Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 533
Filtrer
1.
Science ; 385(6709): eado7010, 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39116252

RÉSUMÉ

Ketamine has been found to have rapid and potent antidepressant activity. However, despite the ubiquitous brain expression of its molecular target, the N-methyl-d-aspartate receptor (NMDAR), it was not clear whether there is a selective, primary site for ketamine's antidepressant action. We found that ketamine injection in depressive-like mice specifically blocks NMDARs in lateral habenular (LHb) neurons, but not in hippocampal pyramidal neurons. This regional specificity depended on the use-dependent nature of ketamine as a channel blocker, local neural activity, and the extrasynaptic reservoir pool size of NMDARs. Activating hippocampal or inactivating LHb neurons swapped their ketamine sensitivity. Conditional knockout of NMDARs in the LHb occluded ketamine's antidepressant effects and blocked the systemic ketamine-induced elevation of serotonin and brain-derived neurotrophic factor in the hippocampus. This distinction of the primary versus secondary brain target(s) of ketamine should help with the design of more precise and efficient antidepressant treatments.


Sujet(s)
Antidépresseurs , Dépression , Habénula , Kétamine , Récepteurs du N-méthyl-D-aspartate , Animaux , Mâle , Souris , Antidépresseurs/pharmacologie , Facteur neurotrophique dérivé du cerveau/métabolisme , Facteur neurotrophique dérivé du cerveau/génétique , Dépression/traitement médicamenteux , Dépression/métabolisme , Habénula/effets des médicaments et des substances chimiques , Habénula/métabolisme , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Kétamine/pharmacologie , Kétamine/administration et posologie , Souris de lignée C57BL , Souris knockout , Cellules pyramidales/effets des médicaments et des substances chimiques , Cellules pyramidales/métabolisme , Récepteurs du N-méthyl-D-aspartate/antagonistes et inhibiteurs , Récepteurs du N-méthyl-D-aspartate/génétique , Sérotonine/métabolisme
3.
Chem Biol Interact ; 400: 111182, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-39098740

RÉSUMÉ

Depression is a severe mental illness affecting patient's physical and mental health. However, long-term effects of existing therapeutic modalities for depression are not satisfactory. Geniposide is an iridoid compound highly expressed in gardenia jasminoides for removing annoyance. The activity of geniposide against depression has been widely studied while most studies concentrated on the expression levels of gene and protein. Herein, the aim of the present study was to employ non-target metabolomic platform of serum to investigate metabolic changes of depression mice and further verify in hippocampus for analyzing the antidepressant mechanism of geniposide. Then we discovered that 9 metabolites of serum were significantly increased in depressive group (prostaglandin E2, leukotriene C4, arachidonic acid, phosphatidylcholine (PC, 16:0/16:0), LysoPC (18:1 (9Z)/0:0), phosphatidylethanolamine (14:0/16:0), creatine, oleamide and aminomalonic acid) and 6 metabolites were decreased (indoxylsulfuric acid, testosterone, lactic acid, glucose 6-phosphate, leucine and valine). The levels of arachidonic acid, LysoPC, lactic acid and glucose 6-phosphate in hippocampus were consistent change with serum in depression mice. Most of them showed significant tendencies to be normal by geniposide treatment. Metabolic pathway analysis indicated that arachidonic acid metabolism and glucose metabolism were the main pathogenesis for the antidepressant effect of geniposide. In addition, the levels of serum tumor necrosis factor-α and interleukin-1 were increased in depressive mice and reversed after geniposide treatment. This study revealed that abnormal metabolism of inflammatory response and glucose metabolism of the serum and hippocampus involved in the occurrence of depressive disorder and antidepressant effect of geniposide.


Sujet(s)
Antidépresseurs , Dépression , Modèles animaux de maladie humaine , Glucose , Hippocampe , Inflammation , Iridoïdes , Animaux , Iridoïdes/pharmacologie , Iridoïdes/usage thérapeutique , Dépression/traitement médicamenteux , Dépression/métabolisme , Souris , Antidépresseurs/pharmacologie , Antidépresseurs/usage thérapeutique , Mâle , Hippocampe/métabolisme , Hippocampe/effets des médicaments et des substances chimiques , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Glucose/métabolisme , Métabolomique
4.
BMC Anesthesiol ; 24(1): 293, 2024 Aug 19.
Article de Anglais | MEDLINE | ID: mdl-39160473

RÉSUMÉ

BACKGROUND: The development of postpartum depression has been linked to fluctuations in the levels of neurotransmitters in the human body, such as 5-hydroxytryptamine (5-HT), dopamine (DA), noradrenaline (Norepinephrine, NE), and brain derived neurotrophic factor (BDNF). Research has indicated that the antidepressant effect of esketamine are mediated by monoamine transmitters and neurotrophic factors. Therefore, we postulate that intravenous administration of esketamine in patients with postpartum depression may alter the serum concentrations of these neurotransmitters. METHODS: Three hundred fifteen patients with postpartum depression were selected and divided into two groups based on randomized numerical expression: esketamine (E) group (0. 25 mg/kg esketamine) and control (C) group (a same volume of 0.9% saline), all the drugs were pumped for 40 min. After the end of drug pumping, all patients were continuously observed for 2 h. Changes in serum levels of 5-HT, DA, NE, BDNF were recorded before drug administration and on the 3rd day after drug administration. The scores of Edinburgh Postnatal Depression Scale (EPDS) were calculated before drug administration, and on the 3rd day and on the 30th day after drug administration. Dizziness, headache, nausea, vomiting, drowsiness, and feeling of detachment occurred were recorded within 2 h after drug administration. RESULTS: Before drug administration, the serum concentrations of 5-HT,DA,BDNF,NE in Group E and Group C were namely (0. 91 ± 0. 19 vs. 0. 98 ± 0. 21, P = 0. 181), (2. 38 ± 0. 35 vs. 2. 32 ± 0. 32, P = 0. 491), (3. 07 ± 0. 89 vs 3. 02 ± 0. 88, P = 0. 828), (39. 79 ± 7. 78 vs 41. 34 ± 10. 03, P = 0. 506). On the third day post-medication, the serum concentrations of 5-HT,DA,BDNF,NE in Group E and Group C were namely (1. 42 ± 0. 35 vs. 0. 96 ± 0. 24, P < 0. 001), (3. 99 ± 0. 17 vs. 2. 41 ± 0. 28, P < 0. 001),(5. 45 ± 0. 81 vs 3. 22 ± 0. 76, P < 0. 001),(44. 36 ± 9. 98 vs 40. 69 ± 11. 75, P = 0. 198). Before medication, the EPDS scores were (16. 15 ± 3. 02 vs 17. 85 ± 3. 89, P = 0. 064). on the third day after medication, the Group E had significantly reduced scores (12. 98 ± 2. 39 vs 16. 73 ± 3. 52, P < 0. 001). On the 30rd day after medication, EPDS scores between the two groups were (16. 34 ± 3. 43 vs 16. 91 ± 4. 02, p = 0. 203). Within 2 h of medication, the rate of adverse events was similar between the two groups. CONCLUSIONS: Small doses of esketamine can increase the serum concentration of 5-HT,DA,BDNF, and in the short term, decrease EPDS scores, and improve postpartum depressive symptoms. TRIAL REGISTRATION: Retrospectively registered in the Chinese Clinical Trial Registry (ChiCTR2300078343, 2023/12/05).


Sujet(s)
Facteur neurotrophique dérivé du cerveau , Dépression du postpartum , Kétamine , Agents neuromédiateurs , Sérotonine , Humains , Femelle , Kétamine/administration et posologie , Kétamine/pharmacologie , Dépression du postpartum/traitement médicamenteux , Dépression du postpartum/sang , Adulte , Agents neuromédiateurs/sang , Facteur neurotrophique dérivé du cerveau/sang , Sérotonine/sang , Antidépresseurs/administration et posologie , Antidépresseurs/usage thérapeutique , Antidépresseurs/pharmacologie , Norépinéphrine/sang , Dopamine/sang
5.
Neurosci Biobehav Rev ; 164: 105833, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39089420

RÉSUMÉ

Major Depressive Disorder (MDD) is characterized by at least one major depressive episode. It requires medical attention typically involving the prescription of antidepressants. Remission in MDD patients is often difficult to achieve because of the limited effectiveness of these drugs. Nowadays, numerous patients undergo various antidepressant treatments, with subjective changes in their personal experiences being regularly monitored. Therefore, it is essential to find clinical and objective tools that offer a more tailored approach to antidepressant selection. The neurochemistry of the retina being similar to the brain, one promising approach would be to use ElectroRetinoGraphy (ERG) measurements on MDD patients requiring antidepressant treatment. Thus, the aim of this scoping review is to highlight effects of different classes of antidepressants on retinal function evaluated by full-field ERG (ffERG), Pattern ERG (PERG) and multifocal ERG (mfERG) waveforms in MDD patients. These ERG measurements could serve as pivotal indicators in defining patient profiles, facilitating a more objective and personalized approach to therapeutic interventions, thereby advancing precision psychiatry.


Sujet(s)
Antidépresseurs , Trouble dépressif majeur , Électrorétinographie , Humains , Trouble dépressif majeur/traitement médicamenteux , Trouble dépressif majeur/physiopathologie , Antidépresseurs/pharmacologie , Antidépresseurs/usage thérapeutique , Rétine/effets des médicaments et des substances chimiques , Rétine/physiopathologie
6.
J Neuroimmune Pharmacol ; 19(1): 45, 2024 Aug 19.
Article de Anglais | MEDLINE | ID: mdl-39158758

RÉSUMÉ

Multiple Sclerosis (MS) is a debilitating disease that severely affects the central nervous system (CNS). Apart from neurological symptoms, it is also characterized by neuropsychiatric comorbidities, such as anxiety and depression. Phosphodiesterase-5 inhibitors (PDE5Is) such as Sildenafil and Tadalafil have been shown to possess antidepressant-like effects, but the mechanisms underpinning such effects are not fully characterized. To address this question, we used the EAE model of MS, behavioral tests, immunofluorescence, immunohistochemistry, western blot, and 16 S rRNA sequencing. Here, we showed that depressive-like behavior in Experimental Autoimmune Encephalomyelitis (EAE) mice is due to neuroinflammation, reduced synaptic plasticity, dysfunction in glutamatergic neurotransmission, glucocorticoid receptor (GR) resistance, increased blood-brain barrier (BBB) permeability, and immune cell infiltration to the CNS, as well as inflammation, increased intestinal permeability, and immune cell infiltration in the distal colon. Furthermore, 16 S rRNA sequencing revealed that behavioral dysfunction in EAE mice is associated with changes in the gut microbiota, such as an increased abundance of Firmicutes and Saccharibacteria and a reduction in Proteobacteria, Parabacteroides, and Desulfovibrio. Moreover, we detected an increased abundance of Erysipelotrichaceae and Desulfovibrionaceae and a reduced abundance of Lactobacillus johnsonii. Surprisingly, we showed that Tadalafil likely exerts antidepressant-like effects by targeting all aforementioned disease aspects. In conclusion, our work demonstrated that anxiety- and depressive-like behavior in EAE is associated with a plethora of neuroimmune and gut microbiota-mediated mechanisms and that Tadalafil exerts antidepressant-like effects probably by targeting these mechanisms. Harnessing the knowledge of these mechanisms of action of Tadalafil is important to pave the way for future clinical trials with depressed patients.


Sujet(s)
Anxiolytiques , Antidépresseurs , Axe cerveau-intestin , Encéphalomyélite auto-immune expérimentale , Microbiome gastro-intestinal , Souris de lignée C57BL , Inhibiteurs de la phosphodiestérase-5 , Tadalafil , Animaux , Tadalafil/pharmacologie , Tadalafil/usage thérapeutique , Inhibiteurs de la phosphodiestérase-5/pharmacologie , Inhibiteurs de la phosphodiestérase-5/usage thérapeutique , Souris , Encéphalomyélite auto-immune expérimentale/traitement médicamenteux , Encéphalomyélite auto-immune expérimentale/immunologie , Antidépresseurs/pharmacologie , Antidépresseurs/usage thérapeutique , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Axe cerveau-intestin/effets des médicaments et des substances chimiques , Axe cerveau-intestin/physiologie , Anxiolytiques/pharmacologie , Anxiolytiques/usage thérapeutique , Femelle , Dépression/traitement médicamenteux , Auto-immunité/effets des médicaments et des substances chimiques
7.
Int J Mol Sci ; 25(15)2024 Jul 29.
Article de Anglais | MEDLINE | ID: mdl-39125857

RÉSUMÉ

Neoponcirin causes anxiolytic-like effects in mice when administered intraperitoneally but not orally. Neoponcirin is non-water-soluble and insoluble in solvents, and in medium acid, it isomerizes, reducing its bioavailability. To improve the pharmacological properties of neoponcirin, we formed a neoponcirin complex with beta-cyclodextrin (NEO/ßCD), which was characterized by FT-IR, UV-Vis, and NMR, and their solubility profile. We evaluated the antidepressant-like effects of NEO/ßCD acutely administered to mice orally in the behavioral paradigms, the tail suspension (TST) and the forced swimming (FST) tests. We also analyzed the benefits of repeated oral doses of NEO/ßCD on depression- and anxiety-like behaviors induced in mice by chronic unpredictable mild stress (CUMS), using the FST, hole board, and open field tests. We determined the stressed mice's expression of stress-related inflammatory cytokines (IL-1ß, IL-6, and TNFα) and corticosterone. Results showed that a single or chronic oral administration of NEO/ßCD caused a robust antidepressant-like effect without affecting the ambulatory activity. In mice under CUMS, NEO/ßCD also produced anxiolytic-like effects and avoided increased corticosterone and IL-1ß levels. The effects of the NEO/ßCD complex were robust in both the acute and the stress chronic models, improving brain neurochemistry and recovering immune responses previously affected by prolonged stress.


Sujet(s)
Antidépresseurs , Dépression , Stress psychologique , Cyclodextrines bêta , Animaux , Cyclodextrines bêta/pharmacologie , Cyclodextrines bêta/composition chimique , Souris , Antidépresseurs/pharmacologie , Antidépresseurs/usage thérapeutique , Mâle , Stress psychologique/traitement médicamenteux , Dépression/traitement médicamenteux , Comportement animal/effets des médicaments et des substances chimiques , Cytokines/métabolisme , Modèles animaux de maladie humaine , Anxiété/traitement médicamenteux , Anxiolytiques/pharmacologie , Natation , Administration par voie orale
8.
PLoS One ; 19(8): e0308413, 2024.
Article de Anglais | MEDLINE | ID: mdl-39116153

RÉSUMÉ

BACKGROUND: Ketamine has recently attracted considerable attention for its rapid effects on patients with major depressive disorder, including treatment-resistant depression (TRD). Despite ketamine's promising results in treating depression, a significant number of patients do not respond to the treatment, and predicting who will benefit remains a challenge. Although its antidepressant effects are known to be linked to its action as an antagonist of the N-methyl-D-aspartate (NMDA) receptor, the precise mechanisms that determine why some patients respond and others do not are still unclear. OBJECTIVE: This study aims to understand the computational mechanisms underlying changes in the auditory mismatch negativity (MMN) response following treatment with intravenous ketamine. Moreover, we aim to link the computational mechanisms to their underlying neural causes and use the parameters of the neurocomputational model to make individual treatment predictions. METHODS: This is a prospective study of 30 patients with TRD who are undergoing intravenous ketamine therapy. Prior to 3 out of 4 ketamine infusions, EEG will be recorded while patients complete the auditory MMN task. Depression, suicidality, and anxiety will be assessed throughout the study and a week after the last ketamine infusion. To translate the effects of ketamine on the MMN to computational mechanisms, we will model changes in the auditory MMN using the hierarchical Gaussian filter, a hierarchical Bayesian model. Furthermore, we will employ a conductance-based neural mass model of the electrophysiological data to link these computational mechanisms to their neural causes. CONCLUSION: The findings of this study may improve understanding of the mechanisms underlying response and resistance to ketamine treatment in patients with TRD. The parameters obtained from fitting computational models to EEG recordings may facilitate single-patient treatment predictions, which could provide clinically useful prognostic information. TRIAL REGISTRATION: Clinicaltrials.gov NCT05464264. Registered June 24, 2022.


Sujet(s)
Trouble dépressif résistant aux traitements , Électroencéphalographie , Kétamine , Kétamine/usage thérapeutique , Kétamine/pharmacologie , Kétamine/administration et posologie , Humains , Trouble dépressif résistant aux traitements/traitement médicamenteux , Trouble dépressif résistant aux traitements/physiopathologie , Adulte , Études prospectives , Antidépresseurs/usage thérapeutique , Antidépresseurs/pharmacologie , Mâle , Femelle , Potentiels évoqués auditifs/effets des médicaments et des substances chimiques , Trouble dépressif majeur/traitement médicamenteux , Trouble dépressif majeur/physiopathologie , Résultat thérapeutique , Récepteurs du N-méthyl-D-aspartate/antagonistes et inhibiteurs , Adulte d'âge moyen , Jeune adulte
9.
Cells ; 13(15)2024 Jul 26.
Article de Anglais | MEDLINE | ID: mdl-39120293

RÉSUMÉ

Major depressive disorder (MDD) is a complex and devastating illness that affects people of all ages. Despite the large use of antidepressants in current medical practice, neither their mechanisms of action nor the aetiology of MDD are completely understood. Experimental evidence supports the involvement of Parvalbumin-positive GABAergic neurons (PV-neurons) in the pathogenesis of MDD. DLX5 and DLX6 (DLX5/6) encode two homeodomain transcription factors involved in cortical GABAergic differentiation and function. In the mouse, the level of expression of these genes is correlated with the cortical density of PV-neurons and with anxiety-like behaviours. The same genomic region generates the lncRNA DLX6-AS1, which, in humans, participates in the GABAergic regulatory module downregulated in schizophrenia and ASD. Here, we show that the expression levels of Dlx5/6 in the adult mouse brain are correlated with the immobility time in the forced swim test, which is used to measure depressive-like behaviours. We show that the administration of the antidepressant fluoxetine (Flx) to normal mice induces, within 24 h, a rapid and stable reduction in Dlx5, Dlx6 and Dlx6-AS1 expression in the cerebral cortex through the activation of the TrkB-CREB pathway. Experimental Dlx5 overexpression counteracts the antidepressant effects induced by Flx treatment. Our findings show that one of the short-term effects of Flx administration is the reduction in Dlx5/6 expression in GABAergic neurons, which, in turn, has direct consequences on PV expression and on behavioural profiles. Variants in the DLX5/6 regulatory network could be implicated in the predisposition to depression and in the variability of patients' response to antidepressant treatment.


Sujet(s)
Antidépresseurs , Cortex cérébral , Fluoxétine , Neurones GABAergiques , Protéines à homéodomaine , Récepteur trkB , Animaux , Neurones GABAergiques/métabolisme , Neurones GABAergiques/effets des médicaments et des substances chimiques , Protéines à homéodomaine/métabolisme , Protéines à homéodomaine/génétique , Fluoxétine/pharmacologie , Fluoxétine/usage thérapeutique , Souris , Antidépresseurs/pharmacologie , Antidépresseurs/usage thérapeutique , Cortex cérébral/effets des médicaments et des substances chimiques , Cortex cérébral/anatomopathologie , Cortex cérébral/métabolisme , Récepteur trkB/métabolisme , Récepteur trkB/génétique , Mâle , Transduction du signal/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Trouble dépressif majeur/traitement médicamenteux , Trouble dépressif majeur/métabolisme , Trouble dépressif majeur/anatomopathologie , Trouble dépressif majeur/génétique
10.
Nutrients ; 16(15)2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39125295

RÉSUMÉ

Type 2 diabetes and depression co-occur in a bidirectional manner. Curcumin supplements exhibit antidepressant effects that may mitigate depression by modulating neurotransmitters and reducing inflammatory and oxidative stress pathways. This study aimed to evaluate the efficacy of curcumin in improving depression severity in obese type 2 diabetes patients. The study employed a randomized, double-blind, placebo-controlled trial design with 227 participants. The primary end-point was depression severity assessed using the Patient Health Questionnaire-9. Biomarkers were measured at baseline and at 3-, 6-, 9-, and 12-month intervals. The biomarkers assessed were serotonin levels, pro-inflammatory cytokines (interleukin-1 beta, interleukin-6, tumor necrosis factor-alpha), antioxidant activities (total antioxidant status, glutathione peroxidase, and superoxide dismutase), and malondialdehyde. After 12 months, the curcumin group exhibited significantly improved depression severity (p = 0.000001). The curcumin group had higher levels of serotonin (p < 0.0001) but lower levels of interleukin-1 beta, interleukin-6, and tumor necrosis factor-alpha (p < 0.001 for all) than the placebo group. Total antioxidant status, glutathione peroxidase activity, and superoxide dismutase activity were elevated in the curcumin group, whereas malondialdehyde levels were greater in the placebo group (p < 0.001 for all). These findings suggest curcumin may have antidepressant effects on obese type 2 diabetes patients.


Sujet(s)
Antioxydants , Marqueurs biologiques , Curcumine , Dépression , Diabète de type 2 , Obésité , Humains , Curcumine/pharmacologie , Curcumine/usage thérapeutique , Diabète de type 2/complications , Diabète de type 2/traitement médicamenteux , Mâle , Obésité/complications , Obésité/traitement médicamenteux , Méthode en double aveugle , Femelle , Adulte d'âge moyen , Dépression/traitement médicamenteux , Dépression/étiologie , Marqueurs biologiques/sang , Malonaldéhyde/sang , Stress oxydatif/effets des médicaments et des substances chimiques , Sérotonine/métabolisme , Sérotonine/sang , Antidépresseurs/usage thérapeutique , Antidépresseurs/pharmacologie , Adulte , Glutathione peroxidase/sang , Glutathione peroxidase/métabolisme , Superoxide dismutase/sang , Superoxide dismutase/métabolisme , Cytokines/sang
11.
Behav Pharmacol ; 35(6): 314-326, 2024 09 01.
Article de Anglais | MEDLINE | ID: mdl-39094014

RÉSUMÉ

Depression is a common mood disorder and many patients do not respond to conventional pharmacotherapy or experience a variety of adverse effects. This work proposed that riparin I (RIP I) and riparin II (RIP II) present neuroprotective effects through modulation of astrocytes and microglia, resulting in the reversal of depressive-like behaviors. To verify our hypothesis and clarify the pathways underlying the effect of RIP I and RIP II on neuroinflammation, we used the chronic unpredictable mild stress (CUMS) depression model in mice. Male Swiss mice were exposed to stressors for 28 days. From 15 th to the 22 nd day, the animals received RIP I or RIP II (50 mg/kg) or fluoxetine (FLU, 10 mg/kg) or vehicle, by gavage. On the 29 th day, behavioral tests were performed. Expressions of microglia (ionized calcium-binding adaptor molecule-1 - Iba-1) and astrocyte (glial fibrillary acidic protein - GFAP) markers and levels of cytokines tumor necrosis factor alfa (TNF-α) and interleukin 1 beta (IL-1ß) were measured in the hippocampus. CUMS induced depressive-like behaviors and cognitive impairment, high TNF-α and IL-1ß levels, decreased GFAP, and increased Iba-1 expressions. RIP I and RIP II reversed these alterations. These results contribute to the understanding the mechanisms underlying the antidepressant effect of RIP I and RIP II, which may be related to neuroinflammatory suppression.


Sujet(s)
Antidépresseurs , Astrocytes , Dépression , Modèles animaux de maladie humaine , Hippocampe , Microglie , Maladies neuro-inflammatoires , Stress psychologique , Animaux , Astrocytes/effets des médicaments et des substances chimiques , Astrocytes/métabolisme , Souris , Mâle , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Antidépresseurs/pharmacologie , Dépression/traitement médicamenteux , Dépression/métabolisme , Maladies neuro-inflammatoires/traitement médicamenteux , Maladies neuro-inflammatoires/métabolisme , Stress psychologique/traitement médicamenteux , Stress psychologique/complications , Stress psychologique/métabolisme , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Fluoxétine/pharmacologie , Facteur de nécrose tumorale alpha/métabolisme , Interleukine-1 bêta/métabolisme , Neuroprotecteurs/pharmacologie , Comportement animal/effets des médicaments et des substances chimiques , Protéine gliofibrillaire acide/métabolisme
12.
Brain Behav ; 14(7): e3586, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38970230

RÉSUMÉ

BACKGROUND: Patients with myocardial infarction (MI) frequently experience a heightened incidence of depression, thereby increasing the risk of adverse cardiovascular events. Consequently, early detection and intervention in depressive symptoms among patients with MI are imperative. Shexiang Baoxin Pills (SBP), a Chinese patent medicine employed for the treatment of MI, exhibits diverse mechanisms targeting this condition. Nevertheless, its therapeutic efficacy on postmyocardial infarction depressive symptoms remains unclear. The aim of this study is to investigate the effectiveness and mechanism of SBP in managing depression during acute myocardial infarction (AMI). METHODS: A rat model combining MI and depression was established, and the rats were randomly divided into four groups: the model (MOD) group, SBP group, Fluoxetine (FLX) group, and Sham group. After 28 days of drug intervention, cardiac function was assessed using echocardiography while behavior was evaluated through sucrose preference test (SPT), forced swimming test (FST), and open-field test (OFT). Additionally, levels of inflammatory factors in serum and hippocampus were measured along with NLRP3 inflammasome-related protein expression via Western blotting and immunofluorescence. RESULTS: SBP can enhance cardiac function in rats with AMI and depression, while significantly ameliorating depressive-like behavior. Compared to the Sham group, levels of IL-1ß, IL-18, TNF-α, and other inflammatory factors were markedly elevated in the MOD group. However, expressions of these inflammatory factors were reduced to varying degrees following treatment with SBP or FLX. Analysis of NLRP3 inflammasome-related proteins in the hippocampus revealed a significant upregulation of IL-1ß, IL-18, NLRP3, ASC, caspase-1, and GSDMD in the MOD group; conversely, these measures were significantly attenuated after SBP intervention. CONCLUSION: We have observed a significant amelioration in depression-like behavior upon SBP administration during the treatment of AMI, suggesting that this effect may be attributed to the inhibition of NLRP3-mediated pyroptosis. (The main findings are summarized in the graphical abstract in the supplementary file.).


Sujet(s)
Antidépresseurs , Dépression , Médicaments issus de plantes chinoises , Inflammasomes , Infarctus du myocarde , Protéine-3 de la famille des NLR contenant un domaine pyrine , Rat Sprague-Dawley , Animaux , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Infarctus du myocarde/traitement médicamenteux , Infarctus du myocarde/métabolisme , Infarctus du myocarde/complications , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/administration et posologie , Rats , Dépression/traitement médicamenteux , Dépression/étiologie , Antidépresseurs/pharmacologie , Antidépresseurs/administration et posologie , Mâle , Inflammasomes/métabolisme , Inflammasomes/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Transduction du signal/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Hippocampe/effets des médicaments et des substances chimiques , Comportement animal/effets des médicaments et des substances chimiques
13.
Int Rev Neurobiol ; 177: 251-293, 2024.
Article de Anglais | MEDLINE | ID: mdl-39029987

RÉSUMÉ

Major depressive disorder (MDD) is a widespread and debilitating condition affecting a significant portion of the global population. Traditional treatment for MDD has primarily involved drugs that increase brain monoamines by inhibiting their uptake or metabolism, which is the basis for the monoaminergic hypothesis of depression. However, these treatments are only partially effective, with many patients experiencing delayed responses, residual symptoms, or complete non-response, rendering the current view of the hypothesis as reductionist. Cannabidiol (CBD) has shown promising results in preclinical models and human studies. Its mechanism is not well-understood, but may involve monoamine and endocannabinoid signaling, control of neuroinflammation and enhanced neuroplasticity. This chapter will explore CBD's effects in preclinical and clinical studies, its molecular mechanisms, and its potential as a treatment for MDD.


Sujet(s)
Cannabidiol , Trouble dépressif majeur , Cannabidiol/usage thérapeutique , Cannabidiol/pharmacologie , Humains , Animaux , Trouble dépressif majeur/traitement médicamenteux , Trouble dépressif majeur/métabolisme , Antidépresseurs/usage thérapeutique , Antidépresseurs/pharmacologie
14.
Mil Med Res ; 11(1): 49, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39044298

RÉSUMÉ

BACKGROUND: The development of ketamine-like rapid antidepressants holds promise for enhancing the therapeutic efficacy of depression, but the underlying cellular and molecular mechanisms remain unclear. Implicated in depression regulation, the neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) is investigated here to examine its role in mediating the rapid antidepressant response. METHODS: The onset of antidepressant response was assessed through depression-related behavioral paradigms. The signaling mechanism of PACAP in the hippocampal dentate gyrus (DG) was evaluated by utilizing site-directed gene knockdown, pharmacological interventions, or optogenetic manipulations. Overall, 446 mice were used for behavioral and molecular signaling testing. Mice were divided into control or experimental groups randomly in each experiment, and the experimental manipulations included: chronic paroxetine treatments (4, 9, 14 d) or a single treatment of ketamine; social defeat or lipopolysaccharides-injection induced depression models; different doses of PACAP (0.4, 2, 4 ng/site; microinjected into the hippocampal DG); pharmacological intra-DG interventions (CALM and PACAP6-38); intra-DG viral-mediated PACAP RNAi; and opotogenetics using channelrhodopsins 2 (ChR2) or endoplasmic natronomonas halorhodopsine 3.0 (eNpHR3.0). Behavioral paradigms included novelty suppressed feeding test, tail suspension test, forced swimming test, and sucrose preference test. Western blotting, ELISA, or quantitative real-time PCR (RT-PCR) analysis were used to detect the expressions of proteins/peptides or genes in the hippocampus. RESULTS: Chronic administration of the slow-onset antidepressant paroxetine resulted in an increase in hippocampal PACAP expression, and intra-DG blockade of PACAP attenuated the onset of the antidepressant response. The levels of hippocampal PACAP expression were reduced in both two distinct depression animal models and intra-DG knockdown of PACAP induced depression-like behaviors. Conversely, a single infusion of PACAP into the DG region produced a rapid and sustained antidepressant response in both normal and chronically stressed mice. Optogenetic intra-DG excitation of PACAP-expressing neurons instantly elicited antidepressant responses, while optogenetic inhibition induced depression-like behaviors. The longer optogenetic excitation/inhibition elicited the more sustained antidepressant/depression-like responses. Intra-DG PACAP infusion immediately facilitated the signaling for rapid antidepressant response by inhibiting calcium/calmodulin-dependent protein kinase II (CaMKII)-eukaryotic elongation factor 2 (eEF2) and activating the mammalian target of rapamycin (mTOR). Pre-activation of CaMKII signaling within the DG blunted PACAP-induced rapid antidepressant response as well as eEF2-mTOR-brain-derived neurotrophic factor (BDNF) signaling. Finally, acute ketamine treatment upregulated hippocampal PACAP expression, whereas intra-DG blockade of PACAP signaling attenuated ketamine's rapid antidepressant response. CONCLUSIONS: Activation of hippocampal PACAP signaling induces a rapid antidepressant response through the regulation of CaMKII inhibition-governed eEF2-mTOR-BDNF signaling.


Sujet(s)
Dépression , Hippocampe , Polypeptide activateur de l'adénylcyclase hypophysaire , Transduction du signal , Animaux , Mâle , Souris , Antidépresseurs/pharmacologie , Antidépresseurs/usage thérapeutique , Comportement animal/effets des médicaments et des substances chimiques , Dépression/traitement médicamenteux , Modèles animaux de maladie humaine , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Paroxétine/pharmacologie , Paroxétine/usage thérapeutique , Polypeptide activateur de l'adénylcyclase hypophysaire/métabolisme , Polypeptide activateur de l'adénylcyclase hypophysaire/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques
15.
Nature ; 632(8026): 921-929, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39048818

RÉSUMÉ

Noradrenaline, also known as norepinephrine, has a wide range of activities and effects on most brain cell types1. Its reuptake from the synaptic cleft heavily relies on the noradrenaline transporter (NET) located in the presynaptic membrane2. Here we report the cryo-electron microscopy (cryo-EM) structures of the human NET in both its apo state and when bound to substrates or antidepressant drugs, with resolutions ranging from 2.5 Å to 3.5 Å. The two substrates, noradrenaline and dopamine, display a similar binding mode within the central substrate binding site (S1) and within a newly identified extracellular allosteric site (S2). Four distinct antidepressants, namely, atomoxetine, desipramine, bupropion and escitalopram, occupy the S1 site to obstruct substrate transport in distinct conformations. Moreover, a potassium ion was observed within sodium-binding site 1 in the structure of the NET bound to desipramine under the KCl condition. Complemented by structural-guided biochemical analyses, our studies reveal the mechanism of substrate recognition, the alternating access of NET, and elucidate the mode of action of the four antidepressants.


Sujet(s)
Antidépresseurs , Cryomicroscopie électronique , Désipramine , Modèles moléculaires , Transporteurs de la norépinéphrine , Norépinéphrine , Humains , Transporteurs de la norépinéphrine/métabolisme , Transporteurs de la norépinéphrine/composition chimique , Transporteurs de la norépinéphrine/antagonistes et inhibiteurs , Désipramine/pharmacologie , Désipramine/composition chimique , Norépinéphrine/métabolisme , Norépinéphrine/composition chimique , Antidépresseurs/composition chimique , Antidépresseurs/pharmacologie , Antidépresseurs/métabolisme , Sites de fixation , Dopamine/métabolisme , Dopamine/composition chimique , Site allostérique , Chlorhydrate d'atomoxétine/composition chimique , Chlorhydrate d'atomoxétine/pharmacologie , Chlorhydrate d'atomoxétine/métabolisme , Potassium/métabolisme , Bupropion/composition chimique , Bupropion/métabolisme , Bupropion/pharmacologie , Citalopram/composition chimique , Citalopram/pharmacologie , Citalopram/métabolisme , Sodium/métabolisme , Spécificité du substrat
16.
J Tradit Chin Med ; 44(4): 670-679, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39066527

RÉSUMÉ

OBJECTIVE: To investigate the effects of luteolin on chronic unpredictable mild stress (CUMS)-induced depressive rats and corticosterone (CORT)-induced depressive primary hippocampal neurons, and to elucidate the mechanism behind the action. METHODS: The antidepressant mechanism of luteolin was studied by using CUMS rat model and primary hippocampal neurons in fetal rats. In vivo, novelty suppressed feeding, open-field and sucrose preference tests as well as Morris water maze were evaluated. The content of brain derived neurotrophic factor (BDNF), 5-hydroxytryptamine (5-HT), norepinephrine (NE), and dopamine (DA) in serum were detected by enzyme-linked immunosorbent assay. The mechanisms of luteolin were explored based on neurotrophin and hippocampal neurogenesis, and proliferation. Survival of the septo-temporal axis in hippocampus was assayed using the 5-bromo-2-deoxyuridine (BrdU), the expression of BDNF, neurotrophin-3 (NT-3), and nerve growth factor (NGF) in hippocampus dentate gyrus region were measured by Western-blotting. In vitro, BDNF, NT-3, tropomyosin receptor kinase B (TrkB), and phosphorylated cyclic adenosine monophosphate responsive element binding protein (p-CREB) were detected through the high content analysis (HCA) to investigate neurotrophin and apoptosis. RESULTS: Induction of CUMS in rats induced depressive symptoms, while luteolin significantly enhanced sucrose consumption, decreased feeding latency, increased locomotor activity, escape latency, distance of target quadrant and regulated the content of depressive-like biomarkers. Histology analysis revealed that luteolin increased the abundance of new born neurons that had been labeled with BrdU, BrdU + neuronal nuclear antigen, and BrdU + doublecortin in septo-temporal axis of S2 (mid-septal) and T3 (mid-temporal). Moreover, expression of BDNF, NT-3, and NGF increased significantly in the septo-temporal axis of S2 and T3. HCA showed increased expression of BDNF, NT-3, TrkB and p-CREB in primary hippocampal neurons. CONCLUSION: The results provided direct evidence that luteolin has an antidepressant effect and could effectively promote the regeneration of the septotemporal axis nerve and hippocampal neuronutrition, which suggested that the antidepressant effect of luteolin may be related to hippocampal neurogenesis.


Sujet(s)
Facteur neurotrophique dérivé du cerveau , Hippocampe , Lutéoline , Neurogenèse , Neurones , Rat Sprague-Dawley , Animaux , Lutéoline/pharmacologie , Rats , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/métabolisme , Neurogenèse/effets des médicaments et des substances chimiques , Mâle , Facteur neurotrophique dérivé du cerveau/métabolisme , Facteur neurotrophique dérivé du cerveau/génétique , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Humains , Stress psychologique/physiopathologie , Stress psychologique/traitement médicamenteux , Femelle , Dépression/traitement médicamenteux , Dépression/métabolisme , Dépression/physiopathologie , Antidépresseurs/pharmacologie , Neurotrophine-3/métabolisme , Neurotrophine-3/génétique
17.
Mol Pharmacol ; 106(3): 107-116, 2024 Aug 16.
Article de Anglais | MEDLINE | ID: mdl-39079719

RÉSUMÉ

Melatonin (N-acetyl-5-methoxytryptamine) is an indoleamine secreted by the pineal gland during the dark phase of the photoperiod. Its main function is the synchronization of different body rhythms with the dark-light cycle. Research on melatonin has significantly advanced since its discovery and we now know that it has considerable significance in various physiological processes, including immunity, aging, and reproduction. Moreover, in recent years evidence of the pharmacological possibilities of melatonin has increased. Indoleamine, on the other hand, has antidepressant-like effects in rodents, which may be mediated by the activation of calcium-calmodulin-dependent kinase II (CaMKII) and are also related to the regulation of neuroplasticity processes, including neurogenesis, synaptic maintenance, and long-term potentiation. Remarkably, patients with major depression show decreased levels of circulating melatonin in plasma. This review presents evidence of the antidepressant-like effects of melatonin in preclinical models and the participation of CaMKII in these actions. CaMKII's role in cognition and memory processes, which are altered in depressive states, are part of the review, and the effects of melatonin in these processes are also reviewed. Furthermore, participation of CaMKII on structural and synaptic plasticity and the effects of melatonin are also described. Finally, the advantages of using melatonin in combination with other antidepressants such as ketamine for neuroplasticity are described. Evidence supports that CaMKII is activated by melatonin and downstream melatonin receptors and may be the common effector in the synergistic effects of melatonin with other antidepressants. SIGNIFICANCE STATEMENT: This review compiled evidence supporting that melatonin causes antidepressant-like effects in mice through calmodulin kinase II stimulation of downstream melatonin receptors as well as the participation of this enzyme in neuroplasticity, memory, and cognition. Finally, we describe evidence about the effectiveness of antidepressant-like effects of melatonin in combination with ketamine.


Sujet(s)
Antidépresseurs , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Mélatonine , Plasticité neuronale , Mélatonine/pharmacologie , Animaux , Antidépresseurs/pharmacologie , Antidépresseurs/usage thérapeutique , Humains , Calcium-Calmodulin-Dependent Protein Kinase Type 2/métabolisme , Plasticité neuronale/effets des médicaments et des substances chimiques , Dépression/traitement médicamenteux , Dépression/métabolisme
18.
Int J Mol Sci ; 25(14)2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-39062817

RÉSUMÉ

Depression is one of the most common psychological disorders nowadays. Studies have shown that 20(S)-protopanaxatriol (PPT) can effectively improve depressive symptoms in mice. However, its mechanism needs to be further explored. In this study, we used an integrated approach combining network pharmacology and transcriptomics to explore the potential mechanisms of PPT for depression. First, the potential targets and pathways of PPT treatment of depression were screened through network pharmacology. Secondly, the BMKCloud platform was used to obtain brain tissue transcription data of chronic unpredictable mild stress (CUMS) model mice and screen PPT-altered differential expression genes (DEGs). Gene ontology (GO) analysis and the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis were performed using network pharmacology and transcriptomics. Finally, the above results were verified by molecular docking, Western blotting, and quantitative real-time polymerase chain reaction (qRT-PCR). In this study, we demonstrated that PPT improved depression-like behavior and brain histopathological changes in CUMS mice, downregulated nitric oxide (NO) and interleukin-6 (IL-6) levels, and elevated serum levels of 5-hydroxytryptamine (5-HT) and brain-derived neurotrophic factor (BDNF) after PPT treatment compared to the CUMS group. Eighty-seven potential targets and 350 DEGs were identified by network pharmacology and transcriptomics. Comprehensive analysis showed that transthyretin (TTR), klotho (KL), FOS, and the phosphatidylinositol 3-kinase-protein kinase B (PI3K-AKT) signaling pathway were closely associated with the therapeutic effects of PPT. Molecular docking results showed that PPT had a high affinity for PI3K, AKT, TTR, KL, and FOS targets. Gene and protein level results showed that PPT could increase the expression of PI3K, phosphorylation of PI3K (p-PI3K), AKT, phosphorylation of AKT (p-AKT), TTR, and KL and inhibit the expression level of FOS in the brain tissue of depressed mice. Our data suggest that PPT may achieve the treatment of depression by inhibiting the expression of FOS, enhancing the expression of TTR and KL, and modulating the PI3K-AKT signaling pathway.


Sujet(s)
Dépression , Pharmacologie des réseaux , Sapogénines , Transcriptome , Animaux , Souris , Dépression/traitement médicamenteux , Dépression/métabolisme , Sapogénines/pharmacologie , Transcriptome/effets des médicaments et des substances chimiques , Mâle , Antidépresseurs/pharmacologie , Antidépresseurs/usage thérapeutique , Simulation de docking moléculaire , Modèles animaux de maladie humaine , Transduction du signal/effets des médicaments et des substances chimiques , Analyse de profil d'expression de gènes , Encéphale/métabolisme , Encéphale/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques
19.
Nutrients ; 16(13)2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38999797

RÉSUMÉ

Astrocyte dysfunction and inflammation play a pivotal role in depression. In this study, we evaluated the antidepressant properties of Heracleum moellendorffii root extract (HME), which is traditionally used for inflammation-related diseases, in a mouse model with astrocyte depletion that resembles the prefrontal cortex pathology of depressive patients. Mice were divided into four groups, with 10 mice per group. To induce astrocyte ablation in the mice's prefrontal cortex (PFC), we used astrocytic toxin L-alpha-aminoadipic acid (L-AAA) and administered HME orally at 200 and 500 mg/kg for 22 days. We utilized the tail suspension test (TST) to assess depression-like behaviors and the open field test (OFT) to evaluate anxiety-like activities. Additionally, astrocytic and inflammatory markers in the PFC were evaluated using immunohistochemistry and ELISA. The results showed that infusion of L-AAA significantly decreased the expression of astrocytic glial fibrillary acidic protein (GFAP), which was accompanied by increased depression and anxiety-like behaviors. However, HME significantly reversed these effects by dose-dependently enhancing GFAP expression and modulating inflammatory markers, such as TNF-α, IL-6, and particularly lipocalin-2, a master proinflammatory mediator. These results imply that HME contributes to the alleviation of depression and anxiety-like behaviors by promoting astrocyte recovery and reducing neuroinflammation, especially through lipocalin-2 inhibition.


Sujet(s)
Antidépresseurs , Astrocytes , Comportement animal , Dépression , Modèles animaux de maladie humaine , Lipocaline-2 , Extraits de plantes , Animaux , Astrocytes/effets des médicaments et des substances chimiques , Astrocytes/métabolisme , Lipocaline-2/métabolisme , Extraits de plantes/pharmacologie , Dépression/traitement médicamenteux , Souris , Antidépresseurs/pharmacologie , Mâle , Comportement animal/effets des médicaments et des substances chimiques , Cortex préfrontal/effets des médicaments et des substances chimiques , Cortex préfrontal/métabolisme , Maladies neuro-inflammatoires/traitement médicamenteux , Protéine gliofibrillaire acide/métabolisme , Souris de lignée C57BL
20.
Int J Mol Sci ; 25(13)2024 Jul 08.
Article de Anglais | MEDLINE | ID: mdl-39000602

RÉSUMÉ

The application of intracerebroventricular injection of streptozotocin (ICV-STZ) is considered a useful animal model to mimic the onset and progression of sporadic Alzheimer's disease (sAD). In rodents, on day 7 of the experiment, the animals exhibit depression-like behaviors. Indoleamine 2,3-dioxygenase (IDO), a rate-limiting enzyme catalyzing the conversion of tryptophan (Trp) to kynurenine (Kyn), is closely related to depression and AD. The present study aimed to investigate the pathophysiological mechanisms of preliminary depression-like behaviors in ICV-STZ rats in two distinct cerebral regions of the medial prefrontal cortex, the prelimbic cortex (PrL) and infralimbic cortex (IL), both presumably involved in AD progression in this model, with a focus on IDO-related Kyn pathways. The results showed an increased Kyn/Trp ratio in both the PrL and IL of ICV-STZ rats, but, intriguingly, abnormalities in downstream metabolic pathways were different, being associated with distinct biological effects. In the PrL, the neuroprotective branch of the Kyn pathway was attenuated, as evidenced by a decrease in the kynurenic acid (KA) level and Kyn aminotransferase II (KAT II) expression, accompanied by astrocyte alterations, such as the decrease in glial fibrillary acidic protein (GFAP)-positive cells and increase in morphological damage. In the IL, the neurotoxicogenic branch of the Kyn pathway was enhanced, as evidenced by an increase in the 3-hydroxy-kynurenine (3-HK) level and kynurenine 3-monooxygenase (KMO) expression paralleled by the overactivation of microglia, reflected by an increase in ionized calcium-binding adaptor molecule 1 (Iba1)-positive cells and cytokines with morphological alterations. Synaptic plasticity was attenuated in both subregions. Additionally, microinjection of the selective IDO inhibitor 1-Methyl-DL-tryptophan (1-MT) in the PrL or IL alleviated depression-like behaviors by reversing these different abnormalities in the PrL and IL. These results suggest that the antidepressant-like effects linked to Trp metabolism changes induced by 1-MT in the PrL and IL occur through different pathways, specifically by enhancing the neuroprotective branch in the PrL and attenuating the neurotoxicogenic branch in the IL, involving distinct glial cells.


Sujet(s)
Antidépresseurs , Dépression , Indoleamine-pyrrole 2,3,-dioxygenase , Cynurénine , Streptozocine , Tryptophane , Animaux , Indoleamine-pyrrole 2,3,-dioxygenase/métabolisme , Streptozocine/toxicité , Rats , Mâle , Cynurénine/métabolisme , Antidépresseurs/pharmacologie , Antidépresseurs/administration et posologie , Tryptophane/métabolisme , Tryptophane/pharmacologie , Dépression/traitement médicamenteux , Dépression/métabolisme , Dépression/induit chimiquement , Injections ventriculaires , Cortex préfrontal/métabolisme , Cortex préfrontal/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Rat Sprague-Dawley
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE