experiences and, in connection, the adequate formation of neuromodulatory systems, particularly serotonergic signaling. Here, we assess the serotonergic background of experience-dependent behavioral responsiveness using male and female zebrafish (Danio rerio). For the first time, we have characterized a period during behavioral metamorphosis in which zebrafish are highly reactive to their environment. Absence of social stimuli during this phase established by isolated rearing fundamentally altered the behavioral phenotype of postmetamorphic zebrafish in a challenge-specific manner, partially due to reduced responsiveness and an inability to develop stress-associated arousal state. In line with this, isolation differentially affected whole-brain serotonergic signaling in resting and stress-induced conditions, an effect that was localized in the dorsal pallium and was negatively associated with responsiveness. Administration of the serotonin receptor 1A partial agonist buspirone prevented the isolation-induced serotonin response to novelty in the level of the whole brain and the forebrain as well, without affecting catecholamine levels, and rescued stress-induced arousal along with challenge-induced behaviors, which together indicates functional connection between these changes. In summary, there is a consistent negative association between behavioral responsiveness and serotonergic signaling in zebrafish, which is well recognizable through the modifying effects of developmental perturbation and pharmacological manipulations as well. Our results imply a conserved serotonergic mechanism that context-dependently modulates environmental reactivity and is highly sensitive to experiences acquired during a specific early-life time window, a phenomenon that was previously only suggested in mammals.SIGNIFICANCE STATEMENT The ability to respond to challenges is a fundamental factor in survival. We show that zebrafish that lack appropriate social stimuli in a sensitive developmental period show exacerbated alertness in nonstressful conditions while failing to react adequately to stressors. This shift is reflected inversely by central serotonergic signaling, a system that is implicated in numerous mental disorders in humans. Serotonergic changes in brain regions modulating responsivity and behavioral impairment were both prevented by the pharmacological blockade of serotonergic function. These results imply a serotonergic mechanism in zebrafish that transmits early-life experiences to the later phenotype by shaping stress-dependent behavioral reactivity, a phenomenon that was previously only suggested in mammals. Zebrafish provide new insights into early-life-dependent neuromodulation of behavioral stress-responses.


Arousal/physiology , Avoidance Learning/physiology , Behavior, Animal/physiology , Receptor, Serotonin, 5-HT1A/physiology , Serotonin Receptor Agonists/pharmacology , Serotonin/physiology , Animals , Arousal/drug effects , Avoidance Learning/drug effects , Behavior, Animal/drug effects , Female , Male , Social Isolation/psychology , Zebrafish
11.
Neuropharmacology ; 170: 108070, 2020 06 15.
Article En | MEDLINE | ID: mdl-32229140

The majority of the fibroblast growth factor receptor 1-serotonin 1 A receptor (FGFR1-5-HT1AR) heterocomplexes in the hippocampus appeared to be located mainly in the neuronal networks and a relevant target for antidepressant drugs. Through a neurochemical and electrophysiological analysis it was therefore tested in the current study if astrocytic FGFR1-5-HT1AR heterocomplexes also exist in hippocampus. They may modulate the structure and function of astroglia in the hippocampus leading to possible changes in the gamma oscillations. Localization of hippocampal FGFR1-5-HT1AR heterocomplexes in astrocytes was found using in situ proximity ligation assay combined with immunohistochemistry using glial fibrillary acidic protein (GFAP) immunoreactivity as a marker for astroglia. Acute i.c.v. treatment with 8-OH-DPAT alone or together with basic fibroblast growth factor (FGF2) significantly increased FGFR1-5-HT1AR heterocomplexes in the GFAP positive cells, especially in the polymorphic layer of the dentate gyrus (PoDG) but also in the CA3 area upon combined treatment. No other hippocampal regions were studied. Also, structural plasticity changes were observed in the astrocytes, especially in the PoDG region, upon these pharmacological treatments. They may also be of relevance for enhancing the astroglial volume transmission with increased modulation of the neuronal networks in the regions studied. The effects of combined FGF2 and 5-HT agonist treatments on gamma oscillations point to a significant antagonistic interaction in astroglial FGFR1-5-HT1AR heterocomplexes that may contribute to counteraction of the 5-HT1AR-mediated decrease of gamma oscillations. This article is part of the special issue entitled 'Serotonin Research: Crossing Scales and Boundaries'.


