Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 5.768
Filtrer
1.
CNS Neurosci Ther ; 30(7): e14831, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38961317

RÉSUMÉ

AIMS: Comorbid anxiodepressive-like symptoms (CADS) in chronic pain are closely related to the overactivation of the lateral habenula (LHb). Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels have been implicated to play a key role in regulating neuronal excitability. However, the role of HCN channels in the LHb during CADS has not yet been characterized. This study aimed to investigate the effect of HCN channels in the LHb on CADS during chronic pain. METHODS: After chronic neuropathic pain induction by spared nerve injury (SNI), mice underwent a sucrose preference test, forced swimming test, tail suspension test, open-field test, and elevated plus maze test to evaluate their anxiodepressive-like behaviors. Electrophysiological recordings, immunohistochemistry, Western blotting, pharmacological experiments, and virus knockdown strategies were used to investigate the underlying mechanisms. RESULTS: Evident anxiodepressive-like behaviors were observed 6w after the SNI surgery, accompanied by increased neuronal excitability, enhanced HCN channel function, and increased expression of HCN2 isoforms in the LHb. Either pharmacological inhibition or virus knockdown of HCN2 channels significantly reduced LHb neuronal excitability and ameliorated both pain and depressive-like behaviors. CONCLUSION: Our results indicated that the LHb neurons were hyperactive under CADS in chronic pain, and this hyperactivation possibly resulted from the enhanced function of HCN channels and up-regulation of HCN2 isoforms.


Sujet(s)
Dépression , Habénula , Canaux contrôlés par les nucléotides cycliques et activés par l'hyperpolarisation , Animaux , Habénula/métabolisme , Habénula/effets des médicaments et des substances chimiques , Canaux contrôlés par les nucléotides cycliques et activés par l'hyperpolarisation/métabolisme , Souris , Mâle , Dépression/métabolisme , Névralgie/métabolisme , Névralgie/psychologie , Souris de lignée C57BL , Douleur chronique/métabolisme , Douleur chronique/psychologie , Canaux potassiques
2.
Theranostics ; 14(9): 3653-3673, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948066

RÉSUMÉ

Rationale: Recent evidence highlights the pivotal role of mitochondrial dysfunction in mood disorders, but the mechanism involved remains unclear. We studied whether the Hippo/YAP/14-3-3η signaling pathway mediates mitochondrial abnormalities that result in the onset of major depressive disorder (MDD) in a mouse model. Methods: The ROC algorithm was used to identify a subpopulation of mice that were exposed to chronic unpredictable mild stress (CUMS) and exhibited the most prominent depressive phenotype (Dep). Electron microscopy, biochemical assays, quantitative PCR, and immunoblotting were used to evaluate synaptic and mitochondrial changes in the basolateral amygdala (BLA). RNA sequencing was used to explore changes in the Hippo pathway and downstream target genes. In vitro pharmacological inhibition and immunoprecipitation was used to confirm YAP/14-3-3η interaction and its role in neuronal mitochondrial dysfunction. We used virus-mediated gene overexpression and knockout in YAP transgenic mice to verify the regulatory effect of the Hippo/YAP/14-3-3η pathway on depressive-like behavior. Results: Transcriptomic data identified a large number of genes and signaling pathways that were specifically altered from the BLA of Dep mice. Dep mice showed notable synaptic impairment in BLA neurons, as well as mitochondrial damage characterized by abnormal mitochondrial morphology, compromised function, impaired biogenesis, and alterations in mitochondrial marker proteins. The Hippo signaling pathway was activated in Dep mice during CUMS, and the transcriptional regulatory activity of YAP was suppressed by phosphorylation of its Ser127 site. 14-3-3η was identified as an important co-regulatory factor of the Hippo/YAP pathway, as it can respond to chronic stress and regulate cytoplasmic retention of YAP. Importantly, the integrated Hippo/YAP/14-3-3η pathway mediated neuronal mitochondrial dysfunction and depressive behavior in Dep mice. Conclusion: The integrated Hippo/YAP/14-3-3η pathway in the BLA neuron is critical in mediating depressive-like behaviors in mice, suggesting a causal role for this pathway in susceptibility to chronic stress-induced depression. This pathway therefore may present a therapeutic target against mitochondrial dysfunction and synaptic impairment in MDD.


Sujet(s)
Groupe nucléaire basolatéral , Modèles animaux de maladie humaine , Voie de signalisation Hippo , Mitochondries , Protein-Serine-Threonine Kinases , Transduction du signal , Protéines de signalisation YAP , Animaux , Souris , Mitochondries/métabolisme , Protéines de signalisation YAP/métabolisme , Groupe nucléaire basolatéral/métabolisme , Groupe nucléaire basolatéral/anatomopathologie , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Mâle , Stress psychologique/complications , Stress psychologique/métabolisme , Protéines 14-3-3/métabolisme , Protéines 14-3-3/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Trouble dépressif majeur/métabolisme , Trouble dépressif majeur/anatomopathologie , Dépression/métabolisme , Souris de lignée C57BL , Neurones/métabolisme , Neurones/anatomopathologie , Souris transgéniques
3.
J Med Chem ; 67(12): 10350-10373, 2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38888140

RÉSUMÉ

Multiple studies have confirmed that acid sphingomyelinase (ASM) activity is associated with depression. The discovery of direct inhibitors against ASM is of great significance for exploring antidepressants and their mechanisms of action. Herein, a series of novel phenylpyrazole analogues were rationally designed and synthesized. Among them, compound 46 exhibited potent inhibitory activity (IC50 = 0.87 µM) and good drug-like properties. In vivo studies demonstrated that compound 46 was involved in multiple antidepressant mechanisms of action, which were associated with a decline of ceramide, including increasing the Bcl-2/Bax ratio and BDNF expression, down-regulating caspase-3 and caspase-9, ameliorating oxidative stress, reducing the levels of proinflammatory cytokines such as TNF-α, IL-1ß, and IL-6, and elevating 5-HT levels in the brains of mice, respectively. These meaningful results reveal for the first time that direct inhibitors exhibit remarkable antidepressant effects in the CUMS-induced mouse model through multiple mechanisms of antidepressant action.


Sujet(s)
Antidépresseurs , Pyrazoles , Sphingomyeline phosphodiesterase , Animaux , Antidépresseurs/pharmacologie , Antidépresseurs/composition chimique , Antidépresseurs/synthèse chimique , Pyrazoles/pharmacologie , Pyrazoles/composition chimique , Pyrazoles/synthèse chimique , Souris , Sphingomyeline phosphodiesterase/antagonistes et inhibiteurs , Sphingomyeline phosphodiesterase/métabolisme , Relation structure-activité , Mâle , Dépression/traitement médicamenteux , Dépression/métabolisme , Découverte de médicament , Antienzymes/pharmacologie , Antienzymes/composition chimique , Antienzymes/synthèse chimique , Humains , Facteur neurotrophique dérivé du cerveau/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques
4.
Sci Rep ; 14(1): 13559, 2024 06 12.
Article de Anglais | MEDLINE | ID: mdl-38866877

RÉSUMÉ

Naringenin (NAR) has various biological activities but low bioavailability. The current study examines the effect of Naringenin-loaded hybridized nanoparticles (NAR-HNPs) and NAR on depression induced by streptozotocin (STZ) in rats. NAR-HNPs formula with the highest in vitro NAR released profile, lowest polydispersity index value (0.21 ± 0.02), highest entrapment efficiency (98.7 ± 2.01%), as well as an acceptable particle size and zeta potential of 415.2 ± 9.54 nm and 52.8 ± 1.04 mV, respectively, was considered the optimum formulation. It was characterized by differential scanning calorimetry, examined using a transmission electron microscope, and a stability study was conducted at different temperatures to monitor its stability efficiency showing that NAR-HNP formulation maintains stability at 4 °C. The selected formulation was subjected to an acute toxicological test, a pharmacokinetic analysis, and a Diabetes mellitus (DM) experimental model. STZ (50 mg/kg) given as a single i.p. rendered rats diabetic. Diabetic rat groups were allocated into 4 groups: one group received no treatment, while the remaining three received oral doses of unloaded HNPs, NAR (50 mg/kg), NAR-HNPs (50 mg/kg) and NAR (50 mg/kg) + peroxisome proliferator-activated receptor-γ (PPAR-γ) antagonist, GW9662 (1mg/kg, i.p.) for three weeks. Additional four non-diabetic rat groups received: distilled water (normal), free NAR, and NAR-HNPs, respectively for three weeks. NAR and NAR-HNPs reduced immobility time in forced swimming test and serum blood glucose while increasing serum insulin level. They also reduced cortical and hippocampal 5-hydroxyindoeacetic acid, 3,4-Dihydroxy-phenylacetic acid, malondialdehyde, NLR family pyrin domain containing-3 (NLRP3) and interleukin-1beta content while raised serotonin, nor-epinephrine, dopamine and glutathione level. PPAR-γ gene expression was elevated too. So, NAR and NAR-HNPs reduced DM-induced depression by influencing brain neurotransmitters and exhibiting anti-oxidant and anti-inflammatory effects through the activation PPAR-γ/ NLRP3 pathway. NAR-HNPs showed the best pharmacokinetic and therapeutic results.


Sujet(s)
Antidépresseurs , Diabète expérimental , Flavanones , Protéine-3 de la famille des NLR contenant un domaine pyrine , Nanoparticules , Récepteur PPAR gamma , Animaux , Flavanones/pharmacologie , Flavanones/administration et posologie , Flavanones/composition chimique , Récepteur PPAR gamma/métabolisme , Diabète expérimental/traitement médicamenteux , Diabète expérimental/métabolisme , Nanoparticules/composition chimique , Rats , Mâle , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Antidépresseurs/pharmacologie , Dépression/traitement médicamenteux , Dépression/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Streptozocine , Rat Wistar , Anilides
5.
J Agric Food Chem ; 72(25): 14199-14215, 2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38871671

RÉSUMÉ

Fucoidan has shown better effects on the improvement of acute ulcerative colitis (UC). However, the specific mechanisms by which fucoidan improves UC-related behavioral disorders in aged mice, especially its effect on the gut-brain axis, remain to be further explored. C57BL/6 male mice aged 8 months were gavaged with 400 or 100 mg/kg bw day fucoidan for five consecutive weeks, with UC being induced by ad libitum to dextran sulfate sodium (DSS) solution in the fifth week. The results showed that fucoidan ameliorated UC and accompanying anxiety- and depressive-like behaviors with downregulated expressions of (NOD)-like receptor family and pyrin domain-containing 3 (NLRP3), apoptosis-associated speck-like protein (ASC), cysteine aspartate-specific protease-1 (Caspase-1) and interlekin-1ß (IL-1ß), and elevated mRNA levels of brain-derived neurotrophic factor (Bdnf) and postsynaptic-density protein 95 (Psd-95) in cortex and hippocampus. Furthermore, fucoidan improved the permeability of intestinal barrier and blood-brain barrier and restored the abnormal structure of the gut microbiota with a significantly decreased ratio of Firmicutes to Bacteroidota (F/B) and obviously increased abundance of Akkermansia. As a diet-derived bioactive ingredient, fucoidan might be a better alternative for the prevention of UC and accompanying anxiety- and depressive-like behaviors.


Sujet(s)
Anxiété , Rectocolite hémorragique , Dépression , Sulfate dextran , Souris de lignée C57BL , Polyosides , Animaux , Polyosides/administration et posologie , Polyosides/pharmacologie , Polyosides/composition chimique , Mâle , Sulfate dextran/effets indésirables , Souris , Rectocolite hémorragique/traitement médicamenteux , Rectocolite hémorragique/métabolisme , Rectocolite hémorragique/induit chimiquement , Dépression/traitement médicamenteux , Dépression/métabolisme , Anxiété/traitement médicamenteux , Humains , Facteur neurotrophique dérivé du cerveau/métabolisme , Facteur neurotrophique dérivé du cerveau/génétique , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Interleukine-1 bêta/génétique , Interleukine-1 bêta/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Caspase-1/métabolisme , Caspase-1/génétique , Modèles animaux de maladie humaine , Hippocampe/métabolisme , Hippocampe/effets des médicaments et des substances chimiques , Comportement animal/effets des médicaments et des substances chimiques
6.
Neurosci Lett ; 835: 137851, 2024 Jul 13.
Article de Anglais | MEDLINE | ID: mdl-38838971

RÉSUMÉ

Chronic psychosocial stress stands as a significant heterogeneous risk factor for psychiatric disorders. The brain's physiological response to such stress varies based on the frequency and intensity of stress episodes. However, whether stress episodes divergently could affect hippocampal cyclic AMP response element-binding protein (CREB)-brain-derived neurotrophic factor (BDNF) signaling remains unclear, a key regulator of psychiatric symptoms. We aimed to assess how two distinct patterns of social defeat stress exposure impact anxiety- and depression-like behaviors, fear, and hippocampal CREB-BDNF signaling in adult male rats. To explore this, adult male Sprague-Dawley rats were subjected to psychosocial stress using a Resident/Intruder paradigm for ten consecutive days (continuous social defeat stress: [CS]) or ten social defeat stress over the course of 21 days (intermittent social defeat stress [IS]). Behavioral tests (including novelty-suppressed feeding test, forced swimming test, and contextually conditioned fear) were conducted. Protein expression levels of phosphorylated CREB and BDNF in the dorsal and ventral hippocampi were examined. CS led to heightened anxiety-like behavior, fear, and increased levels of phosphorylated CREB in both the dorsal and ventral hippocampi. Conversely, IS resulted in increased anxiety-like behavior and behavioral despair alongside decreased levels of phosphorylated CREB and BDNF, particularly in the dorsal hippocampus. These findings indicate that chronic psychosocial stress divergently affects hippocampal CREB-BDNF signaling and emotional regulation depending on the stress episode. Such insights could enhance our understanding of the molecular basis of the heterogeneity of psychiatric disorders and facilitate the development of innovative treatment approaches to patients with psychiatric disorders.


Sujet(s)
Facteur neurotrophique dérivé du cerveau , Protéine de liaison à l'élément de réponse à l'AMP cyclique , Hippocampe , Rat Sprague-Dawley , Stress psychologique , Animaux , Facteur neurotrophique dérivé du cerveau/métabolisme , Hippocampe/métabolisme , Mâle , Stress psychologique/métabolisme , Stress psychologique/psychologie , Phosphorylation , Protéine de liaison à l'élément de réponse à l'AMP cyclique/métabolisme , Défaite sociale , Rats , Anxiété/métabolisme , Anxiété/psychologie , Comportement animal/physiologie , Peur/physiologie , Peur/psychologie , Émotions/physiologie , Dépression/métabolisme , Dépression/psychologie
7.
In Vivo ; 38(4): 1677-1689, 2024.
Article de Anglais | MEDLINE | ID: mdl-38936893

RÉSUMÉ

BACKGROUND/AIM: Depression is associated with metabolic disorders, including non-alcoholic fatty liver disease (NAFLD). However, the mechanisms underlying the interaction between them are still poorly known. MATERIALS AND METHODS: In this study, mice on a choline deficiency, L-amino acid-defined, high-fat diet (CDAHFD) developing steatosis were challenged with chronic restraint stress (CRS), a protocol widely used to induce depression. The development of depression and steatosis was evaluated using histopathology analysis, ELISA, q-PCR and Western Blot. RESULTS: The contribution of the activated HPA axis to hepatic steatosis progress was fully established, which was validated using a hepatocyte model. Histopathological and biochemical analysis indicated that steatosis was exacerbated by CRS challenge, and behavioral tests indicated that the mice developed depression. Among the screened endocrinal pathways, the hypothalamic-pituitary-adrenal (HPA) axis was significantly activated and the synergistic effect of CDAHFD and CRS in activating the HPA axis was observed. In the hypothalamus, expression of corticotropin-releasing hormone (CRH) was increased by 86% and the protein levels of hypothalamic CRH were upregulated by 25% to 33% by CRS treatment. Plasma CRH levels were elevated by 45-56% and plasma adrenocorticotropic hormone (ACTH) levels were elevated by 29-58% by CRS treatment. In the liver, target genes of the HPA axis were activated, accompanied by disruption of the lipid metabolism and progression of steatohepatitis. The lipid metabolism in the Hepa1-6 cell line treated with endogenous corticosterone (CORT) was in accordance with the aforementioned in vivo responses. CONCLUSION: Depression aggravated hepatic steatosis in CDAHFD-fed mice by activating the HPA axis. The risk of NAFLD development should be fully considered in depressive patients and improvement of psychotic disorders could be an etiological treatment strategy for them.


Sujet(s)
Dépression , Modèles animaux de maladie humaine , Axe hypothalamohypophysaire , Stéatose hépatique non alcoolique , Axe hypophyso-surrénalien , Animaux , Axe hypothalamohypophysaire/métabolisme , Axe hypophyso-surrénalien/métabolisme , Souris , Dépression/métabolisme , Dépression/étiologie , Dépression/génétique , Stéatose hépatique non alcoolique/métabolisme , Stéatose hépatique non alcoolique/anatomopathologie , Stéatose hépatique non alcoolique/étiologie , Mâle , Souris de lignée C57BL , Corticolibérine/métabolisme , Corticolibérine/génétique , Alimentation riche en graisse/effets indésirables , Hormone corticotrope/sang , Foie/métabolisme , Foie/anatomopathologie , Stéatose hépatique/métabolisme , Stéatose hépatique/étiologie , Stéatose hépatique/anatomopathologie , Corticostérone/sang
8.
eNeuro ; 11(7)2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38866499

RÉSUMÉ

Previous studies have shown that ligands that bind to sigma-2 receptor/TMEM97 (s2R/TMEM97), a transmembrane protein, have anxiolytic/antidepressant-like properties and relieve neuropathic pain-like effects in rodents. Despite medical interest in s2R/TMEM97, little affective and pain behavioral characterization has been done using transgenic mice, which limits the development of s2R/TMEM97 as a viable therapeutic target. Using wild-type (WT) and global Tmem97 knock-out (KO) mice, we sought to identify the contribution of Tmem97 in modulating affective and pain-like behaviors using a battery of affective and pain assays, including open field, light/dark preference, elevated plus maze, forced swim test, tail suspension test, and the mechanical sensitivity tests. Our results demonstrate that female Tmem97 KO mice show less anxiety-like and depressive-like behaviors in light/dark preference and tail suspension tests but not in an open field, elevated plus maze, and forced swim tests at baseline. We next performed spared nerve injury in WT and Tmem97 KO mice to assess the role of Tmem97 in neuropathic pain-induced anxiety and depression. WT mice, but not Tmem97 KO mice, developed a prolonged neuropathic pain-induced depressive-like phenotype when tested 10 weeks after nerve injury in females. Our results show that Tmem97 plays a role in modulating anxiety-like and depressive-like behaviors in naive animals with a significant change in the presence of nerve injury in female mice. Overall, these data demonstrate that Tmem97 could be a target to alleviate affective comorbidities of pain disorders.


Sujet(s)
Dépression , Protéines membranaires , Souris de lignée C57BL , Souris knockout , Névralgie , Récepteur sigma , Animaux , Récepteur sigma/métabolisme , Femelle , Névralgie/métabolisme , Protéines membranaires/métabolisme , Protéines membranaires/génétique , Dépression/métabolisme , Dépression/étiologie , Comportement animal/physiologie , Souris , Anxiété/métabolisme , Modèles animaux de maladie humaine , Mâle
9.
Pharmacol Biochem Behav ; 241: 173805, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38848976

RÉSUMÉ

Depression, a prevalent mental health condition, significantly impacts global mental impairment rates. While antidepressants are commonly used, treatment-resistant depression (TRD) poses a challenge. Emerging research highlights the role of the gut microbiota in depression through the gut-brain axis. This study identifies key genes associated with depression influenced by specific gut microbiota, Coprococcus and Subdoligranulum. Using bioinformatics tools, potential targets were elucidated, and molecular docking studies were performed. Furthermore, gene expression analysis identified hub-genes related to depression, intersecting with metabolite targets. Protein-protein interaction analysis revealed pivotal targets such as PTGS2 and MMP9. Molecular docking demonstrated 3-Indolepropionic acid's superior affinity over (R)-3-(4-Hydroxyphenyl)lactate. Physicochemical properties and toxicity profiles were compared, suggesting favorable attributes for 3-Indolepropionic acid. Molecular dynamics simulations confirmed stability and interactions of compounds with target proteins. This comprehensive approach sheds light on the complex interplay between gut microbiota, genes, and depression, emphasizing the potential for microbiota-targeted interventions in mental health management.


Sujet(s)
Microbiome gastro-intestinal , Simulation de docking moléculaire , Simulation de dynamique moléculaire , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Humains , Pharmacologie des réseaux , Indoles/pharmacologie , Dépression/traitement médicamenteux , Dépression/microbiologie , Dépression/métabolisme , Propionates/pharmacologie , Propionates/métabolisme , Axe cerveau-intestin/effets des médicaments et des substances chimiques , Antidépresseurs/pharmacologie
10.
Animal Model Exp Med ; 7(3): 347-361, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38895818

RÉSUMÉ

BACKGROUND: Apolipoprotein E4 (ApoE4) allele is the strongest genetic risk factor for late-onset Alzheimer's disease, and it can aggravate depressive symptoms in non-AD patients. However, the impact of ApoE4 on AD-associated depression-like behaviors and its underlying pathogenic mechanisms remain unclear. METHODS: This study developed a 5xFAD mouse model overexpressing human ApoE4 (E4FAD). Behavioral assessments and synaptic function tests were conducted to explore the effects of ApoE4 on cognition and depression in 5xFAD mice. Changes in peripheral and central lipid metabolism, as well as the levels of serotonin (5-HT) and γ-aminobutyric acid (GABA) neurotransmitters in the prefrontal cortex, were examined. In addition, the protein levels of 24-dehydrocholesterol reductase/glycogen synthase kinase-3 beta/mammalian target of rapamycin (DHCR24/GSK3ß/mTOR) and postsynaptic density protein 95/calmodulin-dependent protein kinase II/brain-derived neurotrophic factor (PSD95/CaMK-II/BDNF) were measured to investigate the molecular mechanism underlying the effects of ApoE4 on AD mice. RESULTS: Compared with 5xFAD mice, E4FAD mice exhibited more severe depression-like behaviors and cognitive impairments. These mice also exhibited increased amyloid-beta deposition in the hippocampus, increased astrocyte numbers, and decreased expression of depression-related neurotransmitters 5-HT and GABA in the prefrontal cortex. Furthermore, lipid metabolism disorders were observed in E4FAD, manifesting as elevated low-density lipoprotein cholesterol and reduced high-density lipoprotein cholesterol in peripheral blood, decreased cholesterol level in the prefrontal cortex, and reduced expression of key enzymes and proteins related to cholesterol synthesis and homeostasis. Abnormal expression of proteins related to the DHCR24/GSK3ß/mTOR and PSD95/CaMK-II/BDNF pathways was also observed. CONCLUSION: This study found that ApoE4 overexpression exacerbates depression-like behaviors in 5xFAD mice and confirmed that ApoE4 reduces cognitive function in these mice. The mechanism may involve the induction of central and peripheral lipid metabolism disorders. Therefore, modulating ApoE expression or function to restore cellular lipid homeostasis may be a promising therapeutic target for AD comorbid with depression. This study also provided a better animal model for studying AD comorbid with depression.


Sujet(s)
Apolipoprotéine E4 , Dépression , Modèles animaux de maladie humaine , Métabolisme lipidique , Souris transgéniques , Animaux , Dépression/métabolisme , Apolipoprotéine E4/génétique , Souris , Maladie d'Alzheimer/métabolisme , Mâle , Humains , Cortex préfrontal/métabolisme , Comportement animal
11.
Int J Biol Macromol ; 273(Pt 2): 132854, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38838879

RÉSUMÉ

Depression is a neuropsychiatric disorder characterized by persistent pleasure loss and behavioral despair. However, the potential mechanisms and therapeutic targets for depression treatment remain unclear. Therefore, identifying the underlying pathogenesis of depression would promote the development of novel treatment and provide effective targets for antidepressant drugs. In this study, proteomics analysis showed that the expression level of phosphatase and actin regulator 4 (Phactr4) was significantly increased in the CA1 hippocampus of depressed rats. The upregulated Phactr4 might induce dysfunction of the synaptic structure via suppressing the p-LIMK/p-Cofilin signaling pathway, and promote neuroinflammation via activating the NF-κB/NLRP3 pathway, which ultimately contributes to the pathogenesis of depression. In contrast, the downregulation of Phactr4 in hippocampal CA1 of depressed rats alleviated depression-like behaviors, along with reducing neuroinflammation and improving synaptic plasticity. In conclusion, these findings provide evidence that Phactr4 plays an important role in regulating neuroinflammatory response and impairment of synaptic plasticity, effects seem to involve in the pathogenesis of depression, and Phactr4 may serve as a potential target for antidepressant treatment.


Sujet(s)
Dépression , Maladies neuro-inflammatoires , Plasticité neuronale , Stress psychologique , Animaux , Plasticité neuronale/effets des médicaments et des substances chimiques , Rats , Dépression/traitement médicamenteux , Dépression/métabolisme , Dépression/étiologie , Mâle , Maladies neuro-inflammatoires/métabolisme , Maladies neuro-inflammatoires/traitement médicamenteux , Stress psychologique/complications , Stress psychologique/métabolisme , Protéines des microfilaments/métabolisme , Rat Sprague-Dawley , Comportement animal/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Antidépresseurs/pharmacologie , Hippocampe/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Facteur de transcription NF-kappa B/métabolisme
12.
J Neuroinflammation ; 21(1): 143, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38822367

RÉSUMÉ

The dysregulation of pro- and anti-inflammatory processes in the brain has been linked to the pathogenesis of major depressive disorder (MDD), although the precise mechanisms remain unclear. In this study, we discovered that microglial conditional knockout of Pdcd4 conferred protection against LPS-induced hyperactivation of microglia and depressive-like behavior in mice. Mechanically, microglial Pdcd4 plays a role in promoting neuroinflammatory responses triggered by LPS by inhibiting Daxx-mediated PPARγ nucleus translocation, leading to the suppression of anti-inflammatory cytokine IL-10 expression. Finally, the antidepressant effect of microglial Pdcd4 knockout under LPS-challenged conditions was abolished by intracerebroventricular injection of the IL-10 neutralizing antibody IL-10Rα. Our study elucidates the distinct involvement of microglial Pdcd4 in neuroinflammation, suggesting its potential as a therapeutic target for neuroinflammation-related depression.


Sujet(s)
Protéines corépressives , Interleukine-10 , Souris knockout , Microglie , Maladies neuro-inflammatoires , Récepteur PPAR gamma , Transduction du signal , Animaux , Mâle , Souris , Protéines adaptatrices de la transduction du signal/déficit , Protéines adaptatrices de la transduction du signal/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines régulatrices de l'apoptose/métabolisme , Protéines régulatrices de l'apoptose/génétique , Protéines régulatrices de l'apoptose/déficit , Protéines corépressives/génétique , Protéines corépressives/métabolisme , Dépression/métabolisme , Dépression/étiologie , Interleukine-10/métabolisme , Interleukine-10/déficit , Interleukine-10/génétique , Lipopolysaccharides/toxicité , Souris de lignée C57BL , Microglie/métabolisme , Microglie/effets des médicaments et des substances chimiques , Chaperons moléculaires/génétique , Chaperons moléculaires/métabolisme , Maladies neuro-inflammatoires/métabolisme , Récepteur PPAR gamma/métabolisme , Récepteur PPAR gamma/génétique , Transduction du signal/physiologie , Transduction du signal/effets des médicaments et des substances chimiques
13.
Neuron ; 112(11): 1725-1727, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38843777

RÉSUMÉ

In this issue of Neuron, Li, Zhang, et al.1 find that the bile acid receptor TGR5 in the lateral hypothalamus influences neuronal dynamics underlying stress-induced depression-like behaviors. Inhibition of these neurons produces antidepressant-like effects through a circuit that includes hippocampal CA3 and dorsolateral septum, revealing a novel potential therapeutic for depression.


Sujet(s)
Acides et sels biliaires , Récepteurs couplés aux protéines G , Animaux , Acides et sels biliaires/métabolisme , Récepteurs couplés aux protéines G/métabolisme , Humains , Dépression/métabolisme
14.
Brain Res Bull ; 214: 111008, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38866373

RÉSUMÉ

The infralimbic (IL) cortex dysfunction has been implicated in major depressive disorder (MDD), yet the precise cellular and molecular mechanisms remain poorly understood. In this study, we investigated the role of layer V pyramidal neurons in a mouse model of MDD induced by repeated lipopolysaccharide (LPS) administration. Our results demonstrate that three days of systemic LPS administration induced depressive-like behavior and upregulated mRNA levels of interleukin-1ß (IL-1ß), tumor necrosis factor-alpha (TNF-α), and transforming growth factor-ß (TGF-ß) in the IL cortex. Electrophysiological recordings revealed a significant decrease in the intrinsic excitability of layer V pyramidal neurons in the IL following systemic LPS exposure. Importantly, chemogenetic activation of IL pyramidal neurons ameliorated LPS-induced depressive-like behavior. Additionally, LPS administration significantly increased microglial activity in the IL, as evidenced by a greater number of Ionized calcium binding adaptor molecule-1 (IBA-1)-positive cells. Morphometric analysis further unveiled enlarged soma, decreased branch numbers, and shorter branch lengths of microglial cells in the IL cortex following LPS exposure. Moreover, the activation of pyramidal neurons by clozapine-N-oxide increased the microglia branch length but did not change branch number or cytosolic area. These results collectively suggest that targeted activation of pyramidal neurons in the IL cortex mitigates microglial response and ameliorates depressive-like behaviors induced by systemic LPS administration. Therefore, our findings offer potential therapeutic targets for the development of interventions aimed at alleviating depressive symptoms by modulating IL cortical circuitry and microglial activity.


Sujet(s)
Lipopolysaccharides , Microglie , Cellules pyramidales , Animaux , Cellules pyramidales/effets des médicaments et des substances chimiques , Cellules pyramidales/métabolisme , Lipopolysaccharides/pharmacologie , Souris , Mâle , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Souris de lignée C57BL , Dépression/induit chimiquement , Dépression/métabolisme , Dépression/traitement médicamenteux , Clozapine/pharmacologie , Clozapine/analogues et dérivés , Modèles animaux de maladie humaine , Trouble dépressif majeur/métabolisme
15.
Int J Mol Sci ; 25(11)2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38892086

RÉSUMÉ

Chronic variable mild stress (CVS) in rats is a well-established paradigm for inducing depressive-like behaviors and has been utilized extensively to explore potential therapeutic interventions for depression. While the behavioral and neurobiological effects of CVS have been extensively studied, its impact on myocardial function remains largely unexplored. To induce the CVS model, rats were exposed to various stressors over 40 days. Behavioral assessments confirmed depressive-like behavior. Biochemical analyses revealed alterations in myocardial metabolism, including changes in NAD+ and NADP+, and NADPH concentrations. Free amino acid analysis indicated disturbances in myocardial amino acid metabolism. Evaluation of oxidative DNA damage demonstrated an increased number of abasic sites in the DNA of rats exposed to CVS. Molecular analysis showed significant changes in gene expression associated with glucose metabolism, oxidative stress, and cardiac remodeling pathways. Histological staining revealed minor morphological changes in the myocardium of CVS-exposed rats, including increased acidophilicity of cells, collagen deposition surrounding blood vessels, and glycogen accumulation. This study provides novel insights into the impact of chronic stress on myocardial function and metabolism, highlighting potential mechanisms linking depression and cardiovascular diseases. Understanding these mechanisms may aid in the development of targeted therapeutic strategies to mitigate the adverse cardiovascular effects of depression.


Sujet(s)
Myocarde , Stress oxydatif , Stress psychologique , Animaux , Rats , Myocarde/métabolisme , Myocarde/anatomopathologie , Mâle , Stress psychologique/métabolisme , Dépression/métabolisme , Dépression/anatomopathologie , Modèles animaux de maladie humaine , Altération de l'ADN , Adaptation physiologique , NAD/métabolisme , Glucose/métabolisme
16.
Int J Mol Sci ; 25(11)2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38892090

RÉSUMÉ

Fetal hypoxia and maternal stress frequently culminate in neuropsychiatric afflictions in life. To replicate this condition, we employed a model of prenatal severe hypoxia (PSH) during days 14-16 of rat gestation. Subsequently, both control and PSH rats at 3 months old were subjected to episodes of inescapable stress to induce learned helplessness (LH). The results of the open field test revealed an inclination towards depressive-like behavior in PSH rats. Following LH episodes, control (but not PSH) rats displayed significant anxiety. LH induced an increase in glucocorticoid receptor (GR) levels in extrahypothalamic brain structures, with enhanced nuclear translocation in the hippocampus (HPC) observed both in control and PSH rats. However, only control rats showed an increase in GR nuclear translocation in the amygdala (AMG). The decreased GR levels in the HPC of PSH rats correlated with elevated levels of hypothalamic corticotropin-releasing hormone (CRH) compared with the controls. However, LH resulted in a reduction of the CRH levels in PSH rats, aligning them with those of control rats, without affecting the latter. This study presents evidence that PSH leads to depressive-like behavior in rats, associated with alterations in the glucocorticoid system. Notably, these impairments also contribute to increased resistance to severe stressors.


Sujet(s)
Anxiété , Dépression , Glucocorticoïdes , Effets différés de l'exposition prénatale à des facteurs de risque , Récepteurs aux glucocorticoïdes , Animaux , Rats , Femelle , Anxiété/métabolisme , Grossesse , Glucocorticoïdes/métabolisme , Dépression/métabolisme , Dépression/étiologie , Récepteurs aux glucocorticoïdes/métabolisme , Effets différés de l'exposition prénatale à des facteurs de risque/métabolisme , Stress psychologique/métabolisme , Mâle , Corticolibérine/métabolisme , Hippocampe/métabolisme , Hypoxie/métabolisme , Phénotype , Comportement animal , Impuissance apprise , Modèles animaux de maladie humaine , Amygdale (système limbique)/métabolisme , Hypoxie foetale/métabolisme , Hypoxie foetale/complications
17.
BMC Neurol ; 24(1): 191, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38849737

RÉSUMÉ

BACKGROUND: Depression is a complex mood disorder whose pathogenesis involves multiple cell types and molecular pathways. The prefrontal cortex, as a key brain region for emotional regulation, plays a crucial role in depression. Microglia, as immune cells of the central nervous system, have been closely linked to the development and progression of depression through their dysfunctional states. This study aims to utilize single-cell RNA-seq technology to reveal the pathogenic mechanism of YAP1 in prefrontal cortex microglia in depression. METHODS: Firstly, we performed cell type identification and differential analysis on normal and depressed prefrontal cortex tissues by mining single-cell RNA-seq datasets from public databases. Focusing on microglia, we conducted sub-clustering, differential gene KEGG enrichment analysis, intercellular interaction analysis, and pseudotime analysis. Additionally, a cross-species analysis was performed to explore the similarities and differences between human and rhesus monkey prefrontal cortex microglia. To validate our findings, we combined bulk RNA-Seq and WGCNA analysis to reveal key genes associated with depression and verified the relationship between YAP1 and depression using clinical samples. RESULTS: Our study found significant changes in the proportion and transcriptional profiles of microglia in depressed prefrontal cortex tissues. Further analysis revealed multiple subpopulations of microglia and their associated differential genes and signaling pathways related to depression. YAP1 was identified as a key molecule contributing to the development of depression and was significantly elevated in depression patients. Moreover, the expression level of YAP1 was positively correlated with HAMD scores, suggesting its potential as a biomarker for predicting the onset of depression. CONCLUSION: This study utilized single-cell RNA-seq technology to reveal the pathogenic mechanism of YAP1 in prefrontal cortex microglia in depression, providing a new perspective for a deeper understanding of the pathophysiology of depression and identifying potential targets for developing novel treatment strategies.


Sujet(s)
Macaca mulatta , Microglie , Cortex préfrontal , Analyse sur cellule unique , Protéines de signalisation YAP , Cortex préfrontal/métabolisme , Microglie/métabolisme , Protéines de signalisation YAP/métabolisme , Humains , Animaux , Analyse sur cellule unique/méthodes , RNA-Seq , Dépression/métabolisme , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Protéines adaptatrices de la transduction du signal/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Mâle , Femelle , Analyse de l'expression du gène de la cellule unique
18.
J Neuroinflammation ; 21(1): 159, 2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38898454

RÉSUMÉ

A significant decrease in estrogen levels puts menopausal women at high risk for major depression, which remains difficult to cure despite its relatively clear etiology. With the discovery of abnormally elevated inflammation in menopausal depressed women, immune imbalance has become a novel focus in the study of menopausal depression. In this paper, we examined the characteristics and possible mechanisms of immune imbalance caused by decreased estrogen levels during menopause and found that estrogen deficiency disrupted immune homeostasis, especially the levels of inflammatory cytokines through the ERα/ERß/GPER-associated NLRP3/NF-κB signaling pathways. We also analyzed the destruction of the blood-brain barrier, dysfunction of neurotransmitters, blockade of BDNF synthesis, and attenuation of neuroplasticity caused by inflammatory cytokine activity, and investigated estrogen-immuno-neuromodulation disorders in menopausal depression. Current research suggests that drugs targeting inflammatory cytokines and NLRP3/NF-κB signaling molecules are promising for restoring homeostasis of the estrogen-immuno-neuromodulation system and may play a positive role in the intervention and treatment of menopausal depression.


Sujet(s)
Oestrogènes , Ménopause , Humains , Femelle , Ménopause/immunologie , Ménopause/métabolisme , Oestrogènes/métabolisme , Animaux , Dépression/immunologie , Dépression/métabolisme , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Transduction du signal/physiologie , Cytokines/métabolisme
19.
Brain Behav ; 14(6): e3604, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38898740

RÉSUMÉ

BACKGROUND AND AIM: Social isolation stress (SIS) is a stressor known to trigger depressive behaviors. Psychiatric disorders are associated with neurobiological changes, such as neuroinflammation and an increase in nitric oxide (NO) signaling. Despite the well-established detrimental effects of SIS and the involvement of neuroinflammation and NO in depression, potential management strategies, especially resocialization, remain insufficiently explored. Our aim was to elucidate the effects of resocialization on depressive behaviors in socially isolated mice, with a focus on the possible involvement of neuroinflammation and nitrite in the hippocampus (HIP). METHODS: We utilized 24 Naval Medical Research Institute male mice, maintained under both social and isolation conditions (SC and IC). After the isolation period, the mice were divided into two groups of eight, including the SIS group and a resocialized group. The SC group was kept without exposure to isolation stress. We conducted the open-field test, forced swimming test, and splash test to evaluate depressive behaviors. Additionally, nitrite levels, as well as the gene expression of interleukin (IL)-1ß, tumor necrosis factor (TNF), and toll-like receptor 4 (TLR4) in the HIP, were measured. RESULTS: The study found that resocialization significantly reduces depressive behaviors in SIS mice. The results suggest that the antidepressive effects of resocialization may be partially due to the modulation of the neuroinflammatory response and nitrite levels in the HIP. This is supported by the observed decrease in hippocampal gene expression of IL-1ß, TLR4, and TNF, along with a reduction in nitrite levels following resocialization. CONCLUSION: These insights could pave the way for new management strategies for depression, emphasizing the potential benefits of social interactions.


Sujet(s)
Dépression , Hippocampe , Nitrites , Isolement social , Stress psychologique , Animaux , Hippocampe/métabolisme , Souris , Mâle , Stress psychologique/métabolisme , Dépression/métabolisme , Dépression/étiologie , Dépression/physiopathologie , Nitrites/métabolisme , Maladies neuro-inflammatoires/métabolisme , Comportement animal/physiologie , Interleukine-1 bêta/métabolisme , Récepteur de type Toll-4/métabolisme , Facteur de nécrose tumorale alpha/métabolisme
20.
Ecotoxicol Environ Saf ; 280: 116521, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38850708

RÉSUMÉ

The aim of this study is to investigate the role of estrogen receptor ß (ERß) in nonylphenol (NP) - induced depression - like behavior in rats and its impact on the regulation of the TPH2/5-HT pathway. In the in vitro experiment, rat basophilic leukaemia cells (RBL-2H3) cells were divided into the four groups: blank group, NP group (20 µM), ERß agonist group (0.01 µM), and NP+ERß agonist group (20 µM+0.01 µM). For the in vivo experiment, 72 adult male Sprague-Dawley rats were randomly divided into following six groups: the Control, NP (40 mg/kg) group, ERß agonist (2 mg/kg, Diarylpropionitrile (DPN)) group, ERß inhibitor (0.1 mg/kg, 4-(2-phenyl-5,7-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-3-yl) phenol (PHTPP)) group, NP+ERß agonist (40 mg/kg NP + 2 mg/kg DPN) group, and NP+ERß inhibitor (40 mg/kg NP + 0.1 mg/kg PHTPP) group, with 12 rats in each group. Each rat in drug group were given NP by gavage and/or received a single intraperitoneal injection of DPN 2 mg/kg or PHTPP 0.1 mg/kg. Both in vivo and in vitro, NP group showed a decrease in the expression levels of ERß, tryptophan hydroxylase (TPH1), and tryptophan hydroxylase-2 (TPH2) genes and proteins, and reduced levels of DA, NE, and 5-hydroxytryptophan (5-HT) neurotransmitters. RBL-2H3 cells showed signs of cell shrinkage, with rounded cells, increased suspension and more loosely arranged cells. The effectiveness of the ERß agonist stimulation exhibited an increase exceeding 60% in RBL-2H3 cells. The application of ERß agonist resulted in an alleviation the aforementioned alterations. ERß agonist activated the TPH2/5-HT signaling pathways. Compared to the control group, the NP content in the brain tissue of the NP group was significantly increased. The latency to eat for the rats was longer and the amount of food consumed was lower, and the rats had prolonged immobility time in the behavioral experiment of rats. The expression levels of ERß, TPH1, TPH2, 5-HT and 5-HITT proteins were decreased in the NP group, suggesting NP-induced depression-like behaviours as well as disturbances in the secretion of serum hormones and monoamine neurotransmitters. In the NP group, the midline raphe nucleus showed an elongated nucleus with a dark purplish-blue colour, nuclear atrophy, displacement and pale cytoplasm. ERß might ameliorate NP-induced depression-like behaviors, and secretion disorders of serum hormones and monoamine neurotransmitters via activating TPH2/5-HT signaling pathways.


Sujet(s)
Dépression , Récepteur bêta des oestrogènes , Phénols , Rat Sprague-Dawley , Sérotonine , Tryptophane 5-monooxygenase , Animaux , Tryptophane 5-monooxygenase/métabolisme , Récepteur bêta des oestrogènes/métabolisme , Phénols/toxicité , Mâle , Rats , Sérotonine/métabolisme , Dépression/induit chimiquement , Dépression/traitement médicamenteux , Dépression/métabolisme , Agents neuromédiateurs/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Nitriles/toxicité , Nitriles/pharmacologie , Propionates/toxicité , Propionates/pharmacologie , Pyrazoles , Pyrimidines
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...