Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Proc Natl Acad Sci U S A ; 116(1): 297-302, 2019 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-30559184

RESUMO

Ketamine, a noncompetitive N-methyl-d-aspartate (NMDA) receptor antagonist, produces rapid and long-lasting antidepressant effects in major depressive disorder (MDD) patients. (2R,6R)-Hydroxynorketamine [(2R,6R)-HNK], a metabolite of ketamine, is reported to produce rapid antidepressant effects in rodent models without the side effects of ketamine. Importantly, (2R,6R)-HNK does not block NMDA receptors like ketamine, and the molecular signaling mechanisms for (2R,6R)-HNK remain unknown. Here, we examined the involvement of BDNF/TrkB/mechanistic target of rapamycin complex 1 (mTORC1) signaling in the antidepressant actions of (2R,6R)-HNK. Intramedial prefrontal cortex (intra-mPFC) infusion or systemic (2R,6R)-HNK administration induces rapid and long-lasting antidepressant effects in behavioral tests, identifying the mPFC as a key region for the actions of (2R,6R)-HNK. The antidepressant actions of (2R,6R)-HNK are blocked in mice with a knockin of the BDNF Val66Met allele (which blocks the processing and activity-dependent release of BDNF) or by intra-mPFC microinjection of an anti-BDNF neutralizing antibody. Blockade of L-type voltage-dependent Ca2+ channels (VDCCs), required for activity-dependent BDNF release, also blocks the actions of (2R,6R)-HNK. Intra-mPFC infusion of pharmacological inhibitors of TrkB or mTORC1 signaling, which are downstream of BDNF, also block the actions of (2R,6R)-HNK. Moreover, (2R,6R)-HNK increases synaptic function in the mPFC. These findings indicate that activity-dependent BDNF release and downstream TrkB and mTORC1 signaling, which increase synaptic function in the mPFC, are required for the rapid and long-lasting antidepressant effects of (2R,6R)-HNK, supporting the potential use of this metabolite for the treatment of MDD.


Assuntos
Antidepressivos/farmacologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Ketamina/análogos & derivados , Animais , Células Cultivadas , Depressão/tratamento farmacológico , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ketamina/farmacologia , Alvo Mecanístico do Complexo 1 de Rapamicina/antagonistas & inibidores , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos
2.
Neurobiol Dis ; 134: 104669, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31707118

RESUMO

Dysfunction of medial prefrontal cortex (mPFC) in association with imbalance of inhibitory and excitatory neurotransmission has been implicated in depression. However, the precise cellular mechanisms underlying this imbalance, particularly for GABAergic transmission in the mPFC, and the link with the rapid acting antidepressant ketamine remains poorly understood. Here we determined the influence of chronic unpredictable stress (CUS), an ethologically validated model of depression, on synaptic markers of GABA neurotransmission, and the influence of a single dose of ketamine on CUS-induced synaptic deficits in mPFC of male rodents. The results demonstrate that CUS decreases GABAergic proteins and the frequency of inhibitory post synaptic currents (IPSCs) of layer V mPFC pyramidal neurons, concomitant with depression-like behaviors. In contrast, a single dose of ketamine can reverse CUS-induced deficits of GABA markers, in conjunction with reversal of CUS-induced depressive-like behaviors. These findings provide further evidence of impairments of GABAergic synapses as key determinants of depressive behavior and highlight ketamine-induced synaptic responses that restore GABA inhibitory, as well as glutamate neurotransmission.


Assuntos
Antidepressivos/administração & dosagem , Depressão/fisiopatologia , Ketamina/administração & dosagem , Neurônios/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Estresse Psicológico/fisiopatologia , Transmissão Sináptica/efeitos dos fármacos , Ácido gama-Aminobutírico/fisiologia , Animais , Modelos Animais de Doenças , Potenciais Pós-Sinápticos Inibidores/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Neurônios/fisiologia , Córtex Pré-Frontal/fisiopatologia
3.
Neuropsychopharmacology ; 45(10): 1725-1734, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32396921

RESUMO

We previously reported that the serotonergic system is important for the antidepressant-like effects of ketamine, a non-competitive N-methyl-D-aspartate receptor antagonist, which produces rapid and long-lasting antidepressant effects in patients with major depressive disorder (MDD). In particular, selective stimulation of the 5-HT1A receptor in the medial prefrontal cortex (mPFC), as opposed to the somatic 5-HT1A autoreceptor, has been shown to play a critical role in the antidepressant-like actions of ketamine. However, the detailed mechanisms underlying mPFC 5-HT1A receptor-mediated antidepressant-like effects are not fully understood. Here we examined the involvement of the glutamate AMPA receptor and brain-derived neurotrophic factor (BDNF) in the antidepressant-like effects of 5-HT1A receptor activation in the mPFC. The results show that intra-mPFC infusion of the 5-HT1A receptor agonist 8-OH-DPAT induces rapid and long-lasting antidepressant-like effects in the forced swim, novelty-suppressed feeding, female urine sniffing, and chronic unpredictable stress tests. In addition, the results demonstrate that the antidepressant-like effects of intra-mPFC infusion of 8-OH-DPAT are blocked by co-infusion of an AMPA receptor antagonist or an anti-BDNF neutralizing antibody. In addition, mPFC infusion of 8-OH-DPAT increased the phosphorylation of signaling proteins downstream of BDNF, including mTOR, ERK, 4EBP1, and p70S6K. Finally, selective stimulation of the 5-HT1A receptor increased levels of synaptic proteins and synaptic function in the mPFC. Collectively, these results indicate that selective stimulation of 5-HT1A receptor in the mPFC exerts rapid and sustained antidepressant-like effects via activation of AMPA receptor/BDNF/mTOR signaling in mice, which subsequently increase synaptic function in the mPFC, and provide evidence for the 5-HT1A receptor as a target for the treatment of MDD.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Transtorno Depressivo Maior , Animais , Antidepressivos/farmacologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Depressão/tratamento farmacológico , Transtorno Depressivo Maior/tratamento farmacológico , Feminino , Humanos , Camundongos , Córtex Pré-Frontal/metabolismo , Receptor 5-HT1A de Serotonina , Receptores de AMPA
4.
J Clin Invest ; 130(3): 1336-1349, 2020 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-31743111

RESUMO

A single subanesthetic dose of ketamine, an NMDA receptor (NMDAR) antagonist, produces rapid and sustained antidepressant actions in depressed patients, addressing a major unmet need for the treatment of mood disorders. Ketamine produces a rapid increase in extracellular glutamate and synaptic formation in the prefrontal cortex, but the initial cellular trigger that initiates this increase and ketamine's behavioral actions has not been identified. To address this question, we used a combination of viral shRNA and conditional mutation to produce cell-specific knockdown or deletion of a key NMDAR subunit, GluN2B, implicated in the actions of ketamine. The results demonstrated that the antidepressant actions of ketamine were blocked by GluN2B-NMDAR knockdown on GABA (Gad1) interneurons, as well as subtypes expressing somatostatin (Sst) or parvalbumin (Pvalb), but not glutamate principle neurons in the medial prefrontal cortex (mPFC). Further analysis of GABA subtypes showed that cell-specific knockdown or deletion of GluN2B in Sst interneurons blocked or occluded the antidepressant actions of ketamine and revealed sex-specific differences that are associated with excitatory postsynaptic currents on mPFC principle neurons. These findings demonstrate that GluN2B-NMDARs on GABA interneurons are the initial cellular trigger for the rapid antidepressant actions of ketamine and show sex-specific adaptive mechanisms to GluN2B modulation.


Assuntos
Antidepressivos/farmacologia , Neurônios GABAérgicos/metabolismo , Interneurônios/metabolismo , Ketamina/farmacologia , Caracteres Sexuais , Animais , Feminino , Neurônios GABAérgicos/patologia , Técnicas de Inativação de Genes , Glutamato Descarboxilase/genética , Glutamato Descarboxilase/metabolismo , Interneurônios/patologia , Masculino , Camundongos , Camundongos Transgênicos , Parvalbuminas/genética , Parvalbuminas/metabolismo , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Somatostatina/genética , Somatostatina/metabolismo
5.
Neuropsychopharmacology ; 44(13): 2230-2238, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31454827

RESUMO

Currently available antidepressants have a delayed onset and limited efficacy, highlighting the need for new, rapid and more efficacious agents. Ketamine, an NMDA receptor antagonist, has emerged as a new rapid-acting antidepressant, effective even in treatment resistant patients. However, ketamine induces undesired psychotomimetic and dissociative side effects that limit its clinical use. The d-stereoisomer of methadone (dextromethadone; REL-1017) is a noncompetitive NMDA receptor antagonist with an apparently favorable safety and tolerability profile. The current study examined the rapid and sustained antidepressant actions of d-methadone in several behavioral paradigms, as well as on mTORC1 signaling and synaptic changes in the medial prefrontal cortex (mPFC). A single dose of d-methadone promoted rapid and sustained antidepressant responses in the novelty-suppressed feeding test (NSFT), a measure of anxiety, and in the female urine sniffing test (FUST), a measure of motivation and reward. D-methadone also produced a rapid reversal of the sucrose preference deficit, a measure of anhedonia, in rats exposed to chronic unpredictable stress. D-methadone increased phospho-p70S6 kinase, a downstream target of mTORC1 in the mPFC, and intra-mPFC infusion of the selective mTORC1 inhibitor rapamycin blocked the antidepressant actions of d-methadone in the FUST and NSFT. D-methadone administration also increased levels of the synaptic proteins, PSD95, GluA1, and Synapsin 1 and enhanced synaptic function in the mPFC. Studies in primary cortical cultures show that d-methadone also increases BDNF release, as well as phospho-p70S6 kinase. These findings indicate that d-methadone induces rapid antidepressant actions through mTORC1-mediated synaptic plasticity in the mPFC similar to ketamine.


Assuntos
Antidepressivos/administração & dosagem , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Metadona/administração & dosagem , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Animais , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/fisiologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Ketamina/administração & dosagem , Masculino , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
6.
J Clin Invest ; 129(6): 2542-2554, 2019 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-30990795

RESUMO

Preclinical studies demonstrate that rapid acting antidepressants, including ketamine require stimulation of mTORC1 signaling. This pathway is regulated by neuronal activity, endocrine and metabolic signals, notably the amino acid leucine, which activates mTORC1 signaling via binding to the upstream regulator sestrin. Here, we examined the antidepressant actions of NV-5138, a novel highly selective small molecule modulator of sestrin that penetrates the blood brain barrier. The results demonstrate that a single dose of NV-5138 produced rapid and long-lasting antidepressant effects, and rapidly reversed anhedonia caused by chronic stress exposure. The antidepressant actions of NV-5138 required BDNF release as the behavioral responses are blocked by infusion of a BDNF neutralizing antibody into the medial prefrontal cortex (mPFC) or in mice with a knock-in of a BDNF polymorphism that blocks activity dependent BDNF release. NV-5138 administration also rapidly increased synapse number and function in the mPFC, and reversed the synaptic deficits caused by chronic stress. Together, the results demonstrate that NV-5138 produced rapid synaptic and antidepressant behavioral responses via activation of the mTORC1 pathway and BDNF signaling, indicating that pharmacological modulation of sestrin is a novel approach for development of rapid acting antidepressants.


Assuntos
Antidepressivos , Comportamento Animal/efeitos dos fármacos , Proteínas de Choque Térmico/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Animais , Antidepressivos/química , Antidepressivos/farmacocinética , Antidepressivos/farmacologia , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Proteínas de Choque Térmico/genética , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Camundongos , Camundongos Knockout , Ratos , Ratos Sprague-Dawley , Transmissão Sináptica/genética
7.
Brain Res ; 1199: 148-58, 2008 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-18267317

RESUMO

Reports of beneficial effects of exercise on psychological health in humans are increasingly supported by basic research studies. Exercise is hypothesized to regulate antidepressant-related mechanisms and we therefore characterized the effects of chronic exercise in mouse behavioral paradigms relevant to antidepressant actions. Mice given free access to running wheels showed antidepressant-like behavior in learned helplessness, forced-swim (FST) and tail suspension paradigms. These responses were similar to responses of antidepressant drug-treated animals. When tested under conditions where locomotor activity was not altered, exercising mice also showed reduced anxiety compared to sedentary control mice. In situ hybridization analysis showed that BDNF mRNA was increased in specific subfields of hippocampus after wheel running. We chose one paradigm, the FST, in which to investigate a functional role for brain-derived neurotrophic factor (BDNF) in the behavioral response to exercise. We tested mice heterozygous for a deletion of the BDNF gene in the FST after wheel-running. Exercising wild-type mice showed the expected antidepressant-like behavioral response in the FST but exercise was ineffective in improving FST performance in heterozygous BDNF knockout mice. A possible functional contribution of a BDNF signaling pathway to FST performance in exercising mice was investigated using the specific MEK inhibitor PD184161 to block the MAPK signaling pathway. Subchronic administration of PD184161 to exercising mice blocked the antidepressant-like behavioral response seen in vehicle-treated exercising mice in the FST. In summary, chronic wheel-running exercise in mice results in antidepressant-like behavioral changes that may involve a BDNF related mechanism similar to that hypothesized for antidepressant drug treatment.


Assuntos
Ansiedade/terapia , Depressão/terapia , Condicionamento Físico Animal/métodos , Amitriptilina/uso terapêutico , Análise de Variância , Animais , Antidepressivos Tricíclicos/uso terapêutico , Ansiedade/fisiopatologia , Comportamento Animal , Fator Neurotrófico Derivado do Encéfalo/deficiência , Depressão/fisiopatologia , Modelos Animais de Doenças , Reação de Fuga/efeitos dos fármacos , Desamparo Aprendido , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Medição da Dor , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/fisiologia , Natação , Fatores de Tempo
8.
Biol Psychiatry ; 83(1): 38-49, 2018 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-28697890

RESUMO

BACKGROUND: Chronic stress exposure causes neuronal atrophy and synaptic deficits in the medial prefrontal cortex (PFC), contributing to development of anxiety- and depressive-like behaviors. Concomitantly, microglia in the PFC undergo morphological and functional changes following stress exposure, suggesting that microglia contribute to synaptic deficits underlying behavioral consequences. METHODS: Male and female mice were exposed to chronic unpredictable stress (CUS) to examine the role of neuron-microglia interactions in the medial PFC during development of anxiety- and depressive-like behaviors. Thy1-GFP-M mice were used to assess microglia-mediated neuronal remodeling and dendritic spine density in the medial PFC. Viral-mediated knockdown of neuronal colony stimulating factor 1 (CSF1) was used to modulate microglia function and behavioral consequences after CUS. RESULTS: CUS promoted anxiety- and depressive-like behaviors that were associated with increased messenger RNA levels of CSF1 in the PFC. Increased CSF1 messenger RNA levels were also detected in the postmortem dorsolateral PFC of individuals with depression. Moreover, microglia isolated from the frontal cortex of mice exposed to CUS show elevated CSF1 receptor expression and increased phagocytosis of neuronal elements. Notably, functional alterations in microglia were more pronounced in male mice compared with female mice. These functional changes in microglia corresponded with reduced dendritic spine density on pyramidal neurons in layer 1 of the medial PFC. Viral-mediated knockdown of neuronal CSF1 in the medial PFC attenuated microglia-mediated neuronal remodeling and prevented behavioral deficits caused by CUS. CONCLUSIONS: These findings revealed that stress-induced elevations in neuronal CSF1 provokes microglia-mediated neuronal remodeling in the medial PFC, contributing to synaptic deficits and development of anxiety- and depressive-like behavior.


Assuntos
Transtorno Depressivo/metabolismo , Fator Estimulador de Colônias de Macrófagos/metabolismo , Microglia/metabolismo , Plasticidade Neuronal/fisiologia , Neurônios/metabolismo , Estresse Psicológico/metabolismo , Animais , Ansiedade/metabolismo , Ansiedade/patologia , Doença Crônica , Transtorno Depressivo/patologia , Modelos Animais de Doenças , Feminino , Fator Estimulador de Colônias de Macrófagos/genética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/patologia , Neurônios/patologia , Fagocitose/fisiologia , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/patologia , RNA Mensageiro/metabolismo , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Caracteres Sexuais , Estresse Psicológico/patologia , Incerteza
9.
Biol Psychiatry ; 61(5): 661-70, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-16945347

RESUMO

BACKGROUND: Brain-derived neurotrophic factor (BDNF) is upregulated in the hippocampus by antidepressant treatments, and centrally administered BDNF can produce antidepressant-like effects in rodent behavioral models of depression. BDNF-regulated signaling pathways are thus potential targets for investigation of antidepressant mechanisms. METHODS: We examined the effects of inhibition of MAPK kinase (MEK) in mouse behavioral models for depression including interactions with effects of antidepressant drugs. We also assessed the behavioral consequences of a heterozygous gene deletion for BDNF combined with MEK inhibition or stress. RESULTS: Acute administration of the MEK inhibitor PD184161 produced depressive-like behavior. PD184161 blocked the antidepressant-like effects of desipramine and sertraline in the forced swim test and blocked the effects of desipramine in the tail suspension test. Heterozygous deletion of BDNF alone did not influence behavior in the forced swim test but resulted in a depressive phenotype when combined with a low-dose MEK inhibitor or stress exposure. CONCLUSIONS: We demonstrate that acute blockade of MAPK signaling produces a depressive-like phenotype and blocks behavioral actions of antidepressants. We also demonstrate in BDNF heterozygous knockout mice an example of a how a defined genetic alteration can confer vulnerability to a pharmacologic or environmental challenge resulting in a depressive behavioral phenotype.


Assuntos
Antidepressivos/uso terapêutico , Comportamento Animal/efeitos dos fármacos , Depressão/tratamento farmacológico , Quinases de Proteína Quinase Ativadas por Mitógeno/fisiologia , Transdução de Sinais/fisiologia , Análise de Variância , Compostos de Anilina/farmacologia , Animais , Benzamidas/farmacologia , Fator Neurotrófico Derivado do Encéfalo/deficiência , Depressão/induzido quimicamente , Depressão/fisiopatologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Interações Medicamentosas , Inibidores Enzimáticos/farmacologia , Desamparo Aprendido , Elevação dos Membros Posteriores/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Natação
10.
Neuropsychopharmacology ; 42(6): 1231-1242, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-27634355

RESUMO

GLYX-13 is a putative NMDA receptor modulator with glycine-site partial agonist properties that produces rapid antidepressant effects, but without the psychotomimetic side effects of ketamine. Studies were conducted to examine the molecular, cellular, and behavioral actions of GLYX-13 to further characterize the mechanisms underlying the antidepressant actions of this agent. The results demonstrate that a single dose of GLYX-13 rapidly activates the mTORC1 pathway in the prefrontal cortex (PFC), and that infusion of the selective mTORC1 inhibitor rapamycin into the medial PFC (mPFC) blocks the antidepressant behavioral actions of GLYX-13, indicating a requirement for mTORC1 similar to ketamine. The results also demonstrate that GLYX-13 rapidly increases the number and function of spine synapses in the apical dendritic tuft of layer V pyramidal neurons in the mPFC. Notably, GLYX-13 significantly increased the synaptic responses to hypocretin, a measure of thalamocortical synapses, compared with its effects on 5-HT responses, a measure of cortical-cortical responses mediated by the 5-HT2A receptor. Behavioral studies further demonstrate that GLYX-13 does not influence 5-HT2 receptor induced head twitch response or impulsivity in a serial reaction time task (SRTT), whereas ketamine increased responses in both tests. In contrast, both GLYX-13 and ketamine increased attention in the SRTT task, which is linked to hypocretin-thalamocortical responses. The differences in the 5-HT2 receptor synaptic and behavioral responses may be related to the lack of psychotomimetic side effects of GLYX-13 compared with ketamine, whereas regulation of the hypocretin responses may contribute to the therapeutic benefits of both rapid acting antidepressants.


Assuntos
Antidepressivos/farmacologia , Comportamento Animal/efeitos dos fármacos , Ketamina/farmacologia , Oligopeptídeos/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Animais , Antidepressivos/administração & dosagem , Ketamina/administração & dosagem , Masculino , Camundongos Endogâmicos C57BL , Oligopeptídeos/administração & dosagem , Ratos , Ratos Sprague-Dawley
11.
Biol Psychiatry ; 59(9): 775-85, 2006 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-16458261

RESUMO

BACKGROUND: Increased serum levels of TNFalpha and other pro-inflammatory cytokines have been found in patients with major depression and several other psychiatric conditions. In rodents, these cytokines produce symptoms commonly referred to as "sickness behavior." Some of these, including reduced feeding and decreased social and exploratory behavior, are reminiscent of those seen in depressed patients. Interpretation of these effects is complicated by the malaise caused by acute injections of pro-inflammatory cytokines, however. Thus, it is unclear whether cytokines are involved in the etiology of depressive symptoms. METHODS: We used a panel of behavioral assays to assess TNFR1(-/-) and TNFR2(-/-) mice for anxiety and depression-like behaviors. RESULTS: We show that deletion of either TNFR1 or TNFR2 leads to an antidepressant-like response in the forced swim test and that mice lacking TNFR2 demonstrate a hedonic response in a sucrose drinking test compared with wildtype littermates. In addition, deletion of TNFR1 leads to decreased fear conditioning. There were no differences in behavior in anxiety tests for either null mutant. CONCLUSIONS: These results are consistent with the hypothesis that TNFalpha can induce depression-like symptoms even in the absence of malaise and demonstrate that both receptor subtypes can be involved in this response.


Assuntos
Ansiedade/fisiopatologia , Depressão/fisiopatologia , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/fisiologia , Análise de Variância , Animais , Comportamento Animal/fisiologia , Condicionamento Psicológico , Modelos Animais de Doenças , Comportamento de Ingestão de Líquido/fisiologia , Comportamento Exploratório/fisiologia , Medo , Expressão Gênica/fisiologia , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/fisiologia , RNA Mensageiro/metabolismo , Tempo de Reação/fisiologia , Receptores do Fator de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral , Receptores Tipo II do Fator de Necrose Tumoral/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Sacarose , Natação/fisiologia , Receptores Chamariz do Fator de Necrose Tumoral
12.
Neurosci Lett ; 601: 20-9, 2015 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-25582786

RESUMO

Clinical brain imaging and postmortem studies provide evidence of structural and functional abnormalities of key limbic and cortical structures in depressed patients, suggesting that spine synapse connectivity is altered in depression. Characterization of the cellular determinants underlying these changes in patients are limited, but studies in rodent models demonstrate alterations of dendrite complexity and spine density and function that could contribute to the morphological and functional alterations observed in humans. Rodent studies demonstrate region specific effects in chronic stress models of depression, including reductions in dendrite complexity and spine density in the hippocampus and prefrontal cortex (PFC) but increases in the basolateral amygdala and nucleus accumbens. Alterations of spine synapse connectivity in these regions are thought to contribute to the behavioral symptoms of depression, including disruption of cognition, mood, emotion, motivation, and reward. Studies of the mechanisms underlying these effects demonstrate a role for altered brain derived neurotrophic factor (BDNF) signaling that regulates synaptic protein synthesis. In contrast, there is evidence that chronic antidepressant treatment can block or reverse the spine synapse alterations caused by stress. Notably, the new fast acting antidepressant ketamine, which produces rapid therapeutic actions in treatment resistant MDD patients, rapidly increases spine synapse number in the PFC of rodents and reverses the effects of chronic stress. The rapid synaptic and behavioral actions of ketamine occur via increased BDNF regulation of synaptic protein synthesis. Together these studies provide evidence for a neurotophic and synaptogenic hypothesis of depression and treatment response and indicate that spine synapse connectivity in key cortical and limbic brain regions is critical for control of mood and emotion.


Assuntos
Espinhas Dendríticas/fisiologia , Depressão/fisiopatologia , Sinapses/fisiologia , Animais , Antidepressivos/uso terapêutico , Córtex Cerebral/patologia , Córtex Cerebral/fisiopatologia , Espinhas Dendríticas/ultraestrutura , Depressão/tratamento farmacológico , Depressão/etiologia , Depressão/patologia , Humanos , Sistema Límbico/patologia , Sistema Límbico/fisiopatologia , Neurônios/patologia , Estresse Psicológico/complicações , Estresse Psicológico/patologia , Estresse Psicológico/fisiopatologia
13.
Nat Med ; 20(5): 531-5, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24728411

RESUMO

Major depressive disorder (MDD) affects up to 17% of the population, causing profound personal suffering and economic loss. Clinical and preclinical studies have revealed that prolonged stress and MDD are associated with neuronal atrophy of cortical and limbic brain regions, but the molecular mechanisms underlying these morphological alterations have not yet been identified. Here, we show that stress increases levels of REDD1 (regulated in development and DNA damage responses-1), an inhibitor of mTORC1 (mammalian target of rapamycin complex-1; ref. 10), in rat prefrontal cortex (PFC). This is concurrent with a decrease in phosphorylation of signaling targets of mTORC1, which is implicated in protein synthesis-dependent synaptic plasticity. We also found that REDD1 levels are increased in the postmortem PFC of human subjects with MDD relative to matched controls. Mutant mice with a deletion of the gene encoding REDD1 are resilient to the behavioral, synaptic and mTORC1 signaling deficits caused by chronic unpredictable stress, whereas viral-mediated overexpression of REDD1 in rat PFC is sufficient to cause anxiety- and depressive-like behaviors and neuronal atrophy. Taken together, these postmortem and preclinical findings identify REDD1 as a critical mediator of the atrophy of neurons and depressive behavior caused by chronic stress exposure.


Assuntos
Transtornos de Ansiedade/genética , Transtorno Depressivo Maior/genética , Sinapses/patologia , Fatores de Transcrição/genética , Animais , Transtornos de Ansiedade/etiologia , Transtornos de Ansiedade/patologia , Transtorno Depressivo Maior/etiologia , Transtorno Depressivo Maior/patologia , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/patologia , Ratos , Transdução de Sinais , Sinapses/genética , Sinapses/metabolismo , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Fatores de Transcrição/metabolismo
14.
Methods Mol Biol ; 982: 127-40, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23456866

RESUMO

The cytokine erythropoietin (EPO) is an important regulator of hematopoesis and has well-known tissue protective properties. Neurotrophic action is implicated as mechanistically important in the treatment of depression, and neurotrophic actions of EPO suggest potential therapeutic utility of an EPO-like mechanism in depressive disorder. Rodent behavioral models that are responsive to clinically used antidepressants as well as to neurotrophic compounds can be used to assess potential antidepressant properties of EPO and EPO-like compounds. Rodent models described here are the forced-swim test (FST), a hyponeophagia test and the novel object recognition test. Each of these models provides different information and relevance to depression and each can be tested with EPO and similar compounds.


Assuntos
Depressão/metabolismo , Eritropoetina/metabolismo , Animais , Antidepressivos/uso terapêutico , Depressão/tratamento farmacológico , Modelos Animais de Doenças , Camundongos , Ratos
15.
Physiol Behav ; 104(2): 354-9, 2011 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-21185848

RESUMO

Early life stressors in rodents, including maternal separation and social isolation, have been shown to disrupt brain development and profoundly affect a wide-range of behaviors in adult animals. In this study, we focus on the development of female Sprague-Dawley rats in the presence and absence of conspecifics during the critical period of social play. Similar studies in male rodents have shown that this form of social deprivation results in dysregulated dopaminergic and serotonergic functions in the brain with core features of neuropsychiatric disorders including anxiety disorder and schizophrenia. Here we examined the behavioral and biochemical effects of post-weaning social isolation in female rats. Our findings demonstrated that isolation rearing produced marked deficits in social interaction behaviors and increased anxiety in open-field and novelty-suppressed feeding tests. The expression of synaptic-associated proteins PSD95 and synapsin I as well as glutamate receptors subunits GluR1 and NR1 in the prefrontal cortex (PFC) were significantly reduced in isolation-reared female rats. Current findings provide evidence that in female rats, post-weaning environmental disruption can result in profound dysregulation of synapse-related proteins and behavior.


Assuntos
Sintomas Comportamentais/etiologia , Sintomas Comportamentais/patologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Córtex Pré-Frontal/metabolismo , Isolamento Social/psicologia , Fatores Etários , Animais , Animais Recém-Nascidos , Peso Corporal , Proteína 4 Homóloga a Disks-Large , Estriol/metabolismo , Estro/fisiologia , Comportamento Exploratório/fisiologia , Feminino , Privação de Alimentos/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Córtex Pré-Frontal/patologia , Gravidez , Ratos , Ratos Sprague-Dawley , Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapsinas/metabolismo
16.
Vitam Horm ; 82: 1-21, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20472130

RESUMO

The incidence of depressive illness is high in the United States and worldwide, and the inadequacy of currently available drug treatments contributes to the significant health burden associated with depression. A basic understanding of the underlying disease processes in depression is lacking, and therefore, recreating the disease in animal models is not possible. Currently used models of depression attempt to produce quantifiable correlates of human symptoms in experimental animals. The models differ in the degree to which they produce features that resemble a depressive-like state, and models that include stress exposure are widely used. Paradigms that employ acute or subchronic stress exposure include learned helplessness, forced swim test, and tail suspension test, which employ relatively short-term exposure to inescapable or uncontrollable stress and can reliably detect antidepressant drug response. Longer-term models include chronic mild stress models, early-life stress models, and social conflict models, which may more accurately simulate processes that lead to depression. These models each have varying degrees of face, construct, and predictive validity for depression and contribute differently to our understanding of antidepressant processes.


Assuntos
Transtorno Depressivo/psicologia , Modelos Animais de Doenças , Estresse Psicológico/psicologia , Animais , Comportamento Animal , Desamparo Aprendido , Humanos
17.
Behav Brain Res ; 198(2): 366-71, 2009 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-19056428

RESUMO

Growth factors in the brain are important to depression and it's treatment and we assessed the ability of peripherally administered insulin-like growth factor-I (IGF-I) to influence behavior related to depression. We found that mice that received chronic IGF-I treatment showed antidepressant-like behavior in forced-swim and novelty-induced hypophagia (NIH) tests and increased sucrose consumption after chronic mild unpredictable stress exposure. Additionally, peripheral anti-IGF-I administration blocked exercise-induced antidepressant effects in the forced-swim test (FST). These results support the functional relevance of neurotrophic mechanisms to depression and extend this idea to include neurotrophic factors in the periphery.


Assuntos
Antidepressivos/farmacologia , Comportamento Animal/efeitos dos fármacos , Depressão/tratamento farmacológico , Fator de Crescimento Insulin-Like I/fisiologia , Estresse Fisiológico/efeitos dos fármacos , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Depressão/psicologia , Ensaio de Imunoadsorção Enzimática , Exercício Físico/psicologia , Humanos , Imunoglobulina G/farmacologia , Hibridização In Situ , Injeções Subcutâneas , Fator de Crescimento Insulin-Like I/administração & dosagem , Fator de Crescimento Insulin-Like I/antagonistas & inibidores , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Fator de Crescimento Insulin-Like I/farmacologia , Masculino , Camundongos , Atividade Motora/fisiologia , Natação/fisiologia , Fatores de Tempo
18.
Biol Psychiatry ; 66(3): 267-74, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19185286

RESUMO

BACKGROUND: The neuroprotective and trophic actions of erythropoietin (EPO) have been tested in several animal models of insult, injury, and neurodegeneration. Recent studies in human volunteers demonstrated that EPO improves cognition and also elicits antidepressant effects. It is believed that the behavioral effects are mediated by EPO's trophic effect on neuronal systems. We therefore tested whether EPO is able to alter behavior and brain gene expression in rats. METHODS: The expression of EPO and EPO receptor (EPOR) in multiple brain regions was examined by quantitative polymerase chain reaction, in situ hybridization, and immunohistochemistry. The regulation of EPO and the transcription factor hypoxia-induced factor-alpha (HIF1alpha) after electroconvulsive seizure (ECS) was investigated. Behavioral effects of EPO were tested in the rodent forced swimming and novelty-induced hypophagia (NIH) models. EPO gene profiles were obtained by microarray analysis of the hippocampus after intracerebroventricular infusion. RESULTS: EPO and EPOR were widely expressed in the brain albeit at low levels. Highest level of EPO and EPOR were in the choroid plexus and striatum, respectively. Peripheral administration of EPO was sufficient to produce a robust antidepressant-like effect in the forced swim and NIH tests. Gene expression profiles revealed that EPO induces the expression of neurotrophic genes such as brain-derived neurotrophic factor, VGF (nonacronymic), and neuritin. CONCLUSIONS: EPO is induced by ECS and independently exhibits antidepressant-like efficacy in the forced swim and NIH tests. EPO regulates the expression of genes implicated in antidepressant action and appears to be a candidate molecule for further testing in neuropsychiatry.


Assuntos
Antidepressivos/uso terapêutico , Depressão/tratamento farmacológico , Eritropoetina/metabolismo , Eritropoetina/uso terapêutico , Regulação da Expressão Gênica/fisiologia , Convulsões , Animais , Comportamento Animal/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Modelos Animais de Doenças , Eletrochoque/efeitos adversos , Comportamento Exploratório/efeitos dos fármacos , Perfilação da Expressão Gênica/métodos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Locomoção/efeitos dos fármacos , Masculino , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Ratos , Ratos Sprague-Dawley , Receptores da Eritropoetina/metabolismo , Convulsões/etiologia , Convulsões/metabolismo , Convulsões/fisiopatologia , Natação
19.
Nat Med ; 13(12): 1476-82, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18059283

RESUMO

Exercise has many health benefits, including antidepressant actions in depressed human subjects, but the mechanisms underlying these effects have not been elucidated. We used a custom microarray to identify a previously undescribed profile of exercise-regulated genes in the mouse hippocampus, a brain region implicated in mood and antidepressant response. Pathway analysis of the regulated genes shows that exercise upregulates a neurotrophic factor signaling cascade that has been implicated in the actions of antidepressants. One of the most highly regulated target genes of exercise and of the growth factor pathway is the gene encoding the VGF nerve growth factor, a peptide precursor previously shown to influence synaptic plasticity and metabolism. We show that administration of a synthetic VGF-derived peptide produces a robust antidepressant response in mice and, conversely, that mutation of VGF in mice produces the opposite effects. The results suggest a new role for VGF and identify VGF signaling as a potential therapeutic target for antidepressant drug development.


Assuntos
Antidepressivos/farmacologia , Transtorno Depressivo/metabolismo , Exercício Físico , Perfilação da Expressão Gênica , Hipocampo/metabolismo , Neuropeptídeos/metabolismo , Condicionamento Físico Animal , Animais , Desenho de Fármacos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Crescimento Neural , Análise de Sequência com Séries de Oligonucleotídeos , Células PC12 , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA