Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Psychopharmacology (Berl) ; 239(1): 253-266, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34982171

ABSTRACT

RATIONALE: The voltage-insensitive, small-conductance calcium-activated potassium (SK) channel is a key regulator of neuronal depolarization and is implicated in the pathophysiology of depressive disorders. OBJECTIVE: We ascertained whether the SK channel is impaired in the chronic unpredictable stress (CUS) model and whether it can serve as a molecular target of antidepressant action. METHODS: We assessed the depressive-like behavioral phenotype of CUS-exposed rats and performed post-mortem SK channel binding and activity-dependent zif268 mRNA analyses on their brains. To begin an assessment of SK channel subtypes involved, we examined the effects of genetic and pharmacological inhibition of the SK3 channel using conditional knockout mice and selective SK3 channel negative allosteric modulators (NAMs). RESULTS: We found that [125I]apamin binding to SK channels is increased in the prefrontal cortex and decreased in the hippocampus, an effect that was associated with reciprocal levels of zif268 mRNA transcripts indicating abnormal regional cell activity in this model. We found that genetic and pharmacological manipulations significantly decreased immobility in the forced swim test without altering general locomotor activity, a hallmark of antidepressant-like activity. CONCLUSIONS: Taken together, these findings link depression-related neural and behavioral pathophysiology with abnormal SK channel functioning and suggest that this can be reversed by the selective inhibition of SK3 channels.


Subject(s)
Neurons , Small-Conductance Calcium-Activated Potassium Channels , Animals , Antidepressive Agents/pharmacology , Apamin , Calcium/metabolism , Mice , Neurons/metabolism , Rats , Small-Conductance Calcium-Activated Potassium Channels/genetics
2.
J Psychopharmacol ; 35(8): 1003-1016, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33908307

ABSTRACT

BACKGROUND: Antidepressant drugs in adolescent depression are sometimes mired by efficacy issues and paradoxical effects. Transcranial direct current stimulation (tDCS) could represent an alternative. AIMS/METHODS: We tested the antidepressant action of prefrontal tDCS and paroxetine (20 mg/kg, intraperitoneal) in olfactory bulbectomised (OBX) adolescent rats. Using enzyme-linked immunosorbent assays and in situ hybridisation, we examined treatment-induced changes in plasma brain-derived neurotrophic factor (BDNF) and brain serotonin transporter (SERT) and 5-HT-1A mRNA. RESULTS: OBX-induced anhedonia-like reductions in sucrose preference (SP) correlated with open field (OF) hyperactivity. These were accompanied by decreased zif268 mRNA in the piriform/amygdalopiriform transition area, and increased zif268 mRNA in the hypothalamus. Acute paroxetine (2 days) led to a profound SP reduction, an effect blocked by combined tDCS-paroxetine administration. Chronic (14 days) tDCS attenuated hyperlocomotion and its combination with paroxetine blocked OBX-induced SP reduction. Correlations among BDNF, SP and hyperlocomotion scores were altered by OBX but were normalised by tDCS-paroxetine co-treatment. In the brain, paroxetine increased zif268 mRNA in the hippocampal CA1 subregion and decreased it in the claustrum. This effect was blocked by tDCS co-administration, which also increased zif268 in CA2. tDCS-paroxetine co-treatment had variable effects on 5-HT1A receptors and SERT mRNA. 5-HT1A receptor changes were found exclusively within depression-related parahippocampal/hippocampal subregions, and SERT changes within fear/defensive response-modulating brainstem circuits. CONCLUSION: These findings point towards potential synergistic efficacies of tDCS and paroxetine in the OBX model of adolescent depression via mechanisms associated with altered expression of BDNF, 5-HT1A, SERT and zif268 in discrete corticolimbic areas.


Subject(s)
Depression/therapy , Paroxetine/pharmacology , Selective Serotonin Reuptake Inhibitors/pharmacology , Transcranial Direct Current Stimulation/methods , Animals , Brain-Derived Neurotrophic Factor/blood , Combined Modality Therapy , Depression/physiopathology , Disease Models, Animal , Male , Olfactory Bulb/surgery , Paroxetine/administration & dosage , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT1A/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism , Selective Serotonin Reuptake Inhibitors/administration & dosage
3.
Article in English | MEDLINE | ID: mdl-32725198

ABSTRACT

BACKGROUND: Chronic exposure to the Δ9-tetrahydrocannabinol (THC), the main cannabis pharmacological component, during adolescence has been shown to be associated with an increased risk of depression and suicidality in humans. AIMS: Little is known about the impact of the long-term effects of chronic exposure to low doses of THC in adolescent compared to adult rodents. METHODS: THC (1mg/kg i.p., once a day) or vehicle was administered for 20 days in both adolescent (post-natal day, PND 30-50) and young adult rats (PND 50-70). After a long washout period (20 days), several behavioral paradigms and electrophysiological recordings of serotonin (5-HT) and norepinephrine (NE) neurons were carried out. RESULTS: Adolescent THC exposure resulted in depressive lbehaviors: a significant decrease in latency to first immobility in the forced swim test, increased anhedonia in the sucrose preference test. Decrease entries in the open arm were observed in the elevated plus maze after adolescent and adult exposure, indicating anxiousphenotype. A significant reduction in dorsal raphe serotonergic neural activity without changing locus coeruleus noradrenergic neural activity was found in THC adolescent and adult exposure. CONCLUSIONS: Altogether, these findings suggest that low doses of chronic THC exposure during the developmental period and adulthood could result in increased vulnerability of the 5-HT system and anxiety symptoms; however, depressive phenotypes occur only after adolescent, but not adult exposure, underscoring the higher vulnerability of young ages to the mental effects of cannabis.

4.
Cereb Cortex ; 30(3): 1559-1572, 2020 03 14.
Article in English | MEDLINE | ID: mdl-31504265

ABSTRACT

The muscarinic acetylcholine receptor antagonist scopolamine elicits rapid antidepressant activity, but its underlying mechanism is not fully understood. In a chronic stress model, a single low-dose administration of scopolamine reversed depressive-like reactivity. This antidepressant-like effect was mediated via a muscarinic M1 receptor-SKC pathway because it was mimicked by intra-medial prefrontal cortex (intra-mPFC) infusions of scopolamine, of the M1 antagonist pirenzepine or of the SKC antagonist apamin, but not by the selective serotonin reuptake inhibitor (SSRI) antidepressant fluoxetine. Extracellular and whole-cell recordings revealed that scopolamine and ketamine attenuate the SKC-mediated action potential hyperpolarization current and rapidly enhance mPFC neuronal excitability within the therapeutically relevant time window. The SKC agonist 1-EBIO abrogated scopolamine-induced antidepressant activity at a dose that completely suppressed burst firing activity. Scopolamine also induced a slow-onset activation of raphe serotonergic neurons, which in turn was dependent on mPFC-induced neuroplasticity or excitatory input, since mPFC transection abolished this effect. These early behavioral and mPFC activational effects of scopolamine did not appear to depend on prefrontocortical brain-derived neurotrophic factor and serotonin-1A activity, classically linked to SSRIs, and suggest a novel mechanism associated with antidepressant response onset through SKC-mediated regulation of activity-dependent plasticity.


Subject(s)
Antidepressive Agents/pharmacology , Small-Conductance Calcium-Activated Potassium Channels/drug effects , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Stress, Physiological/drug effects , Action Potentials/drug effects , Animals , Ketamine/pharmacology , Male , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats, Inbred F344 , Scopolamine/pharmacology , Serotonergic Neurons/drug effects , Serotonergic Neurons/metabolism , Serotonin/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Stress, Physiological/physiology
5.
Psychopharmacology (Berl) ; 236(6): 1917-1929, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30796492

ABSTRACT

RATIONALE: The ventral tegmental area (VTA) is implicated in the pathophysiology of depression and addictive disorders and is subject to the detrimental effects of stress. Chronic stress may differentially alter the activity pattern of its different subregions along the rostrocaudal and dorsoventral axes, which may relate to the variable behavioral sensitivity to stress mediated by these subregions. OBJECTIVES: Here, chronic stress-exposed rats were tested for depressive-like reactivity. In situ hybridization for zif268 as a marker of neuronal activation was combined with in vivo single-unit recording of dopaminergic neurons to assess modifications in the activity of the rostral VTA (rVTA) and caudal VTA (cVTA). Changes in the expression of stress-responsive glucocorticoid receptors (GR) and brain-derived neurotrophic factor (BDNF) were also assessed. RESULTS: Stress-induced anhedonia-like, hyper-anxious, and passive-like responding were associated with reductions in dopaminergic burst activity in the cVTA and an increase in local GABAergic activity, particularly in GABAA receptor sensitivity. On the other hand, stress increased single-spiking activity, burst activity, and zif268 mRNA levels in the rVTA, which were associated with increased glutamatergic tonus and enhanced GR and AMPA receptor (AMPAR) expression. rVTA and cVTA activity differentially correlated with sucrose preference and passivity measures. CONCLUSIONS: These data demonstrate that the rVTA and cVTA respond differently to stress and suggest that while cVTA activity may be related to passivity-like states, the activity of both subregions appears to be related to anhedonia and the processing of incentive value. These region-dependent abnormalities indicate the multi-modular composition of the VTA, which could provide multiple substrates for different symptom features.


Subject(s)
Anhedonia/physiology , Stress, Psychological/metabolism , Stress, Psychological/psychology , Ventral Tegmental Area/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Chronic Disease , Depression/genetics , Depression/metabolism , Depression/psychology , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Early Growth Response Protein 1/biosynthesis , Early Growth Response Protein 1/genetics , Male , Rats , Rats, Inbred F344 , Stress, Psychological/genetics
6.
Neurobiol Dis ; 116: 166-178, 2018 08.
Article in English | MEDLINE | ID: mdl-29727711

ABSTRACT

Some evidence suggests that the cerebellum modulates affect via connectivities with mood-regulating corticolimbic structures, such as the prefrontal cortex and monoamine nuclei. In rats exposed to chronic unpredictable stress (CUS), we examined the neuro-behavioural effects of high frequency stimulation and surgical ablation/disconnection of the cerebellar vermis. CUS reduced sucrose preference, increased novelty-induced feeding suppression and passive coping. These depressive-like behaviours were associated with decreased cerebellar zif268 expression, indicating possible cerebellar involvement in stress pathology. These were paralleled by decreased vermal Purkinje simple and complex spiking activity and raphe serotonergic activity. Protracted (24-h) vermal stimulation reversed these behavioural deficits through serotonin-mediated mechanisms since this effect was abrogated by the serotonin-depleting agent pCPA. Vermal stimulation and disconnection lesion also enhanced serotonergic activity, but did not modify prefrontocortical pyramidal firing. This effect was likely mediated by 5-HT1A receptors (5-HT1AR). Indeed, acute vermal stimulation mimicked the effect of the 5-HT1AR agonist 8-OH-DPAT in inhibiting serotonergic activity, which was prevented by pre-treatment with the 5-HT1AR antagonist WAY100,635. These results demonstrate vermal involvement in depressive-type behaviour via its modulatory action on serotonergic neurons. They further suggest that vermal and mPFC stimulation may bestow therapeutic benefits via parallel pathways.


Subject(s)
Cerebellar Vermis/physiology , Deep Brain Stimulation/methods , Dorsal Raphe Nucleus/physiology , Prefrontal Cortex/physiology , Stress, Psychological/physiopathology , Stress, Psychological/psychology , Animals , Chronic Disease , Male , Rats , Rats, Wistar , Stress, Psychological/therapy
7.
Neuropsychopharmacology ; 43(5): 1099-1106, 2018 04.
Article in English | MEDLINE | ID: mdl-28862251

ABSTRACT

Deep brain stimulation (DBS) is being investigated for a number of psychiatric indications, including posttraumatic stress disorder (PTSD). Preclinical studies continue to be a cornerstone for the development of new DBS applications. We investigate whether DBS delivered to the infralimbic cortex (IL), a region involved in mechanisms of stress resiliency, may counter behavioral abnormalities in rats that present persistent extinction deficits and long-term anxiety after exposure to fear conditioning. Rats undergoing fear conditioning/extinction were segregated into weak and strong extinction groups (WE >70% or SE <30% of freezing during extinction). Following 2 weeks of DBS, animals were exposed to novel recall sessions and tested in the open field, novelty-suppressed feeding, and elevated plus maze. zif268 expression was measured in structures involved in mechanisms of fear and stress. In vivo electrophysiology was used to record activity from the basolateral amygdala (BLA). We found that DBS improved extinction deficits and anxiety-like behavior in WE animals, having no significant effects in SE rats. No major differences in absolute zif268 levels were recorded across groups. However, correlation between zif268 expression in the IL and BLA was disrupted in WE animals, a deficit that was countered by DBS treatment. Electrophysiology experiments have shown that DBS reduced BLA firing of both putative principal cells and interneurons in WE rats, with no significant differences being detected between SE and SE DBS animals. In summary, IL DBS mitigated fear, partially improved anxiety-like behavior, reversed neurocircuitry abnormalities, and reduced BLA cell firing in a preclinical model of PTSD.


Subject(s)
Anxiety/physiopathology , Basolateral Nuclear Complex/physiology , Deep Brain Stimulation , Fear/physiology , Prefrontal Cortex/physiology , Stress Disorders, Post-Traumatic/physiopathology , Animals , Basolateral Nuclear Complex/metabolism , Behavior, Animal/physiology , Conditioning, Psychological/physiology , Early Growth Response Protein 1/biosynthesis , Extinction, Psychological/physiology , Immobility Response, Tonic/physiology , Male , Prefrontal Cortex/metabolism , Rats
8.
Int J Neuropsychopharmacol ; 18(3)2015 Jan 31.
Article in English | MEDLINE | ID: mdl-25638817

ABSTRACT

BACKGROUND: Melancholic depression, described also as endogenous depression, is a mood disorder with distinctive specific psychopathological features and biological homogeneity, including anhedonia, circadian variation of mood, psychomotor activation, weight loss, diurnal cortisol changes, and sleep disturbances. Although several hypotheses have been proposed, the etiology of this disorder is still unknown. METHODS: Behavioral, electrophysiological and biochemical approaches were used to characterize the emotional phenotype, serotonergic and noradrenergic electrical activity, and corticosterone in melatonin MT1 receptor knockout mice and their wild type counterparts, during both light and dark phases. RESULTS: Melatonin MT1 receptor knockout mice have decreased mobility in the forced swim and tail suspension tests as well as decreased sucrose consumption, mostly during the dark/inactive phase. These mood variations are reversed by chronic treatment with the tricyclic antidepressant desipramine. In addition, MT1 receptor knockout mice exhibit psychomotor disturbances, higher serum levels of corticosterone the dark phase, and a blunted circadian variation of corticosterone levels. In vivo electrophysiological recordings show a decreased burst-firing activity of locus coeruleus norepinephrine neurons during the dark phase. The circadian physiological variation in the spontaneous firing activity of high-firing neuronal subpopulations of both norepinephrine neurons and dorsal raphe serotonin neurons are abolished in MT1 knockout mice. CONCLUSIONS: These data demonstrate that melatonin MT1 receptor knockout mice recapitulate several behavioral and neurobiological circadian changes of human melancholic depression and, for the first time, suggest that the MT1 receptor may be implicated in the pathogenesis of melancholic depression and is a potential pharmacological target for this mental condition.


Subject(s)
Chronobiology Disorders/genetics , Circadian Rhythm/genetics , Depressive Disorder/genetics , Receptor, Melatonin, MT1/deficiency , Animals , Antidepressive Agents, Tricyclic/therapeutic use , Chronobiology Disorders/drug therapy , Corticosterone/blood , Depressive Disorder/drug therapy , Desipramine/therapeutic use , Disease Models, Animal , Exploratory Behavior/drug effects , Feeding Behavior/drug effects , Food Preferences , Hindlimb Suspension , Male , Maze Learning/drug effects , Mice , Mice, Knockout , Receptor, Melatonin, MT1/genetics , Swimming
9.
Neuropharmacology ; 83: 79-88, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24721625

ABSTRACT

Nandrolone decanoate, an anabolic androgen steroid (AAS) illicitly used by adult and adolescent athletes to enhance physical performance and body image, induces psychiatric side effects, such as aggression, depression as well as a spectrum of adverse physiological impairments. Since adolescence represents a neurodevelopmental window that is extremely sensitive to the detrimental effects of drug abuse, we investigated the long-term behavioral and neurophysiological consequences of nandrolone abuse during adolescence. Adolescent rats received daily injections of nandrolone decanoate (15 mg/kg, i.m.) for 14 days (PND 40-53). At early adulthood (PND 68), forced swim, sucrose preference, open field and elevated plus maze tests were performed to assess behavioral changes. In vivo electrophysiological recordings were carried out to monitor changes in electrical activity of serotonergic neurons of the dorsal raphe nucleus (DRN) and noradrenergic neurons of the locus coeruleus (LC). Our results show that after early exposure to nandrolone, rats display depression-related behavior, characterized by increased immobility in the forced swim test and reduced sucrose intake in the sucrose preference test. In addition, adult rats presented anxiety-like behavior characterized by decreased time and number of entries in the central zone of the open field and decreased time spent in the open arms of the elevated plus maze. Nandrolone decreased the firing rate of spontaneously active serotonergic neurons in the DRN while increasing the firing rate of noradrenergic neurons in the LC. These results provide evidence that nandrolone decanoate exposure during adolescence alters the emotional profile of animals in adulthood and significantly modifies both serotonergic and noradrenergic neurotransmission.


Subject(s)
Adrenergic Neurons/drug effects , Anabolic Agents/toxicity , Anxiety/chemically induced , Depression/chemically induced , Dorsal Raphe Nucleus/drug effects , Nandrolone/analogs & derivatives , Serotonergic Neurons/drug effects , Synaptic Transmission/drug effects , Action Potentials/drug effects , Adrenergic Neurons/physiology , Age Factors , Anabolic Agents/administration & dosage , Animals , Anxiety/physiopathology , Behavior, Animal/drug effects , Dorsal Raphe Nucleus/physiology , Male , Nandrolone/administration & dosage , Nandrolone/toxicity , Nandrolone Decanoate , Rats , Rats, Sprague-Dawley , Serotonergic Neurons/physiology
10.
Neurosci Biobehav Rev ; 38: 173-92, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24300695

ABSTRACT

Chronic stressful life events are risk factors for developing major depression, the pathophysiology of which is strongly linked to impairments in serotonin (5-HT) neurotransmission. Exposure to chronic unpredictable stress (CUS) has been found to induce depressive-like behaviours, including passive behavioural coping and anhedonia in animal models, along with many other affective, cognitive, and behavioural symptoms. The heterogeneity of these symptoms represents the plurality of corticolimbic structures involved in mood regulation that are adversely affected in the disorder. Chronic stress has also been shown to negatively regulate adult hippocampal neurogenesis, a phenomenon that is involved in antidepressant effects and regulates subsequent stress responses. Although there exists an enormous body of data on stress-induced alterations of 5-HT activity, there has not been extensive exploration of 5-HT adaptations occurring presynaptically or at the level of the raphe nuclei after exposure to CUS. Similarly, although hippocampal neurogenesis is known to be negatively regulated by stress and positively regulated by antidepressant treatment, the role of neurogenesis in mediating affective behaviour in the context of stress remains an active area of investigation. The goal of this review is to link the serotonergic and neurogenic hypotheses of depression and antidepressant effects in the context of stress. Specifically, chronic stress significantly attenuates 5-HT neurotransmission and 5-HT1A autoreceptor sensitivity, and this effect could represent an endophenotypic hallmark for mood disorders. In addition, by decreasing neurogenesis, CUS decreases hippocampal inhibition of the hypothalamic-pituitary-adrenal (HPA) axis, exacerbating stress axis overactivity. Similarly, we discuss the possibility that adult hippocampal neurogenesis mediates antidepressant effects via the ventral (in rodents; anterior in humans) hippocampus' influence on the HPA axis, and mechanisms by which antidepressants may reverse chronic stress-induced 5-HT and neurogenic changes. Although data are as yet equivocal, antidepressant modulation of 5-HT neurotransmission may well serve as one of the factors that could drive neurogenesis-dependent antidepressant effects through these stress regulation-related mechanisms.


Subject(s)
Depressive Disorder/physiopathology , Hippocampus/physiopathology , Neurogenesis , Serotonin/metabolism , Stress, Psychological/physiopathology , Animals , Antidepressive Agents/therapeutic use , Brain/physiopathology , Depressive Disorder/drug therapy , Humans , Models, Neurological
11.
Curr Top Behav Neurosci ; 15: 243-91, 2013.
Article in English | MEDLINE | ID: mdl-23271325

ABSTRACT

Major depressive disorder has been associated with manifold pathophysiological changes. These include metabolic abnormalities in discreet brain areas; modifications in the level of stress hormones, neurotransmitters, and neurotrophic factors; impaired spinogenesis and synaptogenesis in crucial brain areas, such as the prefrontal cortex and the hippocampus; and impaired neurogenesis in the hippocampus. Antidepressant therapy facilitates remission by reversing most of these disturbances, indicating that these dysfunctions may participate causally in depressive symptomatology. However, few attempts have been made to integrate these different pathophysiologies into one model. The present chapter endeavors (1) to review the extant literature in the field, with particular focus on the role of neurogenesis and synaptogenesis in depression; (2) and to suggest a possible interplay between these two processes, as well as, describe the ways by which improving both neurogenesis and synaptogenesis may enable effective recovery by acting on a larger neuronal network.


Subject(s)
Antidepressive Agents/pharmacology , Brain , Depressive Disorder, Major , Neurogenesis/drug effects , Synapses/drug effects , Brain/drug effects , Brain/metabolism , Brain/physiopathology , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/metabolism , Depressive Disorder, Major/physiopathology , Humans , Synapses/metabolism , Synapses/pathology
12.
Prog Neuropsychopharmacol Biol Psychiatry ; 38(1): 88-96, 2012 Jul 02.
Article in English | MEDLINE | ID: mdl-22386778

ABSTRACT

Preclinical and clinical studies suggest that direct and indirect cannabinoid agonists, including enhancers of endocannabinoids, engender stress-relieving, anxiolytic and antidepressant effects, mediated by central CB(1) receptors (CB(1)Rs). The effect of the main pharmacologically active principle in cannabis, (-)-trans-Δ(9)-tetrahydrocannabinol (delta-9-THC), on depressive behavior and on the serotonin (5-HT) system, which is implicated in the mechanism of action of antidepressants, has not been extensively clarified. Here, we showed that repeated (5 days), but not single (acute) intraperitoneal (ip) treatment with delta-9-THC (1mg/kg) exerts antidepressant-like properties in the rat forced swim test (FST). This effect was CB(1)R-dependent because it was blocked by the CB(1)R antagonist rimonabant (1mg/kg, ip). Using in vivo electrophysiology, we demonstrated that delta-9-THC modulated dorsal raphe (DR) 5-HT neuronal activity through a CB(1)R-dependent mechanism. Acute intravenous delta-9-THC administration (0.1-1.5mg/kg) elicited a complex response profile, producing excitatory, inhibitory and inert responses of 5-HT neurons. Only excitatory responses were blocked by rimonabant. Finally, repeated but not single delta-9-THC administration (1mg/kg, ip) enhanced tonic 5-HT(1A) receptor activity in the hippocampus, a postsynaptic event commonly elicited by standard antidepressants. These results suggest that delta-9-THC, like other CB(1)R agonists and endocannabinoid enhancers, may possess antidepressant properties at low doses, and could modulate 5-HT transmission in the DR and hippocampus as standard antidepressants such as selective serotonin reuptake inhibitors.


Subject(s)
Adaptation, Psychological/drug effects , Behavior, Animal/drug effects , Dronabinol/pharmacology , Serotonergic Neurons/drug effects , Synaptic Transmission/drug effects , Animals , Hippocampus/drug effects , Hippocampus/metabolism , Male , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT1A/metabolism
13.
Int J Neuropsychopharmacol ; 15(9): 1319-30, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22053980

ABSTRACT

Despite the growing non-medical consumption of amphetamine (Amph) during adolescence, its long-term neurobiological and behavioural effects have remained largely unexplored. The present research sought to characterize the behavioural profile and electrophysiological properties of midbrain monoaminergic neurons in adult rodents after Amph exposure during adolescence. Adolescent rats were administered vehicle, 0.5, 1.5, or 5.0 mg/kg.d Amph from postnatal day (PND) 30-50. At adulthood (PND 70), rats were tested in an open-field test (OFT) and elevated plus maze (EPM), paralleled by in-vivo extracellular recordings of serotonin (5-HT), dopamine (DA) and norepinephrine (NE) neurons from the dorsal raphe nucleus, ventral tegmental area, and locus coeruleus, respectively. 5-HT firing in adulthood was increased in rats that had received Amph (1.5 mg/kg.d) during adolescence. At this regimen, DA firing activity was increased, but not NE firing. Conversely, the highest Amph dose regimen (5.0 mg/kg.d) enhanced NE firing, but not DA or 5-HT firing rates. In the OFT, Amph (1.5 mg/kg.d) significantly increased the total distance travelled, while the other doses were ineffective. In the EPM, all three Amph doses increased time spent in the open arms and central platform, as well as the number of stretch-attend postures made. Repeated adolescent exposure to Amph differentially augments monoaminergic neuronal firing in a dose-specific fashion in adulthood, with corresponding alterations in locomotion, risk assessment (stretch-attend postures and central platform occupancy) and risk-taking behaviours (open-arm exploration). Thus, adolescent Amph exposure induces long-lasting neurophysiological alterations that may have implications for drug-seeking behaviour in the future.


Subject(s)
Amphetamine/pharmacology , Behavior, Animal/drug effects , Biogenic Monoamines/physiology , Central Nervous System Stimulants/pharmacology , Synaptic Transmission/drug effects , Animals , Anxiety/psychology , Dopamine/physiology , Dose-Response Relationship, Drug , Electrophysiological Phenomena/drug effects , Female , Locus Coeruleus/physiology , Motor Activity/drug effects , Neurons/drug effects , Neurons/physiology , Norepinephrine/physiology , Pregnancy , Raphe Nuclei/physiology , Rats , Rats, Sprague-Dawley , Risk-Taking , Serotonin/physiology , Ventral Tegmental Area/physiology
14.
J Psychopharmacol ; 26(6): 830-44, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21730015

ABSTRACT

Melatonin (MLT) and serotonin (5-HT) are two biosynthetically related compounds implicated in several common physiological functions and the etiology of mood disorders. How they interact, though, is not yet fully understood. In this study, single-unit extracellular recordings were used to monitor dorsal raphe nucleus (DR) 5-HT neuronal activity in anesthetized rats, under basal conditions (CTRL), in response to MLT administration, and after pinealectomy (PX) across the light-dark cycle. Under basal conditions, the number of spontaneously active 5-HT neurons and their firing rate were both significantly lower in the dark phase. In the light phase, administration of MLT at low doses (0.5-1 mg/kg, i.v.) decreased 5-HT firing activity. This inhibitory effect of MLT was completely blocked by the MT1/MT2 receptor antagonist luzindole, but not by the selective MT(2) receptor antagonist 4P-PDOT, the selective 5-HT(1A) receptor antagonist WAY100635, or by the α2 adrenoceptor antagonist idazoxan. In the opposite experiment, PX increased 5-HT firing activity in the dark phase, and this was reversed by MLT administration (1 mg/kg, i.v.). Finally, in a forced swim test, MLT (1 mg/kg, i.p.) increased immobility time and decreased swimming behavior. Together, these results suggest that nocturnal MLT secretion imposes tonic inhibitory control over a sub-population of DR 5-HT neurons. This MLT-induced decrease in 5-HT neurotransmission may represent a biological mechanism underlying mood disorders characterized by increased MLT secretion, such as seasonal affective disorder.


Subject(s)
Melatonin/metabolism , Neurons/physiology , Photoperiod , Pineal Gland/metabolism , Raphe Nuclei/physiology , Serotonin/metabolism , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Animals , Idazoxan/pharmacology , Male , Mood Disorders/metabolism , Mood Disorders/physiopathology , Neurons/drug effects , Neurons/metabolism , Pineal Gland/drug effects , Pineal Gland/physiology , Piperazines/pharmacology , Pyridines/pharmacology , Raphe Nuclei/drug effects , Raphe Nuclei/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Melatonin, MT1/antagonists & inhibitors , Receptor, Melatonin, MT1/metabolism , Receptor, Melatonin, MT2/antagonists & inhibitors , Receptor, Melatonin, MT2/metabolism , Serotonin 5-HT1 Receptor Antagonists/pharmacology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Tetrahydronaphthalenes/pharmacology , Tryptamines/pharmacology
15.
J Neurosci ; 31(50): 18439-52, 2011 Dec 14.
Article in English | MEDLINE | ID: mdl-22171046

ABSTRACT

Melatonin activates two brain G-protein coupled receptors, MT(1) and MT(2), whose differential roles in the sleep-wake cycle remain to be defined. The novel MT(2) receptor partial agonist, N-{2-[(3-methoxyphenyl) phenylamino] ethyl} acetamide (UCM765), is here shown to selectively promote non-rapid eye movement sleep (NREMS) in rats and mice. The enhancement of NREMS by UCM765 is nullified by the pharmacological blockade or genetic deletion of MT(2) receptors. MT(2), but not MT(1), knock-out mice show a decrease in NREMS compared to the wild strain. Immunohistochemical labeling reveals that MT(2) receptors are localized in sleep-related brain regions, and notably the reticular thalamic nucleus (Rt). Microinfusion of UCM765 in the Rt promotes NREMS, and its systemic administration induces an increase in firing and rhythmic burst activity of Rt neurons, which is blocked by the MT(2) antagonist 4-phenyl-2-propionamidotetralin. Since developing hypnotics that increase NREMS without altering sleep architecture remains a medical challenge, MT(2) receptors may represent a novel target for the treatment of sleep disorders.


Subject(s)
Acetamides/pharmacology , Aniline Compounds/pharmacology , Neurons/drug effects , Receptor, Melatonin, MT2/metabolism , Sleep/drug effects , Thalamus/drug effects , Animals , Female , Male , Mice , Mice, Knockout , Rats , Rats, Sprague-Dawley , Receptor, Melatonin, MT2/agonists , Receptor, Melatonin, MT2/genetics
16.
Neuropsychopharmacology ; 35(10): 2083-100, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20571484

ABSTRACT

Pharmacological blockade of the anandamide-degrading enzyme, fatty acid amide hydrolase (FAAH), produces CB(1) receptor (CB(1)R)-mediated analgesic, anxiolytic-like and antidepressant-like effects in murids. Using behavioral and electrophysiological approaches, we have characterized the emotional phenotype and serotonergic (5-HT) activity of mice lacking the FAAH gene in comparison to their wild type counterparts, and their response to a challenge of the CB(1)R antagonist, rimonabant. FAAH null-mutant (FAAH(-/-)) mice exhibited reduced immobility in the forced swim and tail suspension tests, predictive of antidepressant activity, which was attenuated by rimonabant. FAAH(-/-) mice showed an increase in the duration of open arm visits in the elevated plus maze, and a decrease in thigmotaxis and an increase in exploratory rearing displayed in the open field, indicating anxiolytic-like effects that were reversed by rimonabant. Rimonabant also prolonged the initiation of feeding in the novelty-suppressed feeding test. Electrophysiological recordings revealed a marked 34.68% increase in dorsal raphe 5-HT neural firing that was reversed by rimonabant in a subset of neurons exhibiting high firing rates (33.15% mean decrease). The response of the prefrontocortical pyramidal cells to the 5-HT(2A/2C) agonist (+/-)-1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane ((+/-)-DOI) revealed desensitized 5-HT(2A/2C) receptors, likely linked to the observed anxiolytic-like behaviors. The hippocampal pyramidal response to the 5-HT(1A) antagonist, WAY-100635, indicates enhanced tonus on the hippocampal 5-HT(1A) heteroreceptors, a hallmark of antidepressant-like action. Together, these results suggest that FAAH genetic deletion enhances anxiolytic-like and antidepressant-like effects, paralleled by altered 5-HT transmission and postsynaptic 5-HT(1A) and 5-HT(2A/2C) receptor function.


Subject(s)
Amidohydrolases/deficiency , Brain/metabolism , Brain/pathology , Mood Disorders/genetics , Mood Disorders/pathology , Serotonin/metabolism , Action Potentials/drug effects , Animals , Behavior, Animal/drug effects , Benzamides/pharmacology , Cannabinoid Receptor Antagonists , Carbamates/pharmacology , Enzyme Inhibitors/pharmacology , Exploratory Behavior/drug effects , Hindlimb Suspension/methods , Hippocampus/metabolism , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Piperidines/pharmacology , Prefrontal Cortex/metabolism , Pyrazoles/pharmacology , Raphe Nuclei/metabolism , Rimonabant , Serotonin Agents/pharmacology , Swimming/psychology
17.
Neurobiol Dis ; 37(3): 641-55, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19969082

ABSTRACT

The pathophysiological neural mechanism underlying the depressogenic and anxiogenic effects of chronic adolescent cannabinoid use may be linked to perturbations in monoaminergic neurotransmission. We tested this hypothesis by administering the CB(1) receptor agonist WIN55,212-2, once daily for 20 days to adolescent and adult rats, subsequently subjecting them to tests for emotional reactivity paralleled by the in vivo extracellular recordings of serotonergic and noradrenergic neurons. Chronic adolescent exposure but not adult exposure to low (0.2 mg/kg) and high (1.0 mg/kg) doses led to depression-like behaviour in the forced swim and sucrose preference test, while the high dose also induced anxiety-like consequences in the novelty-suppressed feeding test. Electrophysiological recordings revealed both doses to have attenuated serotonergic activity, while the high dose also led to a hyperactivity of noradrenergic neurons only after adolescent exposure. These suggest that long-term exposure to cannabinoids during adolescence induces anxiety-like and depression-like behaviours in adulthood and that this may be instigated by serotonergic hypoactivity and noradrenergic hyperactivity.


Subject(s)
Biogenic Monoamines/agonists , Biogenic Monoamines/antagonists & inhibitors , Brain/drug effects , Cannabinoids/toxicity , Marijuana Abuse/physiopathology , Mental Disorders/chemically induced , Age Factors , Aging/physiology , Animals , Animals, Newborn , Anxiety Disorders/chemically induced , Anxiety Disorders/metabolism , Anxiety Disorders/physiopathology , Behavior, Animal/drug effects , Behavior, Animal/physiology , Benzoxazines/toxicity , Brain/growth & development , Brain/metabolism , Brain Chemistry/drug effects , Brain Chemistry/physiology , Calcium Channel Blockers/toxicity , Depressive Disorder/chemically induced , Depressive Disorder/metabolism , Depressive Disorder/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Emotions/drug effects , Emotions/physiology , Male , Marijuana Abuse/metabolism , Marijuana Abuse/psychology , Mental Disorders/metabolism , Morpholines/toxicity , Naphthalenes/toxicity , Norepinephrine/metabolism , Rats , Rats, Sprague-Dawley , Serotonin/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Time
18.
Naunyn Schmiedebergs Arch Pharmacol ; 380(5): 383-97, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19727675

ABSTRACT

New atypical antipsychotics show a greater affinity to serotonergic rather than to dopamine receptors, suggesting that serotonin (5-HT) has a major role in the pathophysiology and treatment of schizophrenia. The goal of this study was to characterise the response of pyramidal neurons in the medial prefrontal cortex (mPFC) to 5-HT and NMDA before and after administration of the NMDA receptor antagonist, MK-801 (dizocilpine), a well-validated pharmacological model of psychosis. mPFC pyramidal (glutamatergic) neurons were recorded in urethane-anaesthetised rats. The responses to NMDA and 5-HT were assessed using in vivo electrophysiology and microiontophoresis. The 5-HT2A/2C antagonist ritanserin and the 5-HT1A antagonist WAY100635 were used to block 5-HT responses. MK-801 decreased the NMDA-induced excitatory responses and increased NMDA-evoked burst activity among mPFC pyramidal neurons. Three subpopulations of pyramidal cells were identified according to their responses to 5-HT: excitation (33%), inhibition (40%) and non-response (27%). The inhibitory responses were blocked by WAY100635 in 100% of cases, but not by ritanserin; the excitatory responses were blocked by ritanserin in 75% of cases, but not by WAY100635. The administration of MK-801 potentiated the firing rate of excitatory responses but did not modify the inhibitory responses induced by microiontophoretic application of 5-HT. These results suggest that MK-801 modifies 5-HT synapses in the mPFC by potentiating the excitatory 5-HT2A/2C responses and attenuating NMDA excitations. These data indicate that 5-HT excitatory transmission is selectively impaired at the mPFC level in this pharmacological model of schizophrenia.


Subject(s)
Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Schizophrenia/physiopathology , Serotonin/metabolism , Animals , Disease Models, Animal , Electrophysiology , Iontophoresis/methods , Male , N-Methylaspartate/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Pyramidal Cells/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT2A/drug effects , Receptor, Serotonin, 5-HT2A/metabolism , Receptor, Serotonin, 5-HT2C/drug effects , Receptor, Serotonin, 5-HT2C/metabolism , Synaptic Transmission/drug effects
19.
Curr Pharm Des ; 15(14): 1623-46, 2009.
Article in English | MEDLINE | ID: mdl-19442178

ABSTRACT

Among all mental disorders, major depression has the highest rate of prevalence and incidence of morbidity. Currently available antidepressant therapies have limited efficacies; consequently, research on new drugs for the treatment of mood disorders has become increasingly critical. Recent preclinical evidences that cannabinoid agonists and endocannabinoid enhancers, such as the fatty acid amide hydrolase (FAAH) inhibitors, can impact mood regulation have opened a new line of research in antidepressant drug discovery. However, the neurobiological mechanisms linking the endocannabinoid system with the pathophysiology of mood disorders and antidepressant action remain unclarified. In this review, we have presented an update on preclinical data indicating the antidepressant potential of cannabinoid agonists and endocannabinoid enhancers in comparison to standard antidepressants. Data obtained from CB(1) knockout (CB(1)-/-) and FAAH knockout (FAAH-/-) mice have also been examined within this context. We have illustrated how the various classes of antidepressants exert their therapeutic action. In particular, all antidepressants increase the neurotransmission of serotonin after long-term treatment, enhance the tonic activity of hippocampal 5-HT(1A) receptors, promote neurogenesis, and modulate (decrease or increase) the firing activity of noradrenergic neurons. Interestingly, cannabinoid agonists and endocannabinoid enhancers increase serotonin and noradrenergic neuronal firing activity, increase serotonin release in the hippocampus, as well as promote neurogenesis. Since cannabinoid-derived drugs potentiate monoaminergic neurotransmission and hippocampal neurogenesis through distinct pathways compared to classical antidepressants, they may represent an alternative drug class in the pharmacotherapy of mood and other neuropsychiatric disorders.


Subject(s)
Antidepressive Agents/pharmacology , Cannabinoids/pharmacology , Depressive Disorder, Major/drug therapy , Animals , Cannabinoid Receptor Modulators/metabolism , Depressive Disorder, Major/physiopathology , Disease Models, Animal , Drug Evaluation, Preclinical , Endocannabinoids , Humans , Mice , Mice, Knockout , Neurogenesis/drug effects , Rats , Receptor, Cannabinoid, CB1/agonists , Serotonin/metabolism
20.
Eur Neuropsychopharmacol ; 19(3): 215-28, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19147333

ABSTRACT

Chronic stressful life events are risk factors for contracting depression, the pathophysiology of which is strongly associated with impairments in serotonergic (5-HT) neurotransmission. Indeed, in rodents, exposure to chronic unpredictable stress (CUS) produces depressive-like behaviours such as behavioural despair and anhedonia. To date, there have not been many studies that especially explore in vivo changes in 5-HT neurotransmission associated with CUS in the rat. Therefore, using in vivo electrophysiology, we evaluated whether CUS that induces anhedonia-like behaviours concurrently impairs midbrain raphe 5-HT neuronal activity. Unlike unstressed and acutely stressed rats, CUS produced progressive reductions in sucrose intake and preference (anhedonia-like). These were associated with a decrease in the spontaneous firing activity (35.4%) as well as in the number of spontaneously active 5-HT neurons, and a desensitization of somatodendritic 5-HT1A autoreceptors in the dorsal raphe. These results suggest that CUS dramatically decreases 5-HT neural activity and 5-HT1A autoreceptor sensitivity, and may represent endophenotypic features of depressive-like states.


Subject(s)
Action Potentials/physiology , Neurons/physiology , Receptor, Serotonin, 5-HT1A/metabolism , Serotonin/metabolism , Stress, Psychological/metabolism , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Action Potentials/drug effects , Analysis of Variance , Animals , Chronic Disease , Disease Models, Animal , Drug Interactions , Electric Stimulation , Food Preferences/drug effects , Male , Neural Inhibition/drug effects , Neurons/drug effects , Piperazines/pharmacology , Pyridines/pharmacology , Raphe Nuclei/pathology , Rats , Rats, Wistar , Serotonin 5-HT1 Receptor Agonists , Serotonin 5-HT1 Receptor Antagonists , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacology , Stress, Psychological/pathology , Sucrose/administration & dosage , Sweetening Agents/administration & dosage , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...