Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Cell ; 184(5): 1299-1313.e19, 2021 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-33606976

RESUMO

It is unclear how binding of antidepressant drugs to their targets gives rise to the clinical antidepressant effect. We discovered that the transmembrane domain of tyrosine kinase receptor 2 (TRKB), the brain-derived neurotrophic factor (BDNF) receptor that promotes neuronal plasticity and antidepressant responses, has a cholesterol-sensing function that mediates synaptic effects of cholesterol. We then found that both typical and fast-acting antidepressants directly bind to TRKB, thereby facilitating synaptic localization of TRKB and its activation by BDNF. Extensive computational approaches including atomistic molecular dynamics simulations revealed a binding site at the transmembrane region of TRKB dimers. Mutation of the TRKB antidepressant-binding motif impaired cellular, behavioral, and plasticity-promoting responses to antidepressants in vitro and in vivo. We suggest that binding to TRKB and allosteric facilitation of BDNF signaling is the common mechanism for antidepressant action, which may explain why typical antidepressants act slowly and how molecular effects of antidepressants are translated into clinical mood recovery.


Assuntos
Antidepressivos/farmacologia , Receptor trkB/metabolismo , Animais , Antidepressivos/química , Antidepressivos/metabolismo , Sítios de Ligação , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Linhagem Celular , Colesterol/metabolismo , Embrião de Mamíferos , Fluoxetina/química , Fluoxetina/metabolismo , Fluoxetina/farmacologia , Hipocampo/metabolismo , Humanos , Camundongos , Modelos Animais , Simulação de Dinâmica Molecular , Domínios Proteicos , Ratos , Receptor trkB/química , Córtex Visual/metabolismo
2.
J Neurosci Res ; 100(3): 897-911, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35088434

RESUMO

Major depressive disorder is one of the most common mental disorders, and more than 300 million of people suffer from depression worldwide. Recent clinical trials indicate that deep brain stimulation of the superolateral medial forebrain bundle (mfb) can have rapid and long-term antidepressant effects in patients with treatment-resistant depression. However, the mechanisms of action are elusive. In this study, using female rats, we demonstrate the antidepressant effects of selective optogenetic stimulation of the ventral tegmental area's dopaminergic (DA) neurons passing through the mfb and compare different stimulation patterns. Chronic mild unpredictable stress (CMUS) induced depressive-like, but not anxiety-like phenotype. Short-term and long-term stimulation demonstrated antidepressant effect (OSST) and improved anxiolytic effect (EPM), while long-term stimulation during CMUS induction prevented depressive-like behavior (OSST and USV) and improved anxiolytic effect (EPM). The results highlight that long-term accumulative stimulation on DA pathways is required for antidepressant and anxiolytic effect.


Assuntos
Estimulação Encefálica Profunda , Transtorno Depressivo Maior , Animais , Estimulação Encefálica Profunda/métodos , Depressão/terapia , Transtorno Depressivo Maior/metabolismo , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Feminino , Humanos , Optogenética , Ratos , Roedores/metabolismo , Área Tegmentar Ventral/fisiologia
3.
J Neurochem ; 151(1): 11-27, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31361031

RESUMO

Adenosine receptor subtypes, first described 40 years ago, are known to regulate diverse biological functions and have a role in various conditions, such as cerebral and cardiac ischemia, immune and inflammatory disorders and cancer. In the brain, they limit potentially dangerous over excitation, but also regulate mechanisms essential in sleep and psychiatric disorders. In this review, we discuss the role of adenosine receptors in mood and anxiety disorders. Activation of A2A receptors is associated with increased depression-like symptoms, while increased A1 receptors signaling elicits rapid antidepressant effects. Indeed, several lines of evidence demonstrate that the therapeutic effects of different non-pharmacological treatments of depression, like sleep deprivation and electroconvulsive therapy are mediated by A1 receptor up-regulation or activation. In addition, A1 receptors may also play a role in the antidepressant effects of transcranial direct current stimulation and deep brain stimulation. As a potential downstream mechanism, which facilitates the antidepressant effects of A1 receptors, we propose a crosstalk between adenosinergic and glutamatergic systems mediated via synaptic plasticity protein Homer1a and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors. Moreover, adenosine receptors are also involved in the control of circadian rhythms, sleep homeostasis and some neuro-immunological mechanisms, all of them implicated in mood regulation. Antagonists of adenosine receptors such as caffeine have general anxiogenic effects. In particular, A2A receptors appear to have an important role in the pathophysiology of anxiety disorders. Taken together, the results discussed here indicate that the adenosinergic system is involved in both the etiology and the treatment of mood and anxiety disorders.


Assuntos
Transtornos de Ansiedade/metabolismo , Transtornos do Humor/metabolismo , Receptores Purinérgicos P1/metabolismo , Animais , Humanos
4.
Biol Chem ; 397(3): 207-14, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26641965

RESUMO

Homer1a is upregulated by several different antidepressant measures, including non-pharmacological treatments, like sleep deprivation (SD) and electroconvulsive therapy (ECT) and antidepressant drugs, such as imipramine, fluoxetine and ketamine. Homer1a induction might thus be a crucial joint mechanism for antidepressant therapy in general. However, the upstream signaling pathways that regulate or induce Homer1a expression are still not well understood. The main focus of the present review is to offer an overview of the current knowledge about the potential role of Homer1a in depression and the signaling pathways responsible for Homer1a regulation. It is suggested here that a detailed characterization of the signaling mechanisms leading to Homer1a expression might provide novel therapeutic targets for antidepressant drug development.


Assuntos
Proteínas de Transporte/genética , Depressão/metabolismo , Regulação da Expressão Gênica , Transdução de Sinais , Animais , Antidepressivos/farmacologia , Antidepressivos/uso terapêutico , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Proteínas de Transporte/metabolismo , Depressão/tratamento farmacológico , Depressão/genética , Transtorno Depressivo/tratamento farmacológico , Transtorno Depressivo/genética , Transtorno Depressivo/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Arcabouço Homer , Humanos , Receptor A1 de Adenosina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas ras/metabolismo
5.
Glia ; 62(4): 592-607, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24470356

RESUMO

ATP is an important regulator of microglia and its effects on microglial cytokine release are currently discussed as important contributors in a variety of brain diseases. We here analyzed the effects of ATP on the production of six inflammatory mediators (IL-6, IL-10, CCL2, IFN-γ, TNF-α, and IL-12p70) in cultured mouse primary microglia. Stimulation of P2X7 receptor by ATP (1 mM) or BzATP (500 µM) evoked the mRNA expression and release of proinflammatory cytokines IL-6, TNF-α, and the chemokine CCL2 in WT cells but not in P2X7(-/-) cells. The effects of ATP and BzATP were inhibited by the nonselective P2 receptor antagonists PPADs and suramin. Various selective P2X7 receptor antagonists blocked the P2X7-dependent release of IL-6 and CCL2, but, surprisingly, had no effect on BzATP-induced release of TNF-α in microglia. Calcium measurements confirmed that P2X7 is the main purine receptor activated by BzATP in microglia and showed that all P2X7 antagonists were functional. It is also presented that pannexin-1 hemichannel function and potential P2X4/P2X7 heterodimers are not involved in P2X7-dependent release of IL-6, CCL2, and TNF-α in microglia. How P2X7-specific antagonists only affect P2X7-dependent IL-6 and CCL2 release, but not TNF-α release is at the moment unclear, but indicates that the P2X7-dependent release of cytokines in microglia is differentially regulated.


Assuntos
Diferenciação Celular/fisiologia , Quimiocina CCL2/metabolismo , Interleucina-6/metabolismo , Microglia/fisiologia , Receptores Purinérgicos P2X7/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/farmacologia , Animais , Animais Recém-Nascidos , Encéfalo/citologia , Cálcio/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Quimiocina CCL2/genética , Conexinas/genética , Conexinas/metabolismo , Relação Dose-Resposta a Droga , Interleucina-6/genética , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Agonistas do Receptor Purinérgico P2X/farmacologia , Antagonistas do Receptor Purinérgico P2X/farmacologia , Receptores Purinérgicos P2X4/deficiência , Receptores Purinérgicos P2X7/deficiência , Receptores Purinérgicos P2X7/genética , Fator de Necrose Tumoral alfa/genética
6.
Nat Commun ; 15(1): 7257, 2024 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-39179578

