Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 149(5): 1152-63, 2012 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-22632977

RESUMO

Our understanding of current treatments for depression, and the development of more specific therapies, is limited by the complexity of the circuits controlling mood and the distributed actions of antidepressants. Although the therapeutic efficacy of serotonin-specific reuptake inhibitors (SSRIs) is correlated with increases in cortical activity, the cell types crucial for their action remain unknown. Here we employ bacTRAP translational profiling to show that layer 5 corticostriatal pyramidal cells expressing p11 (S100a10) are strongly and specifically responsive to chronic antidepressant treatment. This response requires p11 and includes the specific induction of Htr4 expression. Cortex-specific deletion of p11 abolishes behavioral responses to SSRIs, but does not lead to increased depression-like behaviors. Our data identify corticostriatal projection neurons as critical for the response to antidepressants, and suggest that the regulation of serotonergic tone in this single cell type plays a pivotal role in antidepressant therapy.


Assuntos
Antidepressivos/metabolismo , Depressão/tratamento farmacológico , Neurônios/citologia , Córtex Pré-Frontal/citologia , Inibidores Seletivos de Recaptação de Serotonina/metabolismo , Animais , Antidepressivos/farmacologia , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Mutação , Córtex Pré-Frontal/metabolismo , Células Piramidais/metabolismo , Proteínas S100/genética , Proteínas S100/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacologia
2.
Mol Psychiatry ; 26(6): 2334-2349, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33441982

RESUMO

Serotonin receptor 4 (5-HT4R) plays an important role in regulating mood, anxiety, and cognition, and drugs that activate this receptor have fast-acting antidepressant (AD)-like effects in preclinical models. However, 5-HT4R is widely expressed throughout the central nervous system (CNS) and periphery, making it difficult to pinpoint the cell types and circuits underlying its effects. Therefore, we generated a Cre-dependent 5-HT4R knockout mouse line to dissect the function of 5-HT4R in specific brain regions and cell types. We show that the loss of functional 5-HT4R specifically from excitatory neurons of hippocampus led to robust AD-like behavioral responses and an elevation in baseline anxiety. 5-HT4R was necessary to maintain the proper excitability of dentate gyrus (DG) granule cells and cell type-specific molecular profiling revealed a dysregulation of genes necessary for normal neural function and plasticity in cells lacking 5-HT4R. These adaptations were accompanied by an increase in the number of immature neurons in ventral, but not dorsal, dentate gyrus, indicating a broad impact of 5-HT4R loss on the local cellular environment. This study is the first to use conditional genetic targeting to demonstrate a direct role for hippocampal 5-HT4R signaling in modulating mood and anxiety. Our findings also underscore the need for cell type-based approaches to elucidate the complex action of neuromodulatory systems on distinct neural circuits.


Assuntos
Ansiedade , Hipocampo , Animais , Giro Denteado/metabolismo , Hipocampo/metabolismo , Camundongos , Neurônios/metabolismo , Receptores de Serotonina , Receptores 5-HT4 de Serotonina/genética , Receptores 5-HT4 de Serotonina/metabolismo
3.
Hum Mol Genet ; 26(1): 192-209, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-28082376

RESUMO

Local mRNA translation in growing axons allows for rapid and precise regulation of protein expression in response to extrinsic stimuli. However, the role of local translation in mature CNS axons is unknown. Such a mechanism requires the presence of translational machinery and associated mRNAs in circuit-integrated brain axons. Here we use a combination of genetic, quantitative imaging and super-resolution microscopy approaches to show that mature axons in the mammalian brain contain ribosomes, the translational regulator FMRP and a subset of FMRP mRNA targets. This axonal translational machinery is associated with Fragile X granules (FXGs), which are restricted to axons in a stereotyped subset of brain circuits. FXGs and associated axonal translational machinery are present in hippocampus in humans as old as 57 years. This FXG-associated axonal translational machinery is present in adult rats, even when adult neurogenesis is blocked. In contrast, in mouse this machinery is only observed in juvenile hippocampal axons. This differential developmental expression was specific to the hippocampus, as both mice and rats exhibit FXGs in mature axons in the adult olfactory system. Experiments in Fmr1 null mice show that FMRP regulates axonal protein expression but is not required for axonal transport of ribosomes or its target mRNAs. Axonal translational machinery is thus a feature of adult CNS neurons. Regulation of this machinery by FMRP could support complex behaviours in humans throughout life.


Assuntos
Axônios/patologia , Encéfalo/patologia , Grânulos Citoplasmáticos/metabolismo , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Síndrome do Cromossomo X Frágil/patologia , RNA Mensageiro/metabolismo , Ribossomos/patologia , Adulto , Animais , Axônios/metabolismo , Encéfalo/metabolismo , Grânulos Citoplasmáticos/patologia , Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/metabolismo , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Neurogênese/genética , Neurônios/metabolismo , Neurônios/patologia , Ratos , Ratos Sprague-Dawley , Ribossomos/metabolismo
4.
Nat Rev Neurosci ; 14(10): 673-80, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24002251

RESUMO

Studies of the multifunctional protein p11 (also known as S100A10) are shedding light on the molecular and cellular mechanisms underlying depression. Here, we review data implicating p11 in both the amplification of serotonergic signalling and the regulation of gene transcription. We summarize studies demonstrating that levels of p11 are regulated in depression and by antidepressant regimens and, conversely, that p11 regulates depression-like behaviours and/or responses to antidepressants. Current and future studies of p11 may provide a molecular and cellular framework for the development of novel antidepressant therapies.


Assuntos
Anexina A2/metabolismo , Antidepressivos/uso terapêutico , Depressão/tratamento farmacológico , Depressão/metabolismo , Proteínas S100/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Depressão/patologia , Modelos Animais de Doenças , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos
5.
Proc Natl Acad Sci U S A ; 112(31): 9745-50, 2015 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-26195764

RESUMO

Adult neurogenesis in the hippocampus subgranular zone is associated with the etiology and treatment efficiency of depression. Factors that affect adult hippocampal neurogenesis have been shown to contribute to the neuropathology of depression. Glutamate, the major excitatory neurotransmitter, plays a critical role in different aspects of neurogenesis. Of the eight metabotropic glutamate receptors (mGluRs), mGluR5 is the most highly expressed in neural stem cells. We previously identified Norbin as a positive regulator of mGluR5 and showed that its expression promotes neurite outgrowth. In this study, we investigated the role of Norbin in adult neurogenesis and depressive-like behaviors using Norbin-deficient mice. We found that Norbin deletion significantly reduced hippocampal neurogenesis; specifically, the loss of Norbin impaired the proliferation and maturation of newborn neurons without affecting cell-fate specification of neural stem cells/neural progenitor cells (NSCs/NPCs). Norbin is highly expressed in the granular neurons in the dentate gyrus of the hippocampus, but it is undetectable in NSCs/NPCs or immature neurons, suggesting that the effect of Norbin on neurogenesis is likely caused by a nonautonomous niche effect. In support of this hypothesis, we found that the expression of a cell-cell contact gene, Desmoplakin, is greatly reduced in Norbin-deletion mice. Moreover, Norbin-KO mice show an increased immobility in the forced-swim test and the tail-suspension test and reduced sucrose preference compared with wild-type controls. Taken together, these results show that Norbin is a regulator of adult hippocampal neurogenesis and that its deletion causes depressive-like behaviors.


Assuntos
Comportamento Animal , Depressão/metabolismo , Depressão/patologia , Deleção de Genes , Hipocampo/patologia , Neurogênese , Neuropeptídeos/metabolismo , Envelhecimento/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Comunicação Celular , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Giro Denteado/metabolismo , Depressão/fisiopatologia , Hipocampo/fisiopatologia , Camundongos Knockout , Atividade Motora , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/patologia , Fenótipo , Receptor de Glutamato Metabotrópico 5/metabolismo
6.
Proc Natl Acad Sci U S A ; 109(28): 11360-5, 2012 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-22733786

RESUMO

A large number of studies have demonstrated that the nucleus accumbens (NAC) is a critical site in the neuronal circuits controlling reward responses, motivation, and mood, but the neuronal cell type(s) underlying these processes are not yet known. Identification of the neuronal cell types that regulate depression-like states will guide us in understanding the biological basis of mood and its regulation by diseases like major depressive disorder. Taking advantage of recent findings demonstrating that the serotonin receptor chaperone, p11, is an important molecular regulator of depression-like states, here we identify cholinergic interneurons (CINs) as a primary site of action for p11 in the NAC. Depression-like behavior is observed in mice after decrease of p11 levels in NAC CINs. This phenotype is recapitulated by silencing neuronal transmission in these cells, demonstrating that accumbal cholinergic neuronal activity regulates depression-like behaviors and suggesting that accumbal CIN activity is crucial for the regulation of mood and motivation.


Assuntos
Anexina A2/metabolismo , Depressão/fisiopatologia , Interneurônios/metabolismo , Núcleo Accumbens/metabolismo , Proteínas S100/metabolismo , Acetilcolina/metabolismo , Animais , Antidepressivos/farmacologia , Comportamento Animal , Depressão/metabolismo , Imuno-Histoquímica/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Chaperonas Moleculares/metabolismo , Neurônios/metabolismo , Neurotransmissores/metabolismo , Fenótipo , Receptores Colinérgicos/metabolismo
7.
Proc Natl Acad Sci U S A ; 108(22): 9262-7, 2011 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-21518864

RESUMO

Antiinflammatory drugs achieve their therapeutic actions at least in part by regulation of cytokine formation. A "cytokine hypothesis" of depression is supported by the observation that depressed individuals have elevated plasma levels of certain cytokines compared with healthy controls. Here we investigated a possible interaction between antidepressant agents and antiinflammatory agents on antidepressant-induced behaviors and on p11, a biochemical marker of depressive-like states and antidepressant responses. We found that widely used antiinflammatory drugs antagonize both biochemical and behavioral responses to selective serotonin reuptake inhibitors (SSRIs). In contrast to the levels detected in serum, we found that frontal cortical levels of certain cytokines (e.g., TNFα and IFNγ) were increased by serotonergic antidepressants and that these effects were inhibited by antiinflammatory agents. The antagonistic effect of antiinflammatory agents on antidepressant-induced behaviors was confirmed by analysis of a dataset from a large-scale real-world human study, "sequenced treatment alternatives to relieve depression" (STAR*D), underscoring the clinical significance of our findings. Our data indicate that clinicians should carefully balance the therapeutic benefits of antiinflammatory agents versus the potentially negative consequences of antagonizing the therapeutic efficacy of antidepressant agents in patients suffering from depression.


Assuntos
Anexina A2/metabolismo , Anti-Inflamatórios/farmacologia , Antidepressivos/farmacologia , Transtorno Depressivo/tratamento farmacológico , Interações Medicamentosas , Proteínas S100/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Citalopram/farmacologia , Citocinas/metabolismo , Fluoxetina/farmacologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
8.
Front Neurosci ; 18: 1353131, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38389788

RESUMO

Background: Post-traumatic stress disorder (PTSD) is a highly prevalent psychiatric disorder that can become chronic and debilitating when left untreated. Available pharmacotherapies are limited, take weeks to show modest benefit and remain ineffective for up to 40% of patients. Methylone is currently in clinical development for the treatment of PTSD. Preclinical studies show rapid, robust and long-lasting antidepressant-like and anxiolytic effects. The mechanism of action underlying these effects is not yet fully understood. This study investigated the downstream gene expression changes and signaling pathways affected by methylone in key brain areas linked to PTSD and MDD. It also sought to determine whether neuroplasticity-related genes were involved. We compared effects of methylone with MDMA to explore similarities and differences in their brain effects because MDMA-assisted psychotherapy has recently shown benefit in clinical trials for PTSD and methylone is a structural analog of MDMA. Methods: Monoamine binding, uptake and release studies were performed and a high-throughput-screen evaluated agonist/antagonist activities at 168 GPCRs in vitro. We used RNA sequencing (RNA-seq) to probe drug-induced gene expression changes in the amygdala and frontal cortex, two brain areas responsible for emotional learning that are affected by PTSD and MDD. Rats were treated with methylone or MDMA (both 10 mg/kg, IP), and their responses were compared with controls. We performed functional enrichment analysis to identify which pathways were regulated by methylone and/or MDMA. We confirmed changes in gene expression using immunohistochemistry. Results: Methylone, a monoamine uptake inhibitor and releaser, demonstrated no off-target effects at 168 GPCRs, unlike MDMA, which showed activity at 5HT2A and 5HT2C receptors. RNA-seq results revealed significant regulation of myelin-related genes in the amygdala, confirmed by immunohistochemistry. In the frontal cortex, methylone significantly upregulated genes implicated in neuroplasticity. Conclusion: Results suggest that (1) methylone is a rapid-acting neuroplastogen that affects key brain substrates for PTSD and MDD and that (2) methylone appears to exhibit higher specificity and fewer off-target effects than MDMA. Together, these results are consistent with the reported clinical experiences of methylone and MDMA and bolster the potential use of methylone in the treatment of PTSD and, potentially, other neuropsychiatric disorders.

9.
Proc Natl Acad Sci U S A ; 107(9): 4401-6, 2010 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-20145109

RESUMO

Dopamine neurotransmission controls motor and perseverative behavior, is mediated by protein phosphorylation, and may be perturbed in disorders of attention and hyperactivity. To assess the role of casein kinase I (CK1) in the regulation of dopamine signaling, we generated a genetically modified mouse line that overexpresses CK1delta (CK1delta OE) specifically in the forebrain. Overexpression was confirmed both at the mRNA and at the protein levels. Under basal conditions, CK1delta OE mice exhibited horizontal and vertical hyperactivity, reduced anxiety, and nesting behavior deficiencies. The CK1delta OE mice also presented paradoxical responses to dopamine receptor stimulation, showing hypoactivity following injection of d-amphetamine or methylphenidate, indicating that CK1 activity has a profound effect on dopamine signaling in vivo. Interestingly, CK1delta overexpression led to significantly reduced D1R and D2R dopamine receptor levels. All together, under basal conditions and in response to drug stimulation, the behavioral phenotype of CK1delta OE mice is reminiscent of the symptoms and drug responses observed in attention-deficit/hyperactivity disorder and therefore the CK1delta OE mice appear to be a model for this disorder.


Assuntos
Transtorno do Deficit de Atenção com Hiperatividade/metabolismo , Caseína Quinase Idelta/metabolismo , Regulação para Baixo , Locomoção , Prosencéfalo/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Anfetamina/farmacologia , Animais , Transtorno do Deficit de Atenção com Hiperatividade/enzimologia , Comportamento Animal/efeitos dos fármacos , Maleato de Dizocilpina/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Metilfenidato/farmacologia , Camundongos
10.
J Med Chem ; 66(14): 9954-9971, 2023 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-37436942

RESUMO

Chemotherapy-induced peripheral neuropathy (CIPN) is a major unmet medical need with limited treatment options. Despite different mechanisms of action, diverse chemotherapeutics can cause CIPN through a converged pathway─an active axon degeneration program that engages the dual leucine zipper kinase (DLK). DLK is a neuronally enriched kinase upstream in the MAPK-JNK cascade, and while it is dormant under physiological conditions, DLK mediates a core mechanism for neuronal injury response under stress conditions, making it an attractive target for treatment of neuronal injury and neurodegenerative diseases. We have developed potent, selective, brain penetrant DLK inhibitors with excellent PK and activity in mouse models of CIPN. Lead compound IACS-52825 (22) showed strongly effective reversal of mechanical allodynia in a mouse model of CIPN and was advanced into preclinical development.


Assuntos
Antineoplásicos , Doenças do Sistema Nervoso Periférico , Camundongos , Animais , Neurônios , Sistema de Sinalização das MAP Quinases , Encéfalo/metabolismo , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Doenças do Sistema Nervoso Periférico/tratamento farmacológico , Antineoplásicos/efeitos adversos , MAP Quinase Quinase Quinases
11.
Front Psychiatry ; 13: 1041277, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36704743

RESUMO

Introduction: Selective serotonin reuptake inhibitor (SSRI) antidepressants represent first-line pharmacological treatment for a variety of neuropsychiatric illnesses, including major depressive disorder (MDD), anxiety, and post-traumatic stress disorder (PTSD), which show high rates of comorbidity. SSRIs have a delayed onset of action. Most patients do not show significant effects until 4-8 weeks of continuous treatment, have impairing side effects and as many as 40% of patients do not respond. Methylone (3,4-methylenedioxy-N-methylcathinone; MDMC, ßk-MDMA, M1) is a rapid-acting entactogen that showed significant benefit in a clinical case series of PTSD patients and was well-tolerated in two Phase 1 studies of healthy volunteers. Based on these early observations in humans, in the current study we tested the hypothesis that methylone has antidepressant-like and anxiolytic effects in preclinical tests. Methods: For all studies, 6-8-week-old male Sprague Dawley rats (N = 6-16) were used. We employed the Forced Swim Test (FST), a classic and widely used screen for antidepressants, to explore the effects of methylone and to probe dose-response relationships, durability of effect, and potential interactions with combined SSRI treatment. We compared the effect of methylone with the prototypical SSRI fluoxetine. Results: Three doses of fluoxetine (10 mg/kg) given within 24 h before FST testing caused a 50% reduction in immobility compared with controls that lasted less than 24 h. In contrast, a single dose of methylone (5-30 mg/kg) administered 30 min prior to testing produced a rapid, robust, and durable antidepressant-like response in the FST, greater in magnitude than fluoxetine. Immobility was reduced by nearly 95% vs. controls and effects persisted for at least 72 h after a single dose (15 mg/kg). Effects on swimming and climbing behavior in the FST, which reflect serotonergic and noradrenergic activity, respectively, were consistent with studies showing that methylone is less serotoninergic than MDMA. Fluoxetine pretreatment did not change methylone's antidepressant-like effect in the FST, suggesting the possibility that the two may be co-administered. In addition, methylone (5-30 mg/kg) exhibited anxiolytic effects measured as increased time spent in the center of an open field. Discussion: Taken together, and consistent with initial clinical findings, our study suggests that methylone may have potential for treating depression and anxiety.

12.
Proc Natl Acad Sci U S A ; 105(32): 11352-7, 2008 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-18682560

RESUMO

All classes of antidepressants increase hippocampal cell proliferation and neurogenesis, which contributes, in part, to the behavioral actions of these treatments. Among antidepressant treatments, electroconvulsive seizure (ECS) is the most robust stimulator of hippocampal cell proliferation and the most efficacious treatment for depression, but the cellular mechanisms underlying the actions of ECS are unknown. To address this question, we investigated the effect of ECS on proliferation of neural stem-like and/or progenitor cells in the subgranular zone of rat dentate gyrus. We define the neural differentiation cascade from stem-like cells to early neural progenitors (also referred to as quiescent and amplifying neural progenitors, respectively) by coexpression of selective cellular and mitotic activity markers. We find that at an early mitotic phase ECS increases the proliferation of quiescent progenitors and then at a later phase increases the proliferation of amplifying progenitors. We further demonstrate that vascular endothelial growth factor (VEGF) signaling is necessary for ECS induction of quiescent neural progenitor cell proliferation and is sufficient to produce this effect. These findings demonstrate that ECS and subsequent induction of VEGF stimulates the proliferation of neural stem-like cells and neural progenitor cells, thereby accounting for the superior neurogenic actions of ECS compared with chemical antidepressants.


Assuntos
Proliferação de Células , Giro Denteado/metabolismo , Eletrochoque , Neurônios/metabolismo , Células-Tronco/metabolismo , Fator A de Crescimento do Endotélio Vascular/farmacologia , Animais , Antígenos de Diferenciação/biossíntese , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Giro Denteado/patologia , Depressão/terapia , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Neurônios/patologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/patologia , Fator A de Crescimento do Endotélio Vascular/biossíntese
13.
J Neurosci ; 29(6): 1937-46, 2009 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-19211900

RESUMO

p11 (S100A10), a member of a large family of S100 proteins, interacts with serotonin receptor 1B (5-HTR1B), modulates 5-HT1B receptor signal transduction, and is required for antidepressant responses to activation of this receptor. In the current study, we investigated the specificity of the interaction between 5-HTR1B and p11 by screening brain-expressed S100 proteins against serotonin and noradrenergic receptors. The data indicate that p11 is unique among its family members for its interactions with defined serotonin receptors. We identify a novel p11-interacting receptor (5-HTR4) and characterize the interaction between p11 and 5-HTR4, demonstrating that (1) p11 and 5-HTR4 mRNA and protein are coexpressed in brain regions that are relevant for major depression, (2) p11 increases 5-HTR4 surface expression and facilitates 5-HTR4 signaling, and (3) p11 is required for the behavioral antidepressant responses to 5-HTR4 stimulation in vivo. The essential role played by p11 in modulating signaling through 5-HT4 as well as 5-HT1B receptors supports the concept that this protein may be a key determinant of vulnerability to depression.


Assuntos
Anexina A2/fisiologia , Apoptose/fisiologia , Membrana Celular/fisiologia , Comportamento Exploratório/fisiologia , Receptores 5-HT4 de Serotonina/fisiologia , Receptores de Serotonina/fisiologia , Proteínas S100/fisiologia , Animais , Células COS , Células Cultivadas , Chlorocebus aethiops , Transtorno Depressivo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
14.
Curr Opin Pharmacol ; 8(1): 14-9, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18061540

RESUMO

Antidepressants are among the most widely prescribed drugs, however the mechanism underlying their therapeutic efficacy is not known. Neurotrophic factors represent a promising class of targets for antidepressant treatments. We recently characterized a role for vascular endothelial growth factor (VEGF) in cellular and behavioral antidepressant responses. VEGF is a potent mitogen and survival factor for endothelial cells (ECs) and neurons, and modulator of synaptic transmission. Because VEGF has been implicated in a variety of diseases, understanding the molecular and cellular specificity of antidepressant-induced VEGF will be crucial to determine its potential as a therapeutic target in depression.


Assuntos
Antidepressivos/farmacologia , Depressão/tratamento farmacológico , Fator A de Crescimento do Endotélio Vascular/fisiologia , Humanos , Neovascularização Fisiológica , Neurônios/fisiologia
15.
Front Behav Neurosci ; 13: 141, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31293404

RESUMO

Fragile X syndrome (FXS) is the most common inherited form of intellectual disability and autism. FXS is also accompanied by attention problems, hyperactivity, anxiety, aggression, poor sleep, repetitive behaviors, and self-injury. Recent work supports the role of γ-aminobutyric-acid (GABA), the primary inhibitory neurotransmitter in the brain, in mediating symptoms of FXS. Deficits in GABA machinery have been observed in a mouse model of FXS, including a loss of tonic inhibition in the amygdala, which is mediated by extrasynaptic GABAA receptors. Humans with FXS also show reduced GABAA receptor availability. Here, we sought to evaluate the potential of gaboxadol (also called OV101 and THIP), a selective and potent agonist for delta-subunit-containing extrasynaptic GABAA receptors (dSEGA), as a therapeutic agent for FXS by assessing its ability to normalize aberrant behaviors in a relatively uncharacterized mouse model of FXS (Fmr1 KO2 mice). Four behavioral domains (hyperactivity, anxiety, aggression, and repetitive behaviors) were probed using a battery of behavioral assays. The results showed that Fmr1 KO2 mice were hyperactive, had abnormal anxiety-like behavior, were more irritable and aggressive, and had an increased frequency of repetitive behaviors compared to wild-type (WT) littermates, which are all behavioral deficits reminiscent of individuals with FXS. Treatment with gaboxadol normalized all of the aberrant behaviors observed in Fmr1 KO2 mice back to WT levels, providing evidence of its potential benefit for treating FXS. We show that the potentiation of extrasynaptic GABA receptors alone, by gaboxadol, is sufficient to normalize numerous behavioral deficits in the FXS model using endpoints that are directly translatable to the clinical presentation of FXS. Taken together, these data support the future evaluation of gaboxadol in individuals with FXS, particularly with regard to symptoms of hyperactivity, anxiety, irritability, aggression, and repetitive behaviors.

16.
Eur J Neurosci ; 27(6): 1485-93, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18336568

RESUMO

Ongoing neurogenesis in the adult hippocampus is thought to play a role in learning and memory processes, and in response to antidepressant treatments. Low doses of irradiation (IRR) produce a significant long-lasting inhibitory effect on hippocampal neurogenesis that correlates with long-lasting behavioral deficits. Here we report that electroconvulsive seizure (ECS), which robustly increases adult neurogenesis in naïve animals, also reverses the disruption of neurogenesis produced by IRR exposure. Moreover, we find that vascular endothelial growth factor (VEGF) is an essential mediator of this effect. Expression of VEGF in the granule cell layer (GCL) of the hippocampus is decreased by IRR, and ECS administration reverses this deficit in VEGF. There is a corresponding alteration in the number of endothelial cells, which express VEGF, in the hippocampal GCL following IRR and ECS. We also find that blockade of VEGF signaling attenuates ECS-induced proliferation, and VEGF infusion partially restores proliferation in irradiated animals. To examine the functional consequences of IRR and ECS on neurogenesis, hippocampus-dependent contextual fear conditioning was assessed. We found that following disruption by IRR, ECS restores contextual learning to baseline levels at time points consistent with its effects on neurogenesis. These findings demonstrate that ECS, in part via induction of VEGF, can reverse long-term neurogenesis deficits resulting from IRR, and that these effects have functional consequences on hippocampus-dependent fear memory.


Assuntos
Diferenciação Celular/efeitos da radiação , Eletrochoque , Medo/efeitos da radiação , Raios gama , Hipocampo/citologia , Hipocampo/efeitos da radiação , Memória/efeitos da radiação , Neurônios/efeitos da radiação , Animais , Diferenciação Celular/fisiologia , Eletrochoque/métodos , Medo/fisiologia , Hipocampo/fisiologia , Masculino , Memória/fisiologia , Neurônios/citologia , Ratos , Ratos Sprague-Dawley , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/fisiologia
17.
Mol Brain ; 8: 59, 2015 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-26438564

RESUMO

BACKGROUND: Non-steroidal anti-inflammatory drugs such as indomethacin are widely used to treat inflammatory diseases and manage pain, fever and inflammation in several conditions, including neuropsychiatric disorders. Although they predominantly function by inhibiting cyclooxygenase (COX) activity, important COX-independent actions also occur. These actions could be responsible for the adverse side effects associated with chronic and/or high dose usage of this popular drug class. RESULTS: We examined gene regulation in the hippocampus after peripheral administration of indomethacin by employing a microarray approach. Secondary confirmation and the brain expression pattern of regulated genes was examined by in situ hybridization and immunohistochemistry. Transglutaminase 2, serum glucocorticoid inducible kinase, Inhibitor of NF-kappa B and vascular endothelial growth factor were among genes that were prominently upregulated, while G-protein coupled receptor 56 and neuropeptide Y were among genes that were downregulated by indomethacin. Co-localization studies using blood vessel markers revealed that transglutaminase 2 was induced specifically in brain vasculature. CONCLUSIONS: The data demonstrate that COX-inhibitors can differentially regulate gene transcription in multiple, functionally distinctly cell types in the brain. The results provide additional insight into the molecular actions of COX-inhibitors and indicate that their effects on vasculature could influence cerebral blood flow mechanisms.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/metabolismo , Indometacina/farmacologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Celecoxib/farmacologia , Ciclo-Oxigenase 2/metabolismo , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/metabolismo , Hipocampo/irrigação sanguínea , Hipocampo/efeitos dos fármacos , Imuno-Histoquímica , Hibridização In Situ , Óxido Nítrico Sintase Tipo III/metabolismo , Proteína 2 Glutamina gama-Glutamiltransferase , Transporte Proteico/efeitos dos fármacos , Ratos , Transglutaminases/genética , Transglutaminases/metabolismo
18.
Biol Psychiatry ; 76(10): 794-801, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24725970

RESUMO

BACKGROUND: The high rate of comorbidity between depression and cocaine addiction suggests shared molecular mechanisms and anatomical pathways. Limbic structures, such as the nucleus accumbens (NAc), play a crucial role in both disorders, yet how different cell types within these structures contribute to the pathogenesis remains elusive. Downregulation of p11 (S100A10), specifically in the NAc, elicits depressive-like behaviors in mice, but its role in drug addiction is unknown. METHODS: We combined mouse genetics and viral strategies to determine how the titration of p11 levels within the entire NAc affects the rewarding actions of cocaine on behavior (six to eight mice per group) and molecular correlates (three experiments, five to eight mice per group). Finally, the manipulation of p11 expression in distinct NAc dopaminoceptive neuronal subsets distinguished cell-type specific effects of p11 on cocaine reward (five to eight mice per group). RESULTS: We demonstrated that p11 knockout mice have enhanced cocaine conditioned place preference, which is reproduced by the focal downregulation of p11 in the NAc of wild-type mice. In wild-type mice, cocaine reduced p11 expression in the NAc, while p11 overexpression exclusively in the NAc reduced cocaine conditioned place preference. Finally, we identified dopamine receptor-1 expressing medium spiny neurons as key mediators of the effects of p11 on cocaine reward. CONCLUSIONS: Our data provide evidence that disruption of p11 homeostasis in the NAc, particularly in dopamine receptor-1 expressing medium spiny neurons, may underlie pathophysiological mechanisms of cocaine rewarding action. Treatments to counter maladaptation of p11 levels may provide novel therapeutic opportunities for cocaine addiction.


Assuntos
Anexina A2/fisiologia , Cocaína/farmacologia , Condicionamento Psicológico/fisiologia , Neurônios/fisiologia , Núcleo Accumbens/citologia , Recompensa , Proteínas S100/fisiologia , Animais , Anexina A2/metabolismo , Condicionamento Psicológico/efeitos dos fármacos , Regulação para Baixo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/fisiologia , Receptores de Dopamina D1/metabolismo , Proteínas S100/metabolismo
19.
Dev Neurobiol ; 72(6): 937-42, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21976455

RESUMO

Neurogenesis continues in the dentate gyrus of the hippocampus throughout life in mammals. This process is influenced by daily activities such as exercise, learning, and stress and may contribute to certain forms of hippocampus-dependent learning and memory. Adult hippocampal neurogenesis is also subject to regulation by antidepressant treatment, including chronic treatment with antidepressant drugs or electroconvulsive seizure (ECS) therapy. Here we investigated how the connectivity of newborn and mature granule cells is influenced by ECS administration in rats. Specifically, we examined the dendritic spine morphology of newborn and mature granule cells in rats and found that ECS administration promoted the maturation of dendritic spines in newborn cells and increased spine density in mature cells. These changes could potentially lead to alteration in dentate circuitry and may partially contribute to the functional effects of ECS.


Assuntos
Espinhas Dendríticas/fisiologia , Giro Denteado/fisiologia , Neurogênese/fisiologia , Neurônios/fisiologia , Convulsões/fisiopatologia , Animais , Giro Denteado/citologia , Giro Denteado/fisiopatologia , Eletrochoque , Masculino , Neurônios/citologia , Ratos , Ratos Sprague-Dawley
20.
Neuropharmacology ; 61(3): 442-50, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21300076

RESUMO

p11 is an adaptor protein which binds to serotonin 5-HT(1B) receptors and 5-HT(4) receptors and regulates their localization at the cell surface. In the present study, we examined to what extent p11 containing neurons co-expressed 5-HT(1B)R and/or 5-HT(4)R in cerebral cortex, hippocampus, cerebellum and caudate-putamen. A triple-labeling immunohistochemical approach was taken using antibodies to detect native p11 and 5-HT(1B)R combined with visualization of EGFP driven under the 5-HT(4)R promoter in BAC-transgenic mice. In the caudate-putamen, the hippocampal pyramidal cell layer of CA1 and the hippocampal granule cell layer of dentate gyrus, most p11 containing cells co-expressed both 5-HT(1B)R and 5-HT(4)R. In the cingulate cortex, stratum radiatum/oriens of CA1, hilus of the dentate gyrus and cerebellar cortex, many cells co-expressed p11 and 5-HT(1B)R, but not 5-HT(4)R. In the studied brain regions, few cells solely expressed p11 without any significant expression of 5-HT(1B)R or 5-HT(4)R. It can be concluded that p11 is anatomically positioned to modulate serotonin neurotransmission, via 5-HT(1B)R and 5-HT(4)R, in brain regions important for emotionality, cognition and locomotion.


Assuntos
Anexina A2/metabolismo , Cerebelo/metabolismo , Córtex Cerebral/metabolismo , Neostriado/metabolismo , Neurônios/metabolismo , Receptor 5-HT1B de Serotonina/biossíntese , Receptores 5-HT4 de Serotonina/biossíntese , Proteínas S100/metabolismo , Animais , Cerebelo/ultraestrutura , Córtex Cerebral/ultraestrutura , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hipocampo/metabolismo , Hipocampo/ultraestrutura , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Transgênicos , Neostriado/ultraestrutura , Neurônios/diagnóstico por imagem , Especificidade de Órgãos , Regiões Promotoras Genéticas , Receptores 5-HT4 de Serotonina/genética , Proteínas Recombinantes de Fusão/biossíntese , Ultrassonografia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA