Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 301
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Psychiatry ; 29(4): 1046-1062, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38233467

RESUMEN

Serotonergic psychedelics are emerging therapeutics for psychiatric disorders, yet their underlying mechanisms of action in the brain remain largely elusive. Here, we developed a wide-field behavioral tracking system for larval zebrafish and investigated the effects of psilocybin, a psychedelic serotonin receptor agonist. Machine learning analyses of precise body kinematics identified latent behavioral states reflecting spontaneous exploration, visually-driven rapid swimming, and irregular swim patterns following stress exposure. Using this method, we found that acute psilocybin treatment has two behavioral effects: [i] facilitation of spontaneous exploration ("stimulatory") and [ii] prevention of irregular swim patterns following stress exposure ("anxiolytic"). These effects differed from the effect of acute SSRI treatment and were rather similar to the effect of ketamine treatment. Neural activity imaging in the dorsal raphe nucleus suggested that psilocybin inhibits serotonergic neurons by activating local GABAergic neurons, consistent with psychedelic-induced suppression of serotonergic neurons in mammals. These findings pave the way for using larval zebrafish to elucidate neural mechanisms underlying the behavioral effects of serotonergic psychedelics.


Asunto(s)
Ansiolíticos , Conducta Animal , Alucinógenos , Psilocibina , Neuronas Serotoninérgicas , Pez Cebra , Animales , Psilocibina/farmacología , Conducta Animal/efectos de los fármacos , Alucinógenos/farmacología , Ansiolíticos/farmacología , Neuronas Serotoninérgicas/efectos de los fármacos , Larva/efectos de los fármacos , Natación , Núcleo Dorsal del Rafe/efectos de los fármacos , Agonistas de Receptores de Serotonina/farmacología , Ansiedad/tratamiento farmacológico , Neuronas GABAérgicas/efectos de los fármacos , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Ketamina/farmacología , Encéfalo/efectos de los fármacos
2.
Cereb Cortex ; 31(2): 1090-1105, 2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33043981

RESUMEN

Cross-species studies have identified an evolutionarily conserved role for serotonin in flexible behavior including reversal learning. The aim of the current study was to investigate the contribution of serotonin within the orbitofrontal cortex (OFC) and medial prefrontal cortex (mPFC) to visual discrimination and reversal learning. Male Lister Hooded rats were trained to discriminate between a rewarded (A+) and a nonrewarded (B-) visual stimulus to receive sucrose rewards in touchscreen operant chambers. Serotonin was depleted using surgical infusions of 5,7-dihydroxytryptamine (5,7-DHT), either globally by intracebroventricular (i.c.v.) infusions or locally by microinfusions into the OFC or mPFC. Rats that received i.c.v. infusions of 5,7-DHT before initial training were significantly impaired during both visual discrimination and subsequent reversal learning during which the stimulus-reward contingencies were changed (A- vs. B+). Local serotonin depletion from the OFC impaired reversal learning without affecting initial discrimination. After mPFC depletion, rats were unimpaired during reversal learning but slower to respond at the stimuli during all the stages; the mPFC group was also slower to learn during discrimination than the OFC group. These findings extend our understanding of serotonin in cognitive flexibility by revealing differential effects within two subregions of the prefrontal cortex in visual discrimination and reversal learning.


Asunto(s)
Aprendizaje Discriminativo/fisiología , Corteza Prefrontal/metabolismo , Aprendizaje Inverso/fisiología , Neuronas Serotoninérgicas/metabolismo , Serotonina/metabolismo , Percepción Visual/fisiología , 5,6-Dihidroxitriptamina/administración & dosificación , 5,6-Dihidroxitriptamina/análogos & derivados , 5,6-Dihidroxitriptamina/toxicidad , Animales , Creatinina/administración & dosificación , Creatinina/análogos & derivados , Creatinina/toxicidad , Aprendizaje Discriminativo/efectos de los fármacos , Infusiones Intraventriculares , Masculino , Estimulación Luminosa/métodos , Corteza Prefrontal/efectos de los fármacos , Ratas , Aprendizaje Inverso/efectos de los fármacos , Neuronas Serotoninérgicas/efectos de los fármacos , Percepción Visual/efectos de los fármacos
3.
J Neurovirol ; 27(3): 403-421, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34003469

RESUMEN

HIV-1 infection affects approximately 37 million individuals, and approximately 50% of seropositive individuals will develop symptoms of clinical depression and/or apathy. Dysfunctions of both serotonergic and dopaminergic neurotransmission have been implicated in the pathogenesis of motivational alterations. The present study evaluated the efficacy of a SSRI (escitalopram) in the HIV-1 transgenic (Tg) rat. Behavioral, neurochemical, and neuroanatomical outcomes with respect to HIV-1 and sex were evaluated to determine the efficacy of chronic escitalopram treatment. Escitalopram treatment restored function in each of the behavioral tasks that were sensitive to HIV-1-induced impairments. Further, escitalopram treatment restored HIV-1-mediated synaptodendritic damage in the nucleus accumbens; treatment with escitalopram significantly increased dendritic proliferation in HIV-1 Tg rats. However, restoration did not consistently occur with the neurochemical analysis in the HIV-1 rat. Taken together, these results suggest a role for SSRI therapies in repairing long-term HIV-1 protein-mediated neuronal damage and restoring function.


Asunto(s)
Antidepresivos/farmacología , Apatía/efectos de los fármacos , Depresión/tratamiento farmacológico , Escitalopram/farmacología , Infecciones por VIH/tratamiento farmacológico , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Animales , Conducta de Elección/efectos de los fármacos , Dendritas/efectos de los fármacos , Dendritas/patología , Dendritas/virología , Depresión/complicaciones , Depresión/fisiopatología , Depresión/virología , Neuronas Dopaminérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/patología , Neuronas Dopaminérgicas/virología , Femenino , Infecciones por VIH/complicaciones , Infecciones por VIH/fisiopatología , Infecciones por VIH/virología , VIH-1/crecimiento & desarrollo , VIH-1/patogenicidad , Humanos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/patología , Núcleo Accumbens/virología , Ratas , Ratas Transgénicas , Neuronas Serotoninérgicas/efectos de los fármacos , Neuronas Serotoninérgicas/patología , Neuronas Serotoninérgicas/virología , Sinapsis/efectos de los fármacos , Sinapsis/patología , Sinapsis/virología , Transmisión Sináptica/efectos de los fármacos , Resultado del Tratamiento
4.
Int J Neuropsychopharmacol ; 24(7): 570-579, 2021 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-33674836

RESUMEN

BACKGROUND: Clinical studies have shown that the rapid antidepressant effect of the glutamate N-methyl-D-aspartate receptor antagonist ketamine generally disappears within 1 week but can be maintained by repeated administration. Preclinical studies showed that a single ketamine injection immediately increases the firing and burst activity of norepinephrine (NE) neurons, but not that of serotonin (5-HT) neurons. It also enhances the population activity of dopamine (DA) neurons. In the present study, we investigated whether such alterations of monoamine neuronal firing are still present 1 day after a single injection, and whether they can be maintained by repeated injections. METHODS: Rats received a single ketamine injection or 6 over 2 weeks and the firing activity of dorsal raphe nucleus 5-HT, locus coeruleus NE, and ventral tegmental area DA neurons was assessed. RESULTS: One day following a single injection of ketamine, there was no change in the firing activity of 5-HT, NE, or DA neurons. One day after repeated ketamine administration, however, there was a robust increase of the firing activity of NE neurons and an enhancement of burst and population activities of DA neurons, but still no change in firing parameters of 5-HT neurons. The increased activity of NE neurons was no longer present 3 days after the last injection, whereas that of DA neurons was still present. DA neurons were firing normally 7 days after repeated injections. CONCLUSION: These results imply that the enhanced activity of NE and DA neurons may play a significant role in the maintenance of the antidepressant action of ketamine.


Asunto(s)
Neuronas Adrenérgicas/efectos de los fármacos , Neuronas Dopaminérgicas/efectos de los fármacos , Antagonistas de Aminoácidos Excitadores/farmacología , Ketamina/farmacología , Tegmento Mesencefálico/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Animales , Antagonistas de Aminoácidos Excitadores/administración & dosificación , Ketamina/administración & dosificación , Locus Coeruleus/efectos de los fármacos , Masculino , Núcleos del Rafe/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Neuronas Serotoninérgicas/efectos de los fármacos , Área Tegmental Ventral/efectos de los fármacos
5.
Cereb Cortex ; 30(3): 1559-1572, 2020 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-31504265

RESUMEN

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.


Asunto(s)
Antidepresivos/farmacología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/efectos de los fármacos , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Estrés Fisiológico/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Animales , Ketamina/farmacología , Masculino , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Ratas Endogámicas F344 , Escopolamina/farmacología , Neuronas Serotoninérgicas/efectos de los fármacos , Neuronas Serotoninérgicas/metabolismo , Serotonina/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Estrés Fisiológico/fisiología
6.
Int J Mol Sci ; 22(16)2021 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-34445721

RESUMEN

Serotonin modulates several physiological and cognitive pathways throughout the human body that affect emotions, memory, sleep, and thermal regulation. The complex nature of the serotonergic system and interactions with other neurochemical systems indicate that the development of depression may be mediated by various pathomechanisms, the common denominator of which is undoubtedly the disturbed transmission in central 5-HT synapses. Therefore, the deliberate pharmacological modulation of serotonergic transmission in the brain seems to be one of the most appropriate strategies for the search for new antidepressants. As discussed in this review, the serotonergic system offers great potential for the development of new antidepressant therapies based on the combination of SERT inhibition with different pharmacological activity towards the 5-HT system. The aim of this article is to summarize the search for new antidepressants in recent years, focusing primarily on the possibility of benefiting from interactions with various 5-HT receptors in the pharmacotherapy of depression.


Asunto(s)
Antidepresivos/uso terapéutico , Depresión/tratamiento farmacológico , Receptores de Serotonina/metabolismo , Encéfalo/metabolismo , Desarrollo de Medicamentos/tendencias , Humanos , Receptores de Serotonina/efectos de los fármacos , Neuronas Serotoninérgicas/efectos de los fármacos , Neuronas Serotoninérgicas/metabolismo , Serotonina/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo
7.
Molecules ; 26(12)2021 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-34208700

RESUMEN

Cardamonin, a naturally occurring chalcone isolated from Alpinia species has shown to possess strong anti-inflammatory and anti-nociceptive activities. Previous studies have demonstrated that cardamonin exerts antihyperalgesic and antiallodynic properties in chronic constriction injury (CCI)-induced neuropathic pain animal model. However, the mechanisms underlying cardamonin's effect have yet to be fully understood. The present study aims to investigate the involvement of the serotonergic system in cardamonin induced antihyperalgesic and antiallodynic effects in CCI-induced neuropathic pain mice model. The neuropathic pain symptoms in the CCI mice model were assessed using Hargreaves Plantar test and von-Frey filament test on day 14 post-surgery. Central depletion of serotonin along the descending serotonergic pathway was done using ρ-chlorophenylalanine (PCPA, 100 mg/kg, i.p.), an inhibitor of serotonin synthesis for four consecutive days before cardamonin treatment, and was found to reverse the antihyperalgesic and antiallodynic effect produced by cardamonin. Pretreatment of the mice with several 5-HT receptor subtypes antagonists: methiothepin (5-HT1/6/77 receptor antagonist, 0.1 mg/kg), WAY 100635 (5-HT1A receptor antagonist, 1 mg/kg), isamoltane (5-HT1B receptor antagonist, 2.5 mg/kg), ketanserin (5-HT2A receptor antagonist, 0.3 mg/kg), and ondansetron (5-HT3 receptor antagonist, 0.5 mg/kg) were shown to abolish the effect of cardamonin induced antihyperalgesic and antiallodynic effects. Further evaluation of the 5-HT1A receptor subtype protein expressions reveals that cardamonin significantly upregulated its expression in the brainstem and spinal cord. Our results suggest that the serotonergic pathway is essential for cardamonin to exert its antineuropathic effect in CCI mice through the involvement of the 5-HT1A receptor subtype in the central nervous system.


Asunto(s)
Chalconas/farmacología , Neuralgia/tratamiento farmacológico , Receptor de Serotonina 5-HT1A/efectos de los fármacos , Analgésicos/farmacología , Animales , Chalconas/metabolismo , Modelos Animales de Enfermedad , Hiperalgesia/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos ICR , Receptor de Serotonina 5-HT1A/metabolismo , Neuronas Serotoninérgicas/efectos de los fármacos , Neuronas Serotoninérgicas/metabolismo , Serotonina/metabolismo , Médula Espinal/efectos de los fármacos
8.
J Neurosci ; 39(8): 1334-1346, 2019 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-30552180

RESUMEN

Selective serotonin (5-HT) reuptake inhibitors (SSRIs) are first-line antidepressants but require several weeks to elicit their actions. Chronic SSRI treatment induces desensitization of 5-HT1A autoreceptors to enhance 5-HT neurotransmission. Mice (both sexes) with gene deletion of 5-HT1A autoreceptors in adult 5-HT neurons (1AcKO) were tested for response to SSRIs. Tamoxifen-induced recombination in adult 1AcKO mice specifically reduced 5-HT1A autoreceptor levels. The 1AcKO mice showed a loss of 5-HT1A autoreceptor-mediated hypothermia and electrophysiological responses, but no changes in anxiety- or depression-like behavior. Subchronic fluoxetine (FLX) treatment induced an unexpected anxiogenic effect in 1AcKO mice in the novelty suppressed feeding and elevated plus maze tests, as did escitalopram in the novelty suppressed feeding test. No effect was seen in wild-type (WT) mice. Subchronic FLX increased 5-HT metabolism in prefrontal cortex, hippocampus, and raphe of 1AcKO but not WT mice, suggesting hyperactivation of 5-HT release. To detect chronic cellular activation, FosB+ cells were quantified. FosB+ cells were reduced in entorhinal cortex and hippocampus (CA2/3) and increased in dorsal raphe 5-HT cells of 1AcKO mice, suggesting increased raphe activation. In WT but not 1AcKO mice, FLX reduced FosB+ cells in the median raphe, hippocampus, entorhinal cortex, and median septum, which receive rich 5-HT projections. Thus, in the absence of 5-HT1A autoreceptors, SSRIs induce a paradoxical anxiogenic response. This may involve imbalance in activation of dorsal and median raphe to regulate septohippocampal or fimbria-fornix pathways. These results suggest that markedly reduced 5-HT1A autoreceptors may provide a marker for aberrant response to SSRI treatment.SIGNIFICANCE STATEMENT Serotonin-selective reuptake inhibitors (SSRIs) are effective in treating anxiety and depression in humans and mouse models. However, in some cases, SSRIs can increase anxiety, but the mechanisms involved are unclear. Here we show that, rather than enhancing SSRI benefits, adulthood knockout (KO) of the 5-HT1A autoreceptor, a critical negative regulator of 5-HT activity, results in an SSRI-induced anxiety effect that appears to involve a hyperactivation of the 5-HT system in certain brain areas. Thus, subjects with very low levels of 5-HT1A autoreceptors, such as during childhood or adolescence, may be at risk for an SSRI-induced anxiety response.


Asunto(s)
Antidepresivos/efectos adversos , Ansiedad/inducido químicamente , Autorreceptores/efectos de los fármacos , Receptor de Serotonina 5-HT1A/deficiencia , Inhibidores Selectivos de la Recaptación de Serotonina/efectos adversos , Neuronas Serotoninérgicas/efectos de los fármacos , 8-Hidroxi-2-(di-n-propilamino)tetralin/toxicidad , Animales , Antidepresivos/farmacología , Química Encefálica/efectos de los fármacos , Conducta Exploratoria/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Femenino , Fluoxetina/efectos adversos , Fluoxetina/farmacología , Hipotermia/inducido químicamente , Hipotermia/fisiopatología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Red Nerviosa/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/análisis , Receptor de Serotonina 5-HT1A/efectos de los fármacos , Receptor de Serotonina 5-HT1A/fisiología , Neuronas Serotoninérgicas/fisiología , Serotonina/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Natación
9.
J Cell Physiol ; 235(2): 836-847, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31332785

RESUMEN

How are memories stored and retrieved? It was one of the most discussed questions in the past century by neuroscientists. Leading studies of the period brought two different explanations to this question: The first statement considers memory as a physiological change in the brain and suggest that the retrieval of memory is only occurred by the same physiologic changes observed during the memory formation, while the second suggests that memory is a psychic mood stored in mind and the retrieval of memory is occurred by mystical energy fluctuations. Although the exact reason and the pathogenesis of Alzheimer's disease have not yet been fully understood, the approaches that centered the retrieval strategy of lost memory constitutes the basis of the treatment strategies in Alzheimer's disease today. The majority of treatment studies has based on the manipulation of the cholinergic system; however, although serotonin has mnemonic effects, its role in the pathogenesis of Alzheimer's disease has not been investigated as much as the cholinergic system. Here we show how serotonin affects the pathogenesis of Alzheimer's disease in a comprehensive perspective and we suggest that the optogenetics manipulation of serotonin nuclei retrieve the lost memory by closing the inward-rectifier potassium channel Kir2 on the memory engram cells. Also, we raise the possible effects of serotonin on the memory engram cells and the interactions between the amyloid-centric hypothesis of Alzheimer's disease and the memory engram hypothesis to explain the pathophysiology of memory loss in Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Demencia/fisiopatología , Trastornos de la Memoria/fisiopatología , Optogenética/métodos , Serotonina/metabolismo , Enfermedad de Alzheimer/genética , Encéfalo/patología , Humanos , Memoria/fisiología , Neuronas Serotoninérgicas/efectos de los fármacos
10.
J Psychiatry Neurosci ; 45(5): 344-355, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32459080

RESUMEN

Background: Altered function of serotonin receptor 1A (5-HT1AR) has been consistently implicated in anxiety, major depressive disorder and resistance to antidepressants. Mechanisms by which the function of 5-HT1AR (expressed as an autoreceptor in serotonergic raphe neurons and as a heteroreceptor in serotonin [5-HT] projection areas) is altered include regulation of its expression, but 5-HT1AR trafficking may also be involved. Methods: We investigated the consequences of the lack of Yif1B (the 5-HT1AR trafficking protein) on 5-HT neurotransmission in mice, and whether Yif1B expression might be affected under conditions known to alter 5-HT neurotransmission, such as anxious or depressive states or following treatment with fluoxetine (a selective serotonin reuptake inhibitor) in humans, monkeys and mice. Results: Compared with wild-type mice, Yif1B-knockout mice showed a significant decrease in the forebrain density of 5-HT projection fibres and a hypofunctionality of 5-HT1A autoreceptors expressed on raphe 5-HT neurons. In addition, social interaction was less in Yif1B-knockout mice, which did not respond to the antidepressant-like effect of acute fluoxetine injection. In wild-type mice, social defeat was associated with downregulated Yif1B mRNA in the prefrontal cortex, and chronic fluoxetine treatment increased Yif1B expression. The expression of Yif1B was also downregulated in the postmortem prefrontal cortex of people with major depressive disorder and upregulated after chronic treatment with a selective serotonin reuptake inhibitor in monkeys. Limitations: We found sex differences in Yif1B expression in humans and monkeys, but not in mice under the tested conditions. Conclusion: These data support the concept that Yif1B plays a critical role in 5-HT1AR functioning and brain 5-HT homeostasis. The opposite changes in its expression observed in anxious or depressive states and after therapeutic fluoxetine treatment suggest that Yif1B might be involved in vulnerability to anxiety and depression, and fluoxetine efficacy.


Asunto(s)
Trastorno Depresivo Mayor/metabolismo , Corteza Prefrontal/efectos de los fármacos , Corteza Prefrontal/metabolismo , Receptor de Serotonina 5-HT1A/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Serotonina/metabolismo , Conducta Social , Proteínas de Transporte Vesicular/efectos de los fármacos , Proteínas de Transporte Vesicular/metabolismo , Animales , Autopsia , Conducta Animal/fisiología , Modelos Animales de Enfermedad , Femenino , Fluoxetina/farmacología , Humanos , Macaca mulatta , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Núcleos del Rafe/efectos de los fármacos , Núcleos del Rafe/fisiología , Neuronas Serotoninérgicas/efectos de los fármacos , Neuronas Serotoninérgicas/fisiología , Agonistas del Receptor de Serotonina 5-HT1/farmacología , Caracteres Sexuales
11.
Biosci Biotechnol Biochem ; 84(1): 159-170, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31483212

RESUMEN

We tested whether Sake Lees (SL) had inhibitory effects on hyperalgesia in the hindpaw under psychophysical stress conditions. Male rats were subjected to repeated forced swim stress treatments (FST) from Day -3 to Day -1. Intraperiotoneal administration of SL which contained low concentration of ethanol (SLX) was conducted after each FST. On Day 0, formalin-evoked licking behaviors and Fos responses in the lumbar spinal cord (DH) and several areas within the rostral ventromedial medulla (RVM) were quantified as nociceptive responses. FST-induced hyperalgesia in the hindpaw was prevented by repeated SL and SLX treatments. Fos expression was significantly increased in DH and some areas within the RVM under FST, which was prevented by repeated SL or SLX. These findings indicated that daily administration of SL had the potential to alleviate stress-induced hyperalgesia.


Asunto(s)
Fermentación , Miembro Posterior/metabolismo , Hiperalgesia/tratamiento farmacológico , Oryza/química , Extractos Vegetales/farmacología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Asta Dorsal de la Médula Espinal/metabolismo , Estrés Fisiológico/fisiología , Animales , Conducta Animal/efectos de los fármacos , Etanol/química , Formaldehído/administración & dosificación , Formaldehído/farmacología , Hiperalgesia/etiología , Inmunohistoquímica , Masculino , Manejo del Dolor , Dimensión del Dolor , Extractos Vegetales/administración & dosificación , Proteínas Proto-Oncogénicas c-fos/inmunología , Ratas , Ratas Sprague-Dawley , Neuronas Serotoninérgicas/efectos de los fármacos , Neuronas Serotoninérgicas/metabolismo , Serotonina/inmunología , Serotonina/metabolismo , Natación/fisiología , Distribución Tisular
12.
Addict Biol ; 25(4): e12779, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31169340

RESUMEN

Abuse of alcohol is a major clinical problem with far-reaching health consequences. Understanding the environmental and genetic factors that contribute to alcohol-related behaviors is a potential gateway for developing novel therapeutic approaches for patients that abuse the drug. To this end, we have used Drosophila melanogaster as a model to investigate the effect of diet, an environmental factor, on ethanol sedation. Providing flies with diets high in yeast, a routinely used component of fly media, increased their resistance to ethanol sedation. The yeast-induced resistance to ethanol sedation occurred in several different genetic backgrounds, was observed in males and females, was elicited by yeast from different sources, was readily reversible, and was associated with increased nutrient intake as well as decreased internal ethanol levels. Inhibition of serotonergic neuron function using multiple independent genetic manipulations blocked the effect of yeast supplementation on ethanol sedation, nutrient intake, and internal ethanol levels. Our results demonstrate that yeast is a critical dietary component that influences ethanol sedation in flies and that serotonergic signaling is required for the effect of dietary yeast on nutrient intake, ethanol uptake/elimination, and ethanol sedation. Our studies establish the fly as a model for diet-induced changes in ethanol sedation and raise the possibility that serotonin might mediate the effect of diet on alcohol-related behavior in other species.


Asunto(s)
Conducta Animal/efectos de los fármacos , Depresores del Sistema Nervioso Central/farmacología , Dieta , Etanol/farmacología , Neuronas Serotoninérgicas/efectos de los fármacos , Levadura Seca , Animales , Drosophila melanogaster , Femenino , Hipnóticos y Sedantes/farmacología , Masculino , Saccharomyces cerevisiae , Neuronas Serotoninérgicas/metabolismo , Serotonina/metabolismo
13.
Int J Mol Sci ; 21(18)2020 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-32906643

RESUMEN

Depression impacts the lives and daily activities of millions globally. Research into the neurobiology of lateral habenula circuitry and the use of psychedelics for treating depressive states has emerged in the last decade as new directions to devise interventional strategies and therapies. Several clinical trials using deep brain stimulation of the habenula, or using ketamine, and psychedelics that target the serotonergic system such as psilocybin are also underway. The promising early results in these fields require cautious optimism as further evidence from experiments conducted in animal systems in ecologically relevant settings, and a larger number of human studies with improved spatiotemporal neuroimaging, accumulates. Designing optimal methods of intervention will also be aided by an improvement in our understanding of the common genetic and molecular factors underlying disorders comorbid with depression, as well as the characterization of psychedelic-induced changes at a molecular level. Advances in the use of cerebral organoids offers a new approach for rapid progress towards these goals. Here, we review developments in these fast-moving areas of research and discuss potential future directions.


Asunto(s)
Depresión/tratamiento farmacológico , Habénula/efectos de los fármacos , Alucinógenos/farmacología , Encefalopatías/tratamiento farmacológico , Encefalopatías/fisiopatología , Comorbilidad , Depresión/metabolismo , Depresión/fisiopatología , Trastorno Depresivo/metabolismo , Trastorno Depresivo/fisiopatología , Habénula/metabolismo , Alucinógenos/metabolismo , Humanos , Psilocibina/metabolismo , Psilocibina/farmacología , Neuronas Serotoninérgicas/efectos de los fármacos
14.
Int J Mol Sci ; 21(19)2020 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-33003279

RESUMEN

Kv3.1 channel is abundantly expressed in neurons and its dysfunction causes sleep loss, neurodegenerative diseases and depression. Fluoxetine, a serotonin selective reuptake inhibitor commonly used to treat depression, acts also on Kv3.1. To define the relationship between Kv3.1 and serotonin receptors (SR) pharmacological modulation, we showed that 1C11, a serotonergic cell line, expresses different voltage gated potassium (VGK) channels subtypes in the presence (differentiated cells (1C11D)) or absence (not differentiated cells (1C11ND)) of induction. Only Kv1.2 and Kv3.1 transcripts increase even if the level of Kv3.1b transcripts is highest in 1C11D and, after fluoxetine, in 1C11ND but decreases in 1C11D. The Kv3.1 channel protein is expressed in 1C11ND and 1C11D but is enhanced by fluoxetine only in 1C11D. Whole cell measurements confirm that 1C11 cells express (VGK) currents, increasing sequentially as a function of cell development. Moreover, SR 5HT1b is highly expressed in 1C11D but fluoxetine increases the level of transcript in 1C11ND and significantly decreases it in 1C11D. Serotonin dosage shows that fluoxetine at 10 nM blocks serotonin reuptake in 1C11ND but slows down its release when cells are differentiated through a decrease of 5HT1b receptors density. We provide the first experimental evidence that 1C11 expresses Kv3.1b, which confirms its major role during differentiation. Cells respond to the fluoxetine effect by upregulating Kv3.1b expression. On the other hand, the possible relationship between the fluoxetine effect on the kinetics of 5HT1b differentiation and Kv3.1bexpression, would suggest the Kv3.1b channel as a target of an antidepressant drug as well as it was suggested for 5HT1b.


Asunto(s)
Fluoxetina/farmacología , Neuronas Serotoninérgicas/efectos de los fármacos , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Canales de Potasio Shaw/genética , Animales , Células CHO , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Cricetulus , Depresión/tratamiento farmacológico , Depresión/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Canal de Potasio Kv.1.2/genética , Neuronas Serotoninérgicas/metabolismo , Serotonina/genética , Serotonina/metabolismo , Antagonistas de la Serotonina/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología
15.
Int J Mol Sci ; 21(8)2020 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-32326436

RESUMEN

Calcium homeostasis is a cellular process required for proper cell function and survival, maintained by the coordinated action of several transporters, among them members of the Na+/Ca2+-exchanger family, such as SLC8A3. Transforming growth factor beta (TGF-ß) signaling defines neuronal development and survival and may regulate the expression of channels and transporters. We investigated the regulation of SLC8A3 by TGF-ß in a conditional knockout mouse with deletion of TGF-ß signaling from Engrailed 1-expressing cells, i.e., in cells from the midbrain and rhombomere 1, and elucidated the underlying molecular mechanisms. The results show that SLC8A3 is significantly downregulated in developing dopaminergic and dorsal raphe serotonergic neurons in mutants and that low SLC8A3 abundance prevents the expression of the anti-apoptotic protein Bcl-xL. TGF-ß signaling affects SLC8A3 via the canonical and p38 signaling pathway and may increase the binding of Smad4 to the Slc8a3 promoter. Expression of the lipid peroxidation marker malondialdehyde (MDA) was increased following knockdown of Slc8a3 expression in vitro. In neurons lacking TGF-ß signaling, the number of MDA- and 4-hydroxynonenal (4-HNE)-positive cells was significantly increased, accompanied with increased cellular 4-HNE abundance. These results suggest that TGF-ß contributes to the regulation of SLC8A3 expression in developing dopaminergic and dorsal raphe serotonergic neurons, thereby preventing oxidative stress.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Mesencéfalo/metabolismo , Neurogénesis/genética , Estrés Oxidativo/genética , Neuronas Serotoninérgicas/metabolismo , Intercambiador de Sodio-Calcio/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Aldehídos/metabolismo , Animales , Apoptosis/genética , Calcio/metabolismo , Línea Celular , Células Cultivadas , Inmunoprecipitación de Cromatina , Neuronas Dopaminérgicas/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Homeostasis , Humanos , Inmunohistoquímica , Malondialdehído/metabolismo , Mesencéfalo/efectos de los fármacos , Mesencéfalo/crecimiento & desarrollo , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas , Unión Proteica , Neuronas Serotoninérgicas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteína Smad4/metabolismo , Intercambiador de Sodio-Calcio/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/farmacología , Proteína bcl-X/metabolismo
16.
J Neurosci ; 38(8): 1915-1925, 2018 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-29378860

RESUMEN

Arousal from sleep in response to CO2 is a critical protective phenomenon. Dysregulation of CO2-induced arousal contributes to morbidity and mortality from prevalent diseases, such as obstructive sleep apnea and sudden infant death syndrome. Despite the critical nature of this protective reflex, the precise mechanism for CO2-induced arousal is unknown. Because CO2 is a major regulator of breathing, prevailing theories suggest that activation of respiratory chemo- and mechano-sensors is required for CO2-induced arousal. However, populations of neurons that are not involved in the regulation of breathing are also chemosensitive. Among these are serotonin (5-HT) neurons in the dorsal raphe nucleus (DRN) that comprise a component of the ascending arousal system. We hypothesized that direct stimulation of these neurons with CO2 could cause arousal from sleep independently of enhancing breathing. Dialysis of CO2-rich acidified solution into DRN, but not medullary raphe responsible for modulating breathing, caused arousal from sleep. Arousal was lost in mice with a genetic absence of 5-HT neurons, and with acute pharmacological or optogenetic inactivation of DRN 5-HT neurons. Here we demonstrate that CO2 can cause arousal from sleep directly, without requiring enhancement of breathing, and that chemosensitive 5-HT neurons in the DRN critically mediate this arousal. Better understanding mechanisms underlying this protective reflex may lead to interventions to reduce disease-associated morbidity and mortality.SIGNIFICANCE STATEMENT Although CO2-induced arousal is critical to a number of diseases, the specific mechanism is not well understood. We previously demonstrated that serotonin (5-HT) neurons are important for CO2-induced arousal, as mice without 5-HT neurons do not arouse to CO2 Many have interpreted this to mean that medullary 5-HT neurons that regulate breathing are important in this arousal mechanism. Here we found that direct application of CO2-rich aCSF to the dorsal raphe nucleus, but not the medullary raphe, causes arousal from sleep, and that this arousal was lost with genetic ablation or acute inhibition of 5-HT neurons. We propose that 5-HT neurons in the dorsal raphe nucleus can be activated directly by CO2 to cause arousal independently of respiratory activation.


Asunto(s)
Nivel de Alerta/efectos de los fármacos , Nivel de Alerta/fisiología , Dióxido de Carbono/farmacología , Núcleo Dorsal del Rafe/efectos de los fármacos , Neuronas Serotoninérgicas/efectos de los fármacos , Animales , Núcleo Dorsal del Rafe/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Reflejo/efectos de los fármacos , Reflejo/fisiología , Neuronas Serotoninérgicas/fisiología , Sueño/efectos de los fármacos , Sueño/fisiología
17.
Physiol Genomics ; 51(2): 43-50, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30576260

RESUMEN

Valproic acid (VPA) and lithium are widely used in the treatment of bipolar disorder. However, the underlying mechanism of action of these drugs is not clearly understood. We used RNA-Seq analysis to examine the global profile of gene expression in a rat serotonergic cell line (RN46A) after exposure to these two mood stabilizer drugs. Numerous genes were differentially regulated in response to VPA (log2 fold change ≥ 1.0; i.e., odds ratio of ≥2, at false discovery rate <5%), but only two genes ( Dynlrb2 and Cdyl2) showed significant differential regulation after exposure of the cells to lithium, with the same analysis criteria. Both of these genes were also regulated by VPA. Many of the differentially expressed genes had functions of potential relevance to mood disorders or their treatment, such as several serpin family genes (including neuroserpin), Nts (neurotensin), Maob (monoamine oxidase B), and Ap2b1, which is important for synaptic vesicle function. Pathway analysis revealed significant enrichment of Gene Ontology terms such as extracellular matrix remodeling, cell adhesion, and chemotaxis. This study in a cell line derived from the raphe nucleus has identified a range of genes and pathways that provide novel insights into potential therapeutic actions of the commonly used mood stabilizer drugs.


Asunto(s)
Litio/farmacología , Neuronas Serotoninérgicas/efectos de los fármacos , Transcriptoma/efectos de los fármacos , Ácido Valproico/farmacología , Animales , Trastorno Bipolar/tratamiento farmacológico , Adhesión Celular/efectos de los fármacos , Adhesión Celular/genética , Línea Celular , Quimiotaxis/efectos de los fármacos , Quimiotaxis/genética , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/genética , Ontología de Genes , Litio/uso terapéutico , RNA-Seq , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Neuronas Serotoninérgicas/metabolismo , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética , Ácido Valproico/uso terapéutico
18.
Eur J Neurosci ; 50(1): 1712-1726, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30687960

RESUMEN

Glucocorticoid hormones and serotonin (5-HT) are strongly associated with the development and treatment of depression, respectively. Glucocorticoids regulate the function of serotonergic neurons in the dorsal raphe nucleus (DR), which are the major source of 5-HT to the forebrain. DR 5-HT neurons are electrophysiologically heterogeneous, though whether this phenotypic variation aligns with specific brain functions or neuropsychiatric disease states is largely unknown. The goal of this work was to determine if chronic exogenous glucocorticoid administration differentially affects the electrophysiological profile of DR neurons implicated in the regulation of emotion versus visual sensation by comparing properties of cells projecting to medial prefrontal cortex (mPFC) versus lateral geniculate nucleus (LGN). Following retrograde tracer injection into mPFC or LGN, male Sprague-Dawley rats received daily injections of corticosterone (CORT) for 21 days, after which whole-cell patch clamp recordings were made from retrogradely labeled DR neurons. CORT-treatment significantly increased the action potential half-width of LGN-projecting DR neurons, but did not significantly affect the firing frequency or excitatory postsynaptic currents of these cells. CORT-treatment significantly reduced the input resistance, evoked firing frequency, and spontaneous excitatory postsynaptic current frequency of mPFC-projecting DR neurons, indicating a concurrent reduction of both intrinsic excitability and excitatory drive. Our results suggest that the serotonergic regulation of cognitive and emotional networks in the mPFC may be more sensitive to the effects of glucocorticoid excess than visual sensory circuits in the LGN and that reduced 5-HT transmission in the mPFC may underlie the association between glucocorticoid excess and depression.


Asunto(s)
Corticosterona/farmacología , Núcleo Dorsal del Rafe/metabolismo , Potenciales Postsinápticos Excitadores/fisiología , Cuerpos Geniculados/metabolismo , Glucocorticoides/metabolismo , Red Nerviosa/metabolismo , Corteza Prefrontal/metabolismo , Neuronas Serotoninérgicas/metabolismo , Serotonina/metabolismo , Vías Visuales/metabolismo , Animales , Corticosterona/administración & dosificación , Depresión/metabolismo , Núcleo Dorsal del Rafe/efectos de los fármacos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Cuerpos Geniculados/efectos de los fármacos , Masculino , Red Nerviosa/efectos de los fármacos , Técnicas de Trazados de Vías Neuroanatómicas , Técnicas de Placa-Clamp , Corteza Prefrontal/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Neuronas Serotoninérgicas/efectos de los fármacos , Vías Visuales/efectos de los fármacos
19.
Toxicol Appl Pharmacol ; 378: 114604, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31153898

RESUMEN

This study investigated the protective effects of dextromethorphan (DXM) on noise-induced hearing loss (NIHL) in rats. This study aimed to improve the auditory threshold and to understand the protective effects of DXM against N-methyl-d-aspartate (NMDA)-induced neurite degeneration of serotonergic neurons. The animals were exposed to 8-kHz narrowband noise at a 118-dB sound pressure level for 3.5 h. The hearing thresholds were determined by measuring the auditory brainstem response to click stimuli. Serotonin transporter (SERT) expression was determined through micro-positron emission tomography (PET) using N,N-dimethyl-2-(2-amino-4-18F-fluorophenylthio)benzylamine (4-[18F]-ADAM). We also investigated the effects of DXM on NMDA-induced morphological changes in the primary cultures of rat serotonergic neurons. NIHL significantly improved after prophylactic treatment with DXM (p < .05). SERT density in DXM-treated rats was significantly higher than that in non-DXM-treated rats. Because prophylactic medication restored the NMDA-inhibited neurite length of serotonergic neurons and presented SERT density, DXM could be a potential agent in alleviating NIHL.


Asunto(s)
Bencilaminas/farmacología , Encéfalo/efectos de los fármacos , Dextrometorfano/farmacología , Pérdida Auditiva/tratamiento farmacológico , Pérdida Auditiva/metabolismo , Proteínas de Unión al ARN/metabolismo , Radiofármacos/farmacología , Animales , Encéfalo/metabolismo , Tronco Encefálico/efectos de los fármacos , Tronco Encefálico/metabolismo , Modelos Animales de Enfermedad , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Masculino , N-Metilaspartato/farmacología , Tomografía de Emisión de Positrones/métodos , Ratas , Ratas Sprague-Dawley , Neuronas Serotoninérgicas/efectos de los fármacos , Neuronas Serotoninérgicas/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo
20.
Pharmacol Res ; 142: 283-293, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30826457

RESUMEN

The 3,4-methylenedioxymethamphetamine (MDMA) is a popular recreational drug, which ultimately leads to serotonergic (5-HT) neurotoxicity and psychiatric disorders. Previous in vitro studies have consistently demonstrated that MDMA provokes autophagic activation, as well as damage of 5-HT axons and nerve fibers. So far, whether autophagy, a well-conserved cellular process that is critical for cell fate, also participates in MDMA-induced neurotoxicity in vivo remains elusive. Here, we first examined time-course of autophagy-related changes during repeated administration of MDMA (10 mg/kg s.c. twice daily for 4 consecutive days) using immunofluorescent staining for tryptophan hydroxylase and microtubule-associated protein 1 light chain 3 beta in rats. We also evaluated the protective effects of 3-methyadanine (3-MA, an autophagy inhibitor, 15 mg/kg i.p.) against MDMA-induced acute and long-term reductions in serotonin transporters (SERT) density in various brain regions using immunohistochemical staining and positron emission tomography (PET) imaging respectively. Plasma corticosterone measurements and forced swim tests were performed to evaluate the depressive performance. The staining results showed that repeated administration of MDMA increased expression of autophagosome and caused reduction in SERT densities of striatum and frontal cortex, which was ameliorated in the presence of 3-MA. PET imaging data also revealed that 3-MA could ameliorate MDMA-induced long-term decreased SERT availability in various brain regions of rats. Furthermore, immobility time of forced swim tests and plasma corticosterone levels were less in the group of MDMA co-injected with 3-MA compared with that of MDMA group. Together, these findings suggest that autophagy inhibition may confer protection against neurobiological and behavioral changes induced by MDMA.


Asunto(s)
Autofagia , Encéfalo/metabolismo , Depresión/metabolismo , N-Metil-3,4-metilenodioxianfetamina , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Adenina/análogos & derivados , Adenina/farmacología , Animales , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Depresión/tratamiento farmacológico , Masculino , Fármacos Neuroprotectores/farmacología , Ratas Sprague-Dawley , Neuronas Serotoninérgicas/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA