Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Language
Publication year range
1.
Front Integr Neurosci ; 17: 1168640, 2023.
Article in English | MEDLINE | ID: mdl-37377628

ABSTRACT

Introduction: Chronic exposure to social defeat stress (SDS) has been used to investigate the neurobiology of depressive- and anxiety-like responses and mnemonic processes. We hypothesized that these affective, emotional, and cognitive consequences induced by SDS are regulated via glutamatergic neurons located in the bed nucleus of the stria terminalis (BNST), amygdaloid complex, and hippocampus in mice. Methods: Here, we investigated the influence of chronic SDS on (i) the avoidance behavior assessed in the social interaction test, (ii) the anxiety-like behavior (e.g., elevated plus-maze, and open field tests) (iii) depressive-like behaviors (e.g., coat state, sucrose splash, nesting building, and novel object exploration tests), (iv) the short-term memory (object recognition test), (v) ΔFosB, CaMKII as well as ΔFosB + CaMKII labeling in neurons located in the BNST, amygdaloid complex, dorsal (dHPC) and the ventral (vHPC) hippocampus. Results: The main results showed that the exposure of mice to SDS (a) increased defensive and anxiety-like behaviors and led to memory impairment without eliciting clear depressive-like or anhedonic effects; (b) increased ΔFosB + CaMKII labeling in BNST and amygdala, suggesting that both areas are strongly involved in the modulation of this type of stress; and produced opposite effects on neuronal activation in the vHPC and dHPC, i.e., increasing and decreasing, respectively, ΔFosB labeling. The effects of SDS on the hippocampus suggest that the vHPC is likely related to the increase of defensive- and anxiety-related behaviors, whereas the dHPC seems to modulate the memory impairment. Discussion: Present findings add to a growing body of evidence indicating the involvement of glutamatergic neurotransmission in the circuits that modulate emotional and cognitive consequences induced by social defeat stress.

2.
Article in English | MEDLINE | ID: mdl-36870468

ABSTRACT

The endocannabinoid (eCB) anandamide (AEA) is synthesized on-demand in the post-synaptic terminal and can act on presynaptic cannabinoid type 1 (CB1) receptors, decreasing the release of neurotransmitters, including glutamate. AEA action is ended through enzymatic hydrolysis via FAAH (fatty acid amid hydrolase) in the post-synaptic neuron. eCB system molecules are widely expressed in brain areas involved in the modulation of fear and anxiety responses, including the Bed Nucleus of the Stria Terminalis (BNST), which is involved in the integration of autonomic, neuroendocrine, and behavioral regulation. The presence of the CB1 and FAAH was described in the BNST; however, their role in the modulation of defensive reactions is not fully comprehended. In the present work we aimed at investigating the role of AEA and CB1 receptors in the BNST in modulating anxiety-related behaviors. Adult male Wistar rats received local BNST injections of the CB1 receptor antagonist AM251 (0.1-0.6 nmol) and/or the FAAH inhibitor (URB597; 0.001-0.1 nmol) and were evaluated in the elevated plus maze (EPM) test, with or without previous acute restraint stress (2 h) exposure, or in the contextual fear conditioning. We observed that although AM251 and URB597 had no effects on the EPM, they increased and decreased, respectively, the conditioned fear response. Supporting a possible influence of stress in these differences, URB597 was able to prevent the restraint stress-induced anxiogenic effect in the EPM. The present data, therefore, suggest that eCB signaling in the BNST is recruited during more aversive situations to counteract the stress effect.


Subject(s)
Cannabinoids , Septal Nuclei , Animals , Male , Rats , Anxiety/drug therapy , Anxiety/chemically induced , Cannabinoids/pharmacology , Endocannabinoids/pharmacology , Rats, Wistar , Receptor, Cannabinoid, CB1
3.
J Hist Neurosci ; 32(1): 19-38, 2023.
Article in English | MEDLINE | ID: mdl-36476105

ABSTRACT

The amygdaloid complex is a crucial component of the basal forebrain that participates in the modulation of many homeostatic functions, emotional behaviors, and learning. These features require a widespread pattern of connections with several brain structures. In the past, the amygdaloid complex was divided into corticomedial and basolateral groups. The existence of a neuronal continuum linking the central amygdaloid nucleus to the lateral bed nucleus of stria terminalis through the subpallidal area was first revealed by José de Olmos (1932-2008) with the aid of his cupric-silver technique. This observation gave birth to the concept of the extended amygdala, a conceptual framework that is useful for understanding the anatomofunctional organization of the amygdaloid complex, with relevance for basic neuroscience and clinical interventions. Traditional tract-tracing staining methods were complicated and tedious to reproduce. Axonal terminal endings were lost among a myriad of normal fibers. The need to visualize these terminals drove de Olmos to develop cupric-silver methods that revealed disintegrating synaptic terminals, without staining normal fibers. In this article, we describe the historical events leading to the development of the cupric-silver technique that evolved into the amino-cupric-silver technique, which developed hand-in-hand over the years.


Subject(s)
Amygdala , Silver , Humans , Amygdala/physiology , Neurons
4.
Pflugers Arch ; 473(2): 253-271, 2021 02.
Article in English | MEDLINE | ID: mdl-33140200

ABSTRACT

The bed nucleus of the stria terminalis (BNST) is a forebrain structure, involved in the modulation of neuroendocrine, cardiovascular and autonomic responses. One of the responses is baroreflex activity, which consists in a neural mechanism responsible for keeping the blood pressure within a narrow range of variation. It has been reported that blockade of BNST α1-adrenoceptors increased the bradycardic component of baroreflex. In addition, such receptors are able to modulate glutamate release in this structure. Interestingly, BNST NMDA receptor antagonism and neuronal nitric oxide synthase (nNOS) inhibition led to the same effect of the α1-adrenoceptors blockade on baroreflex bradycardic response. Therefore, the hypothesis of the present study is that BNST noradrenergic transmission interacts with NMDA/NO pathway through α1 adrenoceptors to modulate the baroreflex activity. Male Wistar rats had stainless steel guide cannulas bilaterally implanted in the BNST. Subsequently, a catheter was inserted into the femoral artery for cardiovascular recordings, and into the femoral vein for assessing baroreflex activation. Injection of the noradrenaline reuptake inhibitor reboxetine in the BNST did not modify the tachycardic, but significantly decreased the bradycardic component of baroreflex. Administration of an α1, but not an α2 antagonist into the BNST prior to reboxetine prevented this effect. Likewise, previous injection of NMDA/NO pathway blockers inhibited the effect of reboxetine on bradycardic response. In conclusion, it was demonstrated for the first time the existence of an interaction between BNST noradrenergic, glutamatergic and nitrergic neurotransmissions in the modulation of bradycardic baroreflex response.


Subject(s)
Autonomic Nervous System/physiology , Baroreflex , Heart/innervation , Nitric Oxide/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Septal Nuclei/metabolism , Animals , Arterial Pressure , Autonomic Nervous System/drug effects , Baroreflex/drug effects , Heart Rate , Male , Neurotransmitter Agents/pharmacology , Rats, Wistar , Receptors, Adrenergic, alpha-1/drug effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Septal Nuclei/drug effects , Time Factors
5.
J Psychopharmacol ; 34(4): 429-440, 2020 04.
Article in English | MEDLINE | ID: mdl-31913077

ABSTRACT

BACKGROUND: Endocannabinoid neurotransmission in the bed nucleus of the stria terminalis is involved in the control of cardiovascular responses to stress. However, the local mechanisms involved is this regulation are not known. AIMS: The purpose of this study was to assess an interaction of bed nucleus of the stria terminalis endocannabinoid neurotransmission with local nitrergic signaling, as well as to investigate the involvement of local N-methyl-D-aspartate glutamate receptor and nitric oxide signaling in the control of cardiovascular responses to acute restraint stress by bed nucleus of the stria terminalis endocannabinoid neurotransmission in rats. METHODS: The first protocol evaluated the effect of intra-bed nucleus of the stria terminalis microinjection of the selective cannabinoid receptor type 1 receptor antagonist AM251 in nitrite/nitrate content in the bed nucleus of the stria terminalis following restraint stress. The other protocols evaluated the impact of local pretreatment with the selective N-methyl-D-aspartate glutamate receptor antagonist LY235959, the selective neuronal nitric oxide synthase inhibitor Nω-propyl-L-arginine, the soluble guanylate cyclase inhibitor 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, or the protein kinase G inhibitor KT5823 in restraint-evoked cardiovascular changes following bed nucleus of the stria terminalis treatment with AM251. RESULTS: Bilateral microinjection of AM251 into the bed nucleus of the stria terminalis increased local nitric oxide release during restraint stress. Bed nucleus of the stria terminalis treatment with the cannabinoid receptor type 1 receptor antagonist also enhanced the tachycardia caused by restraint stress, but without affecting arterial pressure increase and sympathetic-mediated cutaneous vasoconstriction. The facilitation of restraint-evoked tachycardia following bed nucleus of the stria terminalis treatment with the cannabinoid receptor type 1 receptor antagonist was completely inhibited by local pretreatment with LY235959, Nω-propyl-L-arginine, 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, or KT5823. CONCLUSIONS: Our results provide evidence that bed nucleus of the stria terminalis endocannabinoid neurotransmission inhibits local N-methyl-D-aspartate/neuronal nitric oxide synthase/soluble guanylate cyclase/protein kinase G signaling, and this mechanism is involved in the control of the cardiovascular responses to stress.


Subject(s)
Hemodynamics/drug effects , Receptor, Cannabinoid, CB1/drug effects , Septal Nuclei/drug effects , Signal Transduction/drug effects , Stress, Psychological/complications , Stress, Psychological/drug therapy , Animals , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/drug effects , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/drug effects , Male , Microinjections , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitric Oxide Synthase Type I/drug effects , Piperidines/administration & dosage , Piperidines/pharmacology , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/drug effects , Restraint, Physical , Synaptic Transmission/drug effects
6.
Neuropharmacology ; 117: 249-259, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28232061

ABSTRACT

Here we provide evidence that repeated immobilization stress (RIS) in rats induces a persistent increase in noradrenergic activity in the anterior aspects of the anterolateral bed nucleus of the stria terminalis (alBNST). This increase in noradrenergic activity results from both enhanced synthesis and reuptake of norepinephrine (NE). It leads to a decrease in the synaptic availability of NE, which elicits an augmented noradrenergic response to the inhibitors of NE reuptake (NRIs), such as desipramine (DMI), an antidepressant. The enduring depression-like behavior and the augmentation of the climbing behavior seen in repeatedly stressed rats following subchronic administration of DMI in the forced swimming test (FST) might be explained by a dysregulation of noradrenergic transmission observed in alBNST. Taken together, we propose that dysregulation of noradrenergic transmission such as the one described in the present work may represent a mechanism underlying major depressive disorders (MDD) with melancholic features in humans.


Subject(s)
Antidepressive Agents, Tricyclic/pharmacology , Depressive Disorder/drug therapy , Desipramine/pharmacology , Norepinephrine/metabolism , Restraint, Physical/psychology , Septal Nuclei/drug effects , Amphetamine/pharmacology , Animals , Anxiety/drug therapy , Anxiety/metabolism , Central Nervous System Stimulants/pharmacology , Depressive Disorder/metabolism , Disease Models, Animal , Extracellular Space/drug effects , Extracellular Space/metabolism , Male , Motor Activity/drug effects , Motor Activity/physiology , Random Allocation , Rats, Sprague-Dawley , Septal Nuclei/metabolism , Stress, Psychological/drug therapy , Stress, Psychological/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
7.
J Psychopharmacol ; 31(6): 674-681, 2017 06.
Article in English | MEDLINE | ID: mdl-28198211

ABSTRACT

The bed nucleus of the stria terminalis (BNST) is a forebrain structure that has been implicated on cardiovascular responses evoked by emotional stress. However, the local neurochemical mechanisms mediating the BNST control of stress responses are not fully described. In our study we investigated the involvement of glutamatergic neurotransmission within the BNST in cardiovascular changes evoked by acute restraint stress in rats. For this study, we investigated the effects of bilateral microinjections of selective antagonists of either N-methyl-D-aspartate (NMDA) or non-NMDA glutamate receptors into the BNST on the arterial pressure and heart rate increase and the decrease in tail skin temperature induced by acute restraint stress. Microinjection of the selective NMDA glutamate receptor antagonist LY235959 (1 nmol/100 nL) into the BNST decreased the tachycardiac response to restraint stress, without affecting the arterial pressure increase and the drop in skin temperature. Bilateral BNST treatment with the selective non-NMDA glutamate receptor NBQX (1 nmol/100 nL) decreased the heart rate increase and the fall in tail skin temperature, without affecting the blood pressure increase. These findings indicate a facilitatory influence of BNST glutamatergic neurotransmission via coactivation of local NMDA and non-NMDA receptors on the tachycardiac response to stress, whereas control of sympathetic-mediated cutaneous vasoconstriction is selectively mediated by local non-NMDA glutamate receptors.


Subject(s)
Cardiovascular System/metabolism , N-Methylaspartate/metabolism , Receptors, Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Restraint, Physical/physiology , Septal Nuclei/metabolism , Stress, Psychological/metabolism , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Cardiovascular System/drug effects , Heart Rate/drug effects , Heart Rate/physiology , Isoquinolines/pharmacology , Male , Rats , Rats, Wistar , Septal Nuclei/drug effects , Skin Temperature/physiology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
8.
Horm Behav ; 79: 74-83, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26774463

ABSTRACT

It has been shown that the bed nucleus of the stria terminalis (BNST) of rats contains nitrergic neurons, which are activated during animal exposure to aversive stimuli. The BNST is also populated by glutamatergic and corticotrophin releasing factor (CRFergic) neurons, which in turn are activated under stressful situations. Here we investigated the anxiogenic-like effects of intra-BNST injections of a nitric oxide (NO) donor, NOC-9 in mice. The role of CRFergic and glutamatergic systems on defensive behavior induced by NOC-9 was investigated with previous intra-BNST infusion of different doses of CP376395, a CRF type 1 receptor antagonist (CRF1), or AP-7, an NMDA (N-methyl-D-aspartate) receptor antagonist. Anxiety-like behavior was assessed immediately and 5 min after intra-BNST drug injection, exposing mice to a novel arena and to the elevated plus-maze (EPM; an anxiogenic situation). Results showed that NOC-9 provoked a short period (≈ 150 s) of freezing behavior in the novel arena and increased anxiety in the EPM. Both CP and AP-7 attenuated the anxiogenic-like effects of NOC-9 in the EPM without changing freezing behavior in the novel arena. When given alone (i.e. without prior intra-BNST injection of NOC-9), AP-7 (0.20 nmol), but not CP (0.75, 1.50, or 3.00 nmol), attenuated anxiety in mice exposed to the EPM. These results suggest that CRF1 and NMDA receptors located within the BNST differentially modulate aversive effects induced by NO production in this limbic forebrain structure.


Subject(s)
Anxiety/chemically induced , Nitric Oxide/pharmacology , Receptors, Corticotropin-Releasing Hormone/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Septal Nuclei/drug effects , Aminopyridines/pharmacology , Animals , Anxiety/metabolism , Behavior, Animal/drug effects , Corticotropin-Releasing Hormone/metabolism , Freezing Reaction, Cataleptic/drug effects , Male , Maze Learning/drug effects , Mice , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Corticotropin-Releasing Hormone/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Stress, Psychological/chemically induced , Stress, Psychological/metabolism , Stress, Psychological/psychology , Triazenes/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL