Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Neuropsychopharmacol ; 21(2): 175-186, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29020410

ABSTRACT

Background: Previous studies have shown that a low dose of scopolamine produces rapid-acting antidepressant-like actions in rodents. Understanding the mechanisms underlying this effect and the dose-dependent variations of drug responses remains an important task. L-type voltage-dependent calcium channels were found to mediate rapid-acting antidepressant effects of certain medications (e.g., ketamine). Therefore, it is of great interest to determine the involvement of L-type voltage-dependent calcium channels in the action of scopolamine. Methods: Herein, we investigated the mechanisms underlying behavioral responses to various doses of scopolamine in mice to clarify the involvement of L-type voltage-dependent calcium channels in its modes of action. Open field test, novel object recognition test, and forced swimming test were performed on mice administered varied doses of scopolamine (0.025, 0.05, 0.1, 1, and 3 mg/kg, i.p.) alone or combined with L-type voltage-dependent calcium channel blocker verapamil (5 mg/kg, i.p.). Then, the changes in brain-derived neurotrophic factor and neuropeptide VGF (nonacronymic) levels in the hippocampus and prefrontal cortex of these mice were analyzed. Results: Low doses of scopolamine (0.025 and 0.05 mg/kg) produced significant antidepressant-like effects in the forced swimming test, while higher doses (1 and 3 mg/kg) resulted in significant memory deficits and depressive-like behaviors. Moreover, the behavioral changes in responses to various doses may be related to the upregulation (0.025 and 0.05 mg/kg) and downregulation (1 and 3 mg/kg) of brain-derived neurotrophic factor and VGF in the hippocampus and prefrontal cortex in mice. We further found that the rapid-acting antidepressant-like effects and the upregulation on brain-derived neurotrophic factor and VGF produced by a low dose of scopolamine (0.025 mg/kg) were completely blocked by verapamil. Conclusions: These results indicate that L-type voltage-dependent calcium channels are likely involved in the behavioral changes in response to various doses of scopolamine through the regulation of brain-derived neurotrophic factor and VGF levels.


Subject(s)
Antidepressive Agents/pharmacology , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/drug effects , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/drug effects , Cognitive Dysfunction/chemically induced , Depression/chemically induced , Hippocampus/drug effects , Neuropeptides/drug effects , Prefrontal Cortex/drug effects , Scopolamine/pharmacology , Verapamil/pharmacology , Animals , Antidepressive Agents/administration & dosage , Calcium Channel Blockers/administration & dosage , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Nerve Growth Factors , Scopolamine/administration & dosage , Verapamil/administration & dosage
2.
J Neuroinflammation ; 14(1): 190, 2017 Sep 20.
Article in English | MEDLINE | ID: mdl-28931410

ABSTRACT

BACKGROUND: Recent evidence has suggested that peripheral inflammatory responses induced by lipopolysaccharides (LPS) play an important role in neuropsychiatric dysfunction in rodents. Interleukin-1ß (IL-1ß), a pro-inflammatory cytokine, has been proposed to be a key mediator in a variety of behavioral dysfunction induced by LPS in mice. Thus, inhibition of IL-1ß may have a therapeutic benefit in the treatment of neuropsychiatric disorders. However, the precise underlying mechanism of knock-down of IL-1ß in repairing behavioral changes by LPS remains unclear. METHODS: The mice were treated with either IL-1ß shRNA lentivirus or non-silencing shRNA control (NS shRNA) lentivirus by microinjection into the dentate gyrus (DG) regions of the hippocampus. After 7 days of recovery, LPS (1 mg/kg, i.p.) or saline was administered. The behavioral task for memory deficits was conducted in mice by the novel object recognition test (NORT), the anxiety-like behaviors were evaluated by the elevated zero maze (EZM), and the depression-like behaviors were examined by the sucrose preference test (SPT) and the forced swimming test (FST). Furthermore, the levels of malondialdehyde (MDA), superoxide dismutase (SOD), nuclear factor erythroid-derived 2-like 2 (Nrf2), heme oxygenase 1 (HO1), IL-1ß, tumor necrosis factor (TNF-α), neuropeptide VGF (non-acronymic), and brain-derived neurotrophic factor (BDNF) were assayed. RESULTS: Our results demonstrated that IL-1ß knock-down in the hippocampus significantly attenuated the memory deficits and anxiety- and depression-like behaviors induced by LPS in mice. In addition, IL-1ß knock-down ameliorated the oxidative and neuroinflammatory responses and abolished the downregulation of VGF and BDNF induced by LPS. CONCLUSIONS: Collectively, our findings suggest that IL-1ß is necessary for the oxidative and neuroinflammatory responses produced by LPS and offers a novel drug target in the IL-1ß/oxidative/neuroinflammatory/neurotrophic pathway for treating neuropsychiatric disorders that are closely associated with neuroinflammation, oxidative stress, and the downregulation of VGF and BDNF.


Subject(s)
Behavior, Animal/physiology , Hippocampus/immunology , Hippocampus/metabolism , Inflammation/complications , Interleukin-1beta/metabolism , Animals , Anxiety/immunology , Anxiety/metabolism , Behavior, Animal/drug effects , Depression/immunology , Depression/metabolism , Gene Knockdown Techniques , Inflammation/chemically induced , Lentivirus , Lipopolysaccharides/toxicity , Male , Memory Disorders/immunology , Memory Disorders/metabolism , Mice
3.
ACS Chem Neurosci ; 8(9): 2005-2018, 2017 09 20.
Article in English | MEDLINE | ID: mdl-28594546

ABSTRACT

Peripheral inflammatory responses affect central nervous system (CNS) function, manifesting in symptoms of memory deficits, depression, and anxiety. Previous studies have revealed that neuropeptide VGF (nonacronymic) C-terminal peptide TLQP-62 rapidly reinforces brain-derived neurotrophic factor (BDNF)/tropomyosin receptor kinase B (TrkB) signaling, regulating memory consolidation and antidepressant-like action. However, whether it is beneficial for lipopolysaccharide (LPS)-induced neuropsychiatric dysfunction in mice is unknown. Herein, we explored the involvement of BDNF/TrkB signaling and biochemical alterations in inflammatory or oxidative stress markers in the alleviating effects of TLQP-62 on LPS-induced neuropsychiatric dysfunction. The mice were treated with TLQP-62 (2 µg/side) via intracerebroventricular (i.c.v.) injection 1 h before LPS (0.5 mg/kg, i.p.) administration. Our results showed that a single treatment with LPS (0.5 mg/kg, i.p) is sufficient to produce recognition memory deficits (in the novel object recognition test), depression-like behavior (in the forced swim test and sucrose preference test), and anxiety-like behavior (in the elevated zero maze). However, pretreatment with TLQP-62 prevented LPS-induced behavioral dysfunction, neuroinflammatory, and oxidative responses. In addition, our results further demonstrated that a reduction in BDNF expression mediated by BDNF-shRNA lentivirus significantly blocked the effects of TLQP-62, suggesting the critical role of BDNF/TrkB signaling in the neuroprotective effects of TLQP-62 in the mice. In conclusion, TLQP-62 could be a therapeutic approach for neuropsychiatric disorders, which are closely associated with neuroinflammation and oxidative stress.


Subject(s)
Anti-Anxiety Agents/pharmacology , Anti-Inflammatory Agents/pharmacology , Antidepressive Agents/pharmacology , Nootropic Agents/pharmacology , Peptides/pharmacology , Animals , Anxiety/drug therapy , Anxiety/immunology , Brain/drug effects , Brain/immunology , Brain-Derived Neurotrophic Factor/metabolism , Depressive Disorder/drug therapy , Depressive Disorder/immunology , Disease Models, Animal , Escherichia coli , Lipopolysaccharides , Male , Memory Disorders/drug therapy , Memory Disorders/immunology , Mice, Inbred ICR , Motor Activity/drug effects , Motor Activity/physiology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Receptor, trkB/metabolism , Recognition, Psychology/drug effects , Recognition, Psychology/physiology , Signal Transduction/drug effects
4.
Pharmacol Biochem Behav ; 157: 47-57, 2017 06.
Article in English | MEDLINE | ID: mdl-28216067

ABSTRACT

Preclinical and clinical studies suggest that neuronal muscarinic acetylcholine receptor (M-AchR) antagonists have antidepressant-like properties. Despite the recent interest in bicaudal C homolog 1 gene (BICC1) as a target for the treatment of depression, the upstream signaling molecules that regulate BICC1 are unknown, and very few studies have addressed the involvement of BICC1 in the antidepressant-like effects of the selective M1-AchR inhibitor, biperiden. Growing evidence indicates that activation of brain-derived neurotrophic factor (BDNF)/tropomyosin-related kinase receptor B (TrkB) signaling may be involved in antidepressant-like activities. In this study, we investigated the role of BDNF/TrkB signaling in the regulation of BICC1 expression in the chronic unpredictable stress (CUS) mouse model of depression. Furthermore, we also examined whether BDNF/TrkB signaling contributes to the antidepressant-like effects of biperiden via down-regulation of BICC1 in the hippocampus and prefrontal cortex of mice. Our current data show that CUS exposure induced significant depression-like behaviors, down-regulation of BDNF/TrkB signaling and up-regulation of BICC1 in the hippocampus and prefrontal cortex of mice. However, biperiden significantly alleviated the CUS-induced abnormalities. Moreover, we found that the effects of biperiden were antagonized by pretreatment with the TrkB antagonist K252a. Our results indicate that BDNF/TrkB signaling may be the major upstream mediator of BICC1 involvement in the antidepressant-like effects of biperiden.


Subject(s)
Antidepressive Agents/therapeutic use , Biperiden/therapeutic use , Brain-Derived Neurotrophic Factor/metabolism , Hippocampus/metabolism , Prefrontal Cortex/metabolism , RNA-Binding Proteins/biosynthesis , Receptor, trkB/metabolism , Animals , Antidepressive Agents/pharmacology , Biperiden/pharmacology , Brain-Derived Neurotrophic Factor/antagonists & inhibitors , Carbazoles/pharmacology , Depression/drug therapy , Depression/metabolism , Hippocampus/drug effects , Indole Alkaloids/pharmacology , Locomotion/drug effects , Locomotion/physiology , Male , Mice , Mice, Inbred ICR , Prefrontal Cortex/drug effects , Receptor, trkB/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology , Stress, Psychological/drug therapy , Stress, Psychological/metabolism , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...