Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
PLoS Biol ; 15(12): e2002690, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29283992

RESUMO

Response to antidepressant treatment in major depressive disorder (MDD) cannot be predicted currently, leading to uncertainty in medication selection, increasing costs, and prolonged suffering for many patients. Despite tremendous efforts in identifying response-associated genes in large genome-wide association studies, the results have been fairly modest, underlining the need to establish conceptually novel strategies. For the identification of transcriptome signatures that can distinguish between treatment responders and nonresponders, we herein submit a novel animal experimental approach focusing on extreme phenotypes. We utilized the large variance in response to antidepressant treatment occurring in DBA/2J mice, enabling sample stratification into subpopulations of good and poor treatment responders to delineate response-associated signature transcript profiles in peripheral blood samples. As a proof of concept, we translated our murine data to the transcriptome data of a clinically relevant human cohort. A cluster of 259 differentially regulated genes was identified when peripheral transcriptome profiles of good and poor treatment responders were compared in the murine model. Differences in expression profiles from baseline to week 12 of the human orthologues selected on the basis of the murine transcript signature allowed prediction of response status with an accuracy of 76% in the patient population. Finally, we show that glucocorticoid receptor (GR)-regulated genes are significantly enriched in this cluster of antidepressant-response genes. Our findings point to the involvement of GR sensitivity as a potential key mechanism shaping response to antidepressant treatment and support the hypothesis that antidepressants could stimulate resilience-promoting molecular mechanisms. Our data highlight the suitability of an appropriate animal experimental approach for the discovery of treatment response-associated pathways across species.


Assuntos
Antidepressivos/farmacologia , Transtorno Depressivo Maior/tratamento farmacológico , Paroxetina/farmacologia , Receptores de Glucocorticoides/fisiologia , Animais , Antidepressivos/uso terapêutico , Biomarcadores Farmacológicos , Encéfalo/metabolismo , Corticosterona/sangue , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos DBA , Família Multigênica , Paroxetina/metabolismo , Paroxetina/uso terapêutico , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo
2.
Neuron ; 110(14): 2283-2298.e9, 2022 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-35649415

RESUMO

A single sub-anesthetic dose of ketamine produces a rapid and sustained antidepressant response, yet the molecular mechanisms responsible for this remain unclear. Here, we identified cell-type-specific transcriptional signatures associated with a sustained ketamine response in mice. Most interestingly, we identified the Kcnq2 gene as an important downstream regulator of ketamine action in glutamatergic neurons of the ventral hippocampus. We validated these findings through a series of complementary molecular, electrophysiological, cellular, pharmacological, behavioral, and functional experiments. We demonstrated that adjunctive treatment with retigabine, a KCNQ activator, augments ketamine's antidepressant-like effects in mice. Intriguingly, these effects are ketamine specific, as they do not modulate a response to classical antidepressants, such as escitalopram. These findings significantly advance our understanding of the mechanisms underlying the sustained antidepressant effects of ketamine, with important clinical implications.


Assuntos
Ketamina , Animais , Antidepressivos/farmacologia , Hipocampo , Canal de Potássio KCNQ2/genética , Ketamina/farmacologia , Ketamina/uso terapêutico , Camundongos , Proteínas do Tecido Nervoso , Neurônios
3.
Neurobiol Dis ; 42(3): 300-10, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21296667

RESUMO

Chronic stress evokes profound structural and molecular changes in the hippocampus, which may underlie spatial memory deficits. Corticotropin-releasing hormone (CRH) and CRH receptor 1 (CRHR1) mediate some of the rapid effects of stress on dendritic spine morphology and modulate learning and memory, thus providing a potential molecular basis for impaired synaptic plasticity and spatial memory by repeated stress exposure. Using adult male mice with CRHR1 conditionally inactivated in the forebrain regions, we investigated the role of CRH-CRHR1 signaling in the effects of chronic social defeat stress on spatial memory, the dendritic morphology of hippocampal CA3 pyramidal neurons, and the hippocampal expression of nectin-3, a synaptic cell adhesion molecule important in synaptic remodeling. In chronically stressed wild-type mice, spatial memory was disrupted, and the complexity of apical dendrites of CA3 neurons reduced. In contrast, stressed mice with forebrain CRHR1 deficiency exhibited normal dendritic morphology of CA3 neurons and mild impairments in spatial memory. Additionally, we showed that the expression of nectin-3 in the CA3 area was regulated by chronic stress in a CRHR1-dependent fashion and associated with spatial memory and dendritic complexity. Moreover, forebrain CRHR1 deficiency prevented the down-regulation of hippocampal glucocorticoid receptor expression by chronic stress but induced increased body weight gain during persistent stress exposure. These findings underscore the important role of forebrain CRH-CRHR1 signaling in modulating chronic stress-induced cognitive, structural and molecular adaptations, with implications for stress-related psychiatric disorders.


Assuntos
Dendritos/metabolismo , Aprendizagem em Labirinto/fisiologia , Memória/fisiologia , Prosencéfalo/metabolismo , Receptores de Hormônio Liberador da Corticotropina/genética , Estresse Psicológico/metabolismo , Análise de Variância , Animais , Western Blotting , Peso Corporal/genética , Moléculas de Adesão Celular/metabolismo , Dominação-Subordinação , Hibridização In Situ , Masculino , Camundongos , Camundongos Transgênicos , Nectinas , Neurônios/metabolismo , Estresse Psicológico/genética
4.
Front Behav Neurosci ; 12: 262, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30483074

RESUMO

Despite a growing body of research over the last few decades, mental disorders, including anxiety disorders or depression, are still one of the most prevalent and hardest to treat health burdens worldwide. Since pharmacological treatment with a single drug is often rather ineffective, approaches such as co-medication with functionally diverse antidepressants (ADs) have been discussed and tried more recently. Besides classical ADs, there is a growing number of candidate targets identified as potential starting points for new treatment methods. One of these candidates, the FK506 binding protein 51 (FKBP51) is linked to a number of psychiatric disorders in humans. In this study, we used SAFit2-a newly developed modulator of FKBP51, which has shown promising results in rodent models for stress-related disorders delivered in a depot formulation. We combined SAFit2 with the commonly prescribed selective serotonin reuptake inhibitor (SSRI) escitalopram and performed basic behavioral characterization in a mouse model. Remarkably, co-application of SAFit2 lowered the efficacy of escitalopram in anxiety-related tests but improved stress coping behavior. Given the fact that mental diseases such as anxiety disorders or depression can be divided into different sub-categories, some of which more or less prone to stress, SAFit2 could indeed be a highly beneficial co-medication in very specific cases. This study could be a first, promising step towards the use of FKBP51 modulators as potent and specific enhancers of AD efficiency for subclasses of patients in the future.

5.
Psychoneuroendocrinology ; 55: 128-43, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25765754

RESUMO

Early-life stress is a key risk factor for the development of neuropsychiatric disorders later in life. Neuronal cell adhesion molecules have been strongly implicated in the pathophysiology of psychiatric disorders and in modulating social behaviors associated with these diseases. Neuroligin-2 is a synaptic cell adhesion molecule, located at the postsynaptic membrane of inhibitory GABAergic synapses, and is involved in synaptic stabilization and maturation. Alterations in neuroligin-2 expression have previously been associated with changes in social behavior linked to psychiatric disorders, including schizophrenia and autism. In this study, we show that early-life stress, induced by limited nesting and bedding material, leads to impaired social recognition and increased aggression in adult mice, accompanied by increased expression levels of hippocampal neuroligin-2. Viral overexpression of hippocampal neuroligin-2 in adulthood mimics early-life stress-induced alterations in social behavior and social cognition. Moreover, viral knockdown of neuroligin-2 in the adult hippocampus attenuates the early-life stress-induced behavioral changes. Our results highlight the importance of neuroligin-2 in mediating early-life stress effects on social behavior and social cognition and its promising role as a novel therapeutic target for neuropsychiatric disorders.


Assuntos
Agressão/fisiologia , Comportamento Animal , Moléculas de Adesão Celular Neuronais/genética , Hipocampo/metabolismo , Proteínas do Tecido Nervoso/genética , RNA Mensageiro/metabolismo , Comportamento Social , Percepção Social , Estresse Psicológico/genética , Agressão/psicologia , Animais , Moléculas de Adesão Celular Neuronais/metabolismo , Técnicas de Silenciamento de Genes , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Estresse Psicológico/metabolismo , Estresse Psicológico/psicologia
6.
Neuropsychopharmacology ; 40(5): 1222-33, 2015 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-25409593

RESUMO

Stress-induced psychiatric disorders, such as depression, have recently been linked to changes in glutamate transmission in the central nervous system. Glutamate signaling is mediated by a range of receptors, including metabotropic glutamate receptors (mGluRs). In particular, mGluR subtype 5 (mGluR5) is highly implicated in stress-induced psychopathology. The major scaffold protein Homer1 critically interacts with mGluR5 and has also been linked to several psychopathologies. Yet, the specific role of Homer1 in this context remains poorly understood. We used chronic social defeat stress as an established animal model of depression and investigated changes in transcription of Homer1a and Homer1b/c isoforms and functional coupling of Homer1 to mGluR5. Next, we investigated the consequences of Homer1 deletion, overexpression of Homer1a, and chronic administration of the mGluR5 inverse agonist CTEP (2-chloro-4-((2,5-dimethyl-1-(4-(trifluoromethoxy)phenyl)-1H-imidazol-4-yl)ethynyl)pyridine) on the effects of chronic stress. In mice exposed to chronic stress, Homer1b/c, but not Homer1a, mRNA was upregulated and, accordingly, Homer1/mGluR5 coupling was disrupted. We found a marked hyperactivity behavior as well as a dysregulated hypothalamic-pituitary-adrenal axis activity in chronically stressed Homer1 knockout (KO) mice. Chronic administration of the selective and orally bioavailable mGluR5 inverse agonist, CTEP, was able to recover behavioral alterations induced by chronic stress, whereas overexpression of Homer1a in the hippocampus led to an increased vulnerability to chronic stress, reflected in an increased physiological response to stress as well as enhanced depression-like behavior. Overall, our results implicate the glutamatergic system in the emergence of stress-induced psychiatric disorders, and support the Homer1/mGluR5 complex as a target for the development of novel antidepressant agents.


Assuntos
Proteínas de Transporte/metabolismo , Transtorno Depressivo/metabolismo , Receptor de Glutamato Metabotrópico 5/metabolismo , Resiliência Psicológica , Estresse Psicológico/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Proteínas de Transporte/genética , Doença Crônica , Modelos Animais de Doenças , Dominação-Subordinação , Agonismo Inverso de Drogas , Antagonistas de Aminoácidos Excitatórios/farmacologia , Proteínas de Arcabouço Homer , Imidazóis/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Isoformas de Proteínas , Piridinas/farmacologia , RNA Mensageiro/metabolismo , Receptor de Glutamato Metabotrópico 5/antagonistas & inibidores , Resiliência Psicológica/efeitos dos fármacos
7.
PLoS One ; 9(4): e95796, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24759731

RESUMO

Psychiatric disorders such as depressive disorders and posttraumatic stress disorder are a major disease burden worldwide and have a higher incidence in women than in men. However, the underlying mechanism responsible for the sex-dependent differences is not fully understood. Besides environmental factors such as traumatic life events or chronic stress, genetic variants contribute to the development of such diseases. For instance, variations in the gene encoding the FK506 binding protein 51 (FKBP51) have been repeatedly associated with mood and anxiety. FKBP51 is a negative regulator of the glucocorticoid receptor and thereby of the hypothalamic-pituitary-adrenal axis that also interacts with other steroid hormone receptors such as the progesterone and androgen receptors. Thus, the predisposition of women to psychiatric disorders and the interaction of female hormones with FKBP51 and the glucocorticoid receptor implicate a possible difference in the regulation of the hypothalamic-pituitary-adrenal axis in female FKBP51 knockout (51KO) mice. Therefore, we investigated neuroendocrine, behavioural and physiological alterations relevant to mood disorders in female 51KO mice. Female 51KOs and wild type littermates were subjected to various behavioural tests, including the open field, elevated plus maze and forced swim test. The neuroendocrine profile was investigated under basal conditions and in response to an acute stressor. Furthermore, we analysed the mRNA expression levels of the glucocorticoid receptor and corticotrophin release hormone in different brain regions. Overall, female 51KO mice did not display any overt behavioural phenotype under basal conditions, but showed a reduced basal hypothalamic-pituitary-adrenal axis activity, a blunted response to, and an enhanced recovery from, acute stress. These characteristics strongly overlap with previous studies in male 51KO mice indicating that FKBP51 shapes the behavioural and neuroendocrine phenotype independent of the sex of the individual.


Assuntos
Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Proteínas de Ligação a Tacrolimo/metabolismo , Animais , Comportamento Animal/fisiologia , Encéfalo/metabolismo , Hormônio Liberador da Corticotropina/genética , Ciclo Estral/genética , Ciclo Estral/fisiologia , Feminino , Hipocampo/metabolismo , Hibridização In Situ , Camundongos , Camundongos Knockout , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptores de Glucocorticoides/genética , Estresse Psicológico/genética , Estresse Psicológico/metabolismo , Proteínas de Ligação a Tacrolimo/genética
8.
Horm Behav ; 53(2): 386-94, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18096163

RESUMO

The period of adolescence is characterized by a high vulnerability to stress and trauma, which might result in long-lasting consequences and an increased risk to develop psychiatric disorders. Using a recently developed mouse model for chronic social stress during adolescence, we studied persistent neuroendocrine and behavioral effects of chronic social stress obtained 12 months after cessation of the stressor. As a reference, we investigated immediate effects of chronic stress exposure obtained at the end of the chronic stress period. Immediately after the 7 week chronic stress period stressed animals show significantly increased adrenal weights, decreased thymus weight, increased basal corticosterone secretion and a flattened circadian rhythm. Furthermore, stressed animals display an increased anxiety-like behavior in the elevated plus maze and the novelty-induced suppression of feeding test. Hippocampal mineralocorticoid receptor (MR) and the glucocorticoid receptor (GR) mRNA levels were significantly decreased. To investigate persistent consequences of this early stressful experience, the same parameters were assessed in aged mice 12 months after the cessation of the stressor. Interestingly, we still found differences between formerly stressed and control mice in important stress-related parameters. MR expression levels were significantly lower in stressed animals, suggesting lasting, possibly epigenetic alterations in gene expression regulation. Furthermore, we observed long-term behavioral alterations in animals stressed during adolescence. Thus, we could demonstrate that chronic stress exposure during a crucial developmental time period results in long-term, persistent effects on physiological and behavioral parameters throughout life, which may contribute to an enhanced vulnerability to stress-induced diseases.


Assuntos
Comportamento Animal/fisiologia , Corticosterona/sangue , Hipocampo/metabolismo , Receptores de Mineralocorticoides/metabolismo , Estresse Psicológico/fisiopatologia , Glândulas Suprarrenais/anatomia & histologia , Fatores Etários , Animais , Ansiedade/etiologia , Ansiedade/fisiopatologia , Doença Crônica , Ritmo Circadiano/fisiologia , Modelos Animais de Doenças , Seguimentos , Masculino , Camundongos , Tamanho do Órgão , RNA Mensageiro/análise , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Receptores de Mineralocorticoides/genética , Estresse Psicológico/complicações , Timo/anatomia & histologia , Fatores de Tempo
9.
J Endocrinol ; 197(2): 421-7, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18434372

RESUMO

The function of the hypothalamic-pituitary-adrenal (HPA) axis of the neonatal mouse or rat is characterized by a period of quiescence, where mild stimuli are unable to elicit a pronounced increase in circulating corticosterone. A disruption of this period by maternal separation has been shown to result in a variety of long-term consequences, including neuroendocrine and behavioral disturbances. We have recently shown that peripheral metabolic markers like glucose or ghrelin are altered by maternal separation and that these changes precede the effects on corticosterone secretion. In the current study, we investigated whether the initial activation of the HPA axis is mediated via neuropeptide Y (NPY). To test this hypothesis, we studied the effects of an 8 h maternal separation in NPY-deficient mice. In addition, we compared the effect of the genotype with the previously described pharmacological effect of a ghrelin receptor antagonist. We could show that the peripheral response to maternal separation is decreased in NPY heterozygous and homozygous animals. In addition, maternal separation effects on corticotropin releasing hormone and glucocorticoid receptor expression in the brain were prevented in NPY-deficient pups. These effects were similar to a pharmacological ghrelin receptor blockade. We conclude that metabolic signals via an NPY-mediated pathway play a crucial role in activating the stress system of the neonatal mouse.


Assuntos
Sistema Hipotálamo-Hipofisário/fisiologia , Privação Materna , Neuropeptídeo Y/fisiologia , Sistema Hipófise-Suprarrenal/fisiologia , Hormônio Adrenocorticotrópico/farmacologia , Animais , Animais Recém-Nascidos , Arginina Vasopressina/genética , Sequência de Bases , Glicemia/análise , Corticosterona/sangue , Hormônio Liberador da Corticotropina/genética , Feminino , Camundongos , Dados de Sequência Molecular , Gravidez , RNA Mensageiro/análise , Receptores de Glucocorticoides/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa