Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 18.539
Filter
1.
Alcohol Alcohol ; 59(4)2024 May 14.
Article in English | MEDLINE | ID: mdl-38742547

ABSTRACT

AIMS: Continued alcohol consumption despite negative consequences is a core symptom of alcohol use disorder. This is modeled in mice by pairing negative stimuli with alcohol, such as adulterating alcohol solution with quinine. Mice consuming alcohol under these conditions are considered to be engaging in aversion-resistant intake. Previously, we have observed sex differences in this behavior, with females more readily expressing aversion-resistant consumption. We also identified three brain regions that exhibited sex differences in neuronal activation during quinine-alcohol drinking: ventromedial prefrontal cortex (vmPFC), posterior insular cortex (PIC), and ventral tegmental area (VTA). Specifically, male mice showed increased activation in vmPFC and PIC, while females exhibited increased activation in VTA. In this study, we aimed to identify what specific type of neurons are activated in these regions during quinine-alcohol drinking. METHOD: We assessed quinine-adulterated alcohol intake using the two-bottle choice procedure. We also utilized RNAscope in situ hybridization in the three brain regions that previously exhibited a sex difference to examine colocalization of Fos, glutamate, GABA, and dopamine. RESULT: Females showed increased aversion-resistant alcohol consumption compared to males. We also found that males had higher colocalization of glutamate and Fos in vmPFC and PIC, while females had greater dopamine and Fos colocalization in the VTA. CONCLUSIONS: Collectively, these experiments suggest that glutamatergic output from the vmPFC and PIC may have a role in suppressing, and dopaminergic activity in the VTA may promote, aversion-resistant alcohol consumption. Future experiments will examine neuronal circuits that contribute to sex differences in aversion resistant consumption.


Subject(s)
Alcohol Drinking , Neurons , Quinine , Sex Characteristics , Animals , Quinine/pharmacology , Female , Male , Mice , Neurons/drug effects , Ventral Tegmental Area/drug effects , Mice, Inbred C57BL , Prefrontal Cortex/drug effects , Mesencephalon/metabolism , Mesencephalon/drug effects , Insular Cortex/drug effects , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Ethanol/pharmacology , Glutamic Acid/metabolism
2.
Biomed Khim ; 70(2): 83-88, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38711407

ABSTRACT

The toxic effect of ethanol on the cerebral cortex and protective effects of omega-3 fatty acids against this neurotoxicity were investigated. Twenty eight male Wistar-albino rats were divided into 4 groups. Rats of the ethanol and ethanol withdrawal groups were treated with ethanol (6 g/kg/day) for 15 days. Animals of the ethanol+omega-3 group received omega-3 fatty acids (400 mg/kg daily) and ethanol. In rats of the ethanol group SOD activity was lower than in animals of the control group. In rats treated with omega-3 fatty acids along with ethanol SOD, activity increased. GSH-Px activity and MDA levels in animals of all groups were similar. In ethanol treated rats NO levels significantly decreased as compared to the animals of the control group (6.45±0.24 nmol/g vs 11.05±0.53 nmol/g, p.


Subject(s)
Cerebral Cortex , Ethanol , Fatty Acids, Omega-3 , Nitric Oxide , Rats, Wistar , Superoxide Dismutase , Animals , Male , Rats , Fatty Acids, Omega-3/pharmacology , Cerebral Cortex/metabolism , Cerebral Cortex/drug effects , Nitric Oxide/metabolism , Superoxide Dismutase/metabolism , Glutathione Peroxidase/metabolism , Antioxidants/pharmacology , Malondialdehyde/metabolism
3.
Neuron ; 112(9): 1373-1375, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38697018

ABSTRACT

Maternal well-being is important for the development of the fetus, with a key influence on its nervous system. In this issue of Neuron, Krontira et al.1 implicate glucocorticoids, the stress hormones, in the regulation of neural stem cell identity and proliferation, with long-lasting consequences on brain architecture and educational attainment.


Subject(s)
Glucocorticoids , Neurogenesis , Humans , Glucocorticoids/pharmacology , Neurogenesis/drug effects , Neurogenesis/physiology , Neurons/drug effects , Neurons/physiology , Cerebral Cortex/drug effects , Cerebral Cortex/cytology , Neural Stem Cells/drug effects
4.
Neurobiol Dis ; 195: 106502, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38608784

ABSTRACT

Synaptic changes are early manifestations of neuronal dysfunction in Huntington's disease (HD). However, the mechanisms by which mutant HTT protein impacts synaptogenesis and function are not well understood. Herein we explored HD pathogenesis in the BACHD mouse model by examining synaptogenesis and function in long term primary cortical cultures. At DIV14 (days in vitro), BACHD cortical neurons showed no difference from WT neurons in synaptogenesis as revealed by colocalization of a pre-synaptic (Synapsin I) and a post-synaptic (PSD95) marker. From DIV21 to DIV35, BACHD neurons showed progressively reduced colocalization of Synapsin I and PSD95 relative to WT neurons. The deficits were effectively rescued by treatment of BACHD neurons with BDNF. The recombinant apical domain of CCT1 (ApiCCT1) yielded a partial rescuing effect. BACHD neurons also showed culture age-related significant functional deficits as revealed by multielectrode arrays (MEAs). These deficits were prevented by BDNF, whereas ApiCCT1 showed a less potent effect. These findings are evidence that deficits in BACHD synapse and function can be replicated in vitro and that BDNF or a TRiC-inspired reagent can potentially be protective against these changes in BACHD neurons. Our findings support the use of cellular models to further explicate HD pathogenesis and potential treatments.


Subject(s)
Brain-Derived Neurotrophic Factor , Cerebral Cortex , Disease Models, Animal , Huntington Disease , Neurons , Synapses , Animals , Huntington Disease/metabolism , Huntington Disease/pathology , Brain-Derived Neurotrophic Factor/metabolism , Synapses/metabolism , Synapses/drug effects , Synapses/pathology , Cerebral Cortex/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Mice , Neurons/metabolism , Neurons/drug effects , Neurons/pathology , Mice, Transgenic , Cells, Cultured , Synapsins/metabolism , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Mice, Inbred C57BL
5.
Genes (Basel) ; 15(4)2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38674386

ABSTRACT

Down syndrome (DS) is the most common form of inherited intellectual disability caused by trisomy of chromosome 21, presenting with intellectual impairment, craniofacial abnormalities, cardiac defects, and gastrointestinal disorders. The Ts65Dn mouse model replicates many abnormalities of DS. We hypothesized that investigation of the cerebral cortex of fluoxetine-treated trisomic mice may provide proteomic signatures that identify therapeutic targets for DS. Subcellular fractionation of synaptosomes from cerebral cortices of age- and brain-area-matched samples from fluoxetine-treated vs. water-treated trisomic and euploid male mice were subjected to HPLC-tandem mass spectrometry. Analysis of the data revealed enrichment of trisomic risk genes that participate in regulation of synaptic vesicular traffic, pre-synaptic and post-synaptic development, and mitochondrial energy pathways during early brain development. Proteomic analysis of trisomic synaptic fractions revealed significant downregulation of proteins involved in synaptic vesicular traffic, including vesicular endocytosis (CLTA, CLTB, CLTC), synaptic assembly and maturation (EXOC1, EXOC3, EXOC8), anterograde axonal transport (EXOC1), neurotransmitter transport to PSD (SACM1L), endosomal-lysosomal acidification (ROGDI, DMXL2), and synaptic signaling (NRXN1, HIP1, ITSN1, YWHAG). Additionally, trisomic proteomes revealed upregulation of several trafficking proteins, involved in vesicular exocytosis (Rab5B), synapse elimination (UBE3A), scission of endocytosis (DBN1), transport of ER in dendritic spines (MYO5A), presynaptic activity-dependent bulk endocytosis (FMR1), and NMDA receptor activity (GRIN2A). Chronic fluoxetine treatment of Ts65Dn mice rescued synaptic vesicular abnormalities and prevented abnormal proteomic changes in adult Ts65Dn mice, pointing to therapeutic targets for potential treatment of DS.


Subject(s)
Down Syndrome , Fluoxetine , Proteomics , Synaptic Vesicles , Animals , Fluoxetine/pharmacology , Mice , Down Syndrome/metabolism , Down Syndrome/drug therapy , Down Syndrome/genetics , Down Syndrome/pathology , Male , Proteomics/methods , Synaptic Vesicles/metabolism , Synaptic Vesicles/drug effects , Disease Models, Animal , Proteome/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/drug effects , Synaptosomes/metabolism , Synaptosomes/drug effects , Trisomy/genetics
6.
J Stroke Cerebrovasc Dis ; 33(6): 106578, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38636320

ABSTRACT

BACKGROUND: Notch1 signaling inhibiton with N-[N-(3,5-difluorophenacetyl)-1-alanyl]-S-phenylglycine t-butylester] (DAPT) treatment could promote brain recovery and the intervention effect is different between striatum (STR) and cortex (CTX), which might be accounted for different changes of glial activities, but the in-depth mechanism is still unknown. The purpose of this study was to identify whether DAPT could modulate microglial subtype shifts and astroglial-endfeet aquaporin-4 (AQP4) mediated waste solute drainage. METHODS: Sprague-Dawley rats (n=10) were subjected to 90min of middle cerebral artery occlusion (MCAO) and were treated with DAPT (n=5) or act as control with no treatment (n=5). Two groups of rats underwent MRI scans at 24h and 4 week, and sacrificed at 4 week after stroke for immunofluorescence (IF). RESULTS: Compared with control rats, MRI data showed structural recovery in ipsilateral STR but not CTX. And IF showed decreased pro-inflammatory M1 microglia and increased anti-inflammatory M2 microglia in striatal lesion core and peri-lesions of STR, CTX. Meanwhile, IF showed decreased AQP4 polarity in ischemic brain tissue, however, AQP4 polarity in striatal peri-lesions of DAPT treated rats was higher than that in control rats but shows no difference in cortical peri-lesions between control and treated rats. CONCLUSIONS: The present study indicated that DAPT could promote protective microglia subtype shift and striatal astrocyte mediated waste solute drainage, that the later might be the major contributor of waste solute metabolism and one of the accounts for discrepant recovery of STR and CTX.


Subject(s)
Aquaporin 4 , Astrocytes , Dipeptides , Disease Models, Animal , Infarction, Middle Cerebral Artery , Microglia , Rats, Sprague-Dawley , Receptor, Notch1 , Recovery of Function , Signal Transduction , Animals , Aquaporin 4/metabolism , Receptor, Notch1/metabolism , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/physiopathology , Male , Astrocytes/metabolism , Astrocytes/drug effects , Astrocytes/pathology , Microglia/metabolism , Microglia/drug effects , Microglia/pathology , Dipeptides/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/pathology , Corpus Striatum/metabolism , Corpus Striatum/drug effects , Corpus Striatum/pathology , Time Factors , Neuroprotective Agents/pharmacology , Ischemic Stroke/metabolism , Ischemic Stroke/drug therapy , Ischemic Stroke/physiopathology , Ischemic Stroke/pathology
7.
PLoS One ; 19(4): e0301355, 2024.
Article in English | MEDLINE | ID: mdl-38683825

ABSTRACT

Diabetes mellitus (DM) is a severe metabolic disease that can have significant consequences for cognitive health. Bioflavonoids such as Trifolium alexandrinum (TA), quercetin (Q), and Biochanin-A (BCA) are known to exert a wide range of pharmacological functions including antihyperglycemic activity. This study aimed to investigate the neurotherapeutic effects of quercetin-loaded nanoparticles (Q-LNP) and BCA extracted from TA against diabetes-induced cerebral cortical damage through modulation of PI3K/Akt/GSK-3ß and AMPK signaling pathways. Adult male Wistar albino rats (N = 25) were randomly assigned to one of five groups: control, diabetics fed a high-fat diet (HFD) for 2 weeks and intraperitoneally (i.p.) injected with STZ (40 mg/kg), and diabetics treated with Q-LNP (50 mg/kg BW/day), BCA (10 mg/kg BW/day), or TA extract (200 mg/kg BW/day). Treatments were applied by oral gavage once daily for 35 days. Diabetic rats treated with Q-LNP, BCA, and TA extract showed improvement in cognitive performance, cortical oxidative metabolism, antioxidant parameters, and levels of glucose, insulin, triglyceride, and total cholesterol. In addition, these treatments improved neurochemical levels, including acetylcholine, dopamine, and serotonin levels as well acetylcholinesterase and monoamine oxidase activities. Furthermore, these treatments lowered proinflammatory cytokine production for TNF-α and NF-κB; downregulated the levels of IL-1ß, iNOS, APP, and PPAR-γ; and attenuated the expressions of PSEN2, BACE, IR, PI3K, FOXO 1, AKT, AMPK, GSK-3ß, and GFAP. The histopathological examinations of the cerebral cortical tissues confirmed the biochemical results. Overall, the present findings suggest the potential therapeutic effects of TA bioflavonoids in modulating diabetes-induced cerebral cortical damage.


Subject(s)
Cerebral Cortex , Diabetes Mellitus, Experimental , Glycogen Synthase Kinase 3 beta , Nanoparticles , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Quercetin , Trifolium , Animals , Male , Rats , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/complications , Glycogen Synthase Kinase 3 beta/metabolism , Nanoparticles/chemistry , Phosphatidylinositol 3-Kinases/metabolism , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Quercetin/pharmacology , Quercetin/therapeutic use , Quercetin/administration & dosage , Rats, Wistar , Signal Transduction/drug effects , Trifolium/chemistry
8.
Neurochem Int ; 176: 105742, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38641028

ABSTRACT

Traumatic brain injury (TBI) remains a major health concern which causes long-term neurological disability particularly in war veterans, athletes and young adults. In spite of intense clinical and research investigations, there is no effective therapy to cease the pathogenesis of the disease. It is believed that axonal injury during TBI is potentiated by neuroinflammation and demyelination and/or failure to remyelination. This study highlights the use of naturally available cinnamein, also chemically known as benzyl cinnamate, in inhibiting neuroinflammation, promoting remyelination and combating the disease process of controlled cortical impact (CCI)-induced TBI in mice. Oral delivery of cinnamein through gavage brought down the activation of microglia and astrocytes to decrease the expression of inducible nitric oxide synthase (iNOS), glial fibrillary acidic protein (GFAP) and ionized calcium binding adaptor molecule 1 (Iba1) in hippocampus and cortex of TBI mice. Cinnamein treatment also stimulated remyelination in TBI mice as revealed by PLP and A2B5 double-labeling, luxol fast blue (LFB) staining and axonal double-labeling for neurofilament and MBP. Furthermore, oral cinnamein reduced the size of lesion cavity in the brain, improved locomotor functions and restored memory and learning in TBI mice. These results suggest a new neuroprotective property of cinnamein that may be valuable in the treatment of TBI.


Subject(s)
Brain Injuries, Traumatic , Disease Models, Animal , Animals , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/metabolism , Mice , Male , Mice, Inbred C57BL , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use
9.
Phytochemistry ; 222: 114094, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38604325

ABSTRACT

Safflopentsides A-C (1-3), three highly oxidized rearranged derivatives of quinochalcone C-glycosides, were isolated from the safflower yellow pigments. Their structures were determined based on a detailed spectroscopic analysis (UV, IR, HR-ESI-MS, 1D and 2D NMR), and the absolute configurations were confirmed by the comparison of experimental ECD spectra with calculated ECD spectra. Compounds 1-3 have an unprecedented cyclopentenone or cyclobutenolide ring A containing C-glucosyl group, respectively. The plausible biosynthetic pathways of compounds have been presented. At 10 µM, 2 showed strong inhibitory activity against rat cerebral cortical neurons damage induced by glutamate and oxygen sugar deprivation.


Subject(s)
Carthamus tinctorius , Glycosides , Oxidation-Reduction , Glycosides/chemistry , Glycosides/pharmacology , Glycosides/isolation & purification , Animals , Carthamus tinctorius/chemistry , Rats , Molecular Structure , Neurons/drug effects , Structure-Activity Relationship , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Neuroprotective Agents/isolation & purification , Dose-Response Relationship, Drug , Cerebral Cortex/drug effects , Chalcones/pharmacology , Chalcones/chemistry , Chalcones/isolation & purification
10.
Neuroreport ; 35(8): 542-550, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38597273

ABSTRACT

Wnt signaling plays an important role in adult brain function, and its dysregulation has been implicated in the loss of neuronal homeostasis. Despite the existence of many studies on the participation of the Wnt pathway in adult neurons, its regulation in astrocytes has been scarcely explored. Several reports point to the presence of Wnt ligands in astrocytes and their possible impact on neuronal plasticity or neuronal death. We aimed to analyze the effect of the neurotransmitter glutamate and the inflammatory cytokine TNFα on the mRNA and protein levels of the canonical Wnt agonist Wnt7a and the antagonist Dkk1 in cultured astrocytes. Primary astrocyte cultures from rat cerebral cortices were exposed to glutamate or TNFα. Wnt7a and Dkk1 expression was analyzed by RT-qPCR and its protein abundance and distribution was assessed by immunofluorescence. We found high basal expression and protein levels of Wnt7a and Dkk1 in unstimulated astrocytes and overproduction of Dkk1 mRNA induced by the two stimuli. These results reveal the astrocytic source of the canonical Wnt ligands Wnt7a and Dkk1, whose levels are differentially regulated by glutamate and TNFα. Astrocytes are a significant source of Wnt ligands, the production of which can be differentially regulated under excitatory or proinflammatory conditions, thereby impacting neuronal function.


Subject(s)
Astrocytes , Glutamic Acid , Intercellular Signaling Peptides and Proteins , Proto-Oncogene Proteins , Tumor Necrosis Factor-alpha , Wnt Proteins , Astrocytes/metabolism , Astrocytes/drug effects , Animals , Intercellular Signaling Peptides and Proteins/metabolism , Glutamic Acid/metabolism , Wnt Proteins/metabolism , Tumor Necrosis Factor-alpha/metabolism , Cells, Cultured , Rats , RNA, Messenger/metabolism , Rats, Wistar , Cerebral Cortex/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/cytology
11.
J Neurosci ; 44(18)2024 May 01.
Article in English | MEDLINE | ID: mdl-38508714

ABSTRACT

Drugs of abuse induce neuroadaptations, including synaptic plasticity, that are critical for transition to addiction, and genes and pathways that regulate these neuroadaptations are potential therapeutic targets. Tropomodulin 2 (Tmod2) is an actin-regulating gene that plays an important role in synapse maturation and dendritic arborization and has been implicated in substance abuse and intellectual disability in humans. Here, we mine the KOMP2 data and find that Tmod2 knock-out mice show emotionality phenotypes that are predictive of addiction vulnerability. Detailed addiction phenotyping shows that Tmod2 deletion does not affect the acute locomotor response to cocaine administration. However, sensitized locomotor responses are highly attenuated in these knock-outs, indicating perturbed drug-induced plasticity. In addition, Tmod2 mutant animals do not self-administer cocaine indicating lack of hedonic responses to cocaine. Whole-brain MR imaging shows differences in brain volume across multiple regions, although transcriptomic experiments did not reveal perturbations in gene coexpression networks. Detailed electrophysiological characterization of Tmod2 KO neurons showed increased spontaneous firing rate of early postnatal and adult cortical and striatal neurons. Cocaine-induced synaptic plasticity that is critical for sensitization is either missing or reciprocal in Tmod2 KO nucleus accumbens shell medium spiny neurons, providing a mechanistic explanation of the cocaine response phenotypes. Combined, these data, collected from both males and females, provide compelling evidence that Tmod2 is a major regulator of plasticity in the mesolimbic system and regulates the reinforcing and addictive properties of cocaine.


Subject(s)
Cocaine , Corpus Striatum , Mice, Knockout , Neuronal Plasticity , Animals , Cocaine/pharmacology , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Mice , Male , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Mice, Inbred C57BL , Cerebral Cortex/drug effects , Cerebral Cortex/physiology , Female , Cocaine-Related Disorders/physiopathology , Cocaine-Related Disorders/genetics , Microfilament Proteins/metabolism , Microfilament Proteins/genetics , Cortical Excitability/drug effects , Dopamine Uptake Inhibitors/pharmacology , Dopamine Uptake Inhibitors/administration & dosage
12.
Exp Neurol ; 376: 114759, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38519010

ABSTRACT

Malformations of cortical development (MCDs) are caused by abnormal neuronal migration processes during the fetal period and are a major cause of intractable epilepsy in infancy. However, the timing of hyperexcitability or epileptogenesis in MCDs remains unclear. To identify the early developmental changes in the brain of the MCD rat model, which exhibits increased seizure susceptibility during infancy (P12-15), we analyzed the pathological changes in the brains of MCD model rats during the neonatal period and tested NMDA-induced seizure susceptibility. Pregnant rats were injected with two doses of methylazoxymethanol acetate (MAM, 15 mg/kg, i.p.) to induce MCD, while controls were administered normal saline. The cortical development of the offspring was measured by performing magnetic resonance imaging (MRI) on postnatal days (P) 1, 5, and 8. At P8, some rats were sacrificed for immunofluorescence, Golgi staining, and Western analysis. In another set of rats, the number and latency to onset of spasms were monitored for 90 min after the NMDA (5 mg/kg i.p.) injection at P8. In MCD rats, in vivo MR imaging showed smaller brain volume and thinner cortex from day 1 after birth (p < 0.001). Golgi staining and immunofluorescence revealed abnormal neuronal migration, with a reduced number of neuronal cell populations and less dendritic arborization at P8. Furthermore, MCD rats exhibited a significant reduction in the expression of NMDA receptors and AMPAR4, along with an increase in AMPAR3 expression (p < 0.05). Although there was no difference in the latency to seizure onset between MCD rats and controls, the MCD rats survived significantly longer than the controls. These results provide insights into the early developmental changes in the cortex of a MCD rat model and suggest that delayed and abnormal neuronal development in the immature brain is associated with a blunted response to NMDA-induced excitotoxic injury. These developmental changes may be involved in the sudden onset of epilepsy in patients with MCD or prenatal brain injury.


Subject(s)
Cell Movement , Disease Models, Animal , Malformations of Cortical Development , N-Methylaspartate , Neurons , Rats, Sprague-Dawley , Animals , Rats , N-Methylaspartate/toxicity , Female , Pregnancy , Cell Movement/drug effects , Neurons/pathology , Neurons/drug effects , Malformations of Cortical Development/chemically induced , Malformations of Cortical Development/pathology , Animals, Newborn , Methylazoxymethanol Acetate/toxicity , Methylazoxymethanol Acetate/analogs & derivatives , Cerebral Cortex/pathology , Cerebral Cortex/drug effects , Male , Magnetic Resonance Imaging
13.
Neuron ; 112(9): 1426-1443.e11, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38442714

ABSTRACT

Glucocorticoids are important for proper organ maturation, and their levels are tightly regulated during development. Here, we use human cerebral organoids and mice to study the cell-type-specific effects of glucocorticoids on neurogenesis. We show that glucocorticoids increase a specific type of basal progenitors (co-expressing PAX6 and EOMES) that has been shown to contribute to cortical expansion in gyrified species. This effect is mediated via the transcription factor ZBTB16 and leads to increased production of neurons. A phenome-wide Mendelian randomization analysis of an enhancer variant that moderates glucocorticoid-induced ZBTB16 levels reveals causal relationships with higher educational attainment and altered brain structure. The relationship with postnatal cognition is also supported by data from a prospective pregnancy cohort study. This work provides a cellular and molecular pathway for the effects of glucocorticoids on human neurogenesis that relates to lasting postnatal phenotypes.


Subject(s)
Cerebral Cortex , Glucocorticoids , Neurogenesis , Promyelocytic Leukemia Zinc Finger Protein , Neurogenesis/drug effects , Neurogenesis/physiology , Humans , Animals , Mice , Glucocorticoids/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/cytology , Female , Promyelocytic Leukemia Zinc Finger Protein/metabolism , Pregnancy , Neurons/metabolism , Neurons/drug effects , Organoids/drug effects , Organoids/metabolism , Gene Expression Regulation, Developmental/drug effects , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Male
14.
Phytomedicine ; 128: 155438, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38537443

ABSTRACT

BACKGROUND: Yi-Qi-Huo-Xue Decoction (YQHXD), a traditional Chinese medicine formula, has demonstrated efficacy in the clinical treatment of intracerebral hemorrhage (ICH) for over a decade. Nevertheless, the precise pharmacotherapeutic compounds of YQHXD capable of penetrating into cerebral tissue and the pharmacological underpinnings of YQHXD remain ambiguous. METHODS: The active components of YQHXD in rat brains was analyzed by ultra-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry. The potential targets, pathways and biological progresses of YQHXD ameliorating ICH induced injury was predicted by network pharmacology. Moreover, collagenase-induced ICH rat model, primary cortex neurons exposed to hemin and molecular docking were applied to validate the molecular mechanisms of YQHXD. RESULTS: Eleven active components of YQHXD were identified within the brains. Employing the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases, our investigation concentrated on the roles of autophagy and the BDNF/TrkB signaling pathway in the pharmacological context. The pharmacological results revealed that YQHXD alleviated neurological dysfunction, brain water content, brain swelling, and pathological injury caused by ICH. Meanwhile, YQHXD inhibited autophagy influx and autophagosome in vivo, and regulated cortex neuronal autophagy and TrkB/BDNF pathway both in vivo and in vitro. Subsequently, N-acetyl serotonin (NAS), a selective TrkB agonist, was employed to corroborate the significance of the BDNF/TrkB pathway in this process. The combination of NAS and YQHXD did not further enhance the protective efficacy of YQHXD in ICH rats. Additionally, outcomes of molecular docking analysis revealed that nine compounds of YQHXD exhibited potential regulatory effects on TrkB. CONCLUSIONS: Ipsilateral neuronal autophagy and BDNF/TrkB pathway were activated 72 h after ICH. YQHXD effectively resisted injury induced by ICH, which was related with suppression of ipsilateral neuronal autophagy via BDNF/TrkB pathway. This study provides novel insights into the therapeutic mechanisms of traditional Chinese medicine in the context of ICH treatment.


Subject(s)
Autophagy , Brain-Derived Neurotrophic Factor , Cerebral Hemorrhage , Drugs, Chinese Herbal , Molecular Docking Simulation , Neurons , Rats, Sprague-Dawley , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cerebral Hemorrhage/drug therapy , Drugs, Chinese Herbal/pharmacology , Autophagy/drug effects , Male , Neurons/drug effects , Rats , Signal Transduction/drug effects , Receptor, trkB/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Disease Models, Animal , Neuroprotective Agents/pharmacology
15.
J Neurol ; 271(5): 2149-2158, 2024 May.
Article in English | MEDLINE | ID: mdl-38289534

ABSTRACT

INTRODUCTION: Ocrelizumab (OCR) and Fingolimod (FGL) are two high-efficacy treatments in multiple sclerosis which, besides their strong anti-inflammatory activity, may limit neurodegeneration. AIM: To compare the effect of OCR and FGL on clinical and MRI endpoints. METHODS: 95 relapsing-remitting patients (57 OCR, 38 FGL) clinically followed for 36 months underwent a 3-Tesla MRI at baseline and after 24 months. The annualized relapse rate, EDSS, new cortical/white matter lesions and regional cortical and deep grey matter volume loss were evaluated. RESULTS: OCR reduced the relapse rate from 0.48 to 0.04, FGL from 0.32 to 0.05 (both p < 0.001). Compared to FGL, OCR-group experienced fewer new white matter lesions (12% vs 32%, p = 0.005), no differences in new cortical lesions, lower deep grey matter volume loss (- 0.12% vs - 0.66%; p = 0.002, Cohen's d = 0.54), lower global cortical thickness change (- 0.45% vs - 0.70%; p = 0.036; d = 0.42) and reduced cortical thinning/volume loss in several regions of interests, including those of parietal gyrus (d-range = 0.65-0.71), frontal gyrus (d-range = 0.47-0.60), cingulate (d-range = 0.41-0.72), insula (d = 0.36), cerebellum (cortex d = 0.72, white matter d = 0.44), putamen (d = 0.35) and thalamus (d = 0.31). The effect on some regional thickness changes was confirmed in patients without focal lesions. CONCLUSIONS: When compared with FGL, patients receiving OCR showed greater suppression of focal MRI lesions accumulation and lower cortical and deep grey matter volume loss.


Subject(s)
Antibodies, Monoclonal, Humanized , Fingolimod Hydrochloride , Gray Matter , Magnetic Resonance Imaging , Multiple Sclerosis, Relapsing-Remitting , Humans , Female , Male , Adult , Gray Matter/diagnostic imaging , Gray Matter/pathology , Gray Matter/drug effects , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/therapeutic use , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/diagnostic imaging , Multiple Sclerosis, Relapsing-Remitting/pathology , Middle Aged , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/therapeutic use , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/pathology , Cerebral Cortex/drug effects , Sphingosine 1 Phosphate Receptor Modulators/pharmacology , Immunologic Factors/pharmacology , Immunologic Factors/administration & dosage , Follow-Up Studies
16.
Behav Brain Res ; 460: 114801, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38070690

ABSTRACT

The Wnt/beta-catenin pathway plays a crucial role in regulating cellular processes and has been implicated in neural activity-dependent learning as well as anxiety. However, the role of this pathway in young children with abnormal cortical development is unknown. Cortical malformations at early development, behavioral abnormalities, and a susceptibility to seizures have been reported in rats prenatally exposed to methylazoxymethanol. In this study, we aimed to investigate whether we could improve the behavioral deficits in young rats with malformed cerebral cortices by modulation of the Wnt/beta-catenin pathway. We found a small molecule Wnt/beta-catenin inhibitor (CWP) that increased exploratory behavior in the open field test (P9, CWP 100 ug treatment, peripheral exploration, P = 0.011) and social behavior test (P12, CWP 250 ug treatment, distance traveled in center, P = 0.033) and decreased anxiety in fear conditioning. However, it did not reduce the susceptibility to seizures. After high dose (250 ug) CWP treatment at P12, phosphocreatine and glutathione (GSH) were decreased in the cortex at P15 (P = 0.021). These findings suggest that the role of Wnt/beta-catenin signaling in exploratory behavior and anxiety during early development warrants further investigation.


Subject(s)
Wnt Signaling Pathway , beta Catenin , Animals , Rats , Anxiety/drug therapy , beta Catenin/drug effects , beta Catenin/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Neurogenesis , Seizures , Wnt Signaling Pathway/drug effects
17.
Eur J Pharmacol ; 964: 176306, 2024 Feb 05.
Article in English | MEDLINE | ID: mdl-38145647

ABSTRACT

During the production of orexin A and B from preproorexin, a common precursor protein, in hypothalamic orexin neurons, C-terminal peptide (herein called preproorexin C-peptide) is concomitantly produced via post-translational processing. The predicted three-dimensional structure of preproorexin C-peptide is similar among mammalian species, suggestive of a conserved function in the mammalian brain. However, C-peptide has long been regarded as a non-functional peptide. We herein examined the effects of rat and/or mouse preproorexin C-peptide on gene expression and cell viability in cultured rat cerebrocortical cells and on memory behavior in C57BL/6J mice. Rat and mouse C-peptides both increased brain-derived neurotrophic factor (Bdnf) mRNA levels. Moreover, C-peptide enhanced high K+-, glutamate-, and BDNF-induced increases in Bdnf mRNA levels without affecting forskolin-induced Bdnf expression. H-89, a protein kinase A inhibitor, blocked C-peptide-induced Bdnf expression, whereas rolipram, a phosphodiesterase inhibitor, enhanced this effect. Intracellular cyclic AMP concentrations were elevated by C-peptide. These results demonstrate that preproorexin C-peptide promoted Bdnf mRNA expression by a cyclic AMP-dependent mechanism. Eleven amino acids at the N terminus of rat preproorexin C-peptide exerted similar effects on Bdnf expression as full-length preproorexin C-peptide. Preproorexin C-peptide also exerted protective effects against CoCl2-induced neuronal cell death. An intracerebroventricular injection of mouse preproorexin C-peptide induced c-fos and Bdnf expression in the cerebral cortex and hippocampus and enhanced novel object recognition memory in mice. Collectively, the present results show that preproorexin C-peptide is a functional substance, at least in some pharmacological and neuronal settings.


Subject(s)
Brain-Derived Neurotrophic Factor , Cerebral Cortex , Orexins , Peptide Fragments , Animals , Mice , Rats , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cyclic AMP/metabolism , Mice, Inbred C57BL , Orexins/pharmacology , RNA, Messenger/metabolism , Peptide Fragments/pharmacology , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism
18.
Biomed Res Int ; 2023: 9911397, 2023.
Article in English | MEDLINE | ID: mdl-37564141

ABSTRACT

The anti-dementia effect following ischemic stroke with metabolic syndrome (MetS) of the polyherbal functional ingredient comprising ginger, Chinese date, and wood ear mushroom (GCJ) was hypothesized due to its neuroprotective effect against stroke. This study was performed to test this hypothesis and to explore the underlying mechanism. Male Wistar rats weighing 180-220 g were induced metabolic syndrome (MetS) with a 16-week high-carbohydrate high-fat diet (HCHF) feeding. The rats with MetS characteristics were orally administered GCJ at various doses (GCJ100, GCJ200, and GCJ300 mg kg-1 BW) 21 days pre-induction and 21 days post-induction of reperfusion injury (I/R) at the right middle cerebral artery (MCAO). Memory was evaluated every 7 days during the study period. At the end of the study, neuron density, AChE activity, and the expressions of eNOS, BDNF, and pERK/ERK in the prefrontal cortex, and hippocampus were also determined. MetS rats with GCJ treatment improved memory impairment, enhanced neuron density, and increased the expressions of eNOS, BDNF, and pERK/ERK but suppressed AChE in both areas. Therefore, the anti-dementia effect following ischemic stroke with metabolic syndrome of GCJ may involve the improvement of AChE, eNOS, BDNF, pERK/ERK, and neural plasticity. However, this required confirmation by clinical study.


Subject(s)
Dementia , Drugs, Chinese Herbal , Ischemic Stroke , Metabolic Syndrome , Neuroprotective Agents , Animals , Male , Rats , Agaricales , Brain-Derived Neurotrophic Factor , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Dementia/drug therapy , Dementia/etiology , Dementia/genetics , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Zingiber officinale , Ischemic Stroke/complications , Ischemic Stroke/drug therapy , Metabolic Syndrome/drug therapy , Metabolic Syndrome/genetics , Metabolic Syndrome/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Phoeniceae , Rats, Wistar , Disease Models, Animal
19.
Cell Mol Neurobiol ; 43(6): 2895-2907, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36862242

ABSTRACT

Isolated sulfite oxidase (ISOD) and molybdenum cofactor (MoCD) deficiencies are genetic diseases biochemically characterized by the toxic accumulation of sulfite in the tissues of patients, including the brain. Neurological dysfunction and brain abnormalities are commonly observed soon after birth, and some patients also have neuropathological alterations in the prenatal period (in utero). Thus, we investigated the effects of sulfite on redox and mitochondrial homeostasis, as well as signaling proteins in the cerebral cortex of rat pups. One-day-old Wistar rats received an intracerebroventricular administration of sulfite (0.5 µmol/g) or vehicle and were euthanized 30 min after injection. Sulfite administration decreased glutathione levels and glutathione S-transferase activity, and increased heme oxygenase-1 content in vivo in the cerebral cortex. Sulfite also reduced the activities of succinate dehydrogenase, creatine kinase, and respiratory chain complexes II and II-III. Furthermore, sulfite increased the cortical content of ERK1/2 and p38. These findings suggest that redox imbalance and bioenergetic impairment induced by sulfite in the brain are pathomechanisms that may contribute to the neuropathology of newborns with ISOD and MoCD. Sulfite disturbs antioxidant defenses, bioenergetics, and signaling pathways in the cerebral cortex of neonatal rats. CII: complex II; CII-III: complex II-III; CK: creatine kinase; GST: glutathione S-transferase; HO-1: heme oxygenase-1; SDH: succinate dehydrogenase; SO32-: sulfite.


Subject(s)
Cerebral Cortex , Energy Metabolism , Molybdenum Cofactors , Sulfite Oxidase , Sulfites , Animals , Rats , Animals, Newborn , Oxidation-Reduction , Sulfites/adverse effects , Sulfite Oxidase/metabolism , Molybdenum Cofactors/metabolism , Rats, Wistar , Homeostasis , Mitochondria/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Antioxidants/metabolism
20.
Curr Alzheimer Res ; 20(1): 29-37, 2023.
Article in English | MEDLINE | ID: mdl-36892031

ABSTRACT

OBJECTIVE: The objective of this study is to investigate the neuroprotective effects of ß- sitosterol using the AlCl3 model of Alzheimer's Disease. METHODS: AlCl3 model was used to study cognition decline and behavioral impairments in C57BL/6 mice. Animals were randomly assigned into 4 groups with the following treatments: Group 1 received normal saline for 21 days, Group 2 received AlCl3 (10 mg/kg) for 14 days; Group 3 received AlCl3(10 mg/kg) for 14 days + ß-sitosterol (25mg/kg) for 21 days; while Group 4 was administered ß-sitosterol (25mg/kg) for 21 days. On day 22, we performed the behavioral studies using a Y maze, passive avoidance test, and novel object recognition test for all groups. Then the mice were sacrificed. The corticohippocampal region of the brain was isolated for acetylcholinesterase (AChE), acetylcholine (ACh), and GSH estimation. We conducted histopathological studies using Congo red staining to measure ß -amyloid deposition in the cortex and hippocampal region for all animal groups. RESULTS: AlCl3 successfully induced cognitive decline in mice following a 14-day induction period, as shown by significantly decreased (p < 0.001) in step-through latency, % alterations, and preference index values. These animals also exhibited a substantial decrease in ACh (p <0.001) and GSH (p < 0.001) and a rise in AChE (p < 0.001) compared to the control group. Mice administered with AlCl3 and ß-sitosterol showed significantly higher step-through latency time, % alteration time, and % preference index (p < 0.001) and higher levels of ACh, GSH, and lower levels of AChE in comparison to the AlCl3 model. AlCl3-administered animals also showed higher ß-amyloid deposition, which got significantly reduced in the ß-sitosterol treated group. CONCLUSION: AlCl3 was effectively employed to induce a cognitive deficit in mice, resulting in neurochemical changes and cognitive decline. ß -sitosterol treatment mitigated AlCl3-mediated cognitive impairment.


Subject(s)
Aluminum Chloride , Alzheimer Disease , Cognitive Dysfunction , Neuroprotective Agents , Sitosterols , Animals , Mice , Acetylcholine/metabolism , Acetylcholinesterase/metabolism , Aluminum Chloride/administration & dosage , Aluminum Chloride/toxicity , Alzheimer Disease/chemically induced , Alzheimer Disease/drug therapy , Alzheimer Disease/prevention & control , Avoidance Learning/drug effects , Case-Control Studies , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cognition/drug effects , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/prevention & control , Computer Simulation , Disease Models, Animal , Glutathione/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Maze Learning/drug effects , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Sitosterols/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...