Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Transl Psychiatry ; 13(1): 22, 2023 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-36693858

RESUMO

Loss-of-function mutations of the gene Cul3 have been identified as a risk factor for autism-spectrum disorder (ASD), but the pathogenic mechanisms are not well understood. Conditional Cul3 ablation in cholinergic neurons of mice (ChatCRECul3F/+) recapitulated ASD-like social and sensory gating phenotypes and caused significant cognitive impairments, with diminished activity of cholinergic neurons in the basal forebrain (BF). Chemogenetic inhibition of BF cholinergic neurons in healthy mice induced similar social and cognitive deficits. Conversely, chemogenetic stimulation of BF cholinergic neurons in ChatCRECul3F/+ mice reversed abnormalities in sensory gating and cognition. Cortical hypofunction was also found after ChAT-specific Cul3 ablation and stimulation of cholinergic projections from the BF to the prefrontal cortex (PFC) mitigated cognitive deficits. Overall, we demonstrate that cholinergic dysfunction due to Cul3 deficiency is involved in ASD-like behavioral abnormalities, and that BF cholinergic neurons are particularly critical for cognitive component through their projections to the PFC.


Assuntos
Prosencéfalo Basal , Neurônios Colinérgicos , Disfunção Cognitiva , Proteínas Culina , Córtex Pré-Frontal , Animais , Camundongos , Prosencéfalo Basal/metabolismo , Colinérgicos , Neurônios Colinérgicos/metabolismo , Cognição/fisiologia , Disfunção Cognitiva/genética , Disfunção Cognitiva/metabolismo , Proteínas Culina/genética , Proteínas Culina/metabolismo , Córtex Pré-Frontal/metabolismo , Colina O-Acetiltransferase/genética , Colina O-Acetiltransferase/metabolismo
2.
Front Mol Neurosci ; 15: 1037481, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36504678

RESUMO

Introduction: An inactivating mutation in the histidine decarboxylase gene (Hdc) has been identified as a rare but high-penetrance genetic cause of Tourette syndrome (TS). TS is a neurodevelopmental syndrome characterized by recurrent motor and vocal tics; it is accompanied by structural and functional abnormalities in the cortico-basal ganglia circuitry. Hdc, which is expressed both in the posterior hypothalamus and peripherally, encodes an enzyme required for the biosynthesis of histamine. Hdc knockout mice (Hdc-KO) functionally recapitulate this mutation and exhibit behavioral and neurochemical abnormalities that parallel those seen in patients with TS. Materials and methods: We performed exploratory RNA-seq to identify pathological alterations in several brain regions in Hdc-KO mice. Findings were corroborated with RNA and protein quantification, immunohistochemistry, and ex vivo brain imaging using MRI. Results: Exploratory RNA-Seq analysis revealed, unexpectedly, that genes associated with oligodendrocytes and with myelin production are upregulated in the dorsal striatum of these mice. This was confirmed by qPCR, immunostaining, and immunoblotting. These results suggest an abnormality in myelination in the striatum. To test this in an intact mouse brain, we performed whole-brain ex vivo diffusion tensor imaging (DTI), which revealed reduced fractional anisotropy (FA) in the dorsal striatum. Discussion: While the DTI literature in individuals with TS is sparse, these results are consistent with findings of disrupted descending cortical projections in patients with tics. The Hdc-KO model may represent a powerful system in which to examine the developmental mechanisms underlying this abnormality.

3.
Mol Psychiatry ; 27(8): 3355-3366, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35296809

RESUMO

Large-scale genetic studies have revealed that the most prominent genes disrupted in autism are chromatin regulators mediating histone methylation/demethylation, suggesting the central role of epigenetic dysfunction in this disorder. Here, we show that histone lysine 4 dimethylation (H3K4me2), a histone mark linked to gene activation, is significantly decreased in the prefrontal cortex (PFC) of autistic human patients and mutant mice with the deficiency of top-ranking autism risk factor Shank3 or Cul3. A brief treatment of the autism models with highly potent and selective inhibitors of the H3K4me2 demethylase LSD1 (KDM1A) leads to the robust rescue of core symptoms of autism, including social deficits and repetitive behaviors. Concomitantly, LSD1 inhibition restores NMDA receptor function in PFC and AMPA receptor-mediated currents in striatum of Shank3-deficient mice. Genome-wide RNAseq and ChIPseq reveal that treatment of Shank3-deficient mice with the LSD1 inhibitor restores the expression and H3K4me2 occupancy of downregulated genes enriched in synaptic signaling and developmental processes. The immediate early gene tightly linked to neuronal plasticity, Egr1, is on the top list of rescued genes. The diminished transcription of Egr1 is recapitulated in PFC of autistic human patients. Overexpression of Egr1 in PFC of Shank3-deficient mice ameliorates social preference deficits. These results have for the first time revealed an important role of H3K4me2 abnormality in ASD pathophysiology, and the therapeutic potential of targeting H3K4me2 demethylase LSD1 or the downstream molecule Egr1 for ASD.


Assuntos
Transtorno Autístico , Histonas , Humanos , Camundongos , Animais , Histonas/metabolismo , Histona Desmetilases/genética , Histona Desmetilases/metabolismo , Cromatina , Modelos Animais de Doenças , Proteínas dos Microfilamentos/genética , Proteínas do Tecido Nervoso/metabolismo
4.
Mol Psychiatry ; 26(5): 1491-1504, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-31455858

RESUMO

Cullin 3 (Cul3) gene, which encodes a core component of the E3 ubiquitin ligase complex that mediates proteasomal degradation, has been identified as a true high-risk factor for autism. Here, by combining behavioral, electrophysiological, and proteomic approaches, we have examined how Cul3 deficiency contributes to the etiology of different aspects of autism. Heterozygous mice with forebrain Cul3 deletion displayed autism-like social interaction impairment and sensory-gating deficiency. Region-specific deletion of Cul3 leads to distinct phenotypes, with social deficits linked to the loss of Cul3 in prefrontal cortex (PFC), and stereotypic behaviors linked to the loss of Cul3 in striatum. Correlated with these behavioral alterations, Cul3 deficiency in forebrain or PFC induces NMDA receptor hypofunction, while Cul3 loss in striatum causes a cell type-specific alteration of neuronal excitability in striatal circuits. Large-scale profiling has identified sets of misregulated proteins resulting from Cul3 deficiency in different regions, including Smyd3, a histone methyltransferase involved in gene transcription. Inhibition or knockdown of Smyd3 in forebrain Cul3-deficient mice ameliorates social deficits and restores NMDAR function in PFC. These results have revealed for the first time a potential molecular mechanism underlying the manifestation of different autism-like behavioral deficits by Cul3 deletion in cortico-striatal circuits.


Assuntos
Transtorno Autístico , Proteínas Culina/genética , Animais , Transtorno Autístico/genética , Proteínas Culina/metabolismo , Camundongos , Fenótipo , Proteômica , Receptores de N-Metil-D-Aspartato
5.
Neuroscience ; 392: 172-179, 2018 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-30278251

RESUMO

Histamine dysregulation has been identified as a rare genetic cause of tic disorders; mice with a knockout of the histidine decarboxylase (Hdc) gene represent a promising model of this pathophysiology. How alterations in the histamine system lead to neuropsychiatric disease, however, remains unclear. The H3R histamine receptor is elevated in the striatum of Hdc KO mice, and H3R agonists, acting in the dorsal striatum, trigger tic-like movements in the model. In wild-type mice, H3R in the dorsal striatum differentially regulates mitogen-activated protein kinase (MAPK) and protein kinase B (Akt) signaling in D1R dopamine receptor-expressing striatonigral medium spiny neurons (dMSNs) and D2R dopamine receptor-expressing striatopallidal MSNs (iMSNs), respectively. We examined the effects of H3R agonist treatment on MSN signaling in the Hdc-KO model. In dMSNs, MAPK signaling was elevated at baseline in the Hdc-KO model, resembling what is seen after H3R activation in WT animals. Similarly, in iMSNs, Akt phosphorylation was reduced at baseline in the KO model, resembling what is seen after H3R activation in WT animals. H3R activation in Hdc-KO mice further enhanced the baseline effect on Akt phosphorylation in iMSNs but attenuated the abnormality in MAPK signaling in dMSNs. These observations support the hypothesis that constitutive activity of upregulated H3R receptors in the Hdc-KO model mediates the observed alterations in baseline MSN signaling; but further activation of H3R, which produces tic-like repetitive movements in the model, has more complex effects.


Assuntos
Corpo Estriado/metabolismo , Neurônios/metabolismo , Receptores Histamínicos H3/metabolismo , Transtornos de Tique/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Histidina Descarboxilase/genética , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteína S6 Ribossômica/metabolismo , Transdução de Sinais
6.
Brain Behav Immun ; 69: 304-311, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29233751

RESUMO

Pediatric Autoimmune Neuropsychiatric Disorder Associated with Streptococcus, or PANDAS, is a syndrome of acute childhood onset of obsessive-compulsive disorder and other neuropsychiatric symptoms in the aftermath of an infection with Group A beta-hemolytic Streptococcus (GABHS). Its pathophysiology remains unclear. PANDAS has been proposed to result from cross-reactivity of antibodies raised against GABHS with brain antigens, but the targets of these antibodies are unclear and may be heterogeneous. We developed an in vivo assay in mice to characterize the cellular targets of antibodies in serum from individuals with PANDAS. We focus on striatal interneurons, which have been implicated in the pathogenesis of tic disorders. Sera from children with well-characterized PANDAS (n = 5) from a previously described clinical trial (NCT01281969), and matched controls, were infused into the striatum of mice; antibody binding to interneurons was characterized using immunofluorescence and confocal microscopy. Antibodies from children with PANDAS bound to ∼80% of cholinergic interneurons, significantly higher than the <50% binding seen with matched healthy controls. There was no elevated binding to two different populations of GABAergic interneurons (PV and nNOS-positive), confirming the specificity of this phenomenon. Elevated binding to cholinergic interneurons resolved in parallel with symptom improvement after treatment with intravenous immunoglobulin. Antibody-mediated dysregulation of striatal cholinergic interneurons may be a locus of pathology in PANDAS. Future clarification of the functional consequences of this specific binding may identify new opportunities for intervention in children with this condition.


Assuntos
Anticorpos/imunologia , Doenças Autoimunes/imunologia , Neurônios Colinérgicos/imunologia , Corpo Estriado/imunologia , Interneurônios/imunologia , Infecções Estreptocócicas/imunologia , Animais , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Camundongos , Transtorno Obsessivo-Compulsivo
7.
Trends Neurosci ; 40(7): 397-407, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28578790

RESUMO

The brain includes multiple types of interconnected excitatory and inhibitory neurons that together allow us to move, think, feel, and interact with the environment. Inhibitory interneurons (INs) comprise a small, heterogeneous fraction, but they exert a powerful and tight control over neuronal activity and consequently modulate the magnitude of neuronal output and, ultimately, information processing. IN abnormalities are linked to two pediatric psychiatric disorders with high comorbidity: autism spectrum disorder (ASD) and Tourette syndrome (TS). Studies probing the basis of this link have been contradictory regarding whether the causative mechanism is a reduction in number, dysfunction, or gene aberrant expression (or a combination thereof). Here, we integrate different theories into a more comprehensive view of INs as responsible for the symptomatology observed in these disorders.


Assuntos
Transtorno Autístico/fisiopatologia , Encéfalo/fisiopatologia , Interneurônios/fisiologia , Síndrome de Tourette/fisiopatologia , Animais , Transtorno Autístico/genética , Humanos , Vias Neurais/fisiopatologia , Síndrome de Tourette/genética
8.
Proc Natl Acad Sci U S A ; 114(25): 6599-6604, 2017 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-28584117

RESUMO

Aberrant histaminergic function has been proposed as a cause of tic disorders. A rare mutation in the enzyme that produces histamine (HA), histidine decarboxylase (HDC), has been identified in patients with Tourette syndrome (TS). Hdc knockout mice exhibit repetitive behavioral pathology and neurochemical characteristics of TS, establishing them as a plausible model of tic pathophysiology. Where, when, and how HA deficiency produces these effects has remained unclear: whether the contribution of HA deficiency to pathogenesis is acute or developmental, and where in the brain the relevant consequences of HA deficiency occur. Here, we address these key pathophysiological questions, using anatomically and cellularly targeted manipulations in mice. We report that specific ablation or chemogenetic silencing of histaminergic neurons in the tuberomammillary nucleus (TMN) of the hypothalamus leads to markedly elevated grooming, a form of repetitive behavioral pathology, and to elevated markers of neuronal activity in both dorsal striatum and medial prefrontal cortex. Infusion of HA directly into the striatum reverses this behavioral pathology, confirming that acute HA deficiency mediates the effect. Bidirectional chemogenetic regulation reveals that dorsal striatum neurons activated after TMN silencing are both sufficient to produce repetitive behavioral pathology and necessary for the full expression of the effect. Chemogenetic activation of TMN-regulated medial prefrontal cortex neurons, in contrast, increases locomotion and not grooming. These data confirm the centrality of striatal regulation by neurotransmitter HA in the adult in the production of pathological grooming.


Assuntos
Gânglios da Base/metabolismo , Asseio Animal/fisiologia , Histamina/metabolismo , Animais , Corpo Estriado/metabolismo , Histidina Descarboxilase/metabolismo , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Córtex Pré-Frontal/metabolismo , Corno Dorsal da Medula Espinal/metabolismo , Síndrome de Tourette/metabolismo
9.
Biol Psychiatry ; 82(3): 194-203, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28347488

RESUMO

BACKGROUND: Interneuronal pathology is implicated in many neuropsychiatric disorders, including autism spectrum disorder (ASD) and Tourette syndrome (TS). Interneurons of the striatum, including the parvalbumin-expressing fast-spiking interneurons (FSIs) and the large cholinergic interneurons (CINs), are affected in patients with TS and in preclinical models of both ASD and TS. METHODS: To test the causal importance of these neuronal abnormalities, we recapitulated them in vivo in developmentally normal mice using a combination transgenic-viral strategy for targeted toxin-mediated ablation. RESULTS: We found that conjoint ~50% depletion of FSIs and CINs in the dorsal striatum of male mice produces spontaneous stereotypy and marked deficits in social interaction. Strikingly, these behavioral effects are not seen in female mice; because ASD and TS have a marked male predominance, this observation reinforces the potential relevance of the finding to human disease. Neither of these effects is seen when only one or the other interneuronal population is depleted; ablation of both is required. Depletion of FSIs, but not of CINs, also produces anxiety-like behavior, as has been described previously. Behavioral pathology in male mice after conjoint FSI and CIN depletion is accompanied by increases in activity-dependent signaling in the dorsal striatum; these alterations were not observed after disruption of only one interneuron type or in doubly depleted female mice. CONCLUSIONS: These data indicate that disruption of CIN and FSI interneurons in the dorsal striatum is sufficient to produce network and behavioral changes of potential relevance to ASD, in a sexually dimorphic manner.


Assuntos
Transtorno Autístico/patologia , Corpo Estriado/patologia , Interneurônios/patologia , Caracteres Sexuais , Animais , Ansiedade/patologia , Ansiedade/fisiopatologia , Transtorno Autístico/fisiopatologia , Condicionamento Operante/fisiologia , Corpo Estriado/fisiopatologia , Modelos Animais de Doenças , Comportamento Exploratório/fisiologia , Feminino , Imuno-Histoquímica , Interneurônios/fisiologia , Masculino , Camundongos Transgênicos , Atividade Motora/fisiologia , Inibição Pré-Pulso/fisiologia , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Comportamento Social , Comportamento Estereotipado/fisiologia , Transmissão Sináptica/fisiologia
10.
Artigo em Inglês | MEDLINE | ID: mdl-27773554

RESUMO

Histaminergic dysfunction has been recently linked to tic disorders and to aberrant striatal function. There is a particular interest in the histamine 3 receptor (H3R) due to its clinical implications for treating multiple disorders and its high expression in the brain. Striatal histamine (HA) modulates through the H3R in complex ways the release of striatal neurotransmitters into this brain region. The H3R has been classically described to be coupled to Gi, although there is evidence that revealed that striatal H3R forms heteromers with the dopamine receptors 1 and 2 in the medium spiny neurons (MSNs) than changes this signaling. Moreover, new data described for the first time a complete, segregated and time dependent signaling after H3R activation in the two types of MSNs (D1R-MSNs and D2R-MSNs). The aim of this review is to update the role of HA and H3R in striatal function at a molecular and signaling levels.


Assuntos
Corpo Estriado/metabolismo , Histamina/metabolismo , Rede Nervosa/metabolismo , Receptores Histamínicos H3/metabolismo , Animais , Corpo Estriado/anatomia & histologia , Humanos
11.
J Biol Chem ; 291(40): 21042-21052, 2016 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-27510032

RESUMO

The basal ganglia have a central role in motor patterning, habits, motivated behaviors, and cognition as well as in numerous neuropsychiatric disorders. Receptors for histamine, especially the H3 receptor (H3R), are highly expressed in the striatum, the primary input nucleus of the basal ganglia, but their effects on this circuitry have been little explored. H3R interacts with dopamine (DA) receptors ex vivo; the nature and functional importance of these interactions in vivo remain obscure. We found H3R activation with the agonist R-(-)-α-methylhistamine to produce a unique time- and cell type-dependent profile of molecular signaling events in the striatum. H3 agonist treatment did not detectably alter extracellular DA levels or signaling through the cAMP/DARPP-32 signaling pathway in either D1- or D2-expressing striatal medium spiny neurons (MSNs). In D1-MSNs, H3 agonist treatment transiently activated MAPK signaling and phosphorylation of rpS6 and led to phosphorylation of GSK3ß-Ser9, a novel effect. Consequences of H3 activation in D2-MSNs were completely different. MAPK signaling was unchanged, and GSK3ß-Ser9 phosphorylation was reduced. At the behavioral level, two H3 agonists had no significant effect on locomotion or stereotypy, but they dramatically attenuated the locomotor activation produced by the D1 agonist SKF82958. H3 agonist co-administration blocked the activation of MAPK signaling and the phosphorylation of rpS6 produced by D1 activation in D1-MSNs, paralleling behavioral effects. In contrast, GSK3ß-Ser9 phosphorylation was seen only after H3 agonist treatment, with no interactive effects. H3R signaling has been neglected in models of basal ganglia function and has implications for a range of pathophysiologies.


Assuntos
Corpo Estriado/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Locomoção/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores Histamínicos H3/metabolismo , Animais , Benzazepinas/farmacologia , Fosfoproteína 32 Regulada por cAMP e Dopamina/genética , Fosfoproteína 32 Regulada por cAMP e Dopamina/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/genética , Glicogênio Sintase Quinase 3 beta/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Proteínas Proto-Oncogênicas c-akt/genética , Receptores de Dopamina D1/genética , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Receptores Histamínicos H3/genética , Proteína S6 Ribossômica/genética , Proteína S6 Ribossômica/metabolismo
12.
Brain Behav Immun ; 57: 326-337, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27381299

RESUMO

Microglia mediate neuroinflammation and regulate brain development and homeostasis. Microglial abnormalities are implicated in a range of neuropsychiatric pathology, including Tourette syndrome (TS) and autism. Histamine (HA) is both a neurotransmitter and an immune modulator. HA deficiency has been implicated as a rare cause of TS and may contribute to other neuropsychiatric conditions. In vitro studies suggest that HA can regulate microglia, but this has never been explored in vivo. We used immunohistochemistry to examine the effects of HA deficiency in histidine decarboxylase (Hdc) knockout mice and of HA receptor stimulation in wild-type animals. We find HA to regulate microglia in vivo, via the H4 receptor. Chronic HA deficiency in Hdc knockout mice reduces ramifications of microglia in the striatum and (at trend level) in the hypothalamus, but not elsewhere in the brain. Depletion of histaminergic neurons in the hypothalamus has a similar effect. Microglia expressing IGF-1 are particularly reduced, However, the microglial response to challenge with lipopolysacchariade (LPS) is potentiated in Hdc knockout mice. Genetic abnormalities in histaminergic signaling may produce a vulnerability to inflammatory challenge, setting the state for pathogenically dysregulated neuroimmune responses.


Assuntos
Doenças do Sistema Nervoso Central/metabolismo , Corpo Estriado/metabolismo , Interação Gene-Ambiente , Histamina/metabolismo , Histidina Descarboxilase/metabolismo , Inflamação/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Microglia/metabolismo , Receptores Histamínicos H4/metabolismo , Animais , Histamina/deficiência , Histidina Descarboxilase/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
13.
Neuropharmacology ; 106: 85-90, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26282120

RESUMO

The potential contributions of dysregulation of the brain's histaminergic modulatory system to neuropsychiatric disease, and the potential of histamine-targeting medications as therapeutic agents, are gradually coming into focus. The H3R receptor, which is expressed primarily in the central nervous system, is a promising pharmacotherapeutic target. Recent evidence for a contribution of histamine dysregulation to Tourette syndrome and tic disorders is particularly strong; although specific mutations in histamine-associated genes are rare, they have led to informative studies in animal models that may pave the way for therapeutic advances. A controlled study of an H3R antagonist in Tourette syndrome is ongoing. Preclinical studies of H3R antagonists in schizophrenia, attention deficit disorder, and narcolepsy have all shown promise. Recently reported controlled studies have been disappointing in schizophrenia and attention deficit disorder, but the H3R antagonist pitolisant shows promise in the treatment of narcolepsy and excessive daytime sleepiness and is currently under regulatory review for these conditions. This article is part of the Special Issue entitled 'Histamine Receptors'.


Assuntos
Histamina/metabolismo , Transtornos Mentais/metabolismo , Receptores Histamínicos/metabolismo , Síndrome de Tourette/metabolismo , Animais , Humanos
14.
Behav Brain Res ; 277: 193-203, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24949809

RESUMO

Serotonin (5-HT) has been proposed as a possible encoder of reward. Nevertheless, the role of this neurotransmitter in reward-based tasks is not well understood. Given that the major serotonergic circuit in the rat brain comprises the dorsal raphe nuclei and the medial prefrontal cortex (mPFC), and because the latter structure is involved in the control of complex behaviors and expresses 1A (5-HT1A), 2A (5-HT2A), and 3 (5-HT3) receptors, the aim was to study the role of 5-HT and of these receptors in the acquisition and extinction of a reward-dependent operant conditioning task. Long Evans rats were trained in an operant conditioning task while receiving fluoxetine (serotonin reuptake inhibitor, 10mg/kg), tianeptine (serotonin reuptake enhancer, 10mg/kg), buspirone (5-HT1A partial agonist, 10mg/kg), risperidone (5-HT2A antagonist, 1mg/kg), ondansetron (5-HT3 antagonist, 2mg/kg) or vehicle. Then, animals that acquired the operant conditioning without any treatment were trained to extinct the task in the presence of the pharmacological agents. Fluoxetine impaired acquisition but improved extinction. Tianeptine administration induced the opposite effects. Buspirone induced a mild deficit in acquisition and had no effects during the extinction phase. Risperidone administration resulted in learning deficits during the acquisition phase, although it promoted improved extinction. Ondansetron treatment showed a deleterious effect in the acquisition phase and an overall improvement in the extinction phase. These data showed a differential role of 5-HT in the acquisition and extinction of an operant conditioning task, suggesting that it may have a dual function in reward encoding.


Assuntos
Receptor 5-HT1A de Serotonina/metabolismo , Receptor 5-HT2A de Serotonina/metabolismo , Receptores 5-HT3 de Serotonina/metabolismo , Recompensa , Serotonina/metabolismo , Animais , Buspirona/farmacologia , Fluoxetina/farmacologia , Ratos Long-Evans , Agonistas do Receptor 5-HT1 de Serotonina/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/farmacologia
15.
Behav Brain Res ; 280: 92-100, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25435314

RESUMO

Dopamine encodes reward and its prediction in reinforcement learning. Catechol-O-methyltransferase (COMT) activity in the medial prefrontal cortex (mPFC) has been shown to influence cognitive abilities by modifying dopamine clearance. Nevertheless, it is unknown how COMT in the mPFC influences operant learning. Systemic entacapone (50mg/kg), as well as local entacapone (3 pg) and recombinant COMT (17 µg) in the mPFC were administered to male Long Evans rats prior to training in an operant conditioning task. We found that systemic and local administration of the COMT inhibitor entacapone significantly improves learning performance. Conversely, recombinant COMT administration totally impaired learning. These data have been interpreted through a computational model where the phasic firing of dopaminergic neurons was computed by means of a temporal difference algorithm and dopamine bioavailability in the mPFC was simulated with a gating window. The duration of this window was selected to simulate the effects of inhibited or enhanced COMT activity (by entacapone or recombinant COMT respectively). The model accounts for an improved performance reproducing the entacapone effects, and a detrimental impact on learning when the clearance is increased reproducing the recombinant COMT effects. The experimental and computational results show that learning performance can be deeply influenced by COMT manipulations in the mPFC.


Assuntos
Condicionamento Operante/fisiologia , Dopamina/metabolismo , Córtex Pré-Frontal/fisiologia , Ácido 3,4-Di-Hidroxifenilacético/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Algoritmos , Animais , Catecol O-Metiltransferase/metabolismo , Inibidores de Catecol O-Metiltransferase/farmacologia , Catecóis/farmacologia , Simulação por Computador , Condicionamento Operante/efeitos dos fármacos , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/fisiologia , Masculino , Modelos Neurológicos , Redes Neurais de Computação , Nitrilas/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Ratos Long-Evans , Proteínas Recombinantes/metabolismo
16.
Eur Neuropsychopharmacol ; 24(12): 1896-906, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25464894

RESUMO

Tic disorders produce substantial morbidity, but their pathophysiology remains poorly understood. Convergent evidence suggests that dysregulation of the cortico-basal ganglia circuitry is central to the pathogenesis of tics. Tourette syndrome (TS), the most severe end of the continuum of tic disorders, is substantially genetic, but causative mutations have been elusive. We recently described a mouse model, the histidine decarboxylase (Hdc) knockout mouse, that recapitulates a rare, highly penetrant mutation found in a single family; these mice exhibit TS-like phenomenology. These animals have a global deficit in brain histamine and a consequent dysregulation of DA in the basal ganglia. Histamine modulation of DA effects is increasingly appreciated, but the mechanisms underlying this modulation remain unclear; the consequences of modest DA elevation in the context of profound HA deficiency are difficult to predict, but understanding them in the Hdc knockout mouse may provide generalizable insights into the pathophysiology of TS. Here we characterized signaling pathways in striatal cells in this model system, at baseline and after amphetamine challenge. In vivo microdialysis confirms elevated DA in Hdc-KO mice. We find dephosphorylation of Akt and its target GSK3ß and activation of the MAPK signaling cascade and its target rpS6; these are characteristic of the effects of DA on D2- and D1-expressing striatal neurons, respectively. Strikingly, there is no alteration in mTOR signaling, which can be regulated by DA in both cell types. These cellular effects help elucidate striatal signaling abnormalities in a uniquely validated mouse model of TS and move towards the identification of new potential therapeutic targets for tic disorders.


Assuntos
Corpo Estriado/metabolismo , Dopamina/metabolismo , Histidina Descarboxilase/genética , Transdução de Sinais/efeitos dos fármacos , Síndrome de Tourette/metabolismo , Anfetamina/farmacologia , Animais , Corpo Estriado/efeitos dos fármacos , Modelos Animais de Doenças , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Proteína Oncogênica v-akt/metabolismo , Fosforilação , Proteína S6 Ribossômica/metabolismo , Serina-Treonina Quinases TOR/metabolismo
17.
Neuron ; 81(1): 77-90, 2014 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-24411733

RESUMO

Tourette syndrome (TS) is characterized by tics, sensorimotor gating deficiencies, and abnormalities of cortico-basal ganglia circuits. A mutation in histidine decarboxylase (Hdc), the key enzyme for the biosynthesis of histamine (HA), has been implicated as a rare genetic cause. Hdc knockout mice exhibited potentiated tic-like stereotypies, recapitulating core phenomenology of TS; these were mitigated by the dopamine (DA) D2 antagonist haloperidol, a proven pharmacotherapy, and by HA infusion into the brain. Prepulse inhibition was impaired in both mice and humans carrying Hdc mutations. HA infusion reduced striatal DA levels; in Hdc knockout mice, striatal DA was increased and the DA-regulated immediate early gene Fos was upregulated. DA D2/D3 receptor binding was altered both in mice and in humans carrying the Hdc mutation. These data confirm histidine decarboxylase deficiency as a rare cause of TS and identify HA-DA interactions in the basal ganglia as an important locus of pathology.


Assuntos
Encéfalo/enzimologia , Histidina Descarboxilase/deficiência , Mutação/genética , Síndrome de Tourette/enzimologia , Síndrome de Tourette/genética , Adolescente , Adulto , Anfetamina , Animais , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Criança , Agonistas de Dopamina/uso terapêutico , Antagonistas de Dopamina/farmacocinética , Comportamento Exploratório/fisiologia , Feminino , Histidina Descarboxilase/genética , Humanos , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Oxazinas , Racloprida/farmacocinética , Cintilografia , Comportamento Estereotipado/efeitos dos fármacos , Comportamento Estereotipado/fisiologia , Fatores de Tempo , Síndrome de Tourette/diagnóstico por imagem , Síndrome de Tourette/fisiopatologia , Triptofano/genética , Adulto Jovem
18.
Behav Brain Res ; 257: 156-65, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24095879

RESUMO

Blockade of N-methyl-d-aspartate receptor (NMDA) by the noncompetitive NMDA receptor (NMDAR) antagonist MK-801 produces behavioral abnormalities and alterations in prefrontal cortex (PFC) functioning. Due to the critical role of the PFC in operant conditioning task learning, we evaluated the effects of acute, repeated postnatal injections of MK-801 (0.1mg/kg) on learning performance. We injected Long-Evans rats i.p. with MK-801 (0.1mg/kg) using three different administration schedules: injection 40 min before beginning the task (during) (n=12); injection twice daily for six consecutive days prior to beginning the experimental procedures (prior) (n=12); or twice daily subcutaneous injections from postnatal day 7 to 11 (postnatal) (n=12). Next, we orally administered risperidone (serotonin receptor 2A and dopamine receptor 2 antagonist, 1mg/kg) or buspirone (serotonin receptor 1A partial agonist, 10mg/kg) to animals treated with the MK-801 schedule described above. The postnatal and prior administration schedules produced severe learning deficits, whereas injection of MK-801 just before training sessions had only mild effects on acquisition of an operant conditioning. Risperidone was able to reverse the detrimental effect of MK-801 in the animals that were treated with MK-801 during and prior training sessions. In contrast, buspirone was only effective at mitigating the cognitive deficits induced by MK-801 when administered during the training procedures. The data demonstrates that NMDA antagonism disrupts basic mechanisms of learning in a simple PFC-mediated operant conditioning task, and that buspirone and risperidone failed to attenuate the learning deficits when NMDA neurotransmission was blocked in the early stages of the postnatal period.


Assuntos
Buspirona/uso terapêutico , Maleato de Dizocilpina/toxicidade , Antagonistas de Aminoácidos Excitatórios/toxicidade , Deficiências da Aprendizagem/induzido quimicamente , Deficiências da Aprendizagem/tratamento farmacológico , Risperidona/uso terapêutico , Serotoninérgicos/uso terapêutico , Animais , Buspirona/farmacologia , Condicionamento Operante/efeitos dos fármacos , Esquema de Medicação , Interações Medicamentosas , Masculino , Ratos , Ratos Long-Evans , Tempo de Reação/efeitos dos fármacos , Risperidona/farmacologia , Serotoninérgicos/farmacologia , Estatísticas não Paramétricas
19.
Life Sci ; 92(10): 525-32, 2013 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-23369745

RESUMO

Two decades ago, it was hypothesized that antidepressants could alter the course of neoplastic diseases. However, contradictory findings indicated that antidepressants could either have carcinogenic properties or improve the disease outcome. Intriguingly, controversial results were reported on the action of antidepressant drugs on immune function. Further hypotheses proposed that antidepressants could indirectly affect the cancer prognosis through the modulation of antitumor activity. Here we review the literature in order to elucidate the influence of antidepressants on cancer and immunity.


Assuntos
Antidepressivos/efeitos adversos , Antidepressivos/imunologia , Antidepressivos/metabolismo , Imunidade Celular/efeitos dos fármacos , Metástase Neoplásica , Neoplasias/induzido quimicamente , Neoplasias/epidemiologia , Transdução de Sinais/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Humanos , Modelos Biológicos , Nitrosação/efeitos dos fármacos , Oxirredução/efeitos dos fármacos , Fatores de Risco , Transdução de Sinais/imunologia , Células Tumorais Cultivadas/efeitos dos fármacos
20.
Eur J Pharmacol ; 659(2-3): 265-72, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21497159

RESUMO

Antidepressants have a controversial role with regard to their influence on cancer and immunity. Recently, we showed that fluoxetine administration induces an enhancement of the T-cell mediated immunity in naïve mice, resulting in the inhibition of tumor growth. Here we studied the effects of fluoxetine on lymphoma proliferation/apoptosis and immunity in tumor bearing-mice. We found an increase of apoptotic cells (active Caspase-3(+)) and a decrease of proliferative cells (PCNA(+)) in tumors growing in fluoxetine-treated animals. In addition, differential gene expressions of cell cycle and death markers were observed. Cyclins D3, E and B were reduced in tumors from animals treated with fluoxetine, whereas the tumor suppressor p53 and the cell cycle inhibitors p15/INK4B, p16/INK4A and p27/Kip1 were increased. Besides, the expression of the antiapoptotic factor Bcl-2 and the proapoptotic factor Bad were lower and higher respectively in these animals. These changes were accompanied by increased IFN-γ and TNF-α levels as well as augmented circulating CD8(+) T lymphocytes in tumor-bearing mice treated with the antidepressant. Therefore, we propose that the up-regulation of T-cell mediated antitumor immunity may be contributing to the alterations of tumor cell proliferation and apoptosis thus resulting in the inhibition of tumor progression.


Assuntos
Apoptose/efeitos dos fármacos , Fluoxetina/administração & dosagem , Fluoxetina/farmacologia , Linfoma/imunologia , Linfoma/patologia , Linfócitos T/imunologia , Regulação para Cima/efeitos dos fármacos , Administração Oral , Animais , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Linfoma/diagnóstico , Linfoma/genética , Camundongos , Camundongos Endogâmicos BALB C , Prognóstico , Inibidores Seletivos de Recaptação de Serotonina/administração & dosagem , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Linfócitos T/efeitos dos fármacos , Regulação para Cima/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...