Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 10 de 10
1.
bioRxiv ; 2024 Feb 06.
Article En | MEDLINE | ID: mdl-38370807

Opioid use disorder occurs alongside impaired risk-related decision-making, but the underlying neural correlates are unclear. We developed a novel approach-avoidance conflict model using a modified conditioned place preference paradigm to study neural signals of risky opioid seeking in the prefrontal cortex, a region implicated in executive decision making. Upon establishment of morphine conditioned place preference, rats underwent a subsequent conflict test in which fear-inducing cat odor was introduced in the previously drug-paired side of the apparatus. While the saline control group avoided the cat odor side, the morphine group maintained preference for the paired side despite the presence of cat odor. K-means clustering identified two subsets of morphine-treated rats that exhibited either persistent drug seeking (Risk-Takers) or increased avoidance (Risk-Avoiders) during conflict. Single-unit recordings from the prelimbic cortex (PL) revealed decreased neuronal firing rates upon acute morphine exposure in both Risk-Takers and Risk-Avoiders, but this firing rate suppression was absent after repeated administration. Risk-Avoiders also displayed distinct post-morphine excitation in PL which persisted across conditioning. During the preference test, subpopulations of PL neurons in all groups were either excited or inhibited when rats entered the paired side. Interestingly, while this inhibitory signal was lost during the subsequent conflict test in both saline and Risk-Avoider groups, these inhibitory responses persisted in Risk-Takers. Our results suggest that loss of PL inhibition after opioid conditioning is associated with the formation of contextual reward memory. Furthermore, persistent PL inhibitory signaling in the drug-associated context during conflict may underlie increased risk taking following opioid exposure.

2.
Elife ; 102021 12 16.
Article En | MEDLINE | ID: mdl-34913438

The recollection of environmental cues associated with threat or reward allows animals to select the most appropriate behavioral responses. Neurons in the prelimbic (PL) cortex respond to both threat- and reward-associated cues. However, it remains unknown whether PL regulates threat-avoidance vs. reward-approaching responses when an animals' decision depends on previously associated memories. Using a conflict model in which male Long-Evans rats retrieve memories of shock- and food-paired cues, we observed two distinct phenotypes during conflict: (1) rats that continued to press a lever for food (Pressers) and (2) rats that exhibited a complete suppression in food seeking (Non-pressers). Single-unit recordings revealed that increased risk-taking behavior in Pressers is associated with persistent food-cue responses in PL, and reduced spontaneous activity in PL glutamatergic (PLGLUT) neurons during conflict. Activating PLGLUT neurons in Pressers attenuated food-seeking responses in a neutral context, whereas inhibiting PLGLUT neurons in Non-pressers reduced defensive responses and increased food approaching during conflict. Our results establish a causal role for PLGLUT neurons in mediating individual variability in memory-based risky decision-making by regulating threat-avoidance vs. reward-approach behaviors.


Cues , Neurons/physiology , Prefrontal Cortex/physiology , Reward , Animals , Male , Rats , Rats, Long-Evans
3.
Neurosci Lett ; 734: 135095, 2020 08 24.
Article En | MEDLINE | ID: mdl-32473195

Dehydroepiandrosterone (DHEA) modulates dopaminergic neurotransmission. It takes part in neurologic and psychiatric diseases involving monoamine neurotransmitters. Earlier results show that DHEA (120-min treatment) reduced striatal dopamine (DA) turnover in rats, suggesting a reduced DA release. Some investigations report that DHEA increases DA release but inhibits motor activity, which seems contradictory. This research examines the effect of DHEA on striatal DA release, its metabolism and motor activity. Male Wistar rats were implanted in the striatum with a cannula for in vivo microdialysis. DHEA was administered (120 mg/kg) and dialysates were collected for 280 min. A depolarizing stimulus was applied at 120 min. Samples were analyzed by HPLC-ED to determine the concentration of DA and its metabolites. The effect of DHEA on motor activity was also evaluated during 120 min. Extracellular DA concentration was greater in treated animals both before and after depolarization. In contrast, DHEA reduced the areas below the curves for DA metabolites and DA/metabolite ratios. DHEA also reduced motor activity, remarkably in the first 20 min after treatment. In summary, DHEA yielded a stimulatory effect on striatal DA release that was not reflected in neither DA metabolism nor motor activity. Thus, DHEA resembles the effect of typical antipsychotics, increasing DA release but reducing behavioral activation.


Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dehydroepiandrosterone/pharmacology , Dopamine/metabolism , Motor Activity/drug effects , Animals , Male , Rats , Rats, Wistar
4.
Front Neurosci ; 13: 360, 2019.
Article En | MEDLINE | ID: mdl-31040765

The dopaminergic and histaminergic systems are the first to appear during the development of the nervous system. Through the activation of H1 receptors (H1Rs), histamine increases neurogenesis of the cortical deep layers, while reducing the dopaminergic phenotype (cells immunoreactive to tyrosine hydroxylase, TH+) in embryo ventral mesencephalon. Although the function of histamine in neuronal differentiation has been studied, the role of H1Rs in neurogenesis has not been addressed. For this purpose, the H1R antagonist/inverse agonist chlorpheniramine was systemically administered (5 mg/kg, i.p.) to pregnant Wistar rats (gestational days 12-14, E12-14), and control and experimental embryos (E14 and E16) and pups (21-day-old) were evaluated for changes in nigro-striatal development. Western blot and immunohistochemistry determinations showed a significant increase in the dopaminergic markers' TH and PITX3 in embryos from chlorpheniramine-treated rats at E16. Unexpectedly, 21-day-old pups from the chlorpheniramine-treated group, showed a significant reduction in TH immunoreactivity in the substantia nigra pars compacta and dorsal striatum. Furthermore, striatal dopamine content, evoked [3H]-dopamine release and methamphetamine-stimulated motor activity were significantly lower compared to the control group. These results indicate that H1R blockade at E14-E16 favors the differentiation of dopaminergic neurons, but hampers their migration, leading to a decrease in dopaminergic innervation of the striatum in post-natal life.

5.
Article En | MEDLINE | ID: mdl-31108178

We have investigated the effect of the local activation of histamine H3 receptors (H3Rs) in the rat prefrontal cortex (PFCx) on the impairment of pre-pulse inhibition (PPI) of the startle response induced by the systemic administration of MK-801, antagonist at glutamate N-Methyl-d-Aspartate (NMDA) receptors, and the possible functional interaction between H3Rs and MK-801 on PFCx dopaminergic transmission. Infusion of the H3R agonist RAMH (19.8 ng/1 µl) into the PFCx reduced or prevented the inhibition by MK-801 (0.15 mg/kg, ip) of PPI evoked by different auditory stimulus intensities (5, 10 and 15 dB), and the RAMH effect was blocked by the H3R antagonist/inverse agonist ciproxifan (30.6 ng/1 µl). MK-801 inhibited [3H]-dopamine uptake (-45.4 ±â€¯2.1%) and release (-32.8 ±â€¯2.6%) in PFCx synaptosomes or slices, respectively, and molecular modeling indicated that MK-801 binds to and blocks the rat and human dopamine transporters. However, H3R activation had no effect on the inhibitory action of MK-801 on dopamine uptake and release. In PFCx slices, MK-801 and the activation of H3Rs or dopamine D1 receptors (D1Rs) stimulated ERK-1/2 and Akt phosphorylation. The co-activation of D1Rs and H3Rs prevented ERK-1/2 and Akt phosphorylation, and H3R activation or D1R blockade prevented the effect of MK-801. In ex vivo experiments, the intracortical infusion of the D1R agonist SKF-81297 (37 ng/1 µl) or the H3R agonist RAMH increased Akt phosphorylation, prevented by D1R/H3R co-activation. These results indicate that MK-801 enhances dopaminergic transmission in the PFCx, and that H3R activation counteracts the post-synaptic actions of dopamine.


Dizocilpine Maleate/pharmacology , Prepulse Inhibition/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Histamine H3/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Reflex, Startle/drug effects , Animals , Benzazepines/administration & dosage , Benzazepines/pharmacology , Dizocilpine Maleate/administration & dosage , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Histamine Agonists/administration & dosage , Histamine Agonists/pharmacology , Imidazoles/administration & dosage , Imidazoles/pharmacology , Male , Microinjections , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Molecular Docking Simulation , Phosphorylation/drug effects , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Receptors, N-Methyl-D-Aspartate/metabolism , Tritium/metabolism
6.
Pharmacol Rep ; 70(1): 146-155, 2018 Feb.
Article En | MEDLINE | ID: mdl-29414147

BACKGROUND: Clobenpropit, a potent antagonist/inverse agonist at the histamine H3 receptor (H3R), reduced the cytotoxic action of 6-hydroxydopamine (6-OHDA) in neuroblastoma SH-SY5Y cells transfected with the human H3R. We therefore set out to study whether this effect involved a receptor-independent action on dopamine transport. METHODS: The uptake of [3H]-dopamine was assayed in SH-SY5Y cells and rat striatal or cerebro-cortical isolated nerve terminals (synaptosomes). Clobenpropit binding to the human norepinephrine (NET) and dopamine (DAT) transporters was analyzed by molecular modeling. RESULTS: In SH-SY5Y cells, [3H]-dopamine uptake was inhibited by desipramine (selective NET inhibitor), GBR-12909 (selective DAT inhibitor), and fluoxetine (selective inhibitor of the serotonin transporter, SERT) with IC50 values 37, 537, and 2800nM, respectively. The potency rank order indicates that [3H]-dopamine uptake is primarily performed by NET. Clobenpropit inhibited [3H]-dopamine uptake (maximum inhibition 82.7±2.8%, IC50 490nM), and the effect was reproduced by the H3R antagonist/inverse agonist iodophenpropit, but not by the agonists R-α-methylhistamine and immepip or the antagonists/inverse agonists ciproxifan and A-331440. Clobenpropit also inhibited [3H]-dopamine uptake by rat striatal and cerebro-cortical synaptosomes (-54.6±11.3% and -46.3±9.6%, respectively, at 10µM). Modeling of the human NET and DAT obtained by homology from the crystal of Drosophila melanogaster DAT showed that clobenpropit can bind to a site also recognized in both transporters by nisoxetine, a potent NET inhibitor. CONCLUSION: These data indicate a direct inhibitory effect of clobenpropit on catecholamine transport.


Brain/drug effects , Dopamine Plasma Membrane Transport Proteins/antagonists & inhibitors , Dopamine Uptake Inhibitors/pharmacology , Dopamine/metabolism , Histamine H3 Antagonists/pharmacology , Imidazoles/pharmacology , Receptors, Histamine H3/drug effects , Synaptosomes/drug effects , Thiourea/analogs & derivatives , Animals , Binding Sites , Brain/metabolism , Cell Line, Tumor , Dopamine Plasma Membrane Transport Proteins/chemistry , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopamine Uptake Inhibitors/chemistry , Dopamine Uptake Inhibitors/metabolism , Dose-Response Relationship, Drug , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/metabolism , Drug Inverse Agonism , Histamine H3 Antagonists/chemistry , Histamine H3 Antagonists/metabolism , Humans , Imidazoles/chemistry , Imidazoles/metabolism , Molecular Docking Simulation , Norepinephrine Plasma Membrane Transport Proteins/antagonists & inhibitors , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Protein Binding , Protein Conformation , Rats , Receptors, Histamine H3/metabolism , Synaptosomes/metabolism , Thiourea/chemistry , Thiourea/metabolism , Thiourea/pharmacology
7.
Neuropharmacology ; 106: 91-101, 2016 07.
Article En | MEDLINE | ID: mdl-26169221

We studied the effect of activating histamine H3 receptors (H3Rs) on rat nucleus accumbens (rNAcc) dopaminergic transmission by analyzing [(3)H]-dopamine uptake by synaptosomes, and dopamine synthesis and depolarization-evoked [(3)H]-dopamine release in slices. The uptake of [(3)H]-dopamine by rNAcc synaptosomes was not affected by the H3R agonist RAMH (10(-10)-10(-6) M). In rNAcc slices perfusion with RAMH (1 µM) had no significant effect on [(3)H]-dopamine release evoked by depolarization with 30 mM K(+) (91.4 ± 4.5% of controls). The blockade of dopamine D2 autoreceptors with sulpiride (1 µM) enhanced K(+)-evoked [(3)H]-dopamine release (168.8 ± 15.5% of controls), but under this condition RAMH (1 µM) also failed to affect [(3)H]-dopamine release. Dopamine synthesis was evaluated in rNAcc slices incubated with the l-dihydroxyphenylalanine (DOPA) decarboxylase inhibitor NSD-1015 (1 mM). Forskolin-induced DOPA accumulation (220.1 ± 10.4% of controls) was significantly reduced by RAMH (41.1 ± 6.5% and 43.5 ± 9.1% inhibition at 100 nM and 1 µM, respectively), and this effect was prevented by the H3R antagonist ciproxifan (10 µM). DOPA accumulation induced by preventing cAMP degradation with IBMX (iso-butyl-methylxantine, 1 mM) or by activating receptors for the vasoactive intestinal peptide (VIP)/pituitary adenylate cyclase-activating peptide (PACAP) with PACAP-27 (1 µM) was reduced (IBMX) or prevented (PACAP-27) by RAMH (100 nM). In contrast, DOPA accumulation induced by 8-Bromo-cAMP (1 mM) was not affected by RAMH (100 nM). These results indicate that in rNAcc H3Rs do not modulate dopamine uptake or release, but regulate dopamine synthesis by inhibiting cAMP formation and thus PKA activation. This article is part of the Special Issue entitled 'Histamine Receptors'.


Dopamine/metabolism , Nucleus Accumbens/metabolism , Receptors, Histamine H3/metabolism , Animals , Central Nervous System Agents/pharmacology , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Dihydroxyphenylalanine/metabolism , Histamine/metabolism , Male , Nucleus Accumbens/drug effects , Rats, Wistar , Receptors, Dopamine D2/metabolism , Synaptosomes/drug effects , Synaptosomes/metabolism , Tissue Culture Techniques
8.
Gac Med Mex ; 148(5): 467-75, 2012.
Article Es | MEDLINE | ID: mdl-23128888

Parkinson's disease is a progressive neurodegenerative movement disorder that results primarily from the death of dopaminergic neurons in the substantia nigra pars compacta. However, other neurotransmitter systems (noradrenergic,cholinergic and serotoninergic) are also involved in the disease. On the other hand, there is increasing evidence for a role of histamine as a neuromodulator in the mammalian central nervous system. Histamine-releasing neurons are exclusively located in the tuberomammilary nucleus of the hypothalamus, project to all major areas of the brain and participate in functions such as the regulation of sleep/wakefulness, locomotor activity, autonomic and vestibular functions, feeding and drinking, analgesia, learning and memory. In this work we review the pathophysiological characteristics of Parkinson's disease and the emerging information about alterations in histaminergic transmission reported for parkinsonian patients and animal models of the disease. In particular, we focus on the role of histamine H3 receptors, expressed at high density in the basal ganglia, in the normal function of these nuclei and their possible participation in the pathophysiology of Parkinson's disease.


Basal Ganglia/physiology , Neurotransmitter Agents/metabolism , Parkinson Disease/physiopathology , Receptors, Histamine H3/physiology , Humans
9.
Salud ment ; 35(4): 345-352, jul.-ago. 2012. ilus, tab
Article Es | LILACS-Express | LILACS | ID: lil-675564

Histamine regulates at the pre- and post-synaptic levels several functions of the mammalian Nervous System, in which three (H1, H2, and H3) out of the four G protein-coupled histamine receptors cloned so far are widely distributed. The histamine H3 receptor (H3R) was first identified as an auto-receptor controlling histamine synthesis and release, but several lines of evidence have shown the H3R to regulate as a hetero-receptor the release of a number of neuroactive substances, namely acetylcholine, 5-hydroxytryptamine (5-HT, serotonin), noradrenaline, dopamine, glutamate, y-aminobutyric acid (GABA) and the neuropeptides sustance P and calcitonin gene-related peptide (CGRP). H3R-mediated regulation of the release of these neurotransmitters and neuro-modulators, both in normal and pathological conditions, suggest that drugs acting at the receptor may have therapeutic use in a number of diseases such as sleep disorders, ischemia-induced cardiac arrhythmias, migraine, obesity, Alzheimer's disease and schizophrenia.


La histamina regula a nivel pre y postsináptico diversas funciones del Sistema Nervioso de los mamíferos, el cual expresa de manera abundante tres (H1, H2 y H3) de los cuatro receptores a histamina acoplados a proteínas G descritos a la fecha (H1-H4). El receptor a histamina H3 (H3R) se identificó inicialmente como el autorreceptor responsable del control de la liberación y la síntesis de la histamina. Posteriormente se estableció que este receptor se encuentra también en las terminales axónicas de otras neuronas del Sistema Nervioso Central y periférico, donde regula como heterorreceptor la liberación de varios transmisores. En este trabajo se revisan los efectos de la activación del H3R en la liberación de histamina, acetilcolina, 5-hidroxitriptamina (5-HT, serotonina), noradrenalina, dopamina, glutamato, ácido y-aminobutírico (GABA) y los neuropéptidos sustancia P y el péptido relacionado al gen de la calcitonina (CGRP). La regulación por el receptor H3 de la liberación de estos neurotransmisores y neuromoduladores, tanto en condiciones normales como patológicas, sugiere que los fármacos que actúen sobre dicho receptor pueden tener uso terapéutico en alteraciones diversas como los transtornos del sueño, las arritmias cardiacas causadas por isquemia, la migraña, la obesidad, la enfermedad de Alzheimer y la esquizofrenia.

10.
Neuropharmacology ; 62(2): 1127-33, 2012 Feb.
Article En | MEDLINE | ID: mdl-22115898

We have studied the effect of histamine H(3) receptor (H(3)R) activation on the depolarization-evoked release of labeled neurotransmitters from slices of the rat olfactory bulb (rOB). The presence of pre-synaptic H(3)Rs was evidenced by the specific binding of the H(3)R ligand N-α-[methyl-(3)H]histamine to membranes from rOB synaptosomes (maximum binding, B(max), 106 ± 19 fmol/mg protein; dissociation constant, K(d), 0.68 ± 0.11 nM) which was inhibited by selective H(3)R ligands (immepip, (R)(-)-α-methylhistamine (RAMH) and clobenpropit) with affinities similar to those previously reported for H(3)Rs expressed in other rat brain areas. Perfusion of rOB slices with the selective H(3)R agonist RAMH (0.1 and 1 µM) had no effect on the release of [(3)H]-γ-aminobutyric acid ([(3)H]-GABA), [(3)H]-d-aspartate, [(3)H]-dopamine or [(3)H]-5-hydroxytryptamine ([(3)H]-5-HT) evoked by depolarization with high K(+) (20 or 40 mM). [(3)H]-Noradrenaline release induced by 20 mM K(+) was reduced in a modest but significant manner by RAMH (94.9 ± 1.7% and 83.1 ± 2.1% of control release at 0.1 and 1 µM, respectively). The effect of 1 µM RAMH was blocked by the selective H(3)R antagonist/inverse agonist clobenpropit (5 µM). When tested alone clobenpropit and a second H(3)R antagonist/inverse agonist, ciproxifan (both at 1 µM) significantly increased K(+)-evoked [(3)H]-noradrenaline release to 119.4 ± 4.2% and 120.0 ± 3.7% of K(+) alone, respectively. Ciproxifan (1 µM) had no effect on the depolarization-evoked release of the other labeled neurotransmitters. These data indicate that H(3)Rs with constitutive activity modulate noradrenaline release in rOB, presumably through a pre-synaptic action. This article is part of a Special Issue entitled 'Post-Traumatic Stress Disorder'.


Norepinephrine/metabolism , Olfactory Bulb/metabolism , Receptors, Histamine H3/metabolism , Synaptic Transmission/physiology , Animals , Dopamine/metabolism , Male , Olfactory Bulb/drug effects , Rats , Rats, Wistar , Synaptic Transmission/drug effects , Synaptosomes/drug effects , Synaptosomes/metabolism , gamma-Aminobutyric Acid/metabolism
...