Astrocytes/physiology , Fibroblast Growth Factor 2/physiology , Gamma Rhythm/physiology , Hippocampus/physiology , Receptor, Fibroblast Growth Factor, Type 1/physiology , Receptor, Serotonin, 5-HT1A/physiology , Serotonin/physiology , Animals , Astrocytes/drug effects , Gamma Rhythm/drug effects , Hippocampus/drug effects , Male , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , Receptor, Fibroblast Growth Factor, Type 1/agonists , Serotonin 5-HT1 Receptor Agonists/pharmacology
12.
Elife ; 92020 03 25.
Article En | MEDLINE | ID: mdl-32209227

To facilitate smoking genetics research we determined whether a screen of mutagenized zebrafish for nicotine preference could predict loci affecting smoking behaviour. From 30 screened F3 sibling groups, where each was derived from an individual ethyl-nitrosurea mutagenized F0 fish, two showed increased or decreased nicotine preference. Out of 25 inactivating mutations carried by the F3 fish, one in the slit3 gene segregated with increased nicotine preference in heterozygous individuals. Focussed SNP analysis of the human SLIT3 locus in cohorts from UK (n=863) and Finland (n=1715) identified two variants associated with cigarette consumption and likelihood of cessation. Characterisation of slit3 mutant larvae and adult fish revealed decreased sensitivity to the dopaminergic and serotonergic antagonist amisulpride, known to affect startle reflex that is correlated with addiction in humans, and increased htr1aa mRNA expression in mutant larvae. No effect on neuronal pathfinding was detected. These findings reveal a role for SLIT3 in development of pathways affecting responses to nicotine in zebrafish and smoking in humans.


Conditioning, Classical/physiology , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Nicotine/administration & dosage , Tobacco Smoking/genetics , Zebrafish Proteins/genetics , Amisulpride/pharmacology , Animals , Bupropion/pharmacology , Choice Behavior , Conditioning, Classical/drug effects , Female , Genetic Loci , Humans , Male , Mutation , Polymorphism, Single Nucleotide , Receptor, Serotonin, 5-HT1A/physiology , Zebrafish
13.
J Ethnopharmacol ; 248: 112276, 2020 Feb 10.
Article En | MEDLINE | ID: mdl-31593812

ETHNOPHARMACOLOGICAL RELEVANCE: Salvia divinorum Epling & Játiva is a Mexican plant used not only in rituals but also in traditional medicine for pain relief. One of the most known bioactive compounds is salvinorin A, which acts centrally in kappa-type opioid receptors. AIM OF THE STUDY: Despite its traditional use as a medicinal plant, there is not enough scientific investigation to reinforce its potential as analgesic. In this study, Salvia divinorum antinociceptive activity was evaluated in experimental models of nociceptive pain; the writhing test and formalin-induced licking behavior in mice. MATERIAL AND METHODS: Different Salvia divinorum extracts were prepared by maceration at room temperature in increased polarity (hexane, ethyl acetate and methanol). The ethyl acetate extract (EAEx) was chosen in order to be fractioned and to obtain a mixture of salvinorins. The antinociceptive effect of EAEx (3, 10, 30, and 100 mg/kg, i.p.) was compared with that of tramadol (a partial opioid agonist analgesic drug, 30 mg/kg, i.p.) and the mixture of salvinorins (30 mg/kg, i.p.). In addition, a participation of opioids (naloxone, NX 1 and/or 3 mg/kg, i.p.) and serotonin 5-HT1A receptors (WAY100635, 0.32 mg/kg, i.p.) was investigated as possible inhibitory neurotransmission involved. RESULTS: As a result, the EAEx produced significant and dose-dependent antinociceptive effect concerning salvinorins constituents. This effect was blocked in the presence of NX and WAY100635 in the abdominal test, but only by NX in the formalin-induced licking behavior. Whereas, the effect of salvinorins mixture involved opioids and serotonin 5-HT1A receptors. CONCLUSION: Data provide evidence of the potential of this species, where salvinorin A is in part responsible bioactive constituent involving participation of the opioids and/or 5-HT1A serotonin receptors depending on the kind of pain model explored.


Analgesics/therapeutic use , Diterpenes, Clerodane/therapeutic use , Pain/drug therapy , Salvia , Animals , Disease Models, Animal , Female , Male , Mice , Plant Leaves , Receptor, Serotonin, 5-HT1A/physiology , Receptors, Opioid/physiology
14.
Med Hypotheses ; 133: 109407, 2019 Dec.
Article En | MEDLINE | ID: mdl-31586811

Attention Deficit Hyperactivity Disorder (ADHD) is one of the most common childhood behavioural disorders, the frontline treatments for which are drugs with abuse potential. As a consequence, there is an urgent need to develop non addictive drug treatments with equivalent efficacy. Preclinical evidence suggests that selective serotonin uptake inhibitors (SSRIs) are likely to be effective in ADHD, however clinical reports suggest that SSRIs are of limited therapeutic value for the treatment of ADHD. We propose that this disconnect can be explained by the pattern of drug administration in existing clinical trials (administration for short periods of time, or intermittently) leading to inadequate control of the autoregulatory processes which control 5-HT release, most notably at the level of inhibitory 5-HT1A somatodendritic autoreceptors. These autoreceptors reduce the firing rate of 5-HT neurons (limiting release) unless they are desensitised by a long term, frequent pattern of drug administration. As such, we argue that the participants in earlier trials were not administered SSRIs in a manner which realises any potential benefits of targeting 5-HT in the pharmacotherapy of ADHD. In light of this, we hypothesise that there may be under-researched potential to exploit 5-HT transmission therapeutically in ADHD, either through changing the administration regime, or by pharmacological means. Recent pharmacological research has successfully potentiated the effects of SSRIs in acute animal preparations by antagonising inhibitory 5-HT1A autoreceptors prior to the administration of the SSRI fluoxetine. We suggest that combination therapies linking SSRIs and 5-HT1A antagonists are a potential way forward in the development of efficacious non-addictive pharmacotherapies for ADHD.


Attention Deficit Disorder with Hyperactivity/drug therapy , Selective Serotonin Reuptake Inhibitors/therapeutic use , Superior Colliculi/physiopathology , Animals , Attention Deficit Disorder with Hyperactivity/physiopathology , Clinical Trials as Topic/methods , Dioxanes/administration & dosage , Dioxanes/therapeutic use , Drug Administration Schedule , Drug Synergism , Drug Therapy, Combination , Humans , Pindolol/pharmacology , Pindolol/therapeutic use , Piperazines/administration & dosage , Piperazines/therapeutic use , Rats , Rats, Inbred SHR , Rats, Inbred Strains , Receptor, Serotonin, 5-HT1A/physiology , Saccades/physiology , Serotonergic Neurons/drug effects , Serotonergic Neurons/physiology , Serotonin/physiology , Serotonin 5-HT1 Receptor Antagonists/administration & dosage , Serotonin 5-HT1 Receptor Antagonists/pharmacology , Serotonin 5-HT1 Receptor Antagonists/therapeutic use , Selective Serotonin Reuptake Inhibitors/administration & dosage , Selective Serotonin Reuptake Inhibitors/pharmacology , Superior Colliculi/drug effects
15.
Behav Brain Res ; 376: 112207, 2019 12 30.
Article En | MEDLINE | ID: mdl-31476331

Although the dorsal hippocampus (dHPC) and serotonin1A (5-HT1A) receptor are involved in cognition, their roles in cognitive impairments in Parkinson' disease (PD) are still unclear. In the present study, the effects of the 5-HT1A receptor agonist 8-OH-DPAT and antagonist WAY100635 administrated into the dHPC of rats were assessed in T-maze rewarded alternation test for working memory and in hole-board test for long-term habituation. Unilateral 6-hydroxydopamine (6-OHDA) lesions of the medial forebrain bundle in rats impaired working memory and long-term habituation, decreased dopamine (DA) levels in the medial prefrontal cortex (mPFC), dHPC and ventral hippocampus (vHPC), and decreased the mean density of 5-HT1A receptors and co-localization of 5-HT1A receptor and excitatory amino acid carrier 1-immunoreactive (EAAC1-ir) neurons in the dHPC compared to sham-operated rats. Activation of dHPC 5-HT1A receptors by local infusion of 8-OH-DPAT impaired working memory and long-term habituation in both sham-operated and the 6-OHDA-lesioned rats. Furthermore, blockade of dHPC 5-HT1A receptors by WAY100635 improved the memories in the 6-OHDA-lesioned rats, but had no effects in sham-operated rats. Additionally, dHPC injection of 8-OH-DPAT decreased noradrenaline (NA) levels, increased 5-HT levels in the mPFC, dHPC and vHPC in sham-operated and lesioned rats, while WAY100635 increased DA and NA levels only in lesioned rats. The results of the present study suggest that dHPC 5-HT1A receptors regulate cognitive impairments in PD by changes of monoamines in the related brain regions.


Memory/physiology , Parkinson Disease/metabolism , Receptor, Serotonin, 5-HT1A/metabolism , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Animals , Disease Models, Animal , Dopamine/metabolism , Habituation, Psychophysiologic/physiology , Hippocampus/metabolism , Hippocampus/physiology , Male , Medial Forebrain Bundle/drug effects , Memory, Short-Term/physiology , Neurons/metabolism , Parkinson Disease/physiopathology , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/physiopathology , Piperazines/pharmacology , Prefrontal Cortex/drug effects , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT1A/physiology , Serotonin/metabolism , Serotonin Receptor Agonists/pharmacology , Temporal Lobe/metabolism
16.
Article En | MEDLINE | ID: mdl-31054943

Long-term single housing increases aggressive behavior in mice, a condition named isolation-induced aggression or territorial aggression, which can be attenuated by anxiolytic, antidepressant, and antipsychotic drugs. Preclinical and clinical findings indicate that cannabidiol (CBD), a non-psychotomimetic compound from Cannabis sativa, has anxiolytic, antidepressant, and antipsychotic properties. Few studies, however, have investigated the effects of CBD on aggressive behaviors. Here, we investigated whether CBD (5, 15, 30, and 60 mg/kg; i.p.) could attenuate social isolation-induced aggressive behavior in the resident-intruder test. Male Swiss mice (7-8 weeks) were single-housed for 10 days (resident mice) to induce aggressive behaviors, while conspecific mice of same sex and age (intruder mice) were group-housed. During the test, the intruder was placed into the resident's home-cage and aggressive behaviors initiated by the resident, including the latency for the first attack, number of attacks, and total duration of aggressive encounters, were recorded. The involvement of 5-HT1A and CB1 receptors (CB1R) in the effects of CBD was also investigated. All tested CBD doses induced anti-aggressive effects, indicated by a decrease in the number of attacks. CBD, at intermediary doses (15 and 30 mg/kg), also increased latency to attack the intruder and decreased the duration of aggressive encounters. No CBD dose interfered with locomotor behavior. CBD anti-aggressive effects were attenuated by the 5-HT1A receptor antagonist WAY100635 (0.3 mg/kg) and the CB1 antagonist AM251 (1 mg/kg), suggesting that CBD decreases social isolation-induced aggressive behaviors through a mechanism associated with the activation of 5-HT1A and CB1 receptors. Also, CBD decreased c-Fos protein expression, a neuronal activity marker, in the lateral periaqueductal gray (lPAG) in social-isolated mice exposed to the resident-intruder test, indicating a potential involvement of this brain region in the drug effects. Taken together, our findings suggest that CBD may be therapeutically useful to treat aggressive behaviors that are usually associated with psychiatric disorders.


Aggression/drug effects , Aggression/physiology , Cannabidiol/antagonists & inhibitors , Cannabidiol/pharmacology , Receptor, Cannabinoid, CB1/physiology , Receptor, Serotonin, 5-HT1A/physiology , Social Isolation , Animals , Cannabinoid Receptor Antagonists/pharmacology , Dose-Response Relationship, Drug , Housing, Animal , Male , Mice , Motor Activity/drug effects , Periaqueductal Gray/metabolism , Periaqueductal Gray/physiology , Piperazines/pharmacology , Piperidines/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Pyrazoles/pharmacology , Pyridines/pharmacology , Serotonin 5-HT1 Receptor Antagonists/pharmacology
17.
Brain Res ; 1715: 27-34, 2019 07 15.
Article En | MEDLINE | ID: mdl-30898674

The aim of the present study was to determine the involvement of the VTA 5-HT1A receptors in analgesia induced by the co-administration of morphine and dextromethorphan (DM). Male Wistar rats were bilaterally cannulated in the VTA by the stereotaxic instrument. The tail-flick and formalin tests were performed to assess nociception in the acute and tonic pain conditions respectively. The present data indicated that intraperitoneal (i.p.) administration of morphine increased the tail-flick latency (1-4 mg/kg) and decreased the pain score of formalin test (2-8 mg/kg), showing an analgesic effect. Co-administration of ineffective doses of morphine (1 or 2 mg/kg) with DM (30 mg/kg, i.p.) induced analgesia in both animal models. Interestingly, intra-VTA microinjection of 5HT1A receptors antagonist, S-WAY100-135 (0.5 and 1 µg/kg), inhibited the analgesic effect of morphine plus DM in both acute and tonic pain models. It should be considered that the same doses of DM or S-WAY100-135 by itself had no effects on antinociception in the animal models. Overall, these results indicated that systemic blockade of NMDA receptors via DM administration potentiated the response of a low dose of morphine to induce analgesic effect. Additionally, it seems that the VTA serotonergic system via 5HT1A receptors mediates the potentiative effect of DM on morphine-induced analgesia.


Pain/physiopathology , Receptor, Serotonin, 5-HT1A/metabolism , Ventral Tegmental Area/metabolism , Analgesia/methods , Analgesics, Opioid/pharmacology , Animals , Dextromethorphan/metabolism , Dextromethorphan/pharmacology , Male , Models, Animal , Morphine/metabolism , Morphine/pharmacology , Nociception/drug effects , Nucleus Accumbens/drug effects , Pain Management/methods , Rats , Rats, Wistar , Receptor, Serotonin, 5-HT1A/physiology , Receptors, N-Methyl-D-Aspartate/drug effects , Serotonin/metabolism , Ventral Tegmental Area/physiology
18.
Psychopharmacology (Berl) ; 236(7): 2273-2281, 2019 Jul.
Article En | MEDLINE | ID: mdl-30903210

RATIONALE: Pre-clinical and clinical studies have suggested that the antidepressant efficacy of escitalopram (ESC) can be augmented by co-administration of aripiprazole (ARI). OBJECTIVE: To establish if the effects of ESC + ARI can be altered by modulating the 5-HT1a receptor. METHODS: Sprague-Dawley male rats received ESC + ARI (10 and 2 mg/kg/day, respectively, via osmotic or by cumulative injections), as well as the 5-HT1a antagonist WAY-100635 (WAY; 0.01-1 mg/kg) and the 5-HT1a agonist 8-OH-DPAT (DPAT; 0.3-1 mg/kg) prior to testing in locomotion chambers and in the forced swim test (FST). Expression of the 5-HT1a receptor mRNA in the dorsal raphe nucleus, hippocampus, septum, and entorhinal cortex was also assessed. RESULTS: WAY generally synergized, while DPAT antagonized, the effect of ESC + ARI on motor activity. All groups showed significantly lower 5-HT1a mRNA in the dorsal raphe nucleus. In the hippocampus, ESC + ARI and WAY + ESC + ARI groups displayed equivalent elevations of 5-HT1a mRNA, but this was not observed in groups that received DPAT + ESC + ARI. Finally, the addition of ARI to ESC augmented the effect that ESC alone had on reducing immobility in the FST. Importantly, WAY antagonized this effect, while DPAT had no consequences. CONCLUSIONS: Taken together, these results in rats indicate that the 5-HT1a receptor is involved in the behavioral and brain region-specific mRNA effects of ESC + ARI.


Aripiprazole/administration & dosage , Citalopram/administration & dosage , Receptor, Serotonin, 5-HT1A/physiology , Selective Serotonin Reuptake Inhibitors/administration & dosage , Serotonin 5-HT1 Receptor Agonists/administration & dosage , 8-Hydroxy-2-(di-n-propylamino)tetralin/administration & dosage , Animals , Antidepressive Agents/administration & dosage , Hippocampus/drug effects , Hippocampus/physiology , Locomotion/drug effects , Locomotion/physiology , Male , Rats , Rats, Sprague-Dawley , Serotonin 5-HT1 Receptor Antagonists/pharmacology , Swimming/physiology , Swimming/psychology
19.
Naunyn Schmiedebergs Arch Pharmacol ; 392(6): 743-754, 2019 06.
Article En | MEDLINE | ID: mdl-30783717

Therapy of depression is difficult and still insufficient despite the presence of many antidepressants on the market. Therefore, there is a constant need to search for new, safer, and more effective drugs that could be used in the treatment of depression. Among many methods, chemical modification is an important strategy for new drug development. This study evaluates antidepressant-like effects and possible mechanism of action of two new arylpiperazine derivatives with isonicotinic and picolinic nuclei, compounds 4pN-(3-(4-(piperonyl)piperazin-1-yl)propyl) isonicotinamide and 3oN-(2-(4-(pyrimidin-2-yl)piperazin-1-yl)ethyl) picolinamide. The forced swim test (FST) and tail suspension test (TST), as two predictive tests for antidepressant effect in mice, were used. The possible involvement of serotonergic system in the effects of the new compounds in the FST through pharmacological antagonists/modulators of serotonergic transmission was also investigated. Compounds 4p and 3o were shown to possess antidepressant activity in both tests, FST and TST. The antidepressant-like effects of the new compounds in the FST were prevented by pretreatment of mice with pCPA (serotonin depletor), (-)pindolol (mixed 5-HT1A/1B and ß-adrenergic antagonist), and WAY 100635 (selective 5-HT1A antagonist). Additionally, in drug interaction studies, the 5-HT2A/2C antagonist, ketanserin, and the classic antidepressant, imipramine, potentiated antidepressant-like effect of both new compounds. The obtained results demonstrate that the new compounds 4p and 3o produce an antidepressant-like effect in mice which seems to be mediated by interaction with the serotonin 5-HT1A receptors and in the case of 4p, also with the 5-HT2C receptors.


Antidepressive Agents/pharmacology , Piperazines/pharmacology , Receptors, Serotonin/drug effects , Animals , Hindlimb Suspension , Imipramine/pharmacology , Ketanserin/pharmacology , Male , Mice , Motor Activity/drug effects , Picolines , Receptor, Serotonin, 5-HT1A/physiology , Receptor, Serotonin, 5-HT2C/physiology , Receptors, Serotonin/physiology
20.
Bull Exp Biol Med ; 166(3): 306-309, 2019 Jan.
Article En | MEDLINE | ID: mdl-30656521

Various types of adaptive behavior during the prepubertal period were analyzed in the offspring of rats receiving chronic injections of serotonin (5-HT) reuptake inhibitor fluoxetine, 5-HT1A receptor agonist buspirone, or their combination starting from gestation day 9 and subjected to immobilization stress from the 15th day of pregnancy until delivery. Prenatal stress increased pain sensitivity, prolonged inflammatory pain response, and increased the levels of anxiety and depression. Chronic administration of drugs acting through 5-HT1A receptors to pregnant rats improved the studied behavioral parameters in their offspring. Differences in the pain sensitivity were found between the effect of drug combination and each of them separately, and in the level of depression between combined administration and fluoxetine alone.


Adaptation, Psychological/drug effects , Buspirone/pharmacology , Fluoxetine/pharmacology , Prenatal Exposure Delayed Effects/drug therapy , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin Receptor Agonists/pharmacology , Stress, Psychological/drug therapy , Animals , Animals, Newborn , Anxiety/physiopathology , Anxiety/prevention & control , Anxiety/psychology , Behavior, Animal/drug effects , Depression/physiopathology , Depression/prevention & control , Depression/psychology , Drug Administration Schedule , Drug Combinations , Female , Fetus , Immobilization , Male , Pain/physiopathology , Pain/prevention & control , Pain/psychology , Pain Measurement , Pregnancy , Prenatal Exposure Delayed Effects/physiopathology , Prenatal Exposure Delayed Effects/psychology , Rats , Rats, Wistar , Receptor, Serotonin, 5-HT1A/physiology , Stress, Psychological/physiopathology , Stress, Psychological/psychology
...