Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Epilepsy Res ; 168: 106476, 2020 12.
Article in English | MEDLINE | ID: mdl-33096314

ABSTRACT

BACKGROUND: Epilepsy is a one of the most frequent serious neurological disorders characterized by enduring and unprovoked seizures. The treatments to epilepsy are very limited and many patients are even resistant to current medications due to the elusive pathogenesis. Here, we sought to investigate the functions of lncRNA SNHG1 and miR-154-5p in epilepsy. METHODS: We employed both in vivo mouse model and in vitro cell model to study epilepsy. H&E staining and Nissl staining were used to examine the morphology of hippocampus and measure neuronal injury, respectively. TUNEL staining and flow cytometry were performed to determine cell apoptosis. Caspase-3 activity assay kit was used to assess caspase-3 activity. RT-qPCR and western blot were conducted to measure the levels of SNHG1, miR-154-5p, TLR5, and SP1, respectively. Dual luciferase reporter assay was employed to validate the binding relationship of SNHG1/miR-154-5p and miR-154-5p/TLR5. ChIP assay was performed to confirm the transcriptional regulation of SP1 on SNHG1. RESULTS: Elevated SNHG1 and decreased miR-154-5p were observed in both in vivo mouse model and in vitro cell model of epilepsy. Knockdown of SNHG1 or transfection with miR-154-5p mimics significantly ameliorated Mg2+ free-induced neuronal injury in SH-SY5Y cells. SNHG1 acted as a sponge of miR-154-5p. Moreover, SNHG1 promoted neuronal injury via acting as a miR-154-5p sponge to disinhibit TLR5. Additionally, SP1 activated the transcriptional activity of SNHG1. CONCLUSION: In summary, SP1 transcriptionally activated-SNHG1 contributes to the development of epilepsy via directly regulating miR-154-5p/TLR5 axis, which provides novel targets in treatment of epilepsy.


Subject(s)
Immunoglobulins/metabolism , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Sp1 Transcription Factor/genetics , Toll-Like Receptor 5/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/physiology , Epilepsy/genetics , Gene Expression Regulation/genetics , Male , Mice , RNA, Long Noncoding/metabolism , Sp1 Transcription Factor/metabolism , Toll-Like Receptor 5/genetics
2.
Neurochem Res ; 45(4): 783-795, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32056051

ABSTRACT

Although many advances have been made in the pathogenesis of epilepsy recently, the pathological mechanisms of epilepsy are still largely unknown. Exploring the pathological mechanisms and developing novel therapeutic strategies for epilepsy are urgently needed. A SD rat model of epilepsy was established with lithium chloride-pilocarpine. Astrocytes were isolated, cultured from 8 to 12 week rats and identified by flow cytometry and immunofluorescence. Immunohistochemical staining was used for MEF2C and NF-κB in paraffin-embedded sections. RT-qPCR and western blot were used to analyze gene expression. ELISA was used to analyze the concentration of IL-6, TNF-α and Cox-2. Cells were transfected with pcDNA-MEFC2, sh-MEFC2, pcDNA-UCA1, sh-UCA1, miR-203 mimic or miR-203 inhibitor. Cell viability was assessed by MTT assay. Dual luciferase assay was used to determine the direct interaction of lncRNA UCA1/miR-203 and miR-203/MEF2C. MEF2C was down-regulated and inhibited NF-κB expression and the secretion of IL-6 and TNF-α in epilepsy. LncRNA UCA1 was also down-regulated in epilepsy. LncRNA UCA1 over-expression increased the expression of MEF2C and its knock-down decreased MEF2C expression. Luciferase activity showed lncRNA UCA1 directly targeted miR-203 and miR-203 directly targeted MEF2C. MiR-203 suppressed the expression of MEF2C, and promoted NF-κB, phosphorylated IκB/IKK and inflammatory effectors, which was reversed by MEF2C knock-down. Moreover, lncRNA UCA1 could increase the expression of MEF2C to inhibit NF-κB, phosphorylated IκB/IKK and inflammatory effectors, which was also reversed by miR-203 mimic transfection. LncRNA UCA1 inhibited the inflammation via regulating miR-203 mediated regulation of MEF2C/NF-κB signaling in epilepsy. Our investigation elucidated novel pathological mechanisms and provided potential therapeutic targets for epilepsy.


Subject(s)
Epilepsy/metabolism , Inflammation/metabolism , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Signal Transduction/physiology , Animals , Gene Knockdown Techniques , HEK293 Cells , Humans , MEF2 Transcription Factors/metabolism , NF-kappa B/metabolism , RNA, Long Noncoding/genetics , Rats, Sprague-Dawley
3.
Environ Toxicol ; 34(6): 699-707, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30835941

ABSTRACT

BACKGROUND: The insecticide exposure has been linked to Parkinson's disease (PD). In the present study, we used a most widely used cell line in study of PD, the SH-SY5Y cells, to investigate mechanisms of chlorpyrifos (CPF) induced cell toxicity and the possible roles of cell pyroptosis and oxidative stress in SH-SY5Y cells, as well as role of miR-181/SIRT1/PGC-1α/Nrf2 signaling pathway in this process. METHODS: SH-SY5Y cells were treated with different concentrations of CPF. Cell viability was measured using CCK-8 assay. Cell pyroptosis was determined by immunofluorescence of caspase-1 and TUNEL assay. The miR-181 (has-miR-181-5p) level was determined by qRT-PCR. Expression of SIRT1, PGC-1α, Nrf2, and pyroptosis related proteins NLRP3, caspase-1, IL-1ß, and IL-18 was determined by both qRT-PCR and Western blotting. RESULTS: Cell viability was found to be decreased with the increased CPF concentrations. The pyroptosis related proteins, ROS levels, as well as level of caspase-1 and the TUNEL positive cells were all significantly up-regulated by CPF. Meanwhile, expression of miR-181 and pyroptosis proteins was also enhanced, while the SIRT1/PGC-1α/Nrf2 signaling was inhibited by CPF. Knockdown of Nrf2 significantly up-regulated the expression of pyroptosis related proteins, ROS level, caspase-1, and the TUNEL positive cells, while over-expression of Nrf2 resulted in opposite results. The expression of PGC-1α and Nrf2 was significantly down-regulated when SIRT1 was inhibited, while over-expressed SIRT1 led to increased PGC-1α and Nrf2 levels. Besides, miR-181 promoted the CPF induced activation of pyroptosis and oxidative stress, as well as down-regulated SIRT1/PGC-1α/Nrf2 signaling, while inhibition of miR-181 led to opposite results. CONCLUSIONS: Chlorpyrifos could inhibit cell proliferation, activate cell pyroptosis and increase susceptibility on oxidative stress-induced toxicity by elevating miR-181 through down-regulation of the SIRT1/PGC-1α/Nrf2 pathway in human neuroblastoma SH-SY5Y cells. This study might give deeper insights for mechanisms of CPF induced toxicity and might give some novel research targets for PD treatment.


Subject(s)
Chlorpyrifos/toxicity , Insecticides/toxicity , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Parkinson Disease, Secondary/chemically induced , Pyroptosis/drug effects , Signal Transduction/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Parkinson Disease, Secondary/metabolism , Parkinson Disease, Secondary/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...