RESUMO

Depression is associated with dysregulated circadian rhythms, but the role of intrinsic clocks in mood-controlling brain regions remains poorly understood. We found increased circadian negative loop and decreased positive clock regulators expression in the medial prefrontal cortex (mPFC) of a mouse model of depression, and a subsequent clock countermodulation by the rapid antidepressant ketamine. Selective Bmal1KO in CaMK2a excitatory neurons revealed that the functional mPFC clock is an essential factor for the development of a depression-like phenotype and ketamine effects. Per2 silencing in mPFC produced antidepressant-like effects, while REV-ERB agonism enhanced the depression-like phenotype and suppressed ketamine action. Pharmacological potentiation of clock positive modulator ROR elicited antidepressant-like effects, upregulating plasticity protein Homer1a, synaptic AMPA receptors expression and plasticity-related slow wave activity specifically in the mPFC. Our data demonstrate a critical role for mPFC molecular clock in regulating depression-like behavior and the therapeutic potential of clock pharmacological manipulations influencing glutamatergic-dependent plasticity.


Assuntos
Fatores de Transcrição ARNTL , Antidepressivos , Depressão , Ketamina , Camundongos Knockout , Córtex Pré-Frontal , Animais , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/efeitos dos fármacos , Depressão/tratamento farmacológico , Depressão/metabolismo , Depressão/genética , Camundongos , Antidepressivos/farmacologia , Masculino , Ketamina/farmacologia , Fatores de Transcrição ARNTL/metabolismo , Fatores de Transcrição ARNTL/genética , Ritmo Circadiano/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Proteínas Circadianas Period/metabolismo , Proteínas Circadianas Period/genética , Modelos Animais de Doenças , Fenótipo , Plasticidade Neuronal/efeitos dos fármacos , Receptores de AMPA/metabolismo , Receptores de AMPA/genética , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/metabolismo , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/genética , Proteínas de Arcabouço Homer/metabolismo , Proteínas de Arcabouço Homer/genética , Neurônios/metabolismo , Neurônios/efeitos dos fármacos
7.
Transl Psychiatry ; 14(1): 18, 2024 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-38195548

RESUMO

The partial N-methyl-D-aspartate receptor (NMDAR) agonist D-Cycloserine (DCS) has been evaluated for the treatment of a wide variety of psychiatric disorders, including dementia, schizophrenia, depression and for the augmentation of exposure-based psychotherapy. Most if not all of the potential psychiatric applications of DCS target an enhancement or restitution of cognitive functions, learning and memory. Their molecular correlate is long-term synaptic plasticity; and many forms of synaptic plasticity depend on the activation of NMDA receptors. Here, we comprehensively examined the modulation of different forms of synaptic plasticity in the hippocampus by DCS and its mechanism. We found that DCS positively modulates NMDAR-dependent forms of long-term synaptic plasticity (long-term synaptic potentiation, LTP, and long-term synaptic depression, LTD) in hippocampal brain slices of juvenile rats without affecting basal synaptic transmission. DCS binds to the D-serine/glycine binding site of the NMDAR. Pharmacological inhibition of this site prevented the induction of LTP, whereas agonism at the D-serine/glycine binding site augmented LTP and could functionally substitute for weak LTP induction paradigms. The most probable origin of endogenous D-serine are astrocytes, and its exocytosis is regulated by astrocytic metabotropic glutamate receptors (mGluR1). Functional eradication of astrocytes, inhibition of mGluR1 receptors and G-protein signaling in astrocytes adjacent to postsynaptic neurons prevented the induction of NMDAR-dependent forms of LTP and LTD. Our results support the enhancement of a bidirectional range of NMDAR-dependent hippocampal synaptic plasticity by DCS and D-serine-mediated gliotransmission. Therefore, the D-serine/glycine-binding site in NMDAR is a major target for psychopharmacological interventions targeting plasticity-related disorders.


Assuntos
Ciclosserina , Receptores de N-Metil-D-Aspartato , Humanos , Animais , Ratos , Ciclosserina/farmacologia , Plasticidade Neuronal , Serina , Glicina , Hipocampo
8.
Sci Rep ; 13(1): 20313, 2023 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-37985784

RESUMO

Light exerts powerful and pervasive effects on physiology and behaviour. These effects can be indirect, through clock synchronization and phase adjustment of circadian rhythms, or direct, independent of the circadian process. Exposure to light at inappropriate times, as commonly experienced in today's society, leads to increased prevalence of circadian, sleep and mood disorders as well as cognitive impairments. In mice, exposure to an ultradian 3.5 h light/3.5 h dark cycle (T7) for several days has been shown to impair behaviour through direct, non-circadian, photic effects, a claim we challenge here. We first confirmed that T7 cycle induces a lengthening of the circadian period resulting in a day by day phase-delay of both activity and sleep rhythms. Spatial novelty preference test performed at different circadian time points in mice housed under T7 cycle demonstrated that cognitive deficit was restrained to the subjective night. Mice under the same condition also showed a modification of stress-induced despair-like behaviour in the forced swim test. Therefore, our data demonstrate that ultradian light cycles cause time-of-day-dependent alteration of cognition and mood through clock period lengthening delaying circadian sleep phase, and not through a direct photic influence. These results are of critical importance for the clinical applications of light therapy in the medical field and for today's society to establish lighting recommendations for shift work, schools, hospitals and homes.


Assuntos
Ritmo Circadiano , Fotoperíodo , Camundongos , Animais , Ritmo Circadiano/fisiologia , Sono , Cognição , Afeto
9.
Mol Neurobiol ; 59(11): 6713-6723, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35999325

RESUMO

Several studies have reported separate roles of adenosine receptors and circadian clockwork in major depressive disorder. While less evidence exists for regulation of the circadian clock by adenosine signaling, a small number of studies have linked the adenosinergic system, the molecular circadian clock, and mood regulation. In this article, we review relevant advances and propose that adenosine receptor signaling, including canonical and other alternative downstream cellular pathways, regulates circadian gene expression, which in turn may underlie the pathogenesis of mood disorders. Moreover, we summarize the convergent point of these signaling pathways and put forward a pattern by which Homer1a expression, regulated by both cAMP-response element binding protein (CREB) and circadian clock genes, may be the final common pathogenetic mechanism in depression.


Assuntos
Relógios Circadianos , Transtorno Depressivo Maior , Adenosina , Relógios Circadianos/genética , Ritmo Circadiano/genética , Transtorno Depressivo Maior/genética , Humanos , Transtornos do Humor , Receptores Purinérgicos P1
10.
J Biol Chem ; 285(28): 21223-32, 2010 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-20427288

RESUMO

The present study compares negative Ets transcription factor (Net) and hypoxia-inducible factor 1alpha (HIF1alpha) regulation by hypoxia. Their protein stabilities are differently regulated by hypoxia, defining three periods in the kinetics: normoxia (high Net levels and low HIF1alpha levels), early hypoxia (high levels of Net and HIF1alpha), and late hypoxia (degradation of Net and HIF1alpha). Modulators of prolyl hydroxylase domain protein (PHD) activity induce a mobility shift of Net, similar to HIF1alpha, suggesting that post-translational modifications of both factors depend on PHD activity. The three PHDs have different roles in the regulation of Net protein levels; PHD1 and PHD3 are involved in the stabilization of Net, whereas PHD2 controls its degradation in late hypoxia. Net physically interacts with PHD2 in hypoxia, whereas PHD1 and PHD3 bind to Net in normoxia and hypoxia. Under the same conditions, PHD2 and PHD3 regulate both HIF1alpha stabilization in early hypoxia and its degradation at late hypoxia, whereas PHD1 is involved in HIF1alpha degradation in late hypoxia. We describe interconnections between the regulation of both Net and HIF1alpha at the protein level. Evidence is provided for a direct physical interaction between Net and HIF1alpha and indirect transcriptional regulation loops that involve the PHDs. Taken together our results indicate that Net and HIF1alpha are components of distinct signaling pathways that are intricately linked.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Hipóxia , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Linhagem Celular , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Células HeLa , Humanos , Cinética , Modelos Biológicos , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-ets , Interferência de RNA , Fatores de Tempo
11.
Neurosci Biobehav Rev ; 126: 23-42, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33757815

RESUMO

Psychiatric disorders exhibit an enormous burden on the health care systems worldwide accounting for around one-third of years lost due to disability among adults. Their etiology is largely unknown and diagnostic classification is based on symptomatology and course of illness and not on objective biomarkers. Most psychiatric disorders are moderately to highly heritable. However, it is still unknown what mechanisms may explain the discrepancy between heritability estimates and the present data from genetic analysis. In addition to genetic differences also epigenetic modifications are considered as potentially relevant in the transfer of susceptibility to psychiatric diseases. Though, whether or not epigenetic alterations can be inherited for many generations is highly controversial. In the present article, we will critically summarize both the genetic findings and the results from epigenetic analyses, including also those of noncoding RNAs. We will argue that one possible solution to the "missing heritability" problem in psychiatry is a potential role of retrotransposons, the exploration of which is presently only in its beginnings.


Assuntos
Transtornos Mentais , Psiquiatria , Elementos de DNA Transponíveis/genética , Epigênese Genética , Epigenômica , Humanos , Transtornos Mentais/genética
12.
J Vis Exp ; (175)2021 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-34633373

RESUMO

Major depressive disorder is one of the most prevalent forms of mental illnesses and causes tremendous individual suffering and socioeconomic burden. Despite its importance, current pharmacological treatment is limited, and novel treatment options are urgently needed. One key factor in the search for potential new drugs is evaluating their anti-depressive potency in appropriate animal models. The classical Porsolt forced swim test was used for this purpose for decades to induce and assess a depressive-like state. It consists of two short periods of forced swimming: the first to induce a depressed state and the second on the following day to evaluate the antidepressant effect of the agent given in between the two swim sessions. This model might be suitable as a screening tool for potential antidepressive agents but ignores the delayed onset of action of many antidepressants. The CDM was recently established and represented a modification of the classical test with notable differences. Mice are forced to swim for 5 consecutive days, following the idea that in humans, depression is induced by chronic rather than by acute stress. In a resting period of several days (1-3 weeks), animals develop sustained behavioral despair. The standard read-out method is the measurement of immobility time in an additional delayed swim session, but several alternative methods are proposed to get a broader view of the mood status of the animal. Multiple analysis tools can be used targeting behavioral, molecular, and electrophysiological changes. The depressed phenotype is stable for at least 4 weeks, providing a time window for rapid but also subchronic antidepressant treatment strategies. Furthermore, alterations in the development of a depressive-like state can be addressed using this approach. CDM, therefore, represents a useful tool to better understand depression and to develop novel treatment interventions.


Assuntos
Depressão , Transtorno Depressivo Maior , Animais , Antidepressivos/farmacologia , Depressão/tratamento farmacológico , Transtorno Depressivo Maior/tratamento farmacológico , Camundongos , Modelos Animais , Natação
13.
J Affect Disord ; 279: 501-509, 2021 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-33128940

RESUMO

BACKGROUND: Understanding the neurobiology of depression and the mechanism of action of therapeutic measures is currently a research priority. We have shown that the expression of the synaptic protein Homer1a correlates with depression-like behavior and its induction is a common mechanism of action of different antidepressant treatments. However, the mechanism of Homer1a regulation is still unknown. METHODS: We combined the chronic despair mouse model (CDM) of chronic depression with different antidepressant treatments. Depression-like behavior was characterized by forced swim and tail suspension tests, and via automatic measurement of sucrose preference in IntelliCage. The Homer1 mRNA expression and promoter DNA methylation were analyzed in cortex and peripheral blood by qRT-PCR and pyrosequencing. RESULTS: CDM mice show decreased Homer1a and Homer1b/c mRNA expression in cortex and blood samples, while chronic treatment with imipramine and fluoxetine or acute ketamine application increases their level only in the cortex. The quantitative analyses of the methylation of 7 CpG sites, located on the Homer1 promoter region containing several CRE binding sites, show a significant increase in DNA methylation in the cortex of CDM mice. In contrast, antidepressant treatments reduce the methylation level. LIMITATIONS: Homer1 expression and promotor methylation were not analyzed in different blood cell types. Other CpG sites of Homer1 promoter should be investigated in future studies. Our experimental approach does not distinguish between methylation and hydroxymethylation. CONCLUSIONS: We demonstrate that stress-induced depression-like behavior and antidepressant treatments are associated with epigenetic alterations of Homer1 promoter, providing new insights into the mechanism of antidepressant treatment.


Assuntos
Antidepressivos , Depressão , Animais , Antidepressivos/farmacologia , Depressão/tratamento farmacológico , Depressão/genética , Modelos Animais de Doenças , Epigênese Genética , Proteínas de Arcabouço Homer/metabolismo , Imipramina , Camundongos , Regiões Promotoras Genéticas/genética
14.
Neuropharmacology ; 162: 107834, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31682853

RESUMO

Resilience to stress is critical for the development of depression. Enhanced adenosine A1 receptor (A1R) signaling mediates the antidepressant effects of acute sleep deprivation (SD). However, chronic SD causes long-lasting upregulation of brain A1R and increases the risk of depression. To investigate the effects of A1R on mood, we utilized two transgenic mouse lines with inducible A1R overexpression in forebrain neurons. These two lines have identical levels of A1R increase in the cortex, but differ in the transgenic A1R expression in the hippocampus. Switching on the transgene promotes robust antidepressant and anxiolytic effects in both lines. The mice of the line without transgenic A1R overexpression in the hippocampus (A1Hipp-) show very strong resistance towards development of stress-induced chronic depression-like behavior. In contrast, the mice of the line in which A1R upregulation extends to the hippocampus (A1Hipp+), exhibit decreased resilience to depression as compared to A1Hipp-. Similarly, automatic analysis of reward behavior of the two lines reveals that depression resistant A1Hipp-transgenic mice exhibit high sucrose preference, while mice of the vulnerable A1Hipp + line developed stress-induced anhedonic phenotype. The A1Hipp + mice have increased Homer1a expression in hippocampus, correlating with impaired long-term potentiation in the CA1 region, mimicking the stressed mice. Furthermore, virus-mediated overexpression of Homer1a in the hippocampus decreases stress resilience. Taken together our data indicate for first time that increased expression of A1R and Homer1a in the hippocampus modulates the resilience to stress-induced depression and thus might potentially mediate the detrimental effects of chronic sleep restriction on mood.


Assuntos
Córtex Cerebral/metabolismo , Depressão/genética , Hipocampo/metabolismo , Proteínas de Arcabouço Homer/genética , Receptor A1 de Adenosina/genética , Resiliência Psicológica , Privação do Sono/metabolismo , Estresse Psicológico/genética , Animais , Comportamento Animal , Região CA1 Hipocampal/metabolismo , Depressão/metabolismo , Depressão/psicologia , Teste de Labirinto em Cruz Elevado , Potenciais Pós-Sinápticos Excitadores , Elevação dos Membros Posteriores , Proteínas de Arcabouço Homer/metabolismo , Potenciação de Longa Duração/genética , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Teste de Campo Aberto , Prosencéfalo , Receptor A1 de Adenosina/metabolismo , Recompensa , Privação do Sono/psicologia
15.
J Neurochem ; 108(6): 1453-63, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19284475

RESUMO

Activation of Ras into the GTP-binding, 'ON' state is a key switch in the neurotrophin-mediated neuronal survival and neurite outgrowth, in vitro as well as in vivo. In the current study we explored changes in GTP-Ras levels following facial nerve injury and the ensuing regeneration and the effects of perturbing these changes in vivo using synapsin-promoter mediated neuronal expression of constitutively active Val12H-Ras (synRas). Quantification of GTP-Ras and total Ras revealed a precipitous drop in the relative GTP-Ras levels in the axotomized facial motor nucleus, to 40% of normal levels at 2 days after cut, followed by a partial recovery to 50-65% at 4-28 days. On western blots, control and axotomized nuclei from synRas mutants showed a 2.2- and 2.5-fold elevation in GTP-Ras, respectively, compared with their wild type littermate controls (p < 5%, anova, TUKEY post-hoc), with the levels in the axotomized synRas nucleus slightly but not significantly above that in the uninjured littermate control (p = 9.9%). Similar increase was also observed in the pERK but not pAKT targets of the Ras cascade. This moderate elevation of GTP-Ras strongly curtailed post-traumatic neuronal cell death (-65%), the influx of T-cells (-48%) as well as other parameters of neuroinflammatory response. Although synRas did not affect the speed of axonal regeneration or functional recovery it caused a very pronounced increase in central axonal sprouting. These current data emphasize the role of reduced active Ras, and by extension, the reduced overall level of retrograde neurotrophin signalling after axotomy, in mediating post-traumatic cell death and inflammation and in restricting the sprouting response. Moreover, the neuroprotective and central sprouting-enhancing effects of neuronal Val12H-Ras could help promote recovery in CNS injury.


Assuntos
Traumatismos do Nervo Facial/fisiopatologia , Proteínas de Ligação ao GTP/fisiologia , Regeneração Nervosa/fisiologia , Neurônios/metabolismo , Recuperação de Função Fisiológica/fisiologia , Análise de Variância , Animais , Axotomia/métodos , Complexo CD3/metabolismo , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Modelos Animais de Doenças , Ativação Enzimática/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Traumatismos do Nervo Facial/genética , Proteínas de Ligação ao GTP/genética , Galanina/metabolismo , Histidina/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Regeneração Nervosa/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Recuperação de Função Fisiológica/genética , Fatores de Tempo , Valina/genética , Proteínas ras/genética , Proteínas ras/fisiologia
16.
Front Cell Neurosci ; 13: 566, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31920563

RESUMO

Different studies have demonstrated that inflammation and alterations in glutamate neurotransmission are two events contributing to the pathophysiology of neurodegenerative or neurological disorders. There are evidences that N-arachidonoylphenolamine (AM404), a cannabinoid system modulator and paracetamol metabolite, modulates inflammation and exerts neuroprotective effects on Huntington's (HD) and Parkinson's diseases (PD), and ischemia. However, the effects of AM404 on the production of inflammatory mediators and excitotoxicity in brain tissue stimulated with N-methyl-D-aspartic acid (NMDA) are not elucidated. In this present study, we investigated the effects of AM404 on the production of inflammatory mediators and neuronal cell death induced by NMDA in organotypic hippocampal slices cultures (OHSC) using qPCR, western blot (WB), and immunohistochemistry. Moreover, to comprehend the mechanism of excitotoxicity, we evaluated the effects of AM404 on glutamate release in hippocampal synaptosomes and the NMDA-induced calcium responses in acute hippocampal slices. Our results showed that AM404 led to a significant decrease in cell death induced by NMDA, through a mechanism possibly involving the reduction of glutamate release and the calcium ions responses. Furthermore, it decreased the expression of the interleukin (IL)-1ß. This study provides new significant insights about the anti-inflammatory and neuroprotection effects of AM404 on NMDA-induced excitotoxicity. To understand the effects of AM404 in these processes might contribute to the therapeutic potential of AM404 in diseases with involvement of neuroinflammation and neurodegeneration and might lead to a possible future treatment of neurodegenerative diseases.

17.
Neuron ; 104(2): 338-352.e7, 2019 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-31420117

RESUMO

Conventional antidepressants have limited efficacy and many side effects, highlighting the need for fast-acting and specific medications. Induction of the synaptic protein Homer1a mediates the effects of different antidepressant treatments, including the rapid action of ketamine and sleep deprivation (SD). We show here that mimicking Homer1a upregulation via intravenous injection of cell-membrane-permeable TAT-Homer1a elicits rapid antidepressant effects in various tests. Similar to ketamine and SD, in vitro and in vivo application of TAT-Homer1a enhances mGlu5 signaling, resulting in increased mTOR pathway phosphorylation, and upregulates synaptic AMPA receptor expression and activity. The antidepressant action of SD and Homer1a induction depends on mGlu5 activation specifically in excitatory CaMK2a neurons and requires enhanced AMPA receptor activity, translation, and trafficking. Moreover, our data demonstrate a pronounced therapeutic potential of different TAT-fused peptides that directly modulate mGlu5 and AMPA receptor activity and thus might provide a novel strategy for rapid and effective antidepressant treatment.


Assuntos
Comportamento Animal/efeitos dos fármacos , Encéfalo/metabolismo , Transtorno Depressivo Maior/metabolismo , Proteínas de Arcabouço Homer/farmacologia , Receptor de Glutamato Metabotrópico 5/efeitos dos fármacos , Receptores de AMPA/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Animais , Transtorno Depressivo Maior/genética , Modelos Animais de Doenças , Produtos do Gene tat , Proteínas de Arcabouço Homer/genética , Proteínas de Arcabouço Homer/metabolismo , Camundongos , Camundongos Knockout , Fragmentos de Peptídeos , Receptor de Glutamato Metabotrópico 5/genética , Receptor de Glutamato Metabotrópico 5/metabolismo , Receptores de AMPA/metabolismo , Transdução de Sinais/efeitos dos fármacos , Privação do Sono/metabolismo , Sinapses/metabolismo , Serina-Treonina Quinases TOR/efeitos dos fármacos , Regulação para Cima
18.
Neurosci Biobehav Rev ; 88: 63-72, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29550211

RESUMO

There is an urgent, unmet clinical need for faster and more efficient antidepressant drugs with higher response rates. In animal models of depression it was shown in the last few years that inhibition of three signaling molecules (BDNF, p11 and Homer1a) prevents efficacy of antidepressant therapy. These data not only show the crucial role of these factors for the treatment of depression, but may also point towards a better understanding of the molecular changes responsible for successful antidepressant therapy. Reviewing the literature concerning BNDF, p11 and Homer1a we here describe a molecular network in which these molecules interact with each other finally leading to facilitation of AMPA receptor signaling and plasticity, corroborating the current idea of AMPA receptors being a promising drug target in depression.


Assuntos
Antidepressivos/uso terapêutico , Depressão/tratamento farmacológico , Transtorno Depressivo/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Animais , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Humanos , Transdução de Sinais/fisiologia , Resultado do Tratamento
19.
Biol Psychiatry ; 84(1): 55-64, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29174591

RESUMO

BACKGROUND: Long-term synaptic plasticity is a basic ability of the brain to dynamically adapt to external stimuli and regulate synaptic strength and ultimately network function. It is dysregulated by behavioral stress in animal models of depression and in humans with major depressive disorder. Antidepressants have been shown to restore disrupted synaptic plasticity in both animal models and humans; however, the underlying mechanism is unclear. METHODS: We examined modulation of synaptic plasticity by selective serotonin reuptake inhibitors (SSRIs) in hippocampal brain slices from wild-type rats and serotonin transporter (SERT) knockout mice. Recombinant voltage-gated calcium (Ca2+) channels in heterologous expression systems were used to determine the modulation of Ca2+ channels by SSRIs. We tested the behavioral effects of SSRIs in the chronic behavioral despair model of depression both in the presence and in the absence of SERT. RESULTS: SSRIs selectively inhibited hippocampal long-term depression. The inhibition of long-term depression by SSRIs was mediated by a direct block of voltage-activated L-type Ca2+ channels and was independent of SERT. Furthermore, SSRIs protected both wild-type and SERT knockout mice from behavioral despair induced by chronic stress. Finally, long-term depression was facilitated in animals subjected to the behavioral despair model, which was prevented by SSRI treatment. CONCLUSIONS: These results showed that antidepressants protected synaptic plasticity and neuronal circuitry from the effects of stress via a modulation of Ca2+ channels and synaptic plasticity independent of SERT. Thus, L-type Ca2+ channels might constitute an important signaling hub for stress response and for pathophysiology and treatment of depression.


Assuntos
Antidepressivos/uso terapêutico , Canais de Cálcio Tipo L/metabolismo , Proteínas de Ligação a RNA/metabolismo , Estresse Psicológico/tratamento farmacológico , Transmissão Sináptica/efeitos dos fármacos , Fatores Etários , Animais , Células CHO , Cloreto de Cádmio/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/genética , Cricetulus , Modelos Animais de Doenças , Estimulação Elétrica , Feminino , Fluvoxamina/uso terapêutico , Células HEK293 , Elevação dos Membros Posteriores/psicologia , Hipocampo/citologia , Humanos , Técnicas In Vitro , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Nifedipino/farmacologia , Paroxetina/farmacologia , Técnicas de Patch-Clamp , Piperazinas/farmacologia , Piridinas/farmacologia , Proteínas de Ligação a RNA/genética , Ratos , Ratos Transgênicos , Ratos Wistar , Serotonina/farmacologia , Antagonistas da Serotonina/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/uso terapêutico , Estresse Psicológico/genética , Natação/psicologia , Transmissão Sináptica/genética , Transfecção
20.
Front Neurol ; 8: 264, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28649228

RESUMO

The small GTPase Ras is a universal eukaryotic cytoplasmic membrane-anchored protein, which regulates diverse downstream signal transduction pathways that play an important role in the proper functioning of neurons. Ras activity is a central regulator of structural and functional synaptic plasticity in the adult nervous system, where it channels neuronal responses to various extracellular cues allowing the organism to adapt to complex environmental stimuli. The suprachiasmatic nucleus (SCN) is the principle pacemaker of the circadian clock, and the circadian and photic regulation of Ras activity in the SCN is an important modulator of the clockwork. We have generated transgenic mouse expressing constitutively active V12-H-Ras selectively in neurons via a synapsin I promoter (synRas mice), which serves as a suitable model to study the role of neuronal Ras signaling. Modulation of Ras activity affects ERK1,2/CREB signaling and glycogen synthase kinase-3 beta expression in the SCN, which in turn modify the photoentrainment of the clock and the fine tuning the circadian period length. The main focus of this review is to offer an overview of the function of Ras signaling in the circadian rhythm and its potential role in learning and memory consolidation.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA