Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 8.427
Filtrer
1.
Transl Psychiatry ; 14(1): 400, 2024 Oct 02.
Article de Anglais | MEDLINE | ID: mdl-39358323

RÉSUMÉ

Major depressive disorder (MDD) is a common disease affecting 300 million people worldwide. The existing drugs are ineffective for approximately 30% of patients, so it is urgent to develop new antidepressant drugs with novel mechanisms. Here, we found that norisoboldine (NOR) showed an antidepressant efficacy in the chronic social defeat stress (CSDS) depression model in the tail suspension, forced swimming, and sucrose consumption tests. We then utilized the drug-treated CSDS mice paradigm to segregate and gain differential protein groups of CSDS versus CON (CSDSCON), imipramine (IMI)-treated versus CSDS (IMICSDS), and NOR-treated versus CSDS (NORCSDS) from the prefrontal cortex. These protein expression alterations were first analyzed by ANOVA with p < 0.05. The protein cluster 1 and cluster 3, in which the pattern of protein levels similar to the mood pattern, showed enrichment in functions and localizations related to mitochondrion, ribosome and synapses. Further GO analysis of the common proteins for NORCSDS groups and NORIMI groups supported the findings from ANOVA analysis. We employed Protein-Protein interaction (PPI) analysis to examine the proteins of NORCSDS and NORIMI, revealing an enrichment of the proteins associated with the mitochondrial ribosomal and synaptic functions. Further independent analysis using parallel reaction monitoring (PRM) revealed that Cox7c, Mrp142, Naa30, Ighm, Apoa4, Ssu72, Mrps30, Apoh, Acbd5, and Cdv3, exhibited regulation in the NOR-treated group to support the homeostasis of mitochondrial functions. Additionally, Dcx, Arid1b, Rnf112, and Fam3c, were also observed to undergo modulation in the NOR-treated groups to support the synaptic formation and functions. These findings suggest that the proteins involved in depression treatment exert effects in strengthen the mitochondrial and synaptic functions in the mice PFC. Western blot analysis supported the data that the levels of Mrpl42, Cox7c, Naa30, Rnf112, Dcx Apoa4, Apoh and Fam3c were altered in the CSDS mice, and rescued by NOR treatment, supporting the PRM data. NOR treatment also rescued the NLRP3 inflammasome activation in CSDS mice. In summary, the current proteomic research conducted on the prefrontal cortex has provided valuable insights into the specific and shared molecular mechanisms underlying pathophysiology and treatment to CSDS-induced depression, shedding light on the therapeutic effects of Norisoboldine.


Sujet(s)
Antidépresseurs , Modèles animaux de maladie humaine , Mitochondries , Cortex préfrontal , Protéomique , Stress psychologique , Animaux , Souris , Antidépresseurs/pharmacologie , Antidépresseurs/usage thérapeutique , Mâle , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Cortex préfrontal/métabolisme , Cortex préfrontal/effets des médicaments et des substances chimiques , Stress psychologique/traitement médicamenteux , Stress psychologique/métabolisme , Synapses/effets des médicaments et des substances chimiques , Synapses/métabolisme , Trouble dépressif majeur/traitement médicamenteux , Trouble dépressif majeur/métabolisme , Souris de lignée C57BL , Protéine doublecortine , Dépression/traitement médicamenteux , Dépression/métabolisme , Comportement animal/effets des médicaments et des substances chimiques , Défaite sociale
2.
Transl Psychiatry ; 14(1): 426, 2024 Oct 08.
Article de Anglais | MEDLINE | ID: mdl-39379355

RÉSUMÉ

Importin α4, which is encoded by the Kpna4 gene, is a well-characterized nuclear-cytoplasmic transport factor known to mediate transport of transcription factors including NF-κB. Here, we report that Kpna4 knock-out (KO) mice exhibit psychiatric disorder-related behavioral abnormalities such as anxiety-related behaviors, decreased social interaction, and sensorimotor gating deficits. Contrary to a previous study predicting attenuated NF-κB activity as a result of Kpna4 deficiency, we observed a significant increase in expression levels of NF-κB genes and proinflammatory cytokines such as TNFα, Il-1ß or Il-6 in the prefrontal cortex or basolateral amygdala of the KO mice. Moreover, examination of inflammatory responses in primary cells revealed that Kpna4 deficient cells have an increased inflammatory response, which was rescued by addition of not only full length, but also a nuclear transport-deficient truncation mutant of importin α4, suggesting contribution of its non-transport functions. Furthermore, RNAseq of sorted adult microglia and astrocytes and subsequent transcription factor analysis suggested increases in polycomb repressor complex 2 (PRC2) activity in Kpna4 KO cells. Taken together, importin α4 deficiency induces psychiatric disorder-related behavioral deficits in mice, along with an increased inflammatory response and possible alteration of PRC2 activity in glial cells.


Sujet(s)
Comportement animal , Souris knockout , Maladies neuro-inflammatoires , Cariophérines alpha , Animaux , Souris , Maladies neuro-inflammatoires/métabolisme , Cariophérines alpha/génétique , Cariophérines alpha/métabolisme , Comportement animal/physiologie , Mâle , Anxiété/génétique , Anxiété/métabolisme , Cortex préfrontal/métabolisme , Microglie/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Cytokines/métabolisme , Troubles mentaux/génétique , Troubles mentaux/métabolisme , Souris de lignée C57BL , Modèles animaux de maladie humaine , Astrocytes/métabolisme , Groupe nucléaire basolatéral/métabolisme
3.
Am J Psychiatry ; 181(10): 920-934, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39350613

RÉSUMÉ

OBJECTIVE: In schizophrenia, impaired working memory is associated with transcriptome alterations in layer 3 pyramidal neurons (L3PNs) in the dorsolateral prefrontal cortex (DLPFC). Distinct subtypes of L3PNs that send axonal projections to the DLPFC in the opposite hemisphere (callosal projection [CP] neurons) or the parietal cortex in the same hemisphere (ipsilateral projection [IP] neurons) play critical roles in working memory. However, how the transcriptomes of these L3PN subtypes might shift during late postnatal development when working memory impairments emerge in individuals later diagnosed with schizophrenia is not known. The aim of this study was to characterize and compare the transcriptome profiles of CP and IP L3PNs across developmental transitions from prepuberty to adulthood in macaque monkeys. METHODS: The authors used retrograde labeling to identify CP and IP L3PNs in the DLPFC of prepubertal, postpubertal, and adult macaque monkeys, and used laser microdissection to capture these neurons for RNA sequencing. RESULTS: At all three ages, CP and IP L3PNs had distinct transcriptomes, with the number of genes differentially expressed between neuronal subtypes increasing with age. For IP L3PNs, age-related shifts in gene expression were most prominent between prepubertal and postpubertal animals, whereas for CP L3PNs such shifts were most prominent between postpubertal and adult animals. CONCLUSIONS: These findings demonstrate the presence of cell type-specific profiles and developmental trajectories of the transcriptomes of PPC-projecting IP and DLPFC-projecting CP L3PNs in monkey DLPFC. The evidence that IP L3PNs reach a mature transcriptome earlier than CP L3PNs suggests that these two subtypes differentially contribute to the maturation of working memory performance across late postnatal development and that they may be differentially vulnerable to the disease process of schizophrenia at specific stages of postnatal development.


Sujet(s)
Cellules pyramidales , Schizophrénie , Transcriptome , Animaux , Schizophrénie/génétique , Schizophrénie/anatomopathologie , Schizophrénie/métabolisme , Cellules pyramidales/métabolisme , Mâle , Mémoire à court terme/physiologie , Cortex préfrontal dorsolatéral , Macaca mulatta , Cortex préfrontal/métabolisme , Cortex préfrontal/croissance et développement , Femelle
4.
Addict Biol ; 29(10): e13440, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39380299

RÉSUMÉ

Relapse is a major challenge in treating opioid addiction, including oxycodone. During abstinence, oxycodone seeking progressively increases, a phenomenon termed incubation of oxycodone craving. We previously demonstrated a causal role of orbitofrontal cortex (OFC) in this incubation. Here, we studied the interaction between glutamatergic projections from OFC and dopamine 1-family receptor (D1R) signaling in dorsal striatum (DS) in this incubation in male rats. We first examined the causal role of D1R signalling in DS in incubated oxycodone seeking. Next, we combined fluorescence-conjugated cholera toxin subunit B (CTb-555, a retrograde tracer) with Fos (a neuronal activity marker) to assess whether the activation of OFC→DS projections was associated with incubated oxycodone seeking. We then used a pharmacological asymmetrical disconnection procedure to examine the role of the interaction between projections from OFC and D1R signalling in DS in incubated oxycodone seeking. We also tested the effect of unilateral pharmacological inactivation of OFC or unilateral D1R blockade of DS on incubated oxycodone seeking. Finally, we assessed whether contralateral disconnection of OFC→DS projections impacted non-incubated oxycodone seeking on abstinence day 1. We found that D1R blockade in DS decreased incubated oxycodone seeking and OFC→DS projections were activated during incubated oxycodone seeking. Moreover, anatomical disconnection of OFC→DS projections, but not unilateral inactivation of OFC or unilateral D1R blockade in DS, decreased incubated oxycodone seeking. Lastly, contralateral disconnection of OFC→DS projections had no effect on oxycodone seeking on abstinence day 1. Together, these results demonstrated a causal role of OFC→DS projections in incubation of oxycodone craving.


Sujet(s)
Corps strié , Besoin impérieux , Comportement de recherche de substances , Oxycodone , Cortex préfrontal , Animaux , Oxycodone/pharmacologie , Cortex préfrontal/effets des médicaments et des substances chimiques , Cortex préfrontal/métabolisme , Mâle , Rats , Besoin impérieux/effets des médicaments et des substances chimiques , Besoin impérieux/physiologie , Comportement de recherche de substances/effets des médicaments et des substances chimiques , Corps strié/métabolisme , Corps strié/effets des médicaments et des substances chimiques , Troubles liés aux opiacés/physiopathologie , Récepteur dopamine D1/métabolisme , Voies nerveuses/effets des médicaments et des substances chimiques , Rat Sprague-Dawley , Analgésiques morphiniques/pharmacologie
5.
Transl Psychiatry ; 14(1): 428, 2024 Oct 09.
Article de Anglais | MEDLINE | ID: mdl-39384764

RÉSUMÉ

Structural and functional changes of the brain are assumed to contribute to excessive cocaine intake, craving, and relapse in cocaine use disorder (CUD). Epigenetic and transcriptional changes were hypothesized as a molecular basis for CUD-associated brain alterations. Here we performed a multi-omics study of CUD by integrating epigenome-wide methylomic (N = 42) and transcriptomic (N = 25) data from the same individuals using postmortem brain tissue of Brodmann Area 9 (BA9). Of the N = 1 057 differentially expressed genes (p < 0.05), one gene, ZFAND2A, was significantly upregulated in CUD at transcriptome-wide significance (q < 0.05). Differential alternative splicing (AS) analysis revealed N = 98 alternatively spliced transcripts enriched in axon and dendrite extension pathways. Strong convergent overlap in CUD-associated expression deregulation was found between our BA9 cohort and independent replication datasets. Epigenomic, transcriptomic, and AS changes in BA9 converged at two genes, ZBTB4 and INPP5E. In pathway analyses, synaptic signaling, neuron morphogenesis, and fatty acid metabolism emerged as the most prominently deregulated biological processes. Drug repositioning analysis revealed glucocorticoid receptor targeting drugs as most potent in reversing the CUD expression profile. Our study highlights the value of multi-omics approaches for an in-depth molecular characterization and provides insights into the relationship between CUD-associated epigenomic and transcriptomic signatures in the human prefrontal cortex.


Sujet(s)
Troubles liés à la cocaïne , Méthylation de l'ADN , Transcriptome , Humains , Troubles liés à la cocaïne/génétique , Mâle , Femelle , Adulte , Analyse de profil d'expression de gènes , Épigenèse génétique , Adulte d'âge moyen , Épigénomique , Encéphale/métabolisme , Cortex préfrontal/métabolisme , Multi-omique
6.
J Neurosci Res ; 102(9): e25387, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39314180

RÉSUMÉ

Attention-deficit/hyperactivity disorder (ADHD) and substance use disorders (SUD) are characterized by exacerbated motor and risk-related impulsivities, which are associated with decreased cortical activity. In rodents, the medial prefrontal cortex (mPFC) and nucleus accumbens (NAc) have been separately implicated in impulsive behaviors, but studies on the specific role of the mPFC-NAc pathway in these behaviors are limited. Here, we investigated whether heightened impulsive behaviors are associated with reduced mPFC activity in rodents and determined the involvement of the mPFC-NAc pathway in motor and risk-related impulsivities. We used the Roman High- (RHA) and Low-Avoidance (RLA) rat lines, which display divergent phenotypes in impulsivity. To investigate alterations in cortical activity in relation to impulsivity, regional brain glucose metabolism was measured using positron emission tomography and [18F]-fluorodeoxyglucose ([18F]FDG). Using chemogenetics, the activity of the mPFC-NAc pathway was either selectively activated in high-impulsive RHA rats or inhibited in low-impulsive RLA rats, and the effects of these manipulations on motor and risk-related impulsivity were concurrently assessed using the rat gambling task. We showed that basal [18F]FDG uptake was lower in the mPFC and NAc of RHA compared to RLA rats. Activation of the mPFC-NAc pathway in RHA rats reduced motor impulsivity, without affecting risk-related decision-making. Conversely, inhibition of the mPFC-NAc pathway had no effect in RLA rats. Our results suggest that the mPFC-NAc pathway controls motor impulsivity, but has limited involvement in risk-related decision-making in our current model. Our findings suggest that reducing fronto-striatal activity may help attenuate motor impulsivity in patients with impulse control dysregulation.


Sujet(s)
Prise de décision , Comportement impulsif , Noyau accumbens , Cortex préfrontal , Animaux , Comportement impulsif/physiologie , Cortex préfrontal/métabolisme , Mâle , Noyau accumbens/métabolisme , Rats , Prise de décision/physiologie , Voies nerveuses/physiologie , Prise de risque , Tomographie par émission de positons , Activité motrice/physiologie
7.
Int J Mol Sci ; 25(18)2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-39337262

RÉSUMÉ

Mu opioid receptors (MORs) represent a vital mechanism related to the modulation of stress-induced analgesia (SIA). Previous studies have reported on the gamma-aminobutyric acid (GABA)ergic "disinhibition" mechanisms of MORs on the descending pain modulatory pathway of SIA induced in the midbrain. However, the role of the MORs expressed in the medial prefrontal cortex (mPFC), one of the main cortical areas participating in pain modulation, in SIA remains completely unknown. In this study, we investigated the contributions of MORs expressed on glutamatergic (MORGlut) and GABAergic (MORGABA) neurons of the medial prefrontal cortex (mPFC), as well as the functional role and activity of neurons projecting from the mPFC to the periaqueductal gray (PAG) region, in male mice. We achieved this through a combination of hot-plate tests, c-fos staining, and 1 h acute restraint stress exposure tests. The results showed that our acute restraint stress protocol produced mPFC MOR-dependent SIA effects. In particular, MORGABA was found to play a major role in modulating the effects of SIA, whereas MORGlut seemed to be unconnected to the process. We also found that mPFC-PAG projections were efficiently activated and played key roles in the effects of SIA, and their activation was mediated by MORGABA to a large extent. These results indicated that the activation of mPFC MORGABA due to restraint stress was able to activate mPFC-PAG projections in a potential "disinhibition" pathway that produced analgesic effects. These findings provide a potential theoretical basis for pain treatment or drug screening targeting the mPFC.


Sujet(s)
Analgésie , Cortex préfrontal , Récepteur mu , Contention physique , Stress psychologique , Animaux , Cortex préfrontal/métabolisme , Mâle , Souris , Récepteur mu/métabolisme , Analgésie/méthodes , Stress psychologique/métabolisme , Douleur/métabolisme , Substance grise centrale du mésencéphale/métabolisme , Neurones GABAergiques/métabolisme
8.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(8): 1441-1449, 2024 Aug 20.
Article de Chinois | MEDLINE | ID: mdl-39276039

RÉSUMÉ

OBJECTIVE: To investigate the effect of Kaixinsan (KXS, a traditional Chinese medicine formula) for alleviating adriamycin-induced depression-like behaviors in mice bearing breast cancer xenografts and explore the pharmacological mechanism. METHODS: Forty female BALB/c mice were randomized equally into control group, model group, and low- and high-dose KXS treatment groups, and in the latter 3 groups, mouse models bearing orthotopic breast cancer 4T1 cell xenografts were established and treated with adriamycin along with saline or KXS via gavage. Depression-like behaviors of the mice were assessed using open field test and elevated plus-maze test, and the changes in serum levels of depression-related factors were examined. RNA-seq analysis and transmission electron microscopy were used and ferroptosis-related factors were detected to explore the mechanisms of adriamycin-induced depression and the therapeutic mechanism of KXS. The results were verified in SH-SY5Y cells using ferroptosis inhibitor Fer-1 as the positive control. RESULTS: KXS significantly alleviated depression-like behaviors and depression-related serological changes induced by adriamycin in the mouse models. RNA-seq results suggested that KXS alleviated chemotherapy-induced depression by regulating oxidative stress, lipid metabolism and iron ion binding in the prefrontal cortex. Pathological analysis and detection of ferroptosis-related factors showed that KXS significantly reduced ferroptosis in the prefrontal cortex of adriamycin-treated mice. In SH-SY5Y cells, both KXS-medicated serum and the ferroptosis inhibitor were capable of attenuating adriamycin-induced cell ferroptosis. CONCLUSION: KXS alleviates adriamycininduced depression-like behaviors in mice by reducing ferroptosis in the prefrontal cortex of breast cancer-bearing mice.


Sujet(s)
Dépression , Doxorubicine , Ferroptose , Souris de lignée BALB C , Cortex préfrontal , Animaux , Ferroptose/effets des médicaments et des substances chimiques , Souris , Dépression/traitement médicamenteux , Dépression/induit chimiquement , Doxorubicine/effets indésirables , Femelle , Cortex préfrontal/métabolisme , Cortex préfrontal/effets des médicaments et des substances chimiques , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/usage thérapeutique , Lignée cellulaire tumorale , Comportement animal/effets des médicaments et des substances chimiques , Humains , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme
9.
Nat Commun ; 15(1): 8176, 2024 Sep 18.
Article de Anglais | MEDLINE | ID: mdl-39289358

RÉSUMÉ

The Claustrum/dorsal endopiriform cortex complex (CLA) is an enigmatic brain region with extensive glutamatergic projections to multiple cortical areas. The transcription factor Nurr1 is highly expressed in the CLA, but its role in this region is not understood. By using conditional gene-targeted mice, we show that Nurr1 is a crucial regulator of CLA neuron identity. Although CLA neurons remain intact in the absence of Nurr1, the distinctive gene expression pattern in the CLA is abolished. CLA has been hypothesized to control hallucinations, but little is known of how the CLA responds to hallucinogens. After the deletion of Nurr1 in the CLA, both hallucinogen receptor expression and signaling are lost. Furthermore, functional ultrasound and Neuropixel electrophysiological recordings revealed that the hallucinogenic-receptor agonists' effects on functional connectivity between prefrontal and sensorimotor cortices are altered in Nurr1-ablated mice. Our findings suggest that Nurr1-targeted strategies provide additional avenues for functional studies of the CLA.


Sujet(s)
Claustrum , Hallucinogènes , Neurones , Membre-2 du groupe A de la sous-famille-4 de récepteurs nucléaires , Animaux , Membre-2 du groupe A de la sous-famille-4 de récepteurs nucléaires/métabolisme , Membre-2 du groupe A de la sous-famille-4 de récepteurs nucléaires/génétique , Souris , Hallucinogènes/pharmacologie , Claustrum/métabolisme , Neurones/métabolisme , Mâle , Souris knockout , Souris de lignée C57BL , Cortex préfrontal/métabolisme , Cortex préfrontal/physiologie , Cortex sensorimoteur/métabolisme , Cortex sensorimoteur/physiologie
10.
CNS Neurosci Ther ; 30(9): e70048, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39300492

RÉSUMÉ

AIMS: The study aimed to assess brain metabolite differences in the medial prefrontal cortex (mPFC) between acute and euthymic episodes of bipolar disorder (BD) with both mania and depression over a 6-month medication treatment period. METHODS: We utilized 1H-MRS technology to assess the metabolite levels in 53 individuals with BD (32 in depressive phase, 21 in manic phase) and 34 healthy controls (HCs) at baseline. After 6 months of medication treatment, 40 subjects underwent a follow-up scan in euthymic state. Metabolite levels, including N-acetyl aspartate (NAA), glutamate (Glu), and Glutamine (Gln), were measured in the mPFC. RESULTS: Patients experiencing depressive and manic episodes exhibited a notable reduction in NAA/Cr + PCr ratios at baseline compared to healthy controls (p = 0.004; p = 0.006) in baseline, compared with HCs. Over the 6-month follow-up period, the manic group displayed a significant decrease in Gln/Cr + PCr compared to the initial acute phase (p = 0.03). No significant alterations were found in depressed group between baseline and follow-up. CONCLUSION: This study suggests that NAA/Cr + PCr ratios and Gln/Cr + PCr ratios in the mPFC may be associated with manic and depressive episodes, implicating that Gln and NAA might be useful biomarkers for distinguishing mood phases in BD and elucidating its mechanisms.


Sujet(s)
Acide aspartique , Trouble bipolaire , Acide glutamique , Glutamine , Cortex préfrontal , Spectroscopie par résonance magnétique du proton , Humains , Trouble bipolaire/traitement médicamenteux , Trouble bipolaire/métabolisme , Trouble bipolaire/imagerie diagnostique , Cortex préfrontal/métabolisme , Cortex préfrontal/effets des médicaments et des substances chimiques , Cortex préfrontal/imagerie diagnostique , Mâle , Femelle , Adulte , Acide aspartique/analogues et dérivés , Acide aspartique/métabolisme , Glutamine/métabolisme , Acide glutamique/métabolisme , Adulte d'âge moyen , Études de suivi , Créatine/métabolisme , Jeune adulte , Phosphocréatine/métabolisme
11.
J Cell Mol Med ; 28(17): e18578, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39234952

RÉSUMÉ

Kruppel-like factor 4 (Klf4) is a transcription factor that is involved in neuronal regeneration and the development of glutamatergic systems. However, it is unknown whether Klf4 is involved in acute seizure. To investigate the potential role of Klf4 in pentylenetetrazol (PTZ)-induced seizure, western blotting, immunofluorescence, behaviour test and electrophysiology were conducted in this study. We found that Klf4 protein and mRNA expression were increased in both the hippocampus (HP) and prefrontal cortex (PFC) after PTZ-induced seizure in mice. HP-specific knockout (KO) of Klf4 in mice decreased protein expression of Klf4 and the down-stream Klf4 target tumour protein 53 (TP53/P53). These molecular changes are accompanied by increased seizure latency, reduced immobility time in the forced swimming test and tail suspension test. Reduced hippocampal protein levels for synaptic proteins, including glutamate receptor 1 (GRIA1/GLUA1) and postsynaptic density protein 95 (DLG4/PSD95), were also observed after Klf4-KO, while increased mRNA levels of complement proteins were observed for complement component 1q subcomponent A (C1qa), complement component 1q subcomponent B (C1qb), complement component 1q subcomponent C (C1qc), complement component 3 (C3), complement component 4A (C4a) and complement component 4B (C4b). Moreover, c-Fos expression induced by PTZ was reduced by hippocampal conditional KO of Klf4. Electrophysiology showed that PTZ-induced action potential frequency was decreased by overexpression of Klf4. In conclusion, these findings suggest that Klf4 plays an important role in regulating PTZ-induced seizures and therefore constitutes a new molecular target that should be explored for the development of antiepileptic drugs.


Sujet(s)
Hippocampe , Facteur-4 de type Kruppel , Facteurs de transcription Krüppel-like , Souris knockout , Pentétrazol , Crises épileptiques , Animaux , Facteur-4 de type Kruppel/métabolisme , Crises épileptiques/métabolisme , Crises épileptiques/induit chimiquement , Crises épileptiques/génétique , Facteurs de transcription Krüppel-like/métabolisme , Facteurs de transcription Krüppel-like/génétique , Souris , Hippocampe/métabolisme , Mâle , Cortex préfrontal/métabolisme , Souris de lignée C57BL , Modèles animaux de maladie humaine
12.
Sci Prog ; 107(3): 368504241264994, 2024.
Article de Anglais | MEDLINE | ID: mdl-39228316

RÉSUMÉ

Millions of people suffer from opioid use disorder, because of the ongoing opioid epidemic. The aversive symptoms of withdrawal are a leading factor for drug relapses, yet there are limited therapeutic options to minimize or prevent withdrawal symptoms. The mechanism behind opioid withdrawal is still not fully understood, thus preventing the development of new therapeutics. This study is an extension of our previously proposed mechanism of a toll-like receptor 2 (TLR2) mediated withdrawal response as a result of morphine induced microbial change that occurs during morphine withdrawal. Transcriptome analysis of the pre-frontal cortex indicated that there was increased expression of genes related to TLR2 signaling in morphine withdrawal treated animals compared to placebo controls. Antibiotic treatment further altered TLR2 related genes, recovering some of the morphine induced effect and leading to additional suppression of some genes related to the TLR2 pathway. Morphine withdrawal induced gene expression was attenuated in a whole body TLR2 knockout model. These results provide more support that TLR2 plays an integral role in morphine withdrawal mechanisms and could be a potential therapeutic target to minimize opioid withdrawal associated co-morbidities.


Sujet(s)
Morphine , Cortex préfrontal , Transduction du signal , Syndrome de sevrage , Récepteur de type Toll-2 , Récepteur de type Toll-2/génétique , Récepteur de type Toll-2/métabolisme , Syndrome de sevrage/génétique , Syndrome de sevrage/métabolisme , Cortex préfrontal/métabolisme , Cortex préfrontal/effets des médicaments et des substances chimiques , Animaux , Transduction du signal/effets des médicaments et des substances chimiques , Souris , Mâle , Souris knockout , Souris de lignée C57BL , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Dépendance à la morphine/génétique , Dépendance à la morphine/métabolisme
13.
Gut Microbes ; 16(1): 2401939, 2024.
Article de Anglais | MEDLINE | ID: mdl-39259834

RÉSUMÉ

Early life stress alters gut microbiota and increases the risk of neuropsychiatric disorders, including social deficits and anxiety, in the host. However, the role of gut commensal bacteria in early life stress-induced neurobehavioral abnormalities remains unclear. Using the maternally separated (MS) mice, our research has unveiled a novel aspect of this complex relationship. We discovered that the reduced levels of amino acid transporters in the intestine of MS mice led to low glutamine (Gln) levels in the blood and synaptic dysfunction in the medial prefrontal cortex (mPFC). Abnormally low blood Gln levels limit the brain's availability of Gln, which is required for presynaptic glutamate (Glu) and γ-aminobutyric acid (GABA) replenishment. Furthermore, MS resulted in gut microbiota dysbiosis characterized by a reduction in the relative abundance of Lactobacillus reuteri (L. reuteri). Notably, supplementation with L. reuteri ameliorates neurobehavioral abnormalities in MS mice by increasing intestinal amino acid transport and restoring synaptic transmission in the mPFC. In conclusion, our findings on the role of L. reuteri in regulating intestinal amino acid transport and buffering early life stress-induced behavioral abnormalities provide a novel insight into the microbiota-gut-brain signaling basis for emotional behaviors.


Sujet(s)
Anxiété , Microbiome gastro-intestinal , Stress psychologique , Animaux , Microbiome gastro-intestinal/physiologie , Souris , Anxiété/microbiologie , Anxiété/métabolisme , Stress psychologique/microbiologie , Stress psychologique/métabolisme , Acides aminés/métabolisme , Mâle , Souris de lignée C57BL , Systèmes de transport d'acides aminés/métabolisme , Cortex préfrontal/métabolisme , Comportement animal , Dysbiose/microbiologie , Séparation d'avec la mère , Glutamine/métabolisme , Axe cerveau-intestin/physiologie , Transmission synaptique , Femelle , Acide glutamique/métabolisme
14.
Int J Mol Sci ; 25(17)2024 Aug 31.
Article de Anglais | MEDLINE | ID: mdl-39273422

RÉSUMÉ

Alzheimer's disease (AD), the leading cause of dementia, is a multifactorial disease influenced by aging, genetics, and environmental factors. miRNAs are crucial regulators of gene expression and play significant roles in AD onset and progression. This exploratory study analyzed the expression levels of 28 genes and 5 miRNAs (miR-124-3p, miR-125b-5p, miR-21-5p, miR-146a-5p, and miR-155-5p) related to AD pathology and neuroimmune responses using RT-qPCR. Analyses were conducted in the prefrontal cortex (PFC) and the hippocampus (HPC) of the 5xFAD mouse AD model at 6 and 9 months old. Data highlighted upregulated genes encoding for glial fibrillary acidic protein (Gfap), triggering receptor expressed on myeloid cells (Trem2) and cystatin F (Cst7), in the 5xFAD mice at both regions and ages highlighting their roles as critical disease players and potential biomarkers. Overexpression of genes encoding for CCAAT enhancer-binding protein alpha (Cebpa) and myelin proteolipid protein (Plp) in the PFC, as well as for BCL2 apoptosis regulator (Bcl2) and purinergic receptor P2Y12 (P2yr12) in the HPC, together with upregulated microRNA(miR)-146a-5p in the PFC, prevailed in 9-month-old animals. miR-155 positively correlated with miR-146a and miR-21 in the PFC, and miR-125b positively correlated with miR-155, miR-21, while miR-146a in the HPC. Correlations between genes and miRNAs were dynamic, varying by genotype, region, and age, suggesting an intricate, disease-modulated interaction between miRNAs and target pathways. These findings contribute to our understanding of miRNAs as therapeutic targets for AD, given their multifaceted effects on neurons and glial cells.


Sujet(s)
Maladie d'Alzheimer , Modèles animaux de maladie humaine , Hippocampe , microARN , Névroglie , Neurones , Animaux , microARN/génétique , microARN/métabolisme , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Souris , Neurones/métabolisme , Névroglie/métabolisme , Hippocampe/métabolisme , Souris transgéniques , Récepteurs immunologiques/génétique , Récepteurs immunologiques/métabolisme , Régulation de l'expression des gènes , Glycoprotéines membranaires/génétique , Glycoprotéines membranaires/métabolisme , Cortex préfrontal/métabolisme , Protéine gliofibrillaire acide/métabolisme , Protéine gliofibrillaire acide/génétique , Mâle
15.
Int J Mol Sci ; 25(17)2024 Sep 06.
Article de Anglais | MEDLINE | ID: mdl-39273621

RÉSUMÉ

Depression is a prevalent and debilitating mental disorder that affects millions worldwide. Current treatments, such as antidepressants targeting the serotonergic system, have limitations, including delayed onset of action and high rates of treatment resistance, necessitating novel therapeutic strategies. Ginsenoside Rc (G-Rc) has shown potential anti-inflammatory and neuroprotective effects, but its antidepressant properties remain unexplored. This study investigated the antidepressant effects of G-Rc in an L-alpha-aminoadipic acid (L-AAA)-induced mouse model of depression, which mimics the astrocytic pathology and neuroinflammation observed in major depressive disorder. Mice were administered G-Rc, vehicle, or imipramine orally after L-AAA injection into the prefrontal cortex. G-Rc significantly reduced the immobility time in forced swimming and tail suspension tests compared to vehicle treatment, with more pronounced effects than imipramine. It also attenuated the expression of pro-inflammatory cytokines (TNF-α, IL-6, TGF-ß, lipocalin-2) and alleviated astrocytic degeneration, as indicated by increased GFAP and decreased IBA-1 levels. Additionally, G-Rc modulated apoptosis-related proteins, decreasing caspase-3 and increasing Bcl-2 levels compared to the L-AAA-treated group. These findings suggest that G-Rc exerts antidepressant effects by regulating neuroinflammation, astrocyte-microglia crosstalk, and apoptotic pathways in the prefrontal cortex, highlighting its potential as a novel therapeutic agent for depression.


Sujet(s)
Acide 2-amino-adipique , Antidépresseurs , Astrocytes , Ginsénosides , Animaux , Astrocytes/effets des médicaments et des substances chimiques , Astrocytes/métabolisme , Souris , Antidépresseurs/pharmacologie , Antidépresseurs/usage thérapeutique , Ginsénosides/pharmacologie , Mâle , Acide 2-amino-adipique/pharmacologie , Dépression/traitement médicamenteux , Maladies neuro-inflammatoires/traitement médicamenteux , Maladies neuro-inflammatoires/métabolisme , Modèles animaux de maladie humaine , Cytokines/métabolisme , Souris de lignée C57BL , Cortex préfrontal/effets des médicaments et des substances chimiques , Cortex préfrontal/métabolisme , Cortex préfrontal/anatomopathologie , Apoptose/effets des médicaments et des substances chimiques
16.
Transl Psychiatry ; 14(1): 392, 2024 Sep 28.
Article de Anglais | MEDLINE | ID: mdl-39341799

RÉSUMÉ

Early social isolation (SI) leads to various abnormalities in emotion and behavior during adulthood. However, the negative impact of SI on offspring remains unclear. This study has discovered that paternal early SI causes social memory deficits and anxiety-like behavior in F1 young adult mice, with alterations of myelin and synapses in the medial prefrontal cortex (mPFC). The 2-week SI in the F1 progeny exacerbates social memory impairment and hypomyelination in the mPFC. Furthermore, the down-regulation of miR-124, a key inhibitor of myelinogenesis, or over-expression of its target gene Nr4a1 in the mPFC of the F1 mice improves social interaction ability and enhances oligodendrocyte maturation and myelin formation. Mechanistically, elevated levels of miR-124 in the sperm of paternal SI mice are transmitted epigenetically to offspring, altering the expression levels of miR-124/Nr4a1/glucocorticoid receptors in mPFC oligodendrocytes. This, in turn, impedes the establishment of myelinogenesis-dependent social behavior. This study unveils a novel mechanism through which miR-124 mediates the intergenerational effects of early isolation stress, ultimately impairing the establishment of social behavior and neurodevelopment.


Sujet(s)
microARN , Cortex préfrontal , Comportement social , Isolement social , Animaux , microARN/génétique , microARN/métabolisme , Souris , Mâle , Cortex préfrontal/métabolisme , Oligodendroglie/métabolisme , Souris de lignée C57BL , Troubles de la mémoire/génétique , Troubles de la mémoire/métabolisme , Comportement animal/physiologie , Gaine de myéline/métabolisme , Gaine de myéline/génétique , Anxiété/génétique , Anxiété/métabolisme , Femelle
17.
Brain Behav ; 14(10): e70043, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39344296

RÉSUMÉ

PURPOSE: The infralimbic (IL) subregion of the medial prefrontal cortex (mPFC) regulates the extinction of conditioned fear memory. Glucocorticoid and gamma-aminobutyric acid (GABA) receptors are expressed in the mPFC and are also critical in fear extinction. This study investigated the possible interactive effects of the glucocorticoids and GABAergic system in the IL on the regulation of fear extinction. METHOD: The rats were trained using an auditory fear conditioning task during which they received three conditioned stimuli (tones, 30 s, 4 kHz, 80 dB), co-terminated with the three unconditioned stimuli (footshock, 0.8 mA, 1 s). Extinction testing was conducted over 3 days (Ext 1-3). Thirty minutes before the first extinction trial (Ext 1), the rats received bicuculline (BIC, 1 mg/kg/2 mL, intraperitoneal [i.p.]) as a GABAA receptor antagonist or CGP55845 (CGP, 0.1 mg/kg/2 ML, i.p.) as a GABAB receptor antagonist followed by systemic injection of corticosterone (CORT, 3 mg/kg/2 ML, i.p.). Furthermore, separate groups of rats received a bilateral intra-IL injection of BIC (100 ng/0.3 µL/side) or CGP (10 ng/0.3 µL/side) followed by a systemic injection of CORT (3 mg/kg/2 ML, i.p.) before the first extinction trial (Ext 1). The extracellular signal-regulated kinase (ERK1) and cAMP response element-binding (CREB) activity in the IL was examined by Western blot analysis after Ext 1. FINDING: The results indicated that systemic CORT injection facilitated fear extinction and increased the expression of ERK1 but not CREB in the IL. Both systemic and intra-IL co-injection of BIC or CGP blocked the effects of CORT on fear extinction and ERK1 expression. CONCLUSION: These findings suggest that glucocorticoids and the GABAergic system may modulate fear extinction through the ERK pathway in the IL.


Sujet(s)
Corticostérone , Extinction (psychologie) , Peur , Cortex préfrontal , Récepteurs GABA-A , Récepteurs GABA-B , Animaux , Cortex préfrontal/effets des médicaments et des substances chimiques , Cortex préfrontal/métabolisme , Extinction (psychologie)/effets des médicaments et des substances chimiques , Extinction (psychologie)/physiologie , Mâle , Peur/effets des médicaments et des substances chimiques , Peur/physiologie , Corticostérone/pharmacologie , Corticostérone/sang , Corticostérone/administration et posologie , Rats , Récepteurs GABA-A/métabolisme , Récepteurs GABA-B/métabolisme , Mémoire/effets des médicaments et des substances chimiques , Mémoire/physiologie , Conditionnement classique/effets des médicaments et des substances chimiques , Conditionnement classique/physiologie , Antagonistes du récepteur GABA-A/pharmacologie , Antagonistes du récepteur GABA-A/administration et posologie , Bicuculline/pharmacologie , Bicuculline/administration et posologie , Antagonistes du récepteur GABA-B/pharmacologie , Rat Sprague-Dawley
18.
Exp Mol Med ; 56(9): 1921-1935, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39218973

RÉSUMÉ

Anatomical connectivity and lesion-deficit studies have shown that the dorsal and ventral hippocampi contribute to cognitive and emotional processes, respectively. However, the role of the dorsal hippocampus (dHP) in emotional or stress-related behaviors remains unclear. Here, we showed that neuronal activity in the dHP affects stress-coping behaviors in mice via excitatory projections to the medial prefrontal cortex (mPFC). The antidepressant ketamine rapidly induced c-Fos expression in both the dorsal and ventral hippocampi. The suppression of GABAergic transmission in the dHP-induced molecular changes similar to those induced by ketamine administration, including eukaryotic elongation factor 2 (eEF2) dephosphorylation, brain-derived neurotrophic factor (BDNF) elevation, and extracellular signal-regulated kinase (ERK) phosphorylation. These synaptic and molecular changes in the dHP induced a reduction in the immobility time of the mice in the tail-suspension and forced swim tests without affecting anxiety-related behavior. Conversely, pharmacological and chemogenetic potentiation of inhibitory neurotransmission in the dHP CA1 region induced passive coping behaviors during the tests. Transneuronal tracing and electrophysiology revealed monosynaptic excitatory connections between dHP CA1 neurons and mPFC neurons. Optogenetic stimulation of dHP CA1 neurons in freely behaving mice produced c-Fos induction and spike firing in the mPFC neurons. Chemogenetic activation of the dHP-recipient mPFC neurons reversed the passive coping behaviors induced by suppression of dHP CA1 neuronal activity. Collectively, these results indicate that neuronal activity in the dHP modulates stress-coping strategies to inescapable stress and contributes to the antidepressant effects of ketamine via the dHP-mPFC circuit.


Sujet(s)
Hippocampe , Cortex préfrontal , Stress psychologique , Animaux , Cortex préfrontal/métabolisme , Cortex préfrontal/physiologie , Souris , Hippocampe/métabolisme , Stress psychologique/métabolisme , Mâle , Neurones/métabolisme , Adaptation psychologique/physiologie , Kétamine/pharmacologie , Facteur neurotrophique dérivé du cerveau/métabolisme , Protéines proto-oncogènes c-fos/métabolisme , Transmission synaptique , Souris de lignée C57BL , Comportement animal , Optogénétique/méthodes , Coping Skills
19.
eNeuro ; 11(9)2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39266326

RÉSUMÉ

Rett syndrome (RTT), a severe neurodevelopmental disorder caused by mutations in the MeCP2 gene, is characterized by cognitive and social deficits. Previous studies have noted hypoactivity in the medial prefrontal cortex (mPFC) pyramidal neurons of MeCP2-deficient mice (RTT mice) in response to both social and nonsocial stimuli. To further understand the neural mechanisms behind the social deficits of RTT mice, we monitored excitatory pyramidal neurons in the prelimbic region of the mPFC during social interactions in mice. These neurons' activity was closely linked to social preference, especially in wild-type mice. However, RTT mice showed reduced social interest and corresponding hypoactivity in these neurons, indicating that impaired mPFC activity contributes to their social deficits. We identified six mPFC neural ensembles selectively tuned to various stimuli, with RTT mice recruiting fewer neurons to ensembles responsive to social interactions and consistently showing lower stimulus-ON ensemble transient rates. Despite these lower rates, RTT mice exhibited an increase in the percentage of social-ON neurons in later sessions, suggesting a compensatory mechanism for the decreased firing rate. This highlights the limited plasticity in the mPFC caused by MeCP2 deficiency and offers insights into the neural dynamics of social encoding. The presence of multifunctional neurons and those specifically responsive to social or object stimuli in the mPFC emphasizes its crucial role in complex behaviors and cognitive functions, with selective neuron engagement suggesting efficiency in neural activation that optimizes responses to environmental stimuli.


Sujet(s)
Protéine-2 de liaison au CpG méthylé , Cortex préfrontal , Cellules pyramidales , Syndrome de Rett , Animaux , Cortex préfrontal/physiologie , Cortex préfrontal/métabolisme , Protéine-2 de liaison au CpG méthylé/déficit , Protéine-2 de liaison au CpG méthylé/génétique , Syndrome de Rett/physiopathologie , Syndrome de Rett/génétique , Mâle , Cellules pyramidales/physiologie , Comportement social , Souris , Souris de lignée C57BL , Souris knockout , Neurones/physiologie , Neurones/métabolisme , Modèles animaux de maladie humaine , Potentiels d'action/physiologie , Interaction sociale , Femelle
20.
Biochem Biophys Res Commun ; 733: 150696, 2024 Nov 12.
Article de Anglais | MEDLINE | ID: mdl-39288700

RÉSUMÉ

Major depressive disorder (MDD) is a psychiatric disorder characterized by depressed mood, behavioral despair and anhedonia. Demyelination in specific brain regions underlies the pathology of MDD, raising the alleviating demyelination as a potential strategy for MDD therapy. Nervonic acid (NA) has the potential to improve brain demyelination, offering benefits for various neurological disorders. However, its effects on depression remain undetermined. Mice were subjected to 14 days of chronic restraint stress (CRS) to induce depression-like behaviors, and were injected with NA (70 mg/kg) daily. The administration of NA significantly improved depressive-like behaviors in CRS mice. CRS led to significant demyelination in the medial prefrontal cortex (mPFC), which were reversed by NA treatment. In addition, NA ameliorated the upregulation of inflammatory cytokines and downregulation of brain-derived neurotrophic factor, improved the alternations in axonal spines observed in the mPFC of CRS mice. Our results highlighted the potential of NA as an antidepressant, with its benefits likely attributed to its effects in alleviating demyelination in the mPFC.


Sujet(s)
Antidépresseurs , Maladies démyélinisantes , Dépression , Souris de lignée C57BL , Cortex préfrontal , Contention physique , Stress psychologique , Animaux , Cortex préfrontal/effets des médicaments et des substances chimiques , Cortex préfrontal/métabolisme , Cortex préfrontal/anatomopathologie , Mâle , Stress psychologique/traitement médicamenteux , Souris , Maladies démyélinisantes/traitement médicamenteux , Maladies démyélinisantes/anatomopathologie , Dépression/traitement médicamenteux , Dépression/anatomopathologie , Dépression/métabolisme , Antidépresseurs/pharmacologie , Antidépresseurs/administration et posologie , Antidépresseurs/usage thérapeutique , Comportement animal/effets des médicaments et des substances chimiques , Facteur neurotrophique dérivé du cerveau/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE