Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters











Database
Language
Publication year range
1.
Int J Neuropsychopharmacol ; 22(6): 372-382, 2019 06 03.
Article in English | MEDLINE | ID: mdl-31038173

ABSTRACT

BACKGROUND: Although depression is the leading cause of disability worldwide, its pathophysiology is poorly understood. Our previous study showed that hippocampal peroxisome proliferator-activated receptor δ (PPARδ) overexpression displays antidepressive effect and enhances hippocampal neurogenesis during chronic stress. Herein, we further extended our curiosity to investigate whether downregulating PPARδ could cause depressive-like behaviors through downregulation of neurogenesis. METHODS: Stereotaxic injection of lentiviral vector, expressing short hairpin RNA complementary to the coding exon of PPARδ, was done into the bilateral dentate gyri of the hippocampus, and the depression-like behaviors were observed in mice. Additionally, hippocampal neurogenesis, brain-derived neurotrophic factor and cAMP response element-binding protein were measured both in vivo and in vitro. RESULTS: Hippocampal PPARδ knockdown caused depressive-like behaviors and significantly decreased neurogenesis, neuronal differentiation, levels of mature brain-derived neurotrophic factor and phosphorylated cAMP response element-binding protein in the hippocampus. In vitro study further confirmed that PPARδ knockdown could inhibit proliferation and differentiation of neural stem cells. Furthermore, these effects were mimicked by repeated systemic administration of a PPARδ antagonist, GSK0660 (1 or 3 mg/kg i.p. for 21 d). CONCLUSIONS: These findings suggest that downregulation of hippocampal PPARδ is associated with depressive behaviors in mice through an inhibitory effect on cAMP response element-binding protein/brain-derived neurotrophic factor-mediated adult neurogenesis in the hippocampus, providing new insights into the pathogenesis of depression.


Subject(s)
Behavior, Animal/physiology , Dentate Gyrus/metabolism , Depression/physiopathology , Neurogenesis/drug effects , PPAR delta/physiology , Animals , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/metabolism , CREB-Binding Protein/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Dentate Gyrus/drug effects , Depression/chemically induced , Down-Regulation/drug effects , Gene Knockdown Techniques , Male , Mice , Neural Stem Cells/drug effects , PPAR delta/genetics , RNA, Small Interfering/pharmacology , Sulfones/pharmacology , Thiophenes/pharmacology
2.
Int J Neuropsychopharmacol ; 19(1)2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26362775

ABSTRACT

BACKGROUND: Emerging data have demonstrated that peroxisome proliferator-activated receptor δ (PPARδ) activation confers a potentially neuroprotective role in some neurodegenerative diseases. However, whether PPARδ is involved in depression is unknown. METHODS: In this study, PPARδ was firstly investigated in the chronic mild stress (CMS) and learned helplessness (LH) models of depression. The changes in depressive behaviors and hippocampal neurogenesis were investigated after PPARδ overexpression by microinfusion of the lentiviral vector, containing the coding sequence of mouse PPARδ (LV-PPARδ), into the bilateral dentate gyri of the hippocampus or PPARδ activation by repeated systemic administration of PPARδ agonist GW0742 (5 or 10mg/kg.d, i.p., for 21 d). RESULTS: We found that both CMS and LH resulted in a significant decrease in the PPARδ expression in the hippocampi of mice, and this change was reversed by treatment with the antidepressant fluoxetine. PPARδ overexpression and PPARδ activation each suppressed the CMS- and LH-induced depressive-like behavior and produced an antidepressive effect. In vivo or in vitro studies also showed that both overexpression and activation of PPARδ enhanced proliferation or differentiation of neural stem cells in the hippocampi of mice. CONCLUSIONS: These results suggest that hippocampal PPARδ upregulation represses stress-induced depressive behaviors, accompanied by enhancement of neurogenesis.


Subject(s)
Depression/complications , Depression/pathology , Hippocampus/metabolism , Neurogenesis/physiology , PPAR delta/metabolism , Stress, Psychological/complications , Animals , Cell Differentiation , Disease Models, Animal , Gene Expression Regulation/physiology , Glial Fibrillary Acidic Protein/metabolism , Hindlimb Suspension , Male , Mice , Mice, Inbred ICR , Neural Stem Cells/physiology , PPAR delta/genetics , Phosphopyruvate Hydratase/metabolism , Sincalide/metabolism , Thiazoles/pharmacology , Time Factors , Transduction, Genetic
3.
Neurobiol Aging ; 35(3): 590-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24269024

ABSTRACT

Accumulation of amyloid-ß (Aß) is thought to be associated with the progressive neuronal death observed in Alzheimer's disease, but the mechanisms underlying neurotoxicity triggered by Aß remain elusive. In the current study, we investigated the roles of cysteinyl leukotriene receptor 1 (CysLT1R) in Aß1-42-induced neurotoxicity in vitro or in vivo. In vitro exposure of mouse primary neurons to Aß1-42 caused a gradual increases in CysLT1R expression. In vivo bilateral intrahippocampal injection of Aß1-42 also elicited time-dependent increases of CysLT1R expression in the hippocampus and cortex of mice. The CysLT1R antagonist pranlukast not only reversed Aß1-42-induced upregulation of CysLT1R, but also suppressed Aß1-42-triggered neurotoxicity evidenced by enhanced nuclear factor-kappa B p65, activated caspase-3, decreased B-cell lymphoma-2 and cell viability and impaired memory. Furthermore, chronic treatment with pranlukast produced similar beneficial effects on memory behavior and hippocampal long-term potentiation to memantine or donepezil in intrahippocampal Aß1-42-injected mice. Our data indicate that CysLT1R is involved in Aß1-42-induced neurotoxicity, and that blockade of CysLT1R, such as application of CysLT1R antagonist, could be a novel and promising strategy for the treatment of Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/toxicity , Cerebral Cortex/metabolism , Gene Expression/drug effects , Hippocampus/metabolism , Neurons/drug effects , Neurons/metabolism , Peptide Fragments/toxicity , Receptors, Leukotriene/genetics , Receptors, Leukotriene/physiology , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cells, Cultured , Cerebral Cortex/cytology , Chromones/pharmacology , Chromones/therapeutic use , Hippocampus/cytology , Leukotriene Antagonists/pharmacology , Leukotriene Antagonists/therapeutic use , Male , Memory , Mice , Mice, Inbred ICR , Mice, Transgenic , Molecular Targeted Therapy , NF-kappa B/metabolism , Neurons/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Leukotriene/metabolism , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL