Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 169
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
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
2.
J Neurosci ; 34(1): 282-94, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24381289

RESUMEN

Selective serotonin reuptake inhibitors (SSRI) are aimed at increasing brain 5-HT tone; however, this expected effect has a slow onset after starting SSRI treatment because of initial activation of 5-HT(1A) autoreceptor-mediated negative feedback of 5-HT release. After chronic SSRI treatment, 5-HT(1A) autoreceptors desensitize, which allows 5-HT tone elevation. Because 5-HT(1A) receptor (5-HT(1A)R) internalization has been proposed as a possible mechanism underlying 5-HT(1A) autoreceptor desensitization, we examined whether this receptor could internalize under well controlled in vitro conditions in the LLC-CPK1 cell line and in raphe or hippocampal neurons from rat embryos. To this goal, cells were transfected with recombinant lentiviral vectors encoding N-terminal tagged 5-HT(1A)R, and exposed to various pharmacological conditions. Constitutive endocytosis and plasma membrane recycling of tagged-5-HT(1A)R was observed in LLC-PK1 cells as well as in neurons. Acute exposure (for 1 h) to the full 5-HT(1A)R agonists, 5-HT and 5-carboxamido-tryptamine, but not the partial agonist 8-OH-DPAT, triggered internalization of tagged 5-HT(1A)R in serotonergic neurons only. In contrast, sustained exposure (for 24 h) to all agonists induced tagged-5-HT(1A)R endocytosis in raphe serotonergic neurons and a portion of hippocampal neurons, but not LLC-PK1 cells and partial agonist displayed an effect only in serotonergic neurons. In all cases, agonist-induced tagged 5-HT(1A)R endocytosis was prevented by the 5-HT(1A)R antagonist, WAY-100635, which was inactive on its own. These data showed that agonist-induced 5-HT(1A)R internalization does exist in neurons and depends on agonist efficacy and neuronal phenotype. Its differential occurrence in serotonergic neurons supports the idea that 5-HT(1A)R internalization might underlie 5-HT(1A) autoreceptor desensitization under SSRI antidepressant therapy.


Asunto(s)
Autorreceptores/agonistas , Autorreceptores/metabolismo , Neuronas/metabolismo , Fenotipo , Receptor de Serotonina 5-HT1A/metabolismo , Agonistas del Receptor de Serotonina 5-HT1/metabolismo , Animales , Endocitosis/efectos de los fármacos , Endocitosis/fisiología , Femenino , Células LLC-PK1 , Neuronas/efectos de los fármacos , Unión Proteica/fisiología , Ratas , Ratas Sprague-Dawley , Agonistas del Receptor de Serotonina 5-HT1/farmacología , Porcinos
3.
J Neurosci ; 34(44): 14739-51, 2014 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-25355226

RESUMEN

Painful experiences are multilayered, composed of sensory, affective, cognitive and behavioral facets. Whereas it is well accepted that the development of chronic pain is due to maladaptive neuronal changes, the underlying molecular mechanisms, their relationship to the different pain modalities, and indeed the localization of these changes are still unknown. Brain-derived neurotrophic factor (BDNF) is an activity-dependent neuromodulator in the adult brain, which enhances neuronal excitability. In the spinal cord, BDNF underlies the development and maintenance of inflammatory and neuropathic pain. Here, we hypothesized that BDNF could be a trigger of some of these plastic changes. Our results demonstrate that BDNF is upregulated in the anterior cingulate cortex (ACC) and the primary sensory cortex (S1) in rats with inflammatory pain. Injections of recombinant BDNF (into the ACC) or a viral vector synthesizing BDNF (into the ACC or S1) triggered both neuronal hyperexcitability, as shown by elevated long-term potentiation, and sustained pain hypersensitivity. Finally, pharmacological blockade of BDNF-tropomyosin receptor kinase B (TrkB) signaling in the ACC, through local injection of cyclotraxin-B (a novel, highly potent, and selective TrkB antagonist) prevented neuronal hyperexcitability, the emergence of cold hypersensitivity, and passive avoidance behavior. These findings show that BDNF-dependent neuronal plasticity in the ACC, a structure known to be involved in the affective-emotional aspect of pain, is a key mechanism in the development and maintenance of the emotional aspect of chronic pain.


Asunto(s)
Afecto/fisiología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Giro del Cíngulo/metabolismo , Hiperalgesia/metabolismo , Plasticidad Neuronal/fisiología , Corteza Somatosensorial/metabolismo , Afecto/efectos de los fármacos , Animales , Conducta Animal/efectos de los fármacos , Conducta Animal/fisiología , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/farmacología , Giro del Cíngulo/efectos de los fármacos , Giro del Cíngulo/fisiopatología , Hiperalgesia/inducido químicamente , Hiperalgesia/fisiopatología , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/fisiopatología , Masculino , Plasticidad Neuronal/efectos de los fármacos , Péptidos Cíclicos/farmacología , Ratas , Ratas Sprague-Dawley , Receptor trkB/antagonistas & inhibidores , Corteza Somatosensorial/efectos de los fármacos , Corteza Somatosensorial/fisiopatología , Regulación hacia Arriba/fisiología
4.
J Neurochem ; 131(5): 566-72, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25113583

RESUMEN

Serotonin (5-HT)2C receptors play a role in psychoaffective disorders and often contribute to the antidepressant and anxiolytic effects of psychotropic drugs. During stress, activation of these receptors exerts a negative feedback on 5-HT release, probably by increasing the activity of GABAergic interneurons. However, to date, the GABA receptor types that mediate the 5-HT2C receptor-induced feedback inhibition are still unknown. To address this question, we assessed the inhibition of 5-HT turnover by a 5-HT2C receptor agonist (RO 60-0175) at the hippocampal level and under conditions of stress, after pharmacological or genetic inactivation of either GABA-A or GABA-B receptors in mice. Neither the GABA-B receptor antagonist phaclofen nor the specific genetic ablation of either GABA-B1a or GABA-B1b subunits altered the inhibitory effect of RO 60-0175, although 5-HT turnover was markedly decreased in GABA-B1a knock-out mice in both basal and stress conditions. In contrast, the 5-HT2C receptor-mediated inhibition of 5-HT turnover was reduced by the GABA-A receptor antagonist bicuculline. However, a significant effect of 5-HT2C receptor activation persisted in mutant mice deficient in the α3 subunit of GABA-A receptors. It can be inferred that non-α3 subunit-containing GABA-A receptors, but not GABA-B receptors, mediate the 5-HT2C -induced inhibition of stress-induced increase in hippocampal 5-HT turnover in mice.


Asunto(s)
GABAérgicos/farmacología , Receptor de Serotonina 5-HT2C/metabolismo , Receptores de GABA/genética , Estrés Psicológico/genética , Estrés Psicológico/metabolismo , Animales , Modelos Animales de Enfermedad , Etilaminas/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Ácido Hidroxiindolacético/metabolismo , Indoles/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de GABA/deficiencia , Serotonina/metabolismo , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/patología
5.
Int J Neuropsychopharmacol ; 18(3)2014 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-25522398

RESUMEN

BACKGROUND: Desensitization and blockade of 5-HT2C receptors (5-HT2CR) have long been thought to be central in the therapeutic action of antidepressant drugs. However, besides behavioral pharmacology studies, there is little in vivo data documenting antidepressant-induced 5-HT2CR desensitization in specific brain areas. METHODS: Mice lacking the 5-HT reuptake carrier (5-HTT(-/-)) were used to model the consequences of chronic 5-HT reuptake inhibition with antidepressant drugs. The effect of this mutation on 5-HT2CR was evaluated at the behavioral (social interaction, novelty-suppressed feeding, and 5-HT2CR-induced hypolocomotion tests), the neurochemical, and the cellular (RT-qPCR, mRNA editing, and c-fos-induced expression) levels. RESULTS: Although 5-HTT(-/-) mice had an anxiogenic profile in the novelty-suppressed feeding test, they displayed less 5-HT2CR-mediated anxiety in response to the agonist m-chlorophenylpiperazine in the social interaction test. In addition, 5-HT2CR-mediated inhibition of a stress-induced increase in 5-HT turnover, measured in various brain areas, was markedly reduced in 5-HTT(-/-) mutants. These indices of tolerance to 5-HT2CR stimulation were associated neither with altered levels of 5-HT2CR protein and mRNA nor with changes in pre-mRNA editing in the frontal cortex. However, basal c-fos mRNA production in cells expressing 5-HT2CR was higher in 5-HTT(-/-) mutants, suggesting an altered basal activity of these cells following sustained 5-HT reuptake carrier inactivation. Furthermore, the increased c-fos mRNA expression in 5-HT2CR-like immune-positive cortical cells observed in wild-type mice treated acutely with the 5-HT2CR agonist RO-60,0175 was absent in 5-HTT(-/-) mutants. CONCLUSIONS: Such blunted responsiveness of the 5-HT2CR system, observed at the cell signaling level, probably contributes to the moderation of the anxiety phenotype in 5-HTT(-/-) mice.


Asunto(s)
Ansiedad , Conducta Animal/fisiología , Encéfalo/metabolismo , Receptor de Serotonina 5-HT2C/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/deficiencia , Análisis de Varianza , Animales , Ansiedad/genética , Ansiedad/metabolismo , Ansiedad/patología , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Modelos Animales de Enfermedad , Conducta Alimentaria/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Relaciones Interpersonales , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN Mensajero/metabolismo , Receptor de Serotonina 5-HT2C/genética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Agonistas de Receptores de Serotonina/farmacología
6.
Brain ; 136(Pt 3): 957-70, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23404338

RESUMEN

Myotonic dystrophy type 1 is a complex multisystemic inherited disorder, which displays multiple debilitating neurological manifestations. Despite recent progress in the understanding of the molecular pathogenesis of myotonic dystrophy type 1 in skeletal muscle and heart, the pathways affected in the central nervous system are largely unknown. To address this question, we studied the only transgenic mouse line expressing CTG trinucleotide repeats in the central nervous system. These mice recreate molecular features of RNA toxicity, such as RNA foci accumulation and missplicing. They exhibit relevant behavioural and cognitive phenotypes, deficits in short-term synaptic plasticity, as well as changes in neurochemical levels. In the search for disease intermediates affected by disease mutation, a global proteomics approach revealed RAB3A upregulation and synapsin I hyperphosphorylation in the central nervous system of transgenic mice, transfected cells and post-mortem brains of patients with myotonic dystrophy type 1. These protein defects were associated with electrophysiological and behavioural deficits in mice and altered spontaneous neurosecretion in cell culture. Taking advantage of a relevant transgenic mouse of a complex human disease, we found a novel connection between physiological phenotypes and synaptic protein dysregulation, indicative of synaptic dysfunction in myotonic dystrophy type 1 brain pathology.


Asunto(s)
Conducta Animal/fisiología , Distrofia Miotónica/genética , Distrofia Miotónica/metabolismo , Transmisión Sináptica/fisiología , Vesículas Sinápticas/metabolismo , Adulto , Anciano , Animales , Western Blotting , Electroforesis en Gel Bidimensional , Electrofisiología , Humanos , Hibridación Fluorescente in Situ , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Distrofia Miotónica/complicaciones , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Expansión de Repetición de Trinucleótido
7.
Proc Natl Acad Sci U S A ; 108(16): 6632-7, 2011 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-21467220

RESUMEN

Among the pathogenic processes contributing to dopaminergic neuron (DN) death in Parkinson disease (PD), evidence points to non-cell-autonomous mechanisms, particularly chronic inflammation mounted by activated microglia. Yet little is known about endogenous regulatory processes that determine microglial actions in pathological states. We examined the role of glucocorticoid receptors (GRs), activated by glucocorticoids released in response to stress and known to regulate inflammation, in DN survival. Overall GR level was decreased in substantia nigra of PD patients and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-intoxicated mice. GR changes, specifically in the microglia after MPTP treatment, revealed a rapid augmentation in the number of microglia displaying nuclear localization of GR. Mice with selective inactivation of the GR gene in macrophages/microglia (GR(LysMCre)) but not in DNs (GR(DATCre)) showed increased loss of DNs after MPTP intoxication. This DN loss in GR(LysMCre) mice was not prevented by corticosterone treatment, in contrast to the protection observed in control littermates. Moreover, absence of microglial GRs augmented microglial reactivity and led to their persistent activation. Analysis of inflammatory genes revealed an up-regulation of Toll-like receptors (TLRs) by MPTP treatment, particularly TLR9, the level of which was high in postmortem parkinsonian brains. The regulatory control of GR was reflected by higher expression of proinflammatory genes (e.g., TNF-α) with a concomitant decrease in anti-inflammatory genes (e.g., IL-1R2) in GR(LysMCre) mice. Indeed, in GR(LysMCre) mice, alterations in phosphorylated NF-κB levels indicated its protracted activation. Together, our data indicate that GR is important in curtailing microglial reactivity, and its deregulation in PD could lead to sustained inflammation-mediated DN injury.


Asunto(s)
Intoxicación por MPTP/metabolismo , Microglía/metabolismo , Enfermedad de Parkinson/metabolismo , Receptores de Glucocorticoides/metabolismo , Sustancia Negra/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/patología , Femenino , Humanos , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Intoxicación por MPTP/genética , Intoxicación por MPTP/patología , Masculino , Ratones , Ratones Transgénicos , Microglía/patología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Receptores de Glucocorticoides/genética , Sustancia Negra/patología , Receptores Toll-Like/genética , Receptores Toll-Like/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
8.
Traffic ; 12(11): 1501-20, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21801291

RESUMEN

By analogy to other axonal proteins, transcytotic delivery following spontaneous endocytosis from the somatodendritic membrane is expected to be essential for polarized distribution of axonal G-protein coupled receptors (GPCRs). However, possible contribution from constitutive activation, which may also result in constitutive GPCR endocytosis, is poorly known. Using two closely related but differentially distributed serotonin receptors, here we demonstrate higher constitutive activation and spontaneous endocytosis for the axonal 5-HT(1B) R, as compared to the somatodendritic 5-HT(1A) R, both in non-neuronal cells and neurons. Activation-dependent constitutive endocytosis is crucial for axonal targeting, because inverse-agonist treatment, which prevents constitutive activation, leads to atypical accumulation of newly synthesized 5-HT(1B) Rs on the somatodendritic plasma membrane. Using receptor chimeras composed of different domains from 5-HT(1A) R and 5-HT(1B) R, we show that the complete third intracellular loop of 5-HT(1B) R is necessary and sufficient for constitutive activation and efficient axonal targeting, both sensitive to inverse-agonist treatment. These results suggest that activation and targeting of 5-HT(1B) Rs are intimately interconnected in neurons.


Asunto(s)
Axones/metabolismo , Receptor de Serotonina 5-HT1B/metabolismo , Secuencia de Aminoácidos , Animales , Células COS , Membrana Celular/metabolismo , Células Cultivadas , Chlorocebus aethiops , Endocitosis/fisiología , Células HeLa , Hipocampo/citología , Hipocampo/metabolismo , Humanos , Células LLC-PK1 , Datos de Secuencia Molecular , Neuronas/metabolismo , Estructura Terciaria de Proteína , Transporte de Proteínas , Ratas , Receptor de Serotonina 5-HT1A/metabolismo , Relación Estructura-Actividad , Porcinos , Células Tumorales Cultivadas
9.
J Neurosci ; 32(41): 14227-41, 2012 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-23055492

RESUMEN

Although essential for their neuronal function, the molecular mechanisms underlying the dendritic targeting of serotonin G-protein-coupled receptors are poorly understood. Here, we characterized a Yif1B-dependent vesicular scaffolding complex mediating the intracellular traffic of the rat 5-HT(1A) receptor (5-HT(1A)R) toward dendrites. By combining directed mutagenesis, GST-pull down, and surface plasmon resonance, we identified a tribasic motif in the C-tail of the 5-HT(1A)R on which Yif1B binds directly with high affinity (K(D) ≈ 37 nM). Moreover, we identified Yip1A, Rab6, and Kif5B as new partners of the 5-HT(1A)R/Yif1B complex, and showed that their expression in neurons is also crucial for the dendritic targeting of the 5-HT(1A)R. Live videomicroscopy revealed that 5-HT(1A)R, Yif1B, Yip1A, and Rab6 traffic in vesicles exiting the soma toward the dendritic tree, and also exhibit bidirectional motions, sustaining their role in 5-HT(1A)R dendritic targeting. Hence, we propose a new trafficking pathway model in which Yif1B is the scaffold protein recruiting the 5-HT(1A)R in a complex including Yip1A and Rab6, with Kif5B and dynein as two opposite molecular motors coordinating the traffic of vesicles along dendritic microtubules. This targeting pathway opens new insights for G-protein-coupled receptors trafficking in neurons.


Asunto(s)
Dendritas/fisiología , Regiones de Fijación a la Matriz/fisiología , Proteínas del Tejido Nervioso/fisiología , Neuronas/fisiología , Receptor de Serotonina 5-HT1A/fisiología , Vesículas Sinápticas/metabolismo , Proteínas de Transporte Vesicular/fisiología , Animales , Células Cultivadas , Dendritas/genética , Marcación de Gen/métodos , Humanos , Regiones de Fijación a la Matriz/genética , Microtúbulos/metabolismo , Microtúbulos/fisiología , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Transporte de Proteínas/genética , Transporte de Proteínas/fisiología , Ratas , Ratas Sprague-Dawley , Receptor de Serotonina 5-HT1A/genética , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Vesículas Sinápticas/genética , Proteínas de Transporte Vesicular/genética
10.
Am J Physiol Lung Cell Mol Physiol ; 303(6): L500-8, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22797248

RESUMEN

Decreasing the bioavailability of serotonin (5-HT) by inhibiting its biosynthesis may represent a useful adjunctive treatment of pulmonary hypertension (PH). We assessed this hypothesis using LP533401, which inhibits the rate-limiting enzyme tryptophan hydroxylase 1 (Tph1) expressed in the gut and lung, without inhibiting Tph2 expressed in neurons. Mice treated repeatedly with LP533401 (30-250 mg/kg per day) exhibited marked 5-HT content reductions in the gut, lungs, and blood, but not in the brain. After a single LP533401 dose (250 mg/kg), lung and gut 5-HT contents decreased by 50%, whereas blood 5-HT levels remained unchanged, suggesting gut and lung 5-HT synthesis. Treatment with the 5-HT transporter (5-HTT) inhibitor citalopram decreased 5-HT contents in the blood and lungs but not in the gut. In transgenic SM22-5-HTT+ mice, which overexpress 5-HTT in smooth muscle cells and spontaneously develop PH, 250 mg/kg per day LP533401 or 10 mg/kg per day citalopram for 21 days markedly reduced lung and blood 5-HT levels, right ventricular (RV) systolic pressure, RV hypertrophy, distal pulmonary artery muscularization, and vascular Ki67-positive cells (P < 0.001). Combined treatment with both drugs was more effective in improving PH-related hemodynamic parameters than either drug alone. LP533401 or citalopram treatment partially prevented PH development in wild-type mice exposed to chronic hypoxia. Lung and blood 5-HT levels were lower in hypoxic than in normoxic mice and decreased further after LP533401 or citalopram treatment. These results provide proof of concept that inhibiting Tph1 may represent a new therapeutic strategy for human PH.


Asunto(s)
Citalopram/farmacología , Duodeno/metabolismo , Hipertensión Pulmonar/prevención & control , Pulmón/efectos de los fármacos , Pirimidinas/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Serotonina/metabolismo , Animales , Duodeno/efectos de los fármacos , Hipoxia/fisiopatología , Ratones , Ratones Endogámicos C57BL , Serotonina/biosíntesis , Serotonina/sangre , Proteínas de Transporte de Serotonina en la Membrana Plasmática/biosíntesis , Triptófano Hidroxilasa/antagonistas & inhibidores
11.
J Neurochem ; 121(1): 99-114, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22146001

RESUMEN

The microtubule-associated Stable Tubulie Only Polypeptide (STOP; also known as MAP6) protein plays a key role in neuron architecture and synaptic plasticity, the dysfunctions of which are thought to be implicated in the pathophysiology of psychiatric diseases. The deletion of STOP in mice leads to severe disorders reminiscent of several schizophrenia-like symptoms, which are also associated with differential alterations of the serotonergic tone in somas versus terminals. In STOP knockout (KO) compared with wild-type mice, serotonin (5-HT) markers are found to be markedly accumulated in the raphe nuclei and, in contrast, deeply depleted in all serotonergic projection areas. In the present study, we carefully examined whether the 5-HT imbalance would lead to behavioral consequences evocative of mood and/or cognitive disorders. We showed that STOP KO mice exhibited depression-like behavior, associated with a decreased anxiety-status in validated paradigms. In addition, although STOP KO mice had a preserved very short-term memory, they failed to perform well in all other learning and memory tasks. We also showed that STOP KO mice exhibited regional imbalance of the norepinephrine tone as observed for 5-HT. As a consequence, mutant mice were hypersensitive to acute antidepressants with different selectivity. Altogether, these data indicate that the deletion of STOP protein in mice caused deep alterations in mood and cognitive performances and that STOP protein might have a crucial role in the 5-HT and norepinephrine networks development.


Asunto(s)
Trastornos del Conocimiento/genética , Eliminación de Gen , Proteínas Asociadas a Microtúbulos/deficiencia , Proteínas Asociadas a Microtúbulos/genética , Trastornos del Humor/genética , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Animales , Conducta Animal/fisiología , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/psicología , Femenino , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/fisiología , Trastornos del Humor/metabolismo , Trastornos del Humor/psicología , Proteínas del Tejido Nervioso/fisiología
12.
Int J Neuropsychopharmacol ; 15(3): 363-74, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21557882

RESUMEN

Genetic factors are believed to be involved in the aetiology of unipolar depressive disorders. We have previously described a model built up by selective breeding of mice with different responses in the tail suspension test, a screening test for potential antidepressants. In this model, helpless H/Rouen mice are essentially immobile in this test, as well as in the Porsolt forced-swim test, whereas non-helpless NH/Rouen mice show the opposite behaviour, i.e. very low immobility. However, it is unclear whether or not the other phenotypic differences (forced swim test, locomotor activity, sucrose test, sleep patterns, effect of fluoxetine) observed between H/Rouen and the NH/Rouen mice may be attributed to a genetic drift phenomenon during the selection step, rather than being related to the trait of selection. In this study we used reciprocal crossbreeding between H/Rouen and NH/Rouen mice and obtained a segregating F2 population in order to determine whether phenotypic differences between the two lines co-segregate with the trait of selection. In the segregating F2 population, we found significant and strong genetic correlations between helplessness in the tail suspension test and some phenotypical features associated with depressive disorders such as 'alterations of sleep patterns', behavioural response to fluoxetine, immobility duration in the forced swim test, and anhedonia. Our results converge with clinical observations in depressed humans. These results strengthen the validity of the H/Rouen mouse as a model of depression, notably for preclinical studies with antidepressants. In addition, this model should open the way to identifying genes related to depression-like behaviours.


Asunto(s)
Trastorno Depresivo/genética , Predisposición Genética a la Enfermedad , Personalidad/genética , Anhedonia/fisiología , Animales , Antidepresivos de Segunda Generación/farmacología , Trastorno Depresivo/tratamiento farmacológico , Trastorno Depresivo/fisiopatología , Sacarosa en la Dieta/administración & dosificación , Modelos Animales de Enfermedad , Femenino , Fluoxetina/farmacología , Masculino , Ratones Endogámicos , Actividad Motora/fisiología , Pruebas Neuropsicológicas , Fenotipo , Sueño/genética , Sueño/fisiología , Especificidad de la Especie
13.
J Neurosci ; 30(6): 2198-210, 2010 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-20147547

RESUMEN

Three different subtypes of H(+)-dependent carriers (named VGLUT1-3) concentrate glutamate into synaptic vesicles before its exocytotic release. Neurons using other neurotransmitter than glutamate (such as cholinergic striatal interneurons and 5-HT neurons) express VGLUT3. It was recently reported that VGLUT3 increases acetylcholine vesicular filling, thereby, stimulating cholinergic transmission. This new regulatory mechanism is herein designated as vesicular-filling synergy (or vesicular synergy). In the present report, we found that deletion of VGLUT3 increased several anxiety-related behaviors in adult and in newborn mice as early as 8 d after birth. This precocious involvement of a vesicular glutamate transporter in anxiety led us to examine the underlying functional implications of VGLUT3 in 5-HT neurons. On one hand, VGLUT3 deletion caused a significant decrease of 5-HT(1A)-mediated neurotransmission in raphe nuclei. On the other hand, VGLUT3 positively modulated 5-HT transmission of a specific subset of 5-HT terminals from the hippocampus and the cerebral cortex. VGLUT3- and VMAT2-positive serotonergic fibers show little or no 5-HT reuptake transporter. These results unravel the existence of a novel subset of 5-HT terminals in limbic areas that might play a crucial role in anxiety-like behaviors. In summary, VGLUT3 accelerates 5-HT transmission at the level of specific 5-HT terminals and can exert an inhibitory control at the raphe level. Furthermore, our results suggest that the loss of VGLUT3 expression leads to anxiety-associated behaviors and should be considered as a potential new target for the treatment of this disorder.


Asunto(s)
Sistemas de Transporte de Aminoácidos Acídicos/fisiología , Ansiedad/fisiopatología , Serotonina/fisiología , Sistemas de Transporte de Aminoácidos Acídicos/genética , Animales , Ansiedad/metabolismo , Autorreceptores/fisiología , Corteza Cerebral/fisiopatología , Hipocampo/fisiopatología , Ratones , Ratones Noqueados , Terminales Presinápticos/metabolismo , Núcleos del Rafe/fisiopatología , Receptor de Serotonina 5-HT1A/fisiología , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Transmisión Sináptica , Proteínas de Transporte Vesicular de Monoaminas/metabolismo
14.
J Physiol ; 589(Pt 8): 2079-91, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21486808

RESUMEN

Defence responses triggered experimentally in rats by stimulation of the dorsomedial nucleus of the hypothalamus (DMH) and the dorsolateral periaqueductal grey matter (PAG) inhibit the cardiac baroreflex response (i.e. bradycardia). It has also been proposed that the midbrain cuneiform nucleus (CnF) is involved in active responses. Our aim was to identify the neurocircuitry involved in defence-induced baroreflex inhibition, with a particular focus on the link between DMH, CnF and dorsolateral PAG. Microinjection of the anterograde tracer Phaseolus vulgaris leucoaggutinin into the CnF revealed a dense projection to the dorsolateral PAG. Moreover, activation of neurons in the CnF induced increased expression of Fos protein in the dorsolateral PAG. Inhibition of neurons of the CnF or dorsolateral PAG prevented the inhibition of baroreflex bradycardia induced by DMH or CnF stimulation, respectively. These results provide a detailed description of the brain circuitry underlying acute baroreflex modulation by neurons of the DMH. Our data have shown for the first time that the CnF plays a key role in defence reaction-associated cardiovascular changes; its stimulation, from the DMH, activates the dorsolateral PAG, which, in turn, inhibits baroreflex bradycardia.


Asunto(s)
Barorreflejo , Bradicardia/prevención & control , Frecuencia Cardíaca , Mesencéfalo/fisiopatología , Inhibición Neural , Vías Nerviosas/fisiopatología , Sustancia Gris Periacueductal/fisiopatología , Análisis de Varianza , Animales , Barorreflejo/efectos de los fármacos , Bradicardia/metabolismo , Bradicardia/fisiopatología , Fármacos Cardiovasculares/administración & dosificación , Mecanismos de Defensa , Retroalimentación Fisiológica , Frecuencia Cardíaca/efectos de los fármacos , Masculino , Núcleo Talámico Mediodorsal/fisiopatología , Mesencéfalo/efectos de los fármacos , Mesencéfalo/metabolismo , Microinyecciones , Inhibición Neural/efectos de los fármacos , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/metabolismo , Técnicas de Trazados de Vías Neuroanatómicas , Trazadores del Tracto Neuronal/administración & dosificación , Neurotransmisores/administración & dosificación , Sustancia Gris Periacueductal/efectos de los fármacos , Sustancia Gris Periacueductal/metabolismo , Fitohemaglutininas/administración & dosificación , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Sprague-Dawley
15.
J Neural Transm (Vienna) ; 118(8): 1215-25, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21301897

RESUMEN

There is considerable evidence showing that the neurodegenerative processes that lead to sporadic Parkinson's disease (PD) begin many years before the appearance of the characteristic motor symptoms and that impairments in olfactory, cognitive and motor functions are associated with time-dependent disruption of dopaminergic neurotransmission in different brain areas. Midkine is a 13-kDa retinoic acid-induced heparin-binding growth factor involved in many biological processes in the central nervous system such as cell migration, neurogenesis and tissue repair. The abnormal midkine expression may be associated with neurochemical dysfunction in the dopaminergic system and cognitive impairments in rodents. Here, we employed adult midkine knockout mice (Mdk(-/-)) to further investigate the relevance of midkine in dopaminergic neurotransmission and in olfactory, cognitive and motor functions. Mdk(/-) mice displayed pronounced impairments in their olfactory discrimination ability and short-term social recognition memory with no gross motor alterations. Moreover, the genetic deletion of midkine decreased the expression of the enzyme tyrosine hydroxylase in the substantia nigra reducing partially the levels of dopamine and its metabolites in the olfactory bulb and striatum of mice. These findings indicate that the genetic deletion of midkine causes a partial loss of dopaminergic neurons and depletion of dopamine, resulting in olfactory and memory deficits with no major motor impairments. Therefore, Mdk(-/-) mice may represent a promising animal model for the study of the early stages of PD and for testing new therapeutic strategies to restore sensorial and cognitive processes in PD.


Asunto(s)
Citocinas/deficiencia , Modelos Animales de Enfermedad , Eliminación de Gen , Factor de Crecimiento Nervioso/deficiencia , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/patología , Animales , Encéfalo/patología , Encéfalo/fisiología , Citocinas/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Midkina , Factor de Crecimiento Nervioso/genética , Enfermedad de Parkinson/metabolismo , Reconocimiento en Psicología/fisiología , Olfato/genética
16.
Bull Acad Natl Med ; 195(7): 1551-65; discussion 1565, 2011 Oct.
Artículo en Francés | MEDLINE | ID: mdl-22812160

RESUMEN

By combining brain section/lesion studies and sleep analysis, neurophysiologists have identified the brain areas responsible for regulating sleep and wakefulness during the first half of the 20th century. Identification of the phenotypic nature of the neurons underlying the regulation of vigilance, as well as their anatomical and functional connections led to a theoretical model based on mutual inhibitory interactions between sleep-on neurons (namely GABAergic neurons of the hypothalamic preoptic region) and wake-on neurons (mainly monoaminergic and cholinergic neurons). In addition to the corresponding neurotransmitters (serotonin, acetylcholine and GABA), other neuroactive molecules that play key roles in sleep and wakefulness regulation have recently been discovered, leading to an updated model. Hypocretin, also known as orexin, is a key neuropeptide involved in the sleep disorder narcolepsy. Extensive characterization of the respective roles of these neurotransmitters has led to the identification of novel therapeutic targets for the treatment of sleep disorders. For example, blockade of hypocretin receptors has hypnotic effects.


Asunto(s)
Sueño/fisiología , Vigilia/fisiología , Encéfalo/fisiología , Humanos , Neuronas/fisiología , Receptores de Neurotransmisores/fisiología
17.
Therapie ; 76(2): 63-66, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33358365

RESUMEN

For this issue, Luc Zimmer, professor of pharmacology and chair of the Neuropsychopharmacology Committee of the French Society of Pharmacology and Therapeutics, talked with Michel Hamon, honorary director of research at the French National Institute of Health and Medical Research (INSERM) and honorary professor of neuropharmacology at Paris-Sorbonne (Pierre et Marie Curie) University. Some of the leading names in neuropsychopharmacology research are mentioned, pointing to significant conceptual advances that founded this discipline. The links between psychopharmacology and neuropharmacology are also discussed in the light of past collaborations. Finally, priorities are proposed for the emergence of the psychopharmacology of the future.


Asunto(s)
Investigación Biomédica , Psicofarmacología , Academias e Institutos , Comunicación , Humanos , Neurofarmacología
18.
J Neurosci ; 29(49): 15575-85, 2009 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-20007481

RESUMEN

Restraint stress produces changes in the sleep pattern that are mainly characterized by a delayed increase in rapid eye movement sleep (REMS) amounts. Because the serotonin (5-HT) and the hypocretin (hcrt) systems that regulate REMS are interconnected, we used mutant mice deficient in the 5-HT transporter (5-HTT(-/-)) to examine the role of 5-HT and hcrt neurotransmissions in the sleep response to stress. In contrast to wild-type mice, restraint stress did not induce a delayed increase in REMS amounts in 5-HTT(-/-) mice, indicating impaired sleep homeostasis in mutants. However, pharmacological blockade of the hcrt type 1 receptor (hcrt-R1) before restraint stress restored the REMS increase in 5-HTT(-/-) mice. In line with this finding, 5-HTT(-/-) mutants displayed after restraint stress higher long-lasting activation of hypothalamic preprohcrt neurons than wild-type mice and elevated levels of the hcrt-1 peptide and the hcrt-R1 mRNA in the anterior raphe area. Thus, hypocretinergic neurotransmission was enhanced by stress in 5-HTT(-/-) mice. Furthermore, in 5-HTT(-/-) but not wild-type mice, hypothalamic levels of the 5-HT metabolite 5-hydroxyindole acetic acid significantly increased after restraint stress, indicating a marked enhancement of serotonergic neurotransmission in mutants. Altogether, our data show that increased serotonergic -and in turn hypocretinergic- neurotransmissions exert an inhibitory influence on stress-induced delayed REMS. We propose that the direct interactions between hcrt neurons in the hypothalamus and 5-HT neurons in the anterior raphe nuclei account, at least in part, for the adaptive sleep-wakefulness regulations triggered by acute stress.


Asunto(s)
Homeostasis/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neuropéptidos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropéptido/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Sueño/fisiología , Estrés Psicológico/fisiopatología , Animales , Ácido Hidroxiindolacético/metabolismo , Hipotálamo/fisiopatología , Masculino , Ratones , Ratones Endogámicos , Ratones Noqueados , Neuronas/fisiología , Receptores de Orexina , Orexinas , ARN Mensajero/metabolismo , Núcleos del Rafe/fisiopatología , Restricción Física , Serotonina/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/deficiencia , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Sueño REM/fisiología , Transmisión Sináptica/fisiología
19.
J Neurochem ; 115(2): 438-49, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20796171

RESUMEN

Stress is known to activate the central 5-hydroxytryptamine (5-HT) system, and this is probably part of a coping response involving several 5-HT receptors. Although 5-HT(2C) receptors are well known to be implicated in anxiety, their participation in stress-induced changes had not been investigated in parallel at both behavioral and neurochemical levels. We show here that the preferential 5-HT(2C) receptor agonist, m-chlorophenylpiperazine, as well as restraint stress increased anxiety in the mouse social interaction test. The selective 5-HT(2C) receptor antagonist, SB 242,084, prevented both of these anxiogenic effects. Restraint stress increased 5-HT turnover in various brain areas, and this effect was prevented by the 5-HT(2B/2C) receptor agonist RO 60-0175 (1 mg/kg), but not the preferential 5-HT(2A) agonist 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane (1 mg/kg), and in contrast potentiated by SB 242,084 (1 mg/kg), which also blocked the effect of RO 60-0175. Using microdialysis, RO 60-0175 was shown to inhibit cortical 5-HT overflow in stressed mice when 5-HT reuptake was blocked locally. Chronic paroxetine prevented both the anxiogenic effect of m-chlorophenylpiperazine and the inhibitory effect of RO 60-0175 on locomotion and stress-induced increase in 5-HT turnover. The anxiolytic action of chronic paroxetine might be associated with an enhancement of 5-HT neurotransmission caused by a decreased 5-HT(2C) receptor-mediated inhibition of stress-induced increase in 5-HT release.


Asunto(s)
Antidepresivos de Segunda Generación/farmacología , Encéfalo/efectos de los fármacos , Paroxetina/farmacología , Receptor de Serotonina 5-HT2C/metabolismo , Serotonina/metabolismo , Estrés Psicológico , Anfetaminas/farmacología , Análisis de Varianza , Animales , Conducta Animal , Encéfalo/metabolismo , Etilaminas/farmacología , Líquido Extracelular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Indoles/farmacología , Relaciones Interpersonales , Masculino , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Piperazinas/farmacología , Agonistas de Receptores de Serotonina/farmacología , Estrés Psicológico/tratamiento farmacológico , Estrés Psicológico/metabolismo , Estrés Psicológico/patología
20.
J Neurochem ; 115(6): 1579-94, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20969568

RESUMEN

The deletion of microtubule-associated protein stable tubule only polypeptide (STOP) leads to neuroanatomical, biochemical and severe behavioral alterations in mice, partly alleviated by antipsychotics. Therefore, STOP knockout (KO) mice have been proposed as a model of some schizophrenia-like symptoms. Preliminary data showed decreased brain serotonin (5-HT) tissue levels in STOP KO mice. As literature data demonstrate various interactions between microtubule-associated proteins and 5-HT, we characterized some features of the serotonergic neurotransmission in STOP KO mice. In the brainstem, mutant mice displayed higher tissue 5-HT levels and in vivo synthesis rate, together with marked increases in 5-HT transporter densities and 5-HT1A autoreceptor levels and electrophysiological sensitivity, without modification of the serotonergic soma number. Conversely, in projection areas, STOP KO mice exhibited lower 5-HT levels and in vivo synthesis rate, associated with severe decreases in 5-HT transporter densities, possibly related to reduced serotonergic terminals. Mutant mice also displayed a deficit of adult hippocampal neurogenesis, probably related to both STOP deletion and 5-HT depletion. Finally, STOP KO mice exhibited a reduced anxiety- and, probably, an increased helpness-status, that could be because of the strong imbalance of the serotonin neurotransmission between somas and terminals. Altogether, these data suggested that STOP deletion elicited peculiar 5-HT disconnectivity.


Asunto(s)
Encéfalo/fisiología , Proteínas Asociadas a Microtúbulos/deficiencia , Proteínas Asociadas a Microtúbulos/genética , Red Nerviosa/fisiología , Serotonina/metabolismo , Animales , Femenino , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Noqueados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA