72
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Neurogenesis-dependent and -independent effects of fluoxetine in an animal model of anxiety/depression.

      Neuron
      Analysis of Variance, Animals, Antidepressive Agents, Second-Generation, therapeutic use, Anxiety, chemically induced, diet therapy, pathology, Arrestins, deficiency, genetics, metabolism, Bromodeoxyuridine, Cell Proliferation, drug effects, radiation effects, Cell Survival, Corticosterone, toxicity, Depression, drug therapy, Disease Models, Animal, Drug Administration Schedule, Exploratory Behavior, Feeding Behavior, Fluoxetine, GTP-Binding Protein alpha Subunit, Gi2, Gene Expression Regulation, Hippocampus, Hypothalamus, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Microtubule-Associated Proteins, Neurogenesis, Neuropeptides, RNA, Messenger, Radiation, Reaction Time

      Read this article at

      ScienceOpenPublisherPMC
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Understanding the physiopathology of affective disorders and their treatment relies on the availability of experimental models that accurately mimic aspects of the disease. Here we describe a mouse model of an anxiety/depressive-like state induced by chronic corticosterone treatment. Furthermore, chronic antidepressant treatment reversed the behavioral dysfunctions and the inhibition of hippocampal neurogenesis induced by corticosterone treatment. In corticosterone-treated mice where hippocampal neurogenesis is abolished by X-irradiation, the efficacy of fluoxetine is blocked in some, but not all, behavioral paradigms, suggesting both neurogenesis-dependent and -independent mechanisms of antidepressant action. Finally, we identified a number of candidate genes, the expression of which is decreased by chronic corticosterone and normalized by chronic fluoxetine treatment selectively in the hypothalamus. Importantly, mice deficient in one of these genes, beta-arrestin 2, displayed a reduced response to fluoxetine in multiple tasks, suggesting that beta-arrestin signaling is necessary for the antidepressant effects of fluoxetine.

          Related collections

          Author and article information

          Comments

          Comment on this article