Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Neuropsychiatr Dis Treat ; 20: 523-533, 2024.
Article in English | MEDLINE | ID: mdl-38469210

ABSTRACT

Background: The 4-hydroxysesamin (4-HS, a di-tetrahydrofuran lignin) is a modified sesamin that was prepared in the laboratory. This preclinical study was designed to preliminarily investigate the neuroprotective properties of 4-HS. Methods: In vitro, neuronal injury and inflammation were simulated by oxygen-glucose deprivation and lipopolysaccharide (LPS) exposure in mouse hippocampal neuronal HT22 cell line, and treated with 4-HS and/or metformin (MET, MAPK pathway activator for exploring mechanism). CCK-8, flow cytometry, and enzyme-linked immunosorbent assay were performed to evaluate cell viability, apoptosis, and inflammation. Apoptosis- and pathway-related proteins were detected by Western blotting. Middle cerebral artery occlusion (MCAO) was constructed as a stroke model and treated with 4-HS for in vivo confirmation. Histological staining was used for in vivo evaluation of 4-HS properties. Results: The 4-HS showed similar anti-inflammatory activity to sesamin but did not affect the cell viability of HT22 cells. In vitro, 4-HS improved the cell viability, ameliorated neuronal apoptosis, along with the reversion of apoptotic proteins (Bax, cleaved-caspase 3/9, Bcl-2) expression and inflammatory cytokines (IL-6, TNF-α, IL-10) in LPS-treated HT22 cells. The 4-HS suppressed the phosphorylation of ERK, JNK, and p38 but the addition of MET reversed 4-HS-induced changes of phenotype and protein expression in LPS-treated cells. In vivo, 4-HS showed apparent improvement in cerebral infarction, brain tissue morphology, neuronal architecture, apoptosis, and inflammation of MCAO mice, and also showed inhibiting effects on the phosphorylation of ERK, JNK, and p38, confirming in vivo results. Conclusion: In this first pre-clinical study on 4-HS, we preliminarily demonstrated the neuroprotective properties of 4-HS both in cell and animal models, and proposed that the underlying mechanism might be associated with the MAPK pathway.

2.
Microorganisms ; 11(11)2023 Oct 25.
Article in English | MEDLINE | ID: mdl-38004640

ABSTRACT

Epilepsy (EP) is a complex brain disorder showing a lot of unknows reasons. Recent studies showed that gut microbiota can influence epilepsy via the brain-gut axis. Nevertheless, the mechanism by which gut microbiota affects adult epilepsy still remains unclear. In this study, fecal and serum samples were obtained from patients with epilepsy and normal controls. Using an integrated analysis, sequencing was performed by macrogenomics and high-throughput targeted metabolomics with various bioinformatics approaches. The macrogenomic sequencing revealed significant changes in microbial structure in patients suffering from epilepsy. For example, at the phylum level, the relative abundance of Actinobacteria, Bacteroidetes and Proteobacteria showed an increase in the patients with epilepsy, whereas that of Firmicutes decreased. In addition, the patients with epilepsy had significantly differential metabolite profiles compared to normal controls, and five clusters with 21 metabolites, mainly containing the upregulation of some fatty acids and downregulation of some amino acids. Tryptophan (AUC = 91.81, p < 0.0001), kynurenine (AUC = 79.09, p < 0.01) and 7Z,10Z,13Z,16Z-Docosatetraenoic acid (AUC = 80.95, p < 0.01) may be used as potential diagnostic markers for epilepsy. Differential serum metabolites have effects on tryptophan metabolism, iron death and other pathways. Furthermore, a multiomic joint analysis observed a statistically significant correlation between the differential flora and the differential serum metabolites. In our findings, a macrogenomic analysis revealed the presence of dysregulated intestinal flora species and function in adult epileptic patients. Deeper metabolomic analyses revealed differences in serum metabolites between patients with epilepsy and healthy populations. Meanwhile, the multiomic combination showed connection between the gut microbes and circulating metabolites in the EP patients, which may be potential therapeutic targets.

3.
Comput Math Methods Med ; 2022: 8371492, 2022.
Article in English | MEDLINE | ID: mdl-36238484

ABSTRACT

Electroconvulsive therapy (ECT) is a nonpharmacological treatment for depressive episodes and other psychiatric disorders. It is used to control the condition by causing a transient loss of consciousness through electrical stimulation. Dexmedetomidine (DEX) is a novel and highly selective adrenergic agonist with sedative, sympathetic nerve activity inhibiting and stress-responsive effects. This study focused on the effect of DEX on cerebral protection after ECT treatment. 68 depression patients were enrolled and divided into control group and DEX group. The occurrence of delirium after ECT treatment in depression cases was recorded. In vivo, we constructed chronic mild and unpredictable stress (CUMS) rats to mimic depression model. Meanwhile, ECT treatment and DEX injection were administrated in CUMS rats. Learning and memory in rats were measured by Morris water maze test, open field test (OFT), and forced swimming test (FST). Finally, the expression of miR-146a-5p and NF-κB was determined by RT-qPCR and western blot assay. The incidence of delirium after ECT treatment was prominently reduced in DEX group in relation to control group. In vivo, DEX injection had no effect on ECT treatment efficacy against depression conditions. After ECT treatment, the cognitive impairment was ameliorated in CUMS rats accomplished with decreased miR-146a-5p and increased NF-κB level. Finally, compared with ECT treatment, DEX injection could protect against depression-like behaviors by increasing miR-146a-5p level and inactivated NF-κB pathway. Overall, ECT-induced cognitive impairment in depression rats could be ameliorated by DEX injection via miR-146a-5p/NF-κB axis.


Subject(s)
Cognitive Dysfunction , Delirium , Dexmedetomidine , Electroconvulsive Therapy , MicroRNAs , Adrenergic Agonists , Animals , Dexmedetomidine/pharmacology , Hypnotics and Sedatives , MicroRNAs/genetics , MicroRNAs/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Rats
4.
J Immunol Res ; 2022: 5052609, 2022.
Article in English | MEDLINE | ID: mdl-35497882

ABSTRACT

Invasive surgical cerebrum biopsy results in delayed treatment for the definitive diagnosis of primary central nervous system lymphoma (PCNSL). The existent research was aimed at confirming the underlying diagnostic miRNAs of distinguishing PCNSL from glioma. A publicly available miRNA expression profiles (GSE139031) from adult PCNSL as well as glioma specimens were provided by GEO datasets. Differentially expressed miRNAs (DEMs) were filtered between 42 PCNSL patients and 170 glioma patients. Candidate miRNAs were identified through SVM-RFE analysis and LASSO model. ROC assays were operated to determine the diagnostic value of serum miRNAs in distinguishing PCNSL from glioma. StarBase v2.0 was applied to screen the targeting genes of miRNAs, and KEGG analysis was applied using the targeting genes of miRNAs. In this study, we identified 12 dysregulated miRNAs between PCNSL and glioma samples. The ten critical miRNAs (miR-6820-3p, miR-6803-3p, miR-30a-3p, miR-4751, miR-3918, miR-146a-3p, miR-548am-3p, miR-371a-3p, miR-487a-3p, and miR-4756-5p) between these two algorithms were ultimately identified. The results of KEGG revealed that the targeting genes of hsa-miR-3918 were primarily related to MAPK signal pathway, PI3K-Akt signal pathway, and human papillomavirus infection. Overall, bioinformatics analysis revealed that ten miRNAs are potential biomarker for distinguishing PCNSL from glioma.


Subject(s)
Glioma , Lymphoma , MicroRNAs , Adult , Central Nervous System , Glioma/diagnosis , Glioma/genetics , Humans , Lymphoma/diagnosis , Lymphoma/genetics , MicroRNAs/genetics , Phosphatidylinositol 3-Kinases
5.
Toxicol Lett ; 322: 66-76, 2020 Apr 01.
Article in English | MEDLINE | ID: mdl-31945382

ABSTRACT

Silent Information Regulator 1 (SIRT1), an NAD+-dependent deacetylase, contributes to the neuroprotective effect. However, intracellular signaling pathways that affect SIRT1 function remain unknown. It is well known that N-methyl-D-aspartate (NMDA) receptor activation induces calcium influx which then activates PKC, and SIRT1 is a mRNA target for HuR protein. We hypothesize that Ca2+-PKC-HuR-SIRT1 pathway modulates SIRT1 function. The present study is to investigate the potential pathway of SIRT1 in the SH-SY5Y cell line as an in vitro model of NMDA-induced neurotoxicity. The results showed that: (1) SIRT1 levels were downregulated in NMDA model; (2) NMDA induced an increase in serine phosphorylation of HuR, while inhibition of serine phosphorylation of HuR increased SIRT1 levels, promoting cell survival; (3) PKC inhibitor (Gö 6976) reversed NMDA insults and also suppressed serine phosphorylation of HuR; (4) 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA-AM), an intracellular calcium chelator, fully reversed NMDA insults and also inhibited PKC activity evoked by NMDA. These results indicate that intracellular elevated Ca2+ activates PKC, which phosphorylates HuR and then promotes SIRT1 mRNA decay and subsequent neuronal death in NMDA model. Therefore, the study suggests that inhibition of Ca2+-PKC-HuR-SIRT1 pathway could be an effective strategy for preventing certain neurological diseases related to NMDA excitotoxicity.


Subject(s)
Excitatory Amino Acid Agonists/toxicity , N-Methylaspartate/toxicity , Neurons/drug effects , Neurotoxicity Syndromes/etiology , Sirtuin 1/metabolism , Calcium/metabolism , Calcium Signaling , Cell Death/drug effects , Cell Line, Tumor , Down-Regulation , ELAV-Like Protein 1/metabolism , Humans , Neurons/enzymology , Neurons/pathology , Neurotoxicity Syndromes/enzymology , Neurotoxicity Syndromes/pathology , Phosphorylation , Protein Kinase C/metabolism , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Serine , Sirtuin 1/genetics
6.
Med Sci Monit ; 25: 1749-1758, 2019 Mar 07.
Article in English | MEDLINE | ID: mdl-30843525

ABSTRACT

BACKGROUND Temporal lobe epilepsy (TLE) is the most common type of intractable epilepsy in humans, and it is often accompanied by cognitive impairment. In this study, we examined the effects of (-)-Epigallocatechin-3-gallate (EGCG) after SE on behavior in the rat lithium-pilocarpine model of TLE. MATERIAL AND METHODS The rats were randomly divided into 3 groups: (1) the control group, in which 12 rats received no treatment); (2) the epilepsy (EP) group, in which 15 rats were treated with saline after status epilepticus (SE); and (3) the EP+EGCG group, in which 15 rats were treated with EGCG (25 mg/kg/d, intraperitoneal) after SE. The SE model was induced with lithium chloride-pilocarpine, and electroencephalography and a high-definition camera were used to monitor SRS. The Morris water maze test and hippocampal late-phase long-term potentiation (L-LTP) recordings were used to evaluate cognitive impairment, and TLR4, NF-kappaB, and IL-1ß levels were determined using Western blot analysis. RESULTS We concluded that EGCG treatment after SE (1) markedly reduced SRS frequency in pilocarpine-treated rats, (2) improved epilepsy-induced cognitive impairment and reversed epilepsy-induced synaptic dysfunction in L-LTP in vivo, (3) protected hippocampal neurons from damage after SRS, and (4) significantly attenuated the increase in TRL-4 and IL-1ß hippocampal levels. The above findings clearly show that EGCG exerts antiepileptogenesis and neuroprotective effects on pilocarpine-induced epilepsy. CONCLUSIONS We found that EGCG can suppress seizures and inhibit hippocampal neuronal apoptosis, as well as improving cognitive function of epileptic rats. Our findings suggest that EGCG may a novel adjuvant therapeutic approach in epilepsy by improving epileptic behavior and cognitive dysfunction.


Subject(s)
Catechin/analogs & derivatives , Epilepsy/drug therapy , Epilepsy/physiopathology , Animals , Catechin/metabolism , Catechin/pharmacology , Disease Models, Animal , Epilepsy/metabolism , Epilepsy, Temporal Lobe/metabolism , Epilepsy, Temporal Lobe/physiopathology , Hippocampus/metabolism , Lithium , Long-Term Potentiation/drug effects , Male , NF-kappa B/drug effects , NF-kappa B/metabolism , Neurons/metabolism , Neuroprotective Agents/pharmacology , Pilocarpine , Rats , Rats, Sprague-Dawley , Seizures/physiopathology , Signal Transduction/drug effects , Toll-Like Receptor 4/drug effects , Toll-Like Receptor 4/metabolism
7.
Brain Res Bull ; 148: 63-69, 2019 05.
Article in English | MEDLINE | ID: mdl-30910691

ABSTRACT

Maternal epilepsy during pregnancy is associated with an increased incidence of brain damage and cognitive deficits in offspring. Oxidative stress is believed to play a critical role in this process. Astaxanthin, a natural carotenoid and dietary supplement, possesses potent antioxidant properties. This study was designed to investigate whether astaxanthin ameliorates the hippocampal damage in newborn rats induced by maternal epileptic seizures in utero and to explore the underlying mechanisms. Female Sprague-Dawley rats underwent chronic amygdalar kindling. After being fully kindled, all rats were allowed to mate, and electrical stimulation in the amygdala was performed every other day throughout the pregnancy. Astaxanthin was intraperitoneally injected at a dose of 30 mg/kg/d throughout pregnancy. Prenatal astaxanthin administration ameliorated neuronal lesions, decreased oxidative stress and induced the expression of cAMP response element-binding protein (CREB) and brain-derived neurotrophic factor (BDNF) in the hippocampus of pups. Astaxanthin also ameliorated placental ischemic damage in epileptic mothers. Based on the results of the present study, we concluded that astaxanthin might serve as a therapeutic agent for preventing brain damage in offspring exposed to prenatal maternal seizures.


Subject(s)
Hippocampus/drug effects , Amygdala/drug effects , Animals , Animals, Newborn/metabolism , Antioxidants/pharmacology , Cognition Disorders/metabolism , Epilepsy/metabolism , Female , Kindling, Neurologic/drug effects , Male , Maternal Exposure , Neurons/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Pregnancy , Prenatal Exposure Delayed Effects/pathology , Rats , Rats, Sprague-Dawley , Seizures/metabolism , Temporal Lobe/metabolism , Xanthophylls/metabolism , Xanthophylls/pharmacology
8.
Brain Res ; 1714: 88-98, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30768929

ABSTRACT

Cognitive impairment is one of the most common and disabling co-morbidities of epilepsy. It is therefore imperative to find novel treatment approaches to rescue cognitive function among epilepsy patients. Adult neurogenesis is strongly implicated in cognitive function, and mild hypoxia is known to promote the proliferation and differentiation of both embryonic and adult neural stem cells (NSCs). In the present study, we investigated the effect of mild hypoxia on cognitive function and hippocampal neurogenesis of rats with pilocarpine-induced chronic epilepsy. Chronic epilepsy induced marked spatial learning and memory deficits in the Morris water maze that were rescued by consecutively 28 days mild hypoxia exposure (6 h/d at 3000 m altitude equivalent) during the chronic phase. Moreover, mild hypoxia reversed the suppression of hippocampal neurogenesis and the downregulation of NT-3 and BDNF expression in hippocampus and cortex of epileptic rats. Mild hypoxia in vitro also promoted hippocampus-derived NSC proliferation and neuronal differentiation. In addition, mild hypoxia enhanced Notch1 and Hes1 expression, suggesting that Notch1 signaling may be involved in neuroprotection of hypoxia. Our data may help to pave the way for identifying new therapeutic targets for rescuing cognition conflicts in epileptic patients by using hypoxia to promote hippocampus neurogenesis.


Subject(s)
Epilepsy/metabolism , Hippocampus/metabolism , Hypoxia/metabolism , Neurogenesis/physiology , Receptor, Notch1/metabolism , Animals , Cell Differentiation/physiology , Cell Proliferation/physiology , Cognition/physiology , Epilepsy/pathology , Hippocampus/cytology , Hippocampus/pathology , Male , Memory Disorders/metabolism , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Pilocarpine/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction , Spatial Learning/physiology , Temporal Lobe/metabolism
9.
Oxid Med Cell Longev ; 2017: 2823454, 2017.
Article in English | MEDLINE | ID: mdl-29081884

ABSTRACT

Silent information regulator 1 (SIRT1), an NAD+-dependent deacetylase, is involved in the regulation of gene transcription, energy metabolism, and cellular aging and has become an important therapeutic target across a range of diseases. Recent research has demonstrated that SIRT1 possesses neuroprotective effects; however, it is unknown whether it protects neurons from NMDA-mediated neurotoxicity. In the present study, by activation of SIRT1 using resveratrol (RSV) in cultured cortical neurons or by overexpression of SIRT1 in SH-SY5Y cell, we aimed to evaluate the roles of SIRT1 in NMDA-induced excitotoxicity. Our results showed that RSV or overexpression of SIRT1 elicited inhibitory effects on NMDA-induced excitotoxicity including a decrease in cell viability, an increase in lactate dehydrogenase (LDH) release, and a decrease in the number of living cells as measured by CCK-8 assay, LDH test, and Calcein-AM and PI double staining. RSV or overexpression of SIRT1 significantly improved SIRT1 deacetylase activity in the excitotoxicity model. Further study suggests that overexpression of SIRT1 partly suppressed an NMDA-induced increase in p53 acetylation. These results indicate that SIRT1 activation by either RSV or overexpression of SIRT1 can exert neuroprotective effects partly by inhibiting p53 acetylation in NMDA-induced neurotoxicity.


Subject(s)
Sirtuin 1/biosynthesis , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Excitatory Postsynaptic Potentials/drug effects , Humans , N-Methylaspartate/toxicity , Rats , Rats, Wistar , Resveratrol , Signal Transduction , Stilbenes/pharmacology , Transfection
10.
Neurosci Lett ; 618: 19-24, 2016 Apr 08.
Article in English | MEDLINE | ID: mdl-26940236

ABSTRACT

There are numerous mechanisms by which the brain generates seizures. It is well known that oxidative stress plays a pivotal role in status epilepticus (SE). Salidroside (SDS) extracted from Rhodiola rosea L. shows multiple bioactive properties, such as neuroprotection and antioxidant activity in vitro and in vivo. This study explored the role of SDS in kainic acid (KA)-induced SE and investigated the underlying mechanism. Latency to SE increased in the SDS-pretreated mice compared to the KA group, while the percentage of incidence of SE was significantly reduced. These results suggested that pretreatment with SDS not only delayed SE, but it also decreased the incidence of SE induced by KA. KA increased MDA level and reduced the production of SOD and GSH at multiple timepoints after KA administration. SDS inhibited the change of MDA, SOD and GSH induced by KA prior to SE onset, indicating that SDS protects against KA-induced SE via suppressing oxidative stress. Based on these results, we investigated the possible molecular mechanism of SDS. Pretreatment with SDS reversed the KA-induced decrease in AMP-activated protein kinase (AMPK); increased the sirtuin 1 (SIRT1) deacetylase activity in KA-treated mice, which had no demonstrable effect on SIRT1 mRNA and protein; and suppressed the KA-induced increase in Ace-FoxO1. These results showed that AMPK/SIRT1/FoxO1 signaling is possibly the molecular mechanism of neuroprotection by SDS.


Subject(s)
Antioxidants/therapeutic use , Glucosides/therapeutic use , Kainic Acid , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Phenols/therapeutic use , Status Epilepticus/prevention & control , AMP-Activated Protein Kinases/metabolism , Animals , Antioxidants/pharmacology , Forkhead Transcription Factors/metabolism , Glucosides/pharmacology , Glutathione/metabolism , Hippocampus/metabolism , Male , Malondialdehyde/metabolism , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neuroprotective Agents/pharmacology , Phenols/pharmacology , Seizures/physiopathology , Seizures/prevention & control , Sirtuin 1/metabolism , Status Epilepticus/chemically induced , Status Epilepticus/metabolism , Status Epilepticus/physiopathology , Superoxide Dismutase/metabolism
12.
Sheng Li Xue Bao ; 64(6): 609-16, 2012 Dec 25.
Article in English | MEDLINE | ID: mdl-23258322

ABSTRACT

NMDA-induced excitotoxicity cause severe neuronal damage including apoptosis and necrosis. The present study was aimed to evaluate the proportion of NMDA-induced apoptosis of rat cortical neurons and discover signal transduction mechanism. Caspase inhibitor and lactate dehydrogenase (LDH) assay were used to study the NMDA-induced apoptosis. To explore the involved signal pathways, the primary culture of rat cortical neurons were pretreated by the inhibitors of three MAPK pathways, extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and p38 MAPK. With 2 h of NMDA treatment, cellular apoptosis was measured by caspase-3 activity, terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick-end labeling (TUNEL) and Annexin V staining. The results showed that: (1) Caspase-dependent apoptosis accounted for 22.49% in NMDA-induced neuronal death; (2) Pretreatment with p38 MAPK inhibitor SB203580 (10 µmol/L) significantly decreased NMDA-mediated caspase-3 activity by 30.43% (P < 0.05). However, ERK inhibitor PD98059 (20 µmol/L) or JNK inhibitor SP600125 (20 µmol/L) did not influence caspase-3 activity; (3) Pretreatment with SB203580 significantly reduced the number of NMDA-induced TUNEL-positive cells by 33.10% (P < 0.05). PD98059 (20 µmol/L) or SP600125 (20 µmol/L) did not show obvious effect; (4) Pretreatment with SB203580 (10 µmol/L) significantly reduced the number of NMDA-induced early apoptotic neurons by 55.56% (P < 0.05). Also, SP600125 (20 µmol/L) significantly decreased the amount of late apoptotic/dead cells by 67.59% (P < 0.05). There was no effect of PD98059 (20 µmol/L). These results indicate that: (1) NMDA induces neuronal apoptosis besides necrosis; (2) p38 MAPK, but not JNK and ERK, is involved in NMDA-induced neuronal apoptosis, and inhibition of the apoptotic signaling pathway contributes to neuroprotection; (3) JNK activation might contribute to NMDA-induced neuronal necrosis rather than apoptosis.


Subject(s)
Apoptosis , MAP Kinase Signaling System , N-Methylaspartate/pharmacology , Neurons/cytology , Animals , Anthracenes/pharmacology , Caspase 3/metabolism , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Imidazoles/pharmacology , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Primary Cell Culture , Pyridines/pharmacology , Rats , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL