Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 244
Filter
Add more filters

Complementary Medicines
Publication year range
1.
Peptides ; 126: 170249, 2020 04.
Article in English | MEDLINE | ID: mdl-31911169

ABSTRACT

Serotonergic neurons of the median raphe nucleus (MnR) and hypothalamic melanin-concentrating hormone (MCH)-containing neurons, have been involved in the control of REM sleep and mood. In the present study, we examined in rats and cats the anatomical relationship between MCH-containing fibers and MnR neurons, as well as the presence of MCHergic receptors in these neurons. In addition, by means of in vivo unit recording in urethane anesthetized rats, we determined the effects of MCH in MnR neuronal firing. Our results showed that MCH-containing fibers were present in the central and paracentral regions of the MnR. MCHergic fibers were in close apposition to serotonergic and non-serotonergic neurons. By means of an indirect approach, we also analyzed the presence of MCHergic receptors within the MnR. Accordingly, we microinjected MCH conjugated with the fluorophore rhodamine (R-MCH) into the lateral ventricle. R-MCH was internalized into serotonergic and non-serotonergic MnR neurons; some of these neurons were GABAergic. Furthermore, we determined that intracerebroventricular administration of MCH induced a significant decrease in the firing rate of 53 % of MnR neurons, while the juxtacellular administration of MCH reduced the frequency of discharge in 67 % of these neurons. Finally, the juxtacellular administration of the MCH-receptor antagonist ATC-0175 produced an increase in the firing rate in 78 % of MnR neurons. Hence, MCH produces a strong regulation of MnR neuronal activity. We hypothesize that MCHergic modulation of the MnR neuronal activity may be involved in the promotion of REM sleep and in the pathophysiology of depressive disorders.


Subject(s)
Hypothalamic Hormones/pharmacology , Hypothalamus/drug effects , Melanins/pharmacology , Nerve Fibers/drug effects , Neurons/drug effects , Pituitary Hormones/pharmacology , Raphe Nuclei/drug effects , Receptors, Pituitary Hormone/metabolism , Animals , Cats , Hypothalamus/metabolism , Hypothalamus/physiology , Nerve Fibers/metabolism , Nerve Fibers/physiology , Neurons/metabolism , Neurons/physiology , Raphe Nuclei/metabolism , Raphe Nuclei/physiology , Rats , Rats, Wistar
2.
Nat Neurosci ; 22(8): 1357-1370, 2019 08.
Article in English | MEDLINE | ID: mdl-31285615

ABSTRACT

The medial prefrontal cortex (mPFC) contains populations of GABAergic interneurons that play different roles in cognition and emotion. Their local and long-range inputs are incompletely understood. We used monosynaptic rabies viral tracers in combination with fluorescence micro-optical sectioning tomography to generate a whole-brain atlas of direct long-range inputs to GABAergic interneurons in the mPFC of male mice. We discovered that three subtypes of GABAergic interneurons in two areas of the mPFC are innervated by same upstream areas. Input from subcortical upstream areas includes cholinergic neurons from the basal forebrain and serotonergic neurons (which co-release glutamate) from the raphe nuclei. Reconstruction of single-neuron morphology revealed novel substantia innominata-anteromedial thalamic nucleus-mPFC and striatum-anteromedial thalamic nucleus-mPFC circuits. Based on the projection logic of individual neurons, we classified cortical and hippocampal input neurons into several types. This atlas provides the anatomical foundation for understanding the functional organization of the mPFC.


Subject(s)
Brain Mapping/methods , Interneurons/physiology , Prefrontal Cortex/anatomy & histology , Prefrontal Cortex/cytology , gamma-Aminobutyric Acid/physiology , Animals , Cell Count , Hippocampus/cytology , Hippocampus/physiology , Male , Mice , Mice, Inbred C57BL , Parasympathetic Nervous System/cytology , Parasympathetic Nervous System/physiology , Prosencephalon/anatomy & histology , Prosencephalon/cytology , Raphe Nuclei/cytology , Raphe Nuclei/physiology , Serotonergic Neurons/physiology , Thalamus/cytology , Thalamus/physiology
3.
Sci Rep ; 9(1): 8198, 2019 06 03.
Article in English | MEDLINE | ID: mdl-31160650

ABSTRACT

Stress is one of major factors that cause sleep problems. Hypocretin represents a stress-related neuropeptide and is well known in maintaining physiological wakefulness. The hypocretinergic neurons originate in the lateral hypothalamic area (LHA) and transmit to several brain regions, including the median raphe nuclei (MRNs). The MRNs modulate both fear responses and sleep-wake activity; however, it remains unclear whether stress alters the levels of hypocretin to regulate MRNs and consequently disrupt sleep. In this paper, we employed the inescapable footshock stimuli (IFS) as a stressor and hypothesized that the IFS-induced sleep disruption is mediated by increased hypocretins in the MRNs. Our results demonstrate that the concentrations of hypocretin in the hypothalamus increased after IFS. Rapid eye movement (REM) sleep was reduced after footshock, and microinjection of non-selective hypocretin receptor antagonist TCS-1102 into the MRNs blocked the IFS-induced decrease of REM sleep. Furthermore, administration of hypocretins into the MRNs mimicked the IFS-induced REM sleep reduction. These results conclude that the increased levels of hypocretins in the MRNs mediate the IFS-induced REM sleep reduction.


Subject(s)
Brain Mapping/methods , Orexins/pharmacology , Raphe Nuclei/physiology , Sleep, REM , Animals , Fear , Hypothalamic Area, Lateral/physiology , Hypothalamus/physiology , Intracellular Signaling Peptides and Proteins/metabolism , Male , Neuropeptides/metabolism , Orexin Receptors , Orexins/metabolism , Rats , Rats, Wistar , Sleep , Stress, Physiological , Wakefulness
4.
Behav Brain Res ; 324: 87-95, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28212941

ABSTRACT

Previously, we showed that the blockade of α1-adrenoreceptors in the median raphe nucleus (MnR) increased food intake in free-feeding rats, indicating that adrenergic mechanisms in the MnR participate in the regulation of food intake. However, the impact of such a pharmacological manipulation on other neural circuits related to food intake remains unknown. In the current study, we sought to identify forebrain regions which are responsive to α1-adrenergic receptor blockade and presumably involved in the modulation of the feeding response. For this purpose, we examined the induction of c-Fos immunoreactivity in forebrain structures following injections of the α1-adrenoceptor antagonist prazosin into the MnR of free-feeding rats. To determine the chemical identity of hypothalamic c-Fos-positive cells, we then conducted double-label immunohistochemistry for Fos/orexin (OX) or Fos/melanin-concentrating hormone (MCH). Finally, we combined anterograde tracing from the MnR with immunohistochemical detection of orexin. Prazosin injections into the MnR significantly increased food intake. The ingestive response was accompanied by an increase in Fos expression in the basolateral amygdala (BLA) and lateral hypothalamic area (LHA). In the LHA, Fos expression occurred in neurons expressing OX, but not MCH. Combined anterograde tracing experiments revealed that LHA OX neurons are prominently targeted by MnR axons. These findings suggest that intra-MnR injection of prazosin, via activation of orexinergic neurons in the LHA and non-orexinergic neurons in the BLA, evoked a motivational response toward food intake.


Subject(s)
Adrenergic alpha-1 Receptor Antagonists/administration & dosage , Eating , Neurons/metabolism , Prazosin/administration & dosage , Raphe Nuclei/physiology , Receptors, Adrenergic, alpha-1/physiology , Animals , Basolateral Nuclear Complex/metabolism , Hypothalamic Hormones/metabolism , Hypothalamus/metabolism , Male , Melanins/metabolism , Orexins/metabolism , Pituitary Hormones/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Raphe Nuclei/drug effects , Rats, Wistar
5.
Physiol Behav ; 165: 35-42, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27364433

ABSTRACT

In neonatal rats, hunger and satiety responses occur particularly via dehydration and gastric distention, respectively. The control of food intake in newborns is yet to be fully consolidated, particularly with respect to the participation of the hypothalamic nuclei and their relationship with the serotonergic pathway. Moreover, it is unclear how the environmental stressors in early life, like undernutrition, interfere in these events. Therefore, this study examined the serotonin-system's impact on food intake in rat neonates at postnatal day (P) 10 and P18 and the manner in which protein undernutrition during pregnancy and lactation interferes in this behavior. To accomplish this, Wistar rats were used, nutritionally manipulated by a diet having two protein levels, (8% and 17%) during pregnancy and lactation, to form the Control (n=10) and Low protein groups (n=10). At 10 and 18 postnatal days pups received an acute dose of fenfluramine (3mg/kg) or saline (0.9% NaCl) and subjected to milk consumption testing and then perfused to obtain the brains for the analysis of cell activation of the immunoreactive c-Fos in the hypothalamic and raphe nuclei. At 10days a reduction in weight gain was observed in both groups. On comparison of the neuronal activation for the paraventricular nucleus, an increased activation in response to fenfluramine was observed. At 18days, the weight gain percentage differed between the groups according to the nutritional manipulation, in which the control animals had no significant change while the undernourished presented increased weight gain with the use of fenfluramine. The marking of c-Fos in response to fenfluramine in the hypothalamic and raphe nuclei revealed, an especially lower activation of the PVN, MnR and DR compared intra-group. However when evaluating the effect of undernutrition, marking activation was observed to increase in all the nuclei analyzed, in the hypothalamus and raphe. Data from this study indicate that the action of serotonin via food intake in the neonates may have been delayed by early protein undernutrition.


Subject(s)
Diet, Protein-Restricted/adverse effects , Eating/physiology , Hypothalamus/physiology , Malnutrition/physiopathology , Raphe Nuclei/physiology , Serotonin/metabolism , Animals , Animals, Newborn , Disease Models, Animal , Eating/drug effects , Female , Fenfluramine/pharmacology , Fetal Nutrition Disorders/metabolism , Fetal Nutrition Disorders/physiopathology , Hypothalamus/drug effects , Hypothalamus/growth & development , Lactation , Male , Milk , Neurons/drug effects , Neurons/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Raphe Nuclei/drug effects , Raphe Nuclei/growth & development , Rats, Wistar , Selective Serotonin Reuptake Inhibitors/pharmacology , Weight Gain/drug effects , Weight Gain/physiology
6.
Pathol Biol (Paris) ; 62(5): 276-83, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25104243

ABSTRACT

Reductions in central serotonin activity with aging might be involved in sleep-related disorders in later life. Although the beneficial effects of aerobic exercise on sleep are not new, sleep represents a complex recurring state of unconsciousness involving many lines of transmitters which remains only partly clear despite intense ongoing research. It is known that serotonin released into diencephalon and cerebrum might play a key inhibitory role to help promote sleep, likely through an active inhibition of supraspinal neural networks. Several lines of evidence support the stimulatory effects of exercise on higher serotonergic pathways. Hence, exercise has proved to elicit acute elevations in forebrain serotonin concentrations, an effect that waned upon cessation of exercise. While adequate exercise training might lead to adaptations in higher serotonergic networks (desensitization of forebrain receptors), excessive training has been linked to serious brain serotonergic maladaptations accompanied by insomnia. Dietary supplementation of tryptophan (the only serotonin precursor) is known to stimulate serotonergic activity and promote sleep, whereas acute tryptophan depletion causes deleterious effects on sleep. Regarding sleep-wake regulation, exercise has proved to accelerate resynchronization of the biological clock to new light-dark cycles following imposition of phase shifts in laboratory animals. Noteworthy, the effect of increased serotonergic transmission on wake state appears to be biphasic, i.e. promote wake and thereafter drowsiness. Therefore, it might be possible that acute aerobic exercise would act on sleep by increasing activity of ascending brain serotonergic projections, though additional work is warranted to better understand the implication of serotonin in the exercise-sleep axis.


Subject(s)
Aging/physiology , Exercise/physiology , Serotonin/physiology , Sleep/physiology , Amino Acids/metabolism , Animals , Athletes , Humans , Nerve Net/physiology , Preoptic Area/physiology , Raphe Nuclei/physiology , Reticular Formation/physiology , Serotonergic Neurons/physiology , Serotonin/deficiency , Sleep Disorders, Circadian Rhythm/physiopathology , Sleep Initiation and Maintenance Disorders/physiopathology , Tryptophan/metabolism , Tryptophan/therapeutic use , Wakefulness
7.
PLoS One ; 8(3): e60686, 2013.
Article in English | MEDLINE | ID: mdl-23544161

ABSTRACT

This work addressed the study of subnucleus reticularis dorsalis (SRD) neurons in relation to their supraspinal input and the spinal terminating sites of their descending axons. SRD extracellular unitary recordings from anesthetized cats aimed to specifically test, 1) the rostrocaudal segmental level reached by axons of spinally projecting (SPr) neurons collateralizing or not to or through the ipsilateral nucleus reticularis gigantocellularis (NRGc), 2) whether SPr fibers bifurcate to the thalamus, and 3) the effects exerted on SRD cells by electrically stimulating the locus coeruleus, the periaqueductal grey, the nucleus raphe magnus, and the mesencephalic locomotor region. From a total of 191 SPr fibers tested to cervical 2 (Ce2), thoracic 5 (Th5) and lumbar5 (Lu5) stimulation, 81 ended between Ce2 and Th5 with 39 of them branching to or through the NRGc; 21/49 terminating between Th5 and Lu5 collateralized to or through the same nucleus, as did 34/61 reaching Lu5. The mean antidromic conduction velocity of SPr fibers slowed in the more proximal segments and increased with terminating distance along the cord. None of the 110 axons tested sent collaterals to the thalamus; instead thalamic stimulation induced long-latency polysynaptic responses in most cells but also short-latency, presumed monosynaptic, in 7.9% of the tested neurons (18/227). Antidromic and orthodromic spikes were elicited from the locus coeruleus and nucleus raphe magnus, but exclusively orthodromic responses were observed following stimulation of the periaqueductal gray or mesencephalic locomotor region. The results suggest that information from pain-and-motor-related supraspinal structures converge on SRD cells that through SPr axons having conduction velocities tuned to their length may affect rostral and caudal spinal cord neurons at fixed delays, both directly and in parallel through different descending systems. The SRD will thus play a dual functional role by simultaneously regulating dorsal horn ascending noxious information and pain-related motor responses.


Subject(s)
Electrophysiological Phenomena , Medulla Oblongata/cytology , Neurons/physiology , Periaqueductal Gray/physiology , Reticular Formation/cytology , Spinal Cord/physiology , Animals , Axons/physiology , Cats , Electric Stimulation , Male , Motor Activity/physiology , Neural Conduction/physiology , Pain/physiopathology , Raphe Nuclei/physiology , Spinal Cord/cytology , Thalamus/physiology
8.
J Chem Neuroanat ; 48-49: 29-45, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23337940

ABSTRACT

As is well recognized, serotonergic (5-HT) fibers distribute widely throughout the brain, including the cerebral cortex. Although some early reports described the 5-HT innervation of the prefrontal cortex (PFC) in rats, the focus was on sensorimotor regions and not on the 'limbic' PFC - or on the medial, orbital and insular cortices. In addition, no reports have described the distribution of 5-HT fibers to PFC in rats using antisera to the serotonin transporter (SERT). Using immunostaining for SERT, we examined the pattern of distribution of 5-HT fibers to the medial, orbital and insular cortices in the rat. We show that 5-HT fibers distribute massively throughout all divisions of the PFC, with distinct laminar variations. Specifically, 5-HT fibers were densely concentrated in superficial (layer 1) and deep (layers 5/6) of the PFC but less heavily so in intermediate layers (layers 2/3). This pattern was most pronounced in the orbital cortex, particularly in the ventral and ventrolateral orbital cortices. With the emergence of granular divisions of the insular cortex, the granular cell layer (layer 4) was readily identifiable by a dense band of labeling confined to it, separating layer 4 from less heavily labeled superficial and deep layers. The pattern of 5-HT innervation of medial, orbital and insular cortices significantly differed from that of sensorimotor regions of the PFC. Serotonergic labeling was much denser overall in limbic compared to non-limbic regions of the PFC, as was striking demonstrated by the generally weaker labeling in layers 1-3 of the primary sensory and motor cortices. The massive serotonergic innervation of the medial, orbital and insular divisions of the PFC likely contributes substantially to well established serotonergic effects on affective and cognitive functions, including a key role in many neurological and psychiatric diseases.


Subject(s)
Cerebral Cortex/physiology , Nerve Fibers/physiology , Serotonin/physiology , Animals , Behavior, Animal/physiology , Biomarkers , Cerebral Cortex/cytology , Female , Immunohistochemistry , Male , Prefrontal Cortex/cytology , Prefrontal Cortex/physiology , Raphe Nuclei/cytology , Raphe Nuclei/physiology , Rats , Rats, Sprague-Dawley , Serotonin/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Signal Transduction , Thalamus/cytology , Thalamus/physiology
9.
Br J Nutr ; 109(7): 1323-9, 2013 Apr 14.
Article in English | MEDLINE | ID: mdl-23167968

ABSTRACT

L-Lysine (Lys) is an essential amino acid and plays an important role in anxiogenic behaviour in both human subjects and rodents. Previous studies have shown the existence of neural plasticity between the Lys-deficient state and the normal state. Lys deficiency causes an increase in noradrenaline release from the hypothalamus and serotonin release from the amygdala in rats. However, no studies have used functional MRI (fMRI) to compare the brain response to ingested Lys in normal, Lys-deficient and Lys-recovered states. Therefore, in the present study, using acclimation training, we performed fMRI on conscious rats to investigate the brain response to an intragastric load of Lys. The brain responses to intragastric administration of Lys (3 mmol/kg body weight) were investigated in six rats intermittently in three states: normal, Lys-deficient and recovered state. First, in the normal state, an intragastric load of Lys activated several brain regions, including the raphe pallidus nucleus, prelimbic cortex and the ventral/lateral orbital cortex. Then, after 6 d of Lys deprivation from the normal state, an intragastric load of Lys activated the ventral tegmental area, raphe pallidus nucleus and hippocampus, as well as several hypothalamic areas. After recovering from the Lys-deficient state, brain activation was similar to that in the normal state. These results indicate that neural plasticity in the prefrontal cortex, hypothalamic area and limbic system is related to the internal Lys state and that this plasticity could have important roles in the control of Lys intake.


Subject(s)
Brain/physiology , Dopaminergic Neurons/physiology , Feeding Behavior , Lysine/metabolism , Neuronal Plasticity , Nutritional Status , Animals , Behavior, Animal , Brain/physiopathology , Brain Mapping , Catheters, Indwelling , Hippocampus/physiology , Hippocampus/physiopathology , Hypothalamus/physiology , Hypothalamus/physiopathology , Limbic System/physiology , Limbic System/physiopathology , Lysine/administration & dosage , Lysine/deficiency , Lysine/therapeutic use , Magnetic Resonance Imaging , Male , Raphe Nuclei/physiology , Raphe Nuclei/physiopathology , Rats , Rats, Wistar , Stomach , Ventral Tegmental Area/physiology , Ventral Tegmental Area/physiopathology
10.
Neuroscience ; 220: 201-7, 2012 Sep 18.
Article in English | MEDLINE | ID: mdl-22710065

ABSTRACT

Seasonal affective disorder (SAD), a major depressive disorder recurring in the fall and winter, is caused by the reduction of light in the environment, and its depressive symptoms can be alleviated by bright light therapy. Both circadian and monoaminergic systems have been implicated in the etiology of SAD. However, the underlying neural pathways through which light regulates mood are not well understood. The present study utilized a diurnal rodent model, Arvicanthis niloticus, to explore the neural pathways mediating the effects of light on brain regions involved in mood regulation. Animals kept in constant darkness received light exposure in early subjective day, the time when light therapy is usually applied. The time course of neural activity following light exposure was assessed using Fos protein as a marker in the following brain regions/cells: the suprachiasmatic nucleus (SCN), orexin neurons in the perifornical-lateral hypothalamic area (PF-LHA) and the dorsal raphe nucleus (DRN). A light-induced increase in Fos expression was observed in orexin neurons and the DRN, but not in the SCN. As the DRN is densely innervated by orexinergic inputs, the involvement of orexinergic signaling in mediating the effects of light on the DRN was tested in the second experiment. The animals were injected with the selective orexin receptor type 1 (OXR1) antagonist SB-334867 prior to the light exposure. The treatment of SB-334867 significantly inhibited the Fos induction in the DRN. The results collectively point to the role of orexin neurons in mediating the effects of light on the mood-regulating monoaminergic areas, suggesting an orexinergic pathway that underlies light-dependent mood fluctuation and the beneficial effects of light therapy.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Neural Pathways/physiology , Neuropeptides/metabolism , Raphe Nuclei/physiology , Signal Transduction/physiology , Animals , Circadian Rhythm/physiology , Darkness , Immunohistochemistry , Light , Neurons/metabolism , Orexins , Phototherapy , Proto-Oncogene Proteins c-fos/analysis , Proto-Oncogene Proteins c-fos/biosynthesis , Rats , Seasonal Affective Disorder/metabolism , Seasonal Affective Disorder/physiopathology
11.
Poult Sci ; 91(4): 817-22, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22399719

ABSTRACT

Aggression and cannibalism in laying hens can differ in intensity and degree due to many factors, including genetics. Previous behavioral analysis of 2 strains of White Leghorns, DeKalb XL (DXL) and HGPS (a group-selected line for high group productivity and survivability), revealed high and low aggressive phenotypes, respectively. However, the exact genetic mechanisms mediating aggressiveness are currently unknown. Analysis of serotonin (5-HT) mediation of aggression in subordinate hens of these strains revealed increases in aggression in DXL hens following antagonism of the 5-HT1A receptor and in HGPS hens following antagonism of the 5-HT1B receptor. Here, we investigate the different neurotransmitter response in the hypothalamus and raphe nucleus mediating these aggressive responses to receptor antagonism. Elevated aggressive response to 5-HT1B antagonism by HGPS hens was also accompanied by a decrease in raphe nucleus dopamine (DA) and an increase in DA turnover. Increased aggressiveness in DXL hens did not coincide with a reduction in raphe nucleus 5-HT or turnover (as indicated by 5-hydroxyindoleacetic acid levels) following 5-HT1A antagonism. A reduction in 5-hydroxyindoleacetic acid (but not 5-HT) was seen in HGPS hens treated with 5-HT1A antagonist; however, these hens exhibited no change in aggressive behaviors. Our data show evidence of different heritable mechanisms of neurotransmitter regulation of aggressive response, specifically heritable differences in the interaction between 5-HT and catecholamines in regulating aggression.


Subject(s)
Aggression , Chickens/physiology , Hypothalamus/physiology , Raphe Nuclei/physiology , Serotonin/metabolism , Synaptic Transmission , Animals , Chickens/genetics , Chromatography, High Pressure Liquid/veterinary , Female , Oxadiazoles/pharmacology , Piperazines/pharmacology , Random Allocation , Receptor, Serotonin, 5-HT1A/metabolism , Receptor, Serotonin, 5-HT1B/metabolism , Selection, Genetic , Serotonin 5-HT1 Receptor Antagonists/pharmacology , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Species Specificity
12.
J Psychopharmacol ; 26(4): 575-83, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21427203

ABSTRACT

Mimosa pudica (Linn.) (M. pudica L.) is a plant used in some countries to treat anxiety and depression. In the present study we investigated the effects of an aqueous extract of M. pudica L. on mouse anxiety-like behaviour using the elevated T maze, and on regulation of dorsal raphe nucleus (DRN) 5-hydroxytryptamine (5-HT) neuronal activity using an in-vitro mouse brain slice preparation. Acute treatment with M. pudica L. extract had an anxiolytic effect on behaviour in the elevated T maze, specifically on inhibitory avoidance behaviour. Acute application of the extract alone had no effect on the activity of DRN 5-HT neurones. However, when co-applied with the GABA(A) receptor agonist THIP (4,5,6,7-tetrahydroisoxazolo[5,4-c]pyridin-3-ol), the extract enhanced the inhibitory effect of the THIP on DRN 5-HT neurones. These observed effects of M. pudica L. on both behaviour and GABA modulation of 5-HT neuronal activity are similar to the effects of diazepam, the established anxiolytic and positive modulator of the GABA(A) receptor. This study suggests that the aqueous extract of M. pudica L. contains a positive modulator of GABA(A) receptor function and provides impetus for further investigation of the neuropharmacologically active constituents of the extract.


Subject(s)
Anti-Anxiety Agents/pharmacology , GABA Modulators/pharmacology , Mimosa , Plant Extracts/pharmacology , Raphe Nuclei/drug effects , Animals , Anxiety/drug therapy , Diazepam/pharmacology , Isoxazoles/pharmacology , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , N-Methylaspartate/pharmacology , Phytotherapy , Raphe Nuclei/physiology , Receptors, GABA-A/drug effects , Serotonin/pharmacology , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
13.
Br J Pharmacol ; 165(8): 2620-34, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21827451

ABSTRACT

BACKGROUND AND PURPOSE: To evaluate the hypothesis that activation of somatodendritic 5-HT(1A) autoreceptors in the dorsal raphe nucleus (DRN) produces the anti-emetic/anti-nausea effects of cannabidiol (CBD), a primary non-psychoactive cannabinoid found in cannabis. EXPERIMENTAL APPROACH: The potential of systemic and intra-DRN administration of 5-HT(1A) receptor antagonists, WAY100135 or WAY100635, to prevent the anti-emetic effect of CBD in shrews (Suncus murinus) and the anti-nausea-like effects of CBD (conditioned gaping) in rats were evaluated. Also, the ability of intra-DRN administration of CBD to produce anti-nausea-like effects (and reversal by systemic WAY100635) was assessed. In vitro studies evaluated the potential of CBD to directly target 5-HT(1A) receptors and to modify the ability of the 5-HT(1A) agonist, 8-OH-DPAT, to stimulate [(35) S]GTPγS binding in rat brainstem membranes. KEY RESULTS: CBD suppressed nicotine-, lithium chloride (LiCl)- and cisplatin (20 mg·kg(-1) , but not 40 mg·kg(-1) )-induced vomiting in the S. murinus and LiCl-induced conditioned gaping in rats. Anti-emetic and anti-nausea-like effects of CBD were suppressed by WAY100135 and the latter by WAY100635. When administered to the DRN: (i) WAY100635 reversed anti-nausea-like effects of systemic CBD, and (ii) CBD suppressed nausea-like effects, an effect that was reversed by systemic WAY100635. CBD also displayed significant potency (in a bell-shaped dose-response curve) at enhancing the ability of 8-OH-DPAT to stimulate [(35) S]GTPγS binding to rat brainstem membranes in vitro. Systemically administered CBD and 8-OH-DPAT synergistically suppressed LiCl-induced conditioned gaping. CONCLUSIONS AND IMPLICATIONS: These results suggest that CBD produced its anti-emetic/anti-nausea effects by indirect activation of the somatodendritic 5-HT(1A) autoreceptors in the DRN. LINKED ARTICLES: This article is part of a themed section on Cannabinoids in Biology and Medicine. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2012.165.issue-8. To view Part I of Cannabinoids in Biology and Medicine visit http://dx.doi.org/10.1111/bph.2011.163.issue-7.


Subject(s)
Antiemetics/therapeutic use , Cannabidiol/therapeutic use , Raphe Nuclei/physiology , Receptor, Serotonin, 5-HT1A/physiology , Serotonin 5-HT1 Receptor Agonists/pharmacology , Vomiting/drug therapy , 8-Hydroxy-2-(di-n-propylamino)tetralin/metabolism , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Animals , Behavior, Animal/drug effects , Cannabis , Female , Male , Nausea/drug therapy , Nausea/physiopathology , Piperazines/pharmacology , Pyridines/pharmacology , Raphe Nuclei/drug effects , Rats , Rats, Sprague-Dawley , Serotonin 5-HT1 Receptor Antagonists/pharmacology , Shrews , Vomiting/physiopathology
14.
Pharmacol Biochem Behav ; 99(4): 566-72, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21689675

ABSTRACT

It has been reported that the sedative component of pentobarbital is mediated by GABA receptors in an endogenous sleep pathway and the ventrolateral preoptic area (VLPO)-tuberomammillary nucleus (TMN) or VLPO-dorsal raphe nucleus (DRN) neural circuit is important in the sedative response to pentobarbital. Our previous findings indicated that the VLPO-TMN neuronal circuit may play crucial part in the augmentative effect of diltiazem on pentobarbital sleep and the serotonergic system may be involved. This study was designed to investigate the role of DRN and the serotonergic receptors 5-HT(1A) and 5-HT(2A/2C) in the augmentative effect of diltiazem on pentobarbital-induced hypnosis in rats. The results showed that diltiazem (5mg/kg, i.g.) significantly reversed pentobarbital-induced (35 mg/kg, i.p.) reduction of c-Fos expression in 5-HT neurons of DRNV (at -7.5mm Bregma), DRND, DRNVL and MRN (at -8.0mm Bregma). However it did not influence this reducing effect of pentobarbital on non-5-HT neurons either in DRN or in MRN. Moreover, the effect of diltiazem (1 or 2mg/kg, i.g.) on pentobarbital-induced (35 mg/kg, i.p.) hypnosis was significantly inhibited by 5-HT(1A) agonist 8-OH-DPAT (0.5mg/kg, i.p.) and 5-HT(2A/2C) agonist DOI (0.5mg/kg, i.p.), and potentiated by 5-HT(1A) antagonist p-MPPI (2mg/kg, i.p.) and 5-HT(2A/2C) antagonist ritanserin (2mg/kg, i.p.), respectively. From these results, it should be presumed that the augmentative effect of diltiazem on pentobarbital-induced sleep may be related to 5-HT(1A) and 5-HT(2A/2C) receptors, and DRN may be involved. In addition, it also suggested that the DRN may play a multi-modulating role in sleep-wake regulation rather than being recognized simply as arousal nuclei.


Subject(s)
Calcium Channel Blockers/pharmacology , Diltiazem/pharmacology , Hypnotics and Sedatives/pharmacology , Pentobarbital/pharmacology , Raphe Nuclei/physiology , Receptor, Serotonin, 5-HT1A/drug effects , Receptor, Serotonin, 5-HT2A/drug effects , Receptor, Serotonin, 5-HT2C/drug effects , Animals , Brain/cytology , Brain/drug effects , Cell Count , Drug Synergism , Electroencephalography/drug effects , Electromyography/drug effects , Gene Expression/drug effects , Genes, fos/drug effects , Immunohistochemistry , Male , Polysomnography , Raphe Nuclei/drug effects , Rats , Rats, Sprague-Dawley , Serotonergic Neurons/drug effects , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacology
15.
Int J Neuropsychopharmacol ; 14(2): 211-23, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20149268

ABSTRACT

Triple reuptake inhibitors represent a potential new class of antidepressant drugs that block norepinephrine (NE), dopamine (DA) and serotonin [5-hydroxytryptamine (5-HT)] transporters. The present in-vivo electrophysiological study was undertaken to determine the effects of the triple reuptake inhibitors SEP-225289 and DOV216303 on the neuronal activities of locus coeruleus (LC) NE, ventral tegmental area (VTA) DA and dorsal raphe (DR) 5-HT neurons. Administered acutely, SEP-225289 and DOV216303 dose-dependently decreased the spontaneous firing rate of LC NE, VTA DA and DR 5-HT neurons through the activation of α2, D2 and 5-HT(1A) autoreceptors, respectively. Both compounds predominantly inhibited the firing rate of LC NE neurons while producing only a partial decrease in VTA DA and DR 5-HT neuronal discharge. SEP-225289 was equipotent at inhibiting 5-HT and NE transporters since it prolonged to the same extent the time required for a 50% recovery (RT50) of the firing activity of dorsal hippocampus CA3 pyramidal neurons from the inhibition induced by microiontophoretic application of 5-HT and NE. Finally, in the presence of WAY100635, a 5-HT(1A) receptor antagonist, SEP-225289 activated 5-HT neurons at doses that normally did not inhibit them. Taken together, the present results indicate that reciprocal interactions among NE, DA and 5-HT inputs need to be considered to anticipate the net effect of triple reuptake inhibitors on the enhancement of brain monoamine transmission. The results also suggest that the therapeutic action of triple reuptake inhibitors may be potentiated by antagonizing the cell body 5-HT(1A) autoreceptors.


Subject(s)
Adrenergic Antagonists/pharmacology , Amines/pharmacology , Aza Compounds/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cyclobutanes/pharmacology , Dopamine Antagonists/pharmacology , Electrophysiological Phenomena/drug effects , Neurons/drug effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Animals , Antidepressive Agents/pharmacology , Dopamine/metabolism , Drug Evaluation, Preclinical , Hippocampus/drug effects , Hippocampus/physiology , Locus Coeruleus/drug effects , Locus Coeruleus/physiology , Male , Neurons/metabolism , Norepinephrine/metabolism , Raphe Nuclei/drug effects , Raphe Nuclei/physiology , Rats , Rats, Sprague-Dawley , Serotonin/metabolism , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/physiology
16.
Am J Physiol Regul Integr Comp Physiol ; 299(5): R1369-76, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20720173

ABSTRACT

We have shown that electroacupuncture (EA) inhibits sympathoexcitatory rostral ventrolateral medulla (rVLM) neurons and reflex responses following activation of a long-loop pathway in the arcuate nucleus and ventrolateral periaqueductal gray (vlPAG). Additionally, EA at P 5-6 acupoints (overlying the median nerve) activates serotonin-containing neurons in the nucleus raphé pallidus (NRP), which, in turn, inhibit rVLM neurons. Although direct projections from the vlPAG to the rVLM exist, it is uncertain whether an indirect pathway through the NRP serves an important role in vlPAG-rVLM cardiovascular modulation. Therefore, the splanchnic nerve (SN) was stimulated to induce cardiovascular sympathoexcitatory reflexes, and EA was applied at P 5-6 acupoints in α-chloralose-anesthetized cats. A single-barreled recording electrode was inserted into the NRP or rVLM. Microinjection of DL-homocysteic acid (DLH) into the vlPAG increased the NRP neuronal response to SN stimulation (5 ± 1 to 12 ± 2 spikes/30 stim). Likewise, EA at P 5-6 for 30 min increased the NRP response to SN stimulation (3 ± 1 to 10 ± 2 spikes/30 stim), an effect that could be blocked by microinjection of kynurenic acid (KYN) into the caudal vlPAG. Furthermore, the reflex increase in blood pressure induced by application of bradykinin to the gallbladder and the rVLM cardiovascular presympathetic neuronal response to SN stimulation was inhibited by injection of DLH into the vlPAG, a response that was reversed by injection of KYN into the NRP. These results indicate that EA activates the vlPAG, which excites the NRP to, in turn, inhibit rVLM presympathetic neurons and reflex cardiovascular sympathoexcitatory responses.


Subject(s)
Cardiovascular System/innervation , Electroacupuncture , Medulla Oblongata/physiology , Mesencephalon/physiology , Neural Inhibition , Raphe Nuclei/physiology , Sympathetic Nervous System/physiology , Action Potentials , Animals , Blood Pressure , Bradykinin/administration & dosage , Cardiovascular System/drug effects , Cats , Excitatory Amino Acid Antagonists/administration & dosage , Female , Homocysteine/administration & dosage , Homocysteine/analogs & derivatives , Kynurenic Acid/administration & dosage , Male , Medulla Oblongata/drug effects , Mesencephalon/drug effects , Microinjections , Neural Inhibition/drug effects , Neural Pathways/physiology , Raphe Nuclei/drug effects , Reflex , Splanchnic Nerves/physiology , Sympathetic Nervous System/drug effects , Time Factors
17.
J Psychopharmacol ; 24(8): 1223-35, 2010 Aug.
Article in English | MEDLINE | ID: mdl-19939862

ABSTRACT

Nomifensine potently inhibits the reuptake of norepinephrine and dopamine in vitro. It is one of few antidepressants with marked potency to block dopamine reuptake that has ever been used clinically. Acute and sustained administration of nomifensine was investigated on the firing of monoaminergic neurons to understand its mechanism of action. In vivo extracellular recordings of locus coeruleus, ventral tegmental area and dorsal raphe nucleus neurons were obtained from male Sprague-Dawley rats. The intravenous injection of nomifensine in the locus coeruleus and ventral tegmental area yielded ED(50) values of 40 +/- 1 and 450 +/- 41 microg/kg, respectively, suggesting that nomifensine directly acted upon dopamine and norepinephrine neurons, since these values are proportional to its affinities for norepinephrine and dopamine transporters. There was no effect on 5-HT neurons. Nomifensine (5 mg/kg/day, subcutaneous, using minipumps) potently and significantly inhibited dopamine neuronal firing in the ventral tegmental area after 2 days, with recovery to normal after the 14-day treatment due to D(2) autoreceptor desensitization. Norepinephrine neuronal firing in the locus coeruleus was significantly decreased after 2 and 14 days. A significant increase in dorsal raphe nucleus 5-HT neuronal firing was seen after a two-day regimen, and remained elevated after 14 days. Desensitization of the 5-HT(1A) receptor on 5-HT neurons of the dorsal raphe nucleus occurred after two days of nomifensine administration. Nomifensine likely treated depression by acting on dopamine, norepinephrine and 5-HT neurons, highlighting the importance of the functional connectivity between these three monoaminergic systems.


Subject(s)
Dopamine/physiology , Neurons/drug effects , Nomifensine/administration & dosage , Norepinephrine/physiology , Action Potentials , Adrenergic Uptake Inhibitors/administration & dosage , Animals , Delayed-Action Preparations , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Electrophysiological Phenomena , Locus Coeruleus/drug effects , Locus Coeruleus/metabolism , Male , Morpholines/administration & dosage , Neurons/physiology , Neurotransmitter Uptake Inhibitors/administration & dosage , Neurotransmitter Uptake Inhibitors/pharmacology , Nomifensine/pharmacology , Raphe Nuclei/drug effects , Raphe Nuclei/physiology , Rats , Rats, Sprague-Dawley , Reboxetine , Receptor, Serotonin, 5-HT1A/metabolism , Receptors, Dopamine D2/metabolism , Serotonin/blood , Time Factors , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism
18.
J Neurosci ; 29(38): 11954-64, 2009 Sep 23.
Article in English | MEDLINE | ID: mdl-19776281

ABSTRACT

Sympathetic premotor neurons in the rostral medullary raphe (RMR) regulate heat conservation by tail artery vasoconstriction and brown adipose tissue thermogenesis. These neurons are a critical relay in the pathway that increases body temperature. However, the origins of the inputs that activate the RMR during cold exposure have not been definitively identified. We investigated the afferents to the RMR that were activated during cold by examining Fos expression in retrogradely labeled neurons after injection of cholera toxin B subunit (CTb) in the RMR. These experiments identified a cluster of Fos-positive neurons in the dorsomedial hypothalamic nucleus and dorsal hypothalamic area (DMH/DHA) with projections to the RMR that may mediate cold-induced elevation of body temperature. Also, neurons in the median preoptic nucleus (MnPO) and dorsolateral preoptic area (DLPO) and in the A7 noradrenergic cell group were retrogradely labeled but lacked Fos expression, suggesting that they may inhibit the RMR. To investigate whether individual or common preoptic neurons project to the RMR and DMH/DHA, we injected CTb into the RMR and Fluorogold into the DMH/DHA. We found that projections from the DLPO and MnPO to the RMR and DMH/DHA emerge from largely separate neuronal populations, indicating they may be differentially regulated. Combined cell-specific lesions of MnPO and DLPO, but not lesions of either one alone, caused baseline hyperthermia. Our data suggest that the MnPO and DLPO provide parallel inhibitory pathways that tonically inhibit the DMH/DHA and the RMR at baseline, and that hyperthermia requires the release of this inhibition from both nuclei.


Subject(s)
Body Temperature Regulation/physiology , Hypothalamus/physiology , Neurons/physiology , Preoptic Area/physiology , Raphe Nuclei/physiology , Animals , Body Temperature , Catecholamines/metabolism , Cholera Toxin , Cold Temperature , Fever/chemically induced , Fever/physiopathology , Lipopolysaccharides , Male , Neural Pathways/injuries , Neural Pathways/physiology , Norepinephrine/metabolism , Preoptic Area/injuries , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Time Factors
19.
J Rehabil Res Dev ; 46(1): 109-22, 2009.
Article in English | MEDLINE | ID: mdl-19533524

ABSTRACT

Serotonin in the spinal cord acutely modulates nociceptive transmission and motor reflexes and may also assist functional restoration after spinal cord injury (SCI). It is released there mainly by descending axons of the medulla's nucleus raphe magnus (NRM). We examined whether mechanical allodynia (cutaneous hypersensitivity) after incomplete SCI is sustainably reversed by prolonged, intermittent electrical stimulation of the NRM and whether altered NRM activity accounts for the allodynia. NRM stimulation was given to rats over several days (average 3.2), beginning about 1 hour after moderate thoracic contusion injury. This stimulation reduced mechanical allodynia in forepaws but not hindpaws at 6 weeks after injury (vs nonstimulated controls). Histologically, the stimulation augmented white matter and reduced astrocytosis (glial fibrillary acidic protein immunostaining) in injured segments at 15 weeks. Cavity volume and perilesion neuron numbers were unchanged. Single-cell extracellular recording 12 to 14 weeks after thoracic contusion injury revealed generally higher spontaneous firing and weaker responses to above-injury noxious stimulation in both inhibited and excited NRM neurons; unresponsive neurons were fewer. Neurons inhibited from dermatomes above the injury were excited from below. Altered NRM activity is unlikely to cause SCI allodynia, since inhibited and excited classes are believed to oppositely modulate nociception. Prolonged, early NRM stimulation probably reverses above-injury allodynia by facilitating qualitative recovery of remaining tissue.


Subject(s)
Electric Stimulation Therapy , Pain/physiopathology , Raphe Nuclei/physiology , Spinal Cord Injuries/rehabilitation , Action Potentials , Analgesia , Animals , Electrodes, Implanted , Female , Male , Neurons/physiology , Pain/etiology , Pain Measurement , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Spinal Cord/pathology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology
20.
Brain Res ; 1227: 76-88, 2008 Aug 28.
Article in English | MEDLINE | ID: mdl-18598674

ABSTRACT

CART (cocaine and amphetamine regulated transcript) is a neuropeptide involved in the control of several physiological processes, such as response to psychostimulants, food intake, depressive diseases and neuroprotection. It is robustly expressed in the brain, mainly in regions that control emotional and stress responses and it is regulated by estrogen in the hypothalamus. There is a distinct population of CART neurons located in the vicinity of the Edinger-Westphal nucleus of the midbrain that also colocalize urocortin-1. The aims of this study were 1) to determine the distribution of CART immunoreactive neurons in the monkey midbrain, 2) to examine the effects of estrogen (E) and progesterone (P) on midbrain CART mRNA and peptide expression and 3) to determine whether midbrain CART neurons contain steroid receptors. Adult female rhesus monkeys (Macaca mulatta) were spayed and either treated with placebo (OVX), estrogen alone (E), progesterone alone (P) or E+P. Animals were prepared (a) for RNA extraction followed by microarray analysis and quantitative (q) RT-PCR (n=3/group); (b) for immunohistochemical analysis of CART and CART+tryptophan hydroxylase (TPH), CART+estrogen receptors (ER) or CART+progesterone receptors (n=5/group) and (c) for Western blots (n=3/group). Both E- and E+P-administration decreased CART gene expression on the microarray and with qRT-PCR. Stereological analysis of CART immunostaining at five levels of the Edinger-Westphal nucleus indicated little effect of E or E+P administration on the area of CART immunostaining. However, P administration increased CART-immunopositive area in comparison to the OVX control group with Student's t-test, but not with ANOVA. CART 55-102 detection on Western blot was unchanged by hormone administration. ERbeta and PR were detected in CART neurons and CART fibers appeared to innervate TPH-positive serotonin neurons in the dorsal raphe. In summary, E decreased CART mRNA, but this effect did not translate to the protein level. Moreover, P administration alone had a variable effect on CART mRNA, but it caused an increase in CART immunostaining. Together, the data suggest that CART neurons in the midbrain have a unique steroid response, which may be mediated by nuclear receptors, neuroactive steroids or interneurons.


Subject(s)
Estrogens/metabolism , Macaca mulatta/metabolism , Mesencephalon/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Peptide Fragments/metabolism , Progesterone/metabolism , Animals , Blotting, Western , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Estrogens/pharmacology , Estrogens/physiology , Female , Gene Expression/drug effects , Hypothalamus/drug effects , Hypothalamus/metabolism , Hypothalamus/physiology , Immunohistochemistry , Macaca mulatta/genetics , Macaca mulatta/physiology , Mesencephalon/drug effects , Mesencephalon/physiology , Microarray Analysis/methods , Nerve Tissue Proteins/genetics , Neurons/drug effects , Neurons/physiology , Ovariectomy/methods , Ovary/metabolism , Peptide Fragments/genetics , Progesterone/pharmacology , Progesterone/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Raphe Nuclei/drug effects , Raphe Nuclei/metabolism , Raphe Nuclei/physiology , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Serotonin/metabolism , Tryptophan Hydroxylase/genetics , Tryptophan Hydroxylase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL