Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.394
Filter
1.
Sci Prog ; 107(3): 368504241264994, 2024.
Article in English | MEDLINE | ID: mdl-39228316

ABSTRACT

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.


Subject(s)
Morphine , Prefrontal Cortex , Signal Transduction , Substance Withdrawal Syndrome , Toll-Like Receptor 2 , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Substance Withdrawal Syndrome/genetics , Substance Withdrawal Syndrome/metabolism , Prefrontal Cortex/metabolism , Prefrontal Cortex/drug effects , Animals , Signal Transduction/drug effects , Mice , Male , Mice, Knockout , Mice, Inbred C57BL , Gene Expression Profiling , Gene Expression Regulation/drug effects , Morphine Dependence/genetics , Morphine Dependence/metabolism
2.
J Cell Mol Med ; 28(17): e18578, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39234952

ABSTRACT

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.


Subject(s)
Hippocampus , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors , Mice, Knockout , Pentylenetetrazole , Seizures , Animals , Kruppel-Like Factor 4/metabolism , Seizures/metabolism , Seizures/chemically induced , Seizures/genetics , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Mice , Hippocampus/metabolism , Male , Prefrontal Cortex/metabolism , Mice, Inbred C57BL , Disease Models, Animal
3.
Int J Mol Sci ; 25(17)2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39273422

ABSTRACT

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.


Subject(s)
Alzheimer Disease , Disease Models, Animal , Hippocampus , MicroRNAs , Neuroglia , Neurons , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Mice , Neurons/metabolism , Neuroglia/metabolism , Hippocampus/metabolism , Mice, Transgenic , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Gene Expression Regulation , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Prefrontal Cortex/metabolism , Glial Fibrillary Acidic Protein/metabolism , Glial Fibrillary Acidic Protein/genetics , Male
4.
Int J Mol Sci ; 25(17)2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39273621

ABSTRACT

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.


Subject(s)
2-Aminoadipic Acid , Antidepressive Agents , Astrocytes , Ginsenosides , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Mice , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Ginsenosides/pharmacology , Male , 2-Aminoadipic Acid/pharmacology , Depression/drug therapy , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/metabolism , Disease Models, Animal , Cytokines/metabolism , Mice, Inbred C57BL , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Apoptosis/drug effects
5.
Nat Commun ; 15(1): 8176, 2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39289358

ABSTRACT

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.


Subject(s)
Claustrum , Hallucinogens , Neurons , Nuclear Receptor Subfamily 4, Group A, Member 2 , Animals , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Mice , Hallucinogens/pharmacology , Claustrum/metabolism , Neurons/metabolism , Male , Mice, Knockout , Mice, Inbred C57BL , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiology , Sensorimotor Cortex/metabolism , Sensorimotor Cortex/physiology
6.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(8): 1441-1449, 2024 Aug 20.
Article in Chinese | MEDLINE | ID: mdl-39276039

ABSTRACT

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.


Subject(s)
Depression , Doxorubicin , Ferroptosis , Mice, Inbred BALB C , Prefrontal Cortex , Animals , Ferroptosis/drug effects , Mice , Depression/drug therapy , Depression/chemically induced , Doxorubicin/adverse effects , Female , Prefrontal Cortex/metabolism , Prefrontal Cortex/drug effects , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Cell Line, Tumor , Behavior, Animal/drug effects , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism
7.
Gut Microbes ; 16(1): 2401939, 2024.
Article in English | MEDLINE | ID: mdl-39259834

ABSTRACT

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.


Subject(s)
Anxiety , Gastrointestinal Microbiome , Stress, Psychological , Animals , Gastrointestinal Microbiome/physiology , Mice , Anxiety/microbiology , Anxiety/metabolism , Stress, Psychological/microbiology , Stress, Psychological/metabolism , Amino Acids/metabolism , Male , Mice, Inbred C57BL , Amino Acid Transport Systems/metabolism , Prefrontal Cortex/metabolism , Behavior, Animal , Dysbiosis/microbiology , Maternal Deprivation , Glutamine/metabolism , Brain-Gut Axis/physiology , Synaptic Transmission , Female , Glutamic Acid/metabolism
8.
CNS Neurosci Ther ; 30(9): e70048, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39300492

ABSTRACT

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.


Subject(s)
Aspartic Acid , Bipolar Disorder , Glutamic Acid , Glutamine , Prefrontal Cortex , Proton Magnetic Resonance Spectroscopy , Humans , Bipolar Disorder/drug therapy , Bipolar Disorder/metabolism , Bipolar Disorder/diagnostic imaging , Prefrontal Cortex/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/diagnostic imaging , Male , Female , Adult , Aspartic Acid/analogs & derivatives , Aspartic Acid/metabolism , Glutamine/metabolism , Glutamic Acid/metabolism , Middle Aged , Follow-Up Studies , Creatine/metabolism , Young Adult , Phosphocreatine/metabolism
9.
Brain Res ; 1844: 149196, 2024 Dec 01.
Article in English | MEDLINE | ID: mdl-39181223

ABSTRACT

This work was done to investigate the ameliorating impact of 4-methylumbilliferon (4-MU) on spatial learning and memory dysfunction and restraint stress (STR)-induced anxiety-like behaviors in male Wistar rats and the underlying mechanisms. Thirty-two animals were assigned into 4 cohorts: control, 4-MU, STR, and STR+4-MU. Animals were exposed to STR for 4 h per day for 14 consecutive days or kept in normal conditions (healthy animals without exposure to stress). 4-MU (25 mg/kg) was intraperitoneally administered once daily to STR rats before restraint stress for 14 consecutive days. The behavioral tests were performed through Morris water maze tests and elevated-plus maze to examine learning/memory function, and anxiety levels, respectively. The levels of the antioxidant defense biomarkers (GPX, SOD) and MDA as an oxidant molecule in the brain tissues were measured using commercial ELISA kits. Neuronal loss or density of neurons was evaluated using Nissl staining. STR exposure could cause significant alterations in the levels of the antioxidant defense biomarkers (MDA, GPX, and SOD) in the prefrontal cortex and hippocampus, induce anxiety, and impair spatial learning and memory function. Treatment with 4-MU markedly reduced anxiety levels and improved spatial learning and memory dysfunction via restoring the antioxidant defense biomarkers to normal values and reducing MDA levels. Moreover, more intact cells with normal morphologies were detected in STR-induced animals treated with 4-MU. 4-MU could attenuate the STR-induced anxiety-like behaviors and spatial learning and memory dysfunction by reducing oxidative damage and neuronal loss in the prefrontal cortex and hippocampus region. Taken together, our findings provide new insights regarding the potential therapeutic effects of 4-MU against neurobehavioral disorders induced by STR.


Subject(s)
Anxiety , Cell Death , Memory Disorders , Neurons , Oxidative Stress , Rats, Wistar , Animals , Oxidative Stress/drug effects , Male , Anxiety/drug therapy , Anxiety/metabolism , Memory Disorders/drug therapy , Memory Disorders/metabolism , Neurons/drug effects , Neurons/metabolism , Rats , Cell Death/drug effects , Stress, Psychological/drug therapy , Stress, Psychological/metabolism , Stress, Psychological/complications , Maze Learning/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Antioxidants/pharmacology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism
10.
Behav Brain Res ; 475: 115220, 2024 Oct 18.
Article in English | MEDLINE | ID: mdl-39214422

ABSTRACT

BACKGROUND: The pathophysiology and molecular mechanisms of schizophrenia (SCZ) remain unclear, and the effective treatment resources are still limited. The goal of this study is to identify the expression of AQP4 in SCZ patients and explore whether AQP4 inhibition could ameliorate schizophrenia-like behaviors and its mechanisms. METHODS: Microarray datasets of PFC compared with healthy control were searched in the Gene Expression Omnibus (GEO) database, and differentially expressed genes (DEGs) were analyzed with the GEO2R online tool. The Venny online tool and metascape online software were used to identify common abnormally expressed genes and conduct cell type signature enrichment analysis. SCZ mouse models were induced with MK-801, an NMDA receptor antagonist (intraperitoneal injection, 0.1 mg/kg/day for 7 days), and C6 cell models were treated with 100 µM MK-801. RT-qPCR, Western Blotting, and immunofluorescence were employed to determine the expression of AQP4, proinflammatory cytokines, and GFAP. Open field tests and social interaction tests were performed to evaluate the schizophrenia-like behaviors. RESULTS: Bioinformatics analysis identified upregulation of AQP4 in the PFC of SCZ patients compared with healthy controls. Cell type signature enrichment analysis showed that all three DEGs lists were strongly enriched in the FAN EMBRYONIC CTX ASTROCYTE 2 category. Upregulation of AQP4 was also observed in MK-801-treated C6 cells and the PFC of MK-801-induced SCZ mouse model. Moreover, AQP4 inhibition with TGN-020 (an inhibitor of AQP4) improved anxiety-like behavior and social novelty preference defects in MK-801-treated mice. AQP4 inhibition also reduced the expression of IL-1ß, IL-6, and TNF-α in MK-801-treated C6 cells and mouse model. CONCLUSIONS: AQP4 is upregulated in the PFC of SCZ patients compared with healthy controls. AQP4 inhibition could alleviate the anxiety-like behavior and social novelty defects in MK-801-treated mice, this may be due to the role of AQP4 in the regulation of the expression of proinflammatory cytokines.


Subject(s)
Aquaporin 4 , Disease Models, Animal , Dizocilpine Maleate , Schizophrenia , Up-Regulation , Animals , Schizophrenia/drug therapy , Schizophrenia/metabolism , Dizocilpine Maleate/pharmacology , Mice , Up-Regulation/drug effects , Aquaporin 4/metabolism , Aquaporin 4/antagonists & inhibitors , Humans , Male , Behavior, Animal/drug effects , Mice, Inbred C57BL , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Female , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Amino Acid Antagonists/administration & dosage
11.
Pharmacol Biochem Behav ; 244: 173849, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39142357

ABSTRACT

Selective serotonin (5-HT) reuptake inhibitors (SSRIs) are commonly prescribed to women during pregnancy and breastfeeding despite posing a risk of adverse cognitive outcomes and affective disorders for the child. The consequences of SSRI-induced excess of 5-HT during development for the brain neuromodulatory 5-HT system remain largely unexplored. In this study, an SSRI - fluoxetine (FLX) - was administered to C57BL/6 J mouse dams during pregnancy and lactation to assess its effects on the offspring. We found that maternal FLX decreased field potentials, impaired long-term potentiation, facilitated long-term depression and tended to increase the density of 5-HTergic fibers in the medial prefrontal cortex (mPFC) of female but not male adolescent offspring. These effects were accompanied by deteriorated performance in the temporal order memory task and reduced sucrose preference with no change in marble burying behavior in FLX-exposed female offspring. We also found that maternal FLX reduced the axodendritic tree complexity of 5-HT dorsal raphe nucleus (DRN) neurons in female but not male offspring, with no changes in the excitability of DRN neurons of either sex. While no effects of maternal FLX on inhibitory postsynaptic currents (sIPSCs) in DRN neurons were found, we observed a significant influence of FLX exposure on kinetics of spontaneous excitatory postsynaptic currents (sEPSCs) in DRN neurons. Finally, we report that no changes in field potentials and synaptic plasticity were evident in the mPFC of the offspring after maternal exposure during pregnancy and lactation to a new antidepressant, vortioxetine. These findings show that in contrast to the mPFC, long-term consequences of maternal FLX exposure on the structure and function of DRN 5-HT neurons are mild and suggest a sex-dependent, distinct sensitivity of cortical and brainstem neurons to FLX exposure in early life. Vortioxetine appears to exert fewer side effects with regards to the mPFC when compared with FLX.


Subject(s)
Dorsal Raphe Nucleus , Fluoxetine , Mice, Inbred C57BL , Neuronal Plasticity , Prefrontal Cortex , Prenatal Exposure Delayed Effects , Selective Serotonin Reuptake Inhibitors , Synaptic Transmission , Animals , Fluoxetine/pharmacology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Female , Mice , Dorsal Raphe Nucleus/drug effects , Dorsal Raphe Nucleus/metabolism , Pregnancy , Male , Neuronal Plasticity/drug effects , Synaptic Transmission/drug effects , Prenatal Exposure Delayed Effects/chemically induced , Selective Serotonin Reuptake Inhibitors/pharmacology , Neurons/drug effects , Serotonin/metabolism , Long-Term Potentiation/drug effects
12.
Neurosci Lett ; 841: 137944, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39154843

ABSTRACT

Depression and anxiety are prominent symptoms of withdrawal syndrome, often caused by the abuse of addictive drugs like morphine. N-palmitoylethanolamide (PEA), a biologically active lipid, is utilized as an anti-inflammatory and analgesic medication. Recent studies have highlighted PEA's role in mitigating cognitive decline and easing depression resulting from chronic pain. However, it remains unknown whether PEA can influence negative emotions triggered by morphine withdrawal. This study seeks to explore the impact of PEA on such emotions and investigate the underlying mechanisms. Mice subjected to morphine treatment underwent a 10-day withdrawal period, followed by assessments of the effect of PEA on anxiety- and depression-like behaviors using various tests. Enzyme-linked immunosorbent assay was conducted to measure levels of monoamine neurotransmitters in specific brain regions. The findings indicate that PEA mitigated anxiety and depression symptoms and reduced 5-hydroxytryptamine, noradrenaline, and dopamine levels in the hippocampus and prefrontal cortex. In summary, PEA demonstrates a significant positive effect on negative emotions associated with morphine withdrawal, accompanied with the reduction in levels of monoamine neurotransmitters in key brain regions. These insights could be valuable for managing negative emotions arising from morphine withdrawal.


Subject(s)
Amides , Anxiety , Depression , Ethanolamines , Morphine , Palmitic Acids , Substance Withdrawal Syndrome , Animals , Substance Withdrawal Syndrome/psychology , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/drug therapy , Ethanolamines/pharmacology , Palmitic Acids/pharmacology , Mice , Male , Morphine/pharmacology , Depression/metabolism , Depression/drug therapy , Depression/psychology , Depression/etiology , Amides/pharmacology , Anxiety/drug therapy , Anxiety/psychology , Anxiety/metabolism , Hippocampus/metabolism , Hippocampus/drug effects , Emotions/drug effects , Serotonin/metabolism , Morphine Dependence/metabolism , Morphine Dependence/psychology , Prefrontal Cortex/metabolism , Prefrontal Cortex/drug effects , Norepinephrine/metabolism , Brain/metabolism , Brain/drug effects
13.
J Pharm Biomed Anal ; 251: 116446, 2024 Dec 15.
Article in English | MEDLINE | ID: mdl-39197207

ABSTRACT

In traditional Chinese medicinal practices, Gegen (GG) and Tianma (TM) are widely utilized for headache relief, but their material basis has not been comprehensively characterized. This research utilized ultra performance liquid chromatography-quadrupole-time of flight-mass spectrometry (UPLC-Q-TOF-MS) for precise determination of Gegen-Tianma's (GGTM) material composition, and employed desorption electrospray ionization-mass spectrometry imaging (DESI-MSI) to pinpoint the brain-absorbed components and various metabolites post oral administration to rats. A total of 80 chemical constituents were identified from GGTM, 11 prototypes and 18 metabolites were identified from plasma. The brain tissue was identified in total 4 prototypes and 5 metabolites, these constituents were basically located in the prefrontal cortex and thalamus. The absorption patterns of components in the rat brain aligned with the varied distribution of metabolites within the brain. This study provides a solid theoretical basis for in-depth exploration of potential drug targets and elucidation of the specific mechanism of action of GGTM in the treatment of migraine.


Subject(s)
Brain , Drugs, Chinese Herbal , Rats, Sprague-Dawley , Spectrometry, Mass, Electrospray Ionization , Animals , Drugs, Chinese Herbal/pharmacokinetics , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/analysis , Rats , Chromatography, High Pressure Liquid/methods , Male , Brain/metabolism , Spectrometry, Mass, Electrospray Ionization/methods , Administration, Oral , Prefrontal Cortex/metabolism , Tissue Distribution
14.
J Psychiatry Neurosci ; 49(4): E265-E281, 2024.
Article in English | MEDLINE | ID: mdl-39209459

ABSTRACT

BACKGROUND: Schizophrenia is characterized by a complex interplay of genetic and environmental factors, leading to alterations in various molecular pathways that may contribute to its pathogenesis. Recent studies have shown that exosomal microRNAs could play essential roles in various brain disorders; thus, we sought to explore the potential molecular mechanisms through which microRNAs in plasma exosomes are involved in schizophrenia. METHODS: We obtained sequencing data sets (SUB12404730, SUB12422862, and SUB12421357) and transcriptome sequencing data sets (GSE111708, GSE108925, and GSE18981) from mouse models of schizophrenia using the Sequence Read Archive and the Gene Expression Omnibus databases, respectively. We performed differential expression analysis on mRNA to identify differentially expressed genes. We conducted Gene Ontology (GO) functional and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses to determine differentially expressed genes. Subsequently, we determined the intersection of differentially expressed microRNAs in plasma exosomes and in prefrontal cortex tissue. We retrieved downstream target genes of mmu-miR-146a-5p from TargetScan and used Cytoscape to visualize and map the microRNA-target gene regulatory network. We conducted in vivo experiments using MK-801-induced mouse schizophrenia models and in vitro experiments using cultured mouse neurons. The role of plasma exosomal miR-146a-5p in schizophrenia was validated using a cell counting kit, detection of lactate dehydrogenase, dual-luciferase assay, quantitative reverse transcription polymerase chain reaction, and Western blot analysis. RESULTS: Differential genes were mainly enriched in synaptic regulation-related functions and pathways and were associated with neuronal degeneration. We found that mmu-miR-146a-5p was highly expressed in both prefrontal cortical tissue and plasma exosomes, which may be transferred to lobe cortical vertebral neurons, leading to the synergistic dysregulation of gene network functions and, therefore, promoting schizophrenia development. We found that mmu-miR-146a-5p may inhibit the Notch signalling pathway-mediated synaptic activity of mouse pyramidal neurons in the lobe cortex by targeting NOTCH1, which in turn could promote the onset and development of schizophrenia in mice. LIMITATIONS: The study's findings are based on animal models and in vitro experiments, which may not fully replicate the complexity of human schizophrenia. CONCLUSION: Our findings suggest that mmu-miR-146a-5p in plasma-derived exosomes may play an important role in the pathogenesis of schizophrenia. Our results provide new insights into the underlying molecular mechanisms of the disease.


Subject(s)
Disease Models, Animal , Exosomes , Mice, Inbred C57BL , MicroRNAs , Prefrontal Cortex , Schizophrenia , Signal Transduction , Animals , Mice , Dizocilpine Maleate/pharmacology , Exosomes/metabolism , Gene Regulatory Networks , MicroRNAs/genetics , MicroRNAs/metabolism , MicroRNAs/blood , Neurons/metabolism , Prefrontal Cortex/metabolism , Receptors, Notch/metabolism , Receptors, Notch/genetics , Schizophrenia/metabolism , Schizophrenia/genetics , Synapses/metabolism
15.
Neurosci Lett ; 841: 137953, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39214331

ABSTRACT

The behavioral sensitization, characterized by escalated behavioral responses triggered by recurrent exposure to psychostimulants, involves neurobiological mechanisms that are brain-region and cell-type specific. Enduring neuroadaptive changes have been observed in response to methamphetamine (METH) within the orbitofrontal cortex (OFC), the cell-type specific transcriptional alterations in response to METH sensitization remain understudied. In this study, we utilized Single-nucleus RNA-sequencing (snRNA-seq) to profile the gene expression changes in the OFC of a rat METH sensitization model. The analyses of differentially expressed genes (DEGs) unveiled cell-type specific transcriptional reactions associated with METH sensitization, with the most significant alterations documented in microglial cells. Bioinformatic investigations revealed that distinct functional and signaling pathways enriched in microglia-specific DEGs majorly involved in macroautophagy processes and the activation of N-methyl-D-aspartate ionotropic glutamate receptors (NMDAR). To validate the translational relevance of our findings, we analyzed our snRNA-seq data in conjunction with a transcriptomic study of individuals with opioid use disorder (OUD) and a large-scale Genome-Wide Association Studies (GWAS) from multiple externalizing phenotypes related to drug addiction. The validation analysis confirmed the consistent expression changes of key microglial DEGs in human METH addiction. Moreover, the integration with GWAS data revealed associations between addiction risk genes and the DEGs observed in specific cell types, particularly microglia and excitatory neurons. Our study highlights the importance of cell-type specific transcriptional alterations in the OFC in the context of METH sensitization and their potential translational relevance to human drug addiction.


Subject(s)
Central Nervous System Stimulants , Methamphetamine , Prefrontal Cortex , Rats, Sprague-Dawley , Methamphetamine/pharmacology , Animals , Male , Rats , Prefrontal Cortex/metabolism , Prefrontal Cortex/drug effects , Central Nervous System Stimulants/pharmacology , Microglia/metabolism , Microglia/drug effects , Sequence Analysis, RNA/methods
16.
J Psychiatr Res ; 178: 180-187, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39146821

ABSTRACT

Schizophrenia is a complex neuropsychiatric disorder with positive, negative, and cognitive symptoms. In rats, sub-chronic administration of ketamine is used for the induction of schizophrenia model. Increased locomotor activity is one of the most important features of psychotic-like symptoms in rodents. On the other hand, risperidone is a potent antipsychotic medication that is approved for the treatment of schizophrenia and bipolar disorder. In the present research, we aimed to investigate the effect of sub-chronic treatment of ketamine on cognitive and behavioral functions, and brain-derived neurotrophic factor (BDNF) expression level in the prefrontal cortex. Also, we assessed the efficacy of risperidone on cognitive and behavioral impairments induced by ketamine. Possible sex differences were also measured. Ketamine was intraperitoneally injected at the dose of 30 mg/kg for five consecutive days. Risperidone was also intraperitoneally injected at the dose of 2 mg/kg. Novel object recognition memory, pain threshold, locomotor activity, rearing behavior, and BDNF level were evaluated. The results showed that ketamine injection for five consecutive days impaired the acquisition of long-term recognition memory and decreased BDNF level in the prefrontal cortex in both sexes. Also, it decreased pain threshold in females, increased rearing behavior in males, and induced hyperlocomotion with greater effect in females. On the other hand, risperidone restored or attenuated the effect of ketamine on all the behavioral effects and BDNF level. In conclusion, we suggested that there were sex differences in the effects of ketamine on pain perception, locomotion, and rearing behavior in a rat model of schizophrenia.


Subject(s)
Brain-Derived Neurotrophic Factor , Disease Models, Animal , Ketamine , Prefrontal Cortex , Risperidone , Schizophrenia , Sex Characteristics , Animals , Ketamine/pharmacology , Ketamine/administration & dosage , Schizophrenia/drug therapy , Schizophrenia/chemically induced , Schizophrenia/physiopathology , Male , Female , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/drug effects , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Risperidone/pharmacology , Risperidone/administration & dosage , Rats , Antipsychotic Agents/pharmacology , Antipsychotic Agents/administration & dosage , Recognition, Psychology/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Amino Acid Antagonists/administration & dosage , Rats, Wistar , Behavior, Animal/drug effects , Pain Threshold/drug effects , Motor Activity/drug effects
17.
Neurosci Lett ; 839: 137936, 2024 Sep 14.
Article in English | MEDLINE | ID: mdl-39151573

ABSTRACT

Synucleins, including α-synuclein (α-syn), ß-syn, and γ-syn, have been implicated in various synucleinopathies, notably Parkinson's disease (PD), which has generated increased interest in understanding their roles. Although α-syn and ß-syn have contrasting neuropathological consequences, the precise role of γ-syn remains unclear. This study validated non-motor symptoms, specifically anxiety-like behavior, along with the degradation of dopaminergic (DAergic) neurons in the nigrostriatal system and DAergic neurites in the prefrontal cortex and hippocampus of rats infused with striatal 6-hydroxydopamine (6-OHDA). Our study further investigated the alterations in γ-syn expression levels in the prefrontal cortices and hippocampi of these 6-OHDA-treated rats, aiming to establish foundational insights into the neuropathophysiology of DA depletion, a central feature of PD. Our findings revealed a significant increase in the expression of γ-syn mRNA and protein in these brain regions, in contrast to unaltered α- and ß-syn expression levels. This suggests a distinct role of γ-syn within the neurobiological milieu under conditions of DA deficiency. Overall, our data shed light on the neurobiological changes observed in the hemiparkinsonian rat model induced with 6-OHDA, underscoring the potential significance of γ-syn in PD pathology.


Subject(s)
Dopamine , Hippocampus , Oxidopamine , Prefrontal Cortex , Up-Regulation , gamma-Synuclein , Animals , Prefrontal Cortex/metabolism , Oxidopamine/toxicity , Male , Hippocampus/metabolism , Dopamine/metabolism , gamma-Synuclein/metabolism , gamma-Synuclein/genetics , Rats , Rats, Sprague-Dawley , Parkinsonian Disorders/metabolism , Parkinsonian Disorders/chemically induced , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Corpus Striatum/metabolism , Disease Models, Animal , alpha-Synuclein/metabolism , alpha-Synuclein/genetics
18.
Physiol Rep ; 12(16): e70001, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39161054

ABSTRACT

Brain-derived neurotrophic factor (BDNF) content and signaling has been identified as one potential regulator of amyloid precursor protein (APP) processing. Recently published work has demonstrated that BDNF reduces BACE1 activity while also elevating the inhibition of GSK3ß in the prefrontal cortex of male C57BL/6J mice. These results provide evidence that BDNF alters APP processing by reducing BACE1 activity, which may act through GSK3ß inhibition. The purpose of this study was to further explore the role of GSK3ß in BDNF-induced regulation on BACE1 activity. We utilized a cell culture and an in vitro activity assay model to pharmacologically target BDNF and GSK3ß signaling to confirm its involvement in the BDNF response. Treatment of differentiated SH-SY5Y neuronal cells with 75 ng/mL BDNF resulted in elevated pTrkB content, pAkt content, pGSK3ß content, and reduced BACE1 activity. An in vitro BACE1 activity assay utilizing mouse prefrontal cortex (n = 6/group) supplemented with BDNF, BDNF + ANA12 (Trkb antagonist), or BDNF + wortmannin (Akt inhibitor) demonstrated that BDNF reduced BACE1 activity; however, in the presence of TrkB or Akt inhibition, this effect was abolished. An in vitro ADAM10 activity assay utilizing mouse prefrontal cortex (n = 6/group) supplemented with BDNF, BDNF + ANA12 (Trkb antagonist), or BDNF + wortmannin (Akt inhibitor) demonstrated that BDNF did not alter ADAM10 activity. However, inhibiting BDNF signaling reduced ADAM10 activity. Collectively these studies suggest that GSK3ß inhibition may be necessary for BDNF-induced reductions in BACE1 activity. These findings will allow for the optimization of future therapeutic strategies by selectively targeting TrkB activation and GSK3ß inhibition.


Subject(s)
Amyloid Precursor Protein Secretases , Aspartic Acid Endopeptidases , Brain-Derived Neurotrophic Factor , Glycogen Synthase Kinase 3 beta , Mice, Inbred C57BL , Neurons , Proto-Oncogene Proteins c-akt , Amyloid Precursor Protein Secretases/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Brain-Derived Neurotrophic Factor/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Neurons/metabolism , Neurons/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Humans , Aspartic Acid Endopeptidases/metabolism , Aspartic Acid Endopeptidases/antagonists & inhibitors , Mice , Male , Prefrontal Cortex/metabolism , Prefrontal Cortex/drug effects , Signal Transduction , Cell Line, Tumor , Receptor, trkB/metabolism , Receptor, trkB/antagonists & inhibitors , Membrane Glycoproteins/metabolism
19.
Brain Res ; 1843: 149136, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39098577

ABSTRACT

Sensory experience affects not only the corresponding primary sensory cortex, but also synaptic and neural circuit functions in other brain regions in a cross-modal manner. However, it remains unclear whether oligodendrocyte (OL) generation and myelination can also undergo cross-modal modulation. Here, we report that while early life short-term whisker deprivation from birth significantly reduces in the number of mature of OLs and the degree of myelination in the primary somatosensory cortex(S1) at postnatal day 14 (P14), it also simultaneously affects the primary visual cortex (V1), but not the medial prefrontal cortex (mPFC) with a similar reduction. Interestingly, when mice were subjected to long-term early whisker deprivation from birth (P0) to P35, they exhibited dramatically impaired myelination and a deduced number of differentiated OLs in regions including the S1, V1, and mPFC, as detected at P60. Meanwhile, the process complexity of OL precursor cells (OPCs) was also rduced, as detected in the mPFC. However, when whisker deprivation occurred during the mid-late postnatal period (P35 to P50), myelination was unaffected in both V1 and mPFC brain regions at P60. In addition to impaired OL and myelin development in the mPFC, long-term early whisker-deprived mice also showed deficits in social novelty, accompanied by abnormal activation of c-Fos in the mPFC. Thus, our results reveal a novel form of cross-modal modulation of myelination by sensory experience that can lead to abnormalities in social behavioral, suggesting a possible similar mechanism underlying brain pathological conditions that suffer from both sensory and social behavioral deficits, such as autism spectrum disorders.


Subject(s)
Mice, Inbred C57BL , Myelin Sheath , Prefrontal Cortex , Sensory Deprivation , Somatosensory Cortex , Vibrissae , Animals , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiology , Vibrissae/physiology , Sensory Deprivation/physiology , Myelin Sheath/physiology , Myelin Sheath/metabolism , Somatosensory Cortex/physiology , Mice , Oligodendroglia/physiology , Oligodendroglia/metabolism , Animals, Newborn , Male , Exploratory Behavior/physiology , Visual Cortex/growth & development , Visual Cortex/metabolism , Visual Cortex/physiology , Social Behavior , Female
20.
Neuroscience ; 556: 96-113, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39103042

ABSTRACT

The aim of the study is to understand the rationale behind the application of deep brain stimulation (DBS) in the treatment of depression. Male Wistar rats, rendered depressive with chronic unpredictable mild stress (CUMS) were implanted with electrode in the lateral hypothalamus-medial forebrain bundle (LH-MFB) and subjected to deep brain stimulation (DBS) for 4 h each day for 14 days. DBS rats, as well as controls, were screened for a range of parameters indicative of depressive state. Symptomatic features noticed in CUMS rats like the memory deficit, anhedonia, reduction in body weight and 5-hydroxytryptamine (5-HT) and 5-hydroxyindoleacetic acid (5-HIAA) levels in mPFC and elevated plasma corticosterone were reversed in rats subjected to DBS. DBS arrested CUMS induced degeneration of 5-HT cells in interfascicular region of dorsal raphe nucleus (DRif) and fibers in LH-MFB and induced dendritic proliferation in mPFC neurons. MFB is known to serve as a major conduit for the DRif-mPFC serotoninergic pathway. While the density of serotonin fibers in the LH-MFB circuit was reduced in CUMS, it was upregulated in DBS-treated rats. Furthermore, microinjection of 5-HT1A receptor antagonist, WAY100635 into mPFC countered the positive effects of DBS like the antidepressant and memory-enhancing action. In this background, we suggest that DBS at LH-MFB may exercise positive effect in depressive rats via upregulation of the serotoninergic system. While these data drawn from the experiments on rat provide meaningful clues, we suggest that further studies aimed at understanding the usefulness of DBS at LH-MFB in humans may be rewarding.


Subject(s)
Deep Brain Stimulation , Depression , Medial Forebrain Bundle , Rats, Wistar , Serotonin , Animals , Deep Brain Stimulation/methods , Male , Serotonin/metabolism , Depression/therapy , Depression/metabolism , Hypothalamic Area, Lateral/metabolism , Stress, Psychological/metabolism , Stress, Psychological/therapy , Cognitive Dysfunction/therapy , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/etiology , Disease Models, Animal , Rats , Corticosterone/blood , Hydroxyindoleacetic Acid/metabolism , Prefrontal Cortex/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL