Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Language
Publication year range
1.
Mikrochim Acta ; 191(6): 325, 2024 05 13.
Article in English | MEDLINE | ID: mdl-38739279

ABSTRACT

Glial fibrillary acidic protein (GFAP) in serum has been shown as a biomarker of traumatic brain injury (TBI) which is a significant global public health concern. Accurate and rapid detection of serum GFAP is critical for TBI diagnosis. In this study, a time-resolved fluorescence immunochromatographic test strip (TRFIS) was proposed for the quantitative detection of serum GFAP. This TRFIS possessed excellent linearity ranging from 0.05 to 2.5 ng/mL for the detection of serum GFAP and displayed good linearity (Y = 598723X + 797198, R2 = 0.99), with the lowest detection limit of 16 pg/mL. This TRFIS allowed for quantitative detection of serum GFAP within 15 min and showed high specificity. The intra-batch coefficient of variation (CV) and the inter-batch CV were both < 4.0%. Additionally, this TRFIS was applied to detect GFAP in the serum samples from healthy donors and patients with cerebral hemorrhage, and the results of TRFIS could efficiently discern the patients with cerebral hemorrhage from the healthy donors. Our developed TRFIS has the characteristics of high sensitivity, high accuracy, and a wide linear range and is suitable for rapid and quantitative determination of serum GFAP on-site.


Subject(s)
Chromatography, Affinity , Glial Fibrillary Acidic Protein , Humans , Biomarkers/blood , Cerebral Hemorrhage/blood , Cerebral Hemorrhage/diagnosis , Chromatography, Affinity/methods , Glial Fibrillary Acidic Protein/blood , Limit of Detection , Reagent Strips
2.
Comb Chem High Throughput Screen ; 24(7): 947-956, 2021.
Article in English | MEDLINE | ID: mdl-32819229

ABSTRACT

AIM AND OBJECTIVE: Cell death is a main pathological change in brain ischemia. Astragalus membranaceus (Ast) and ligustrazine (Lig), as traditional Chinese herbs, have a protective effect against ischemia-reperfusion injury. We aim to find whether the underlying protective mechanism of Astragalus membranaceus and ligustrazine against Oxygen-glucose deprivation/reoxygenation (OGD/R) -induced injury in RBMECs is related to PKCδ/MARCKS pathway. MATERIALS AND METHODS: OGD/R preconditioning was instituted in rat brain microvascular endothelial cells (RBMECs). The survival and apoptosis of RBMECs were detected by a Cell Counting Kit-8 and TUNEL staining; PKCδ/MARCKS and MMP9 expression were examined by immunofluorescence, western blot and quantitative real-time PCR. RESULTS: OGD/R stimulation significantly increased RBMEC apoptosis, whereas Ast+Lig, Rottlerin or Ast+Lig+Rottlerin treatment evidently reduced cellular apoptosis and increased cell viability (P <0.05). Furthermore, Ast+Lig, Rottlerin or Ast+Lig+Rottlerin treatment significantly reduced mRNA expression levels of PKCδ/MARCKS and MMP9 (P <0.05), compared to OGD/R control group. Moreover, Ast+Lig, Rottlerin or Ast+Lig+Rottlerin treatment evidently reduced protein expression levels of PKCδ, MMP9, and MARCKS (P <0.05), compared to OGD/R control group, detected by western blotting or immunofluorescence. CONCLUSION: The administration of Astragalus membranaceus and ligustrazine protected RBMECs against OGD/R-induced apoptosis. PKCδ/MARCKS and MMP9 expression were significantly increased after OGD/R stimulation, while Astragalus membranaceus and ligustrazine treatment evidently suppressed. Collectively, Astragalus membranaceus and ligustrazine play protective effects against OGD/R-induced injury in RBMECs through regulating PKCδ/MARCKS pathway to inhibit MMP9 activation.


Subject(s)
Astragalus propinquus/chemistry , Brain/drug effects , Drugs, Chinese Herbal/pharmacology , Endothelial Cells/drug effects , Protective Agents/pharmacology , Pyrazines/pharmacology , Animals , Brain/metabolism , Brain/pathology , Cells, Cultured , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/isolation & purification , Endothelial Cells/metabolism , Endothelial Cells/pathology , Glucose/metabolism , Medicine, Chinese Traditional , Myristoylated Alanine-Rich C Kinase Substrate/antagonists & inhibitors , Myristoylated Alanine-Rich C Kinase Substrate/metabolism , Oxygen/metabolism , Protective Agents/chemistry , Protective Agents/isolation & purification , Protein Kinase C-delta/antagonists & inhibitors , Protein Kinase C-delta/metabolism , Pyrazines/chemistry , Rats
3.
Exp Cell Res ; 396(2): 112332, 2020 11 15.
Article in English | MEDLINE | ID: mdl-33065113

ABSTRACT

DEAD-box (DDX) helicases are critical for recognizing viral nucleic acids to regulate antiviral innate immunity. Although DDX5 has been reported to participate in various virus infection, whether DDX5 regulates innate immune responses and its underlying mechanisms are still unknown. Here, we report that DDX5 is a negative regulator of type I IFN (IFN-I) production in antiviral responses. DDX5 knockdown significantly promoted DNA or RNA virus infection-induced IFN-I production and IFN-stimulated genes (ISGs) expression, while ectopic expression of DDX5 inhibited IFN-I production and promoted viral replication. Furthermore, we found that DDX5 specifically interacted with serine/threonine-protein phosphatase 2 A catalytic subunit beta (PP2A-Cß) and viral infection enhanced the interaction between DDX5 and PP2A-Cß. Besides, PP2A-Cß interacted with IFN regulatory factor 3 (IRF3), and PP2A-Cß knockdown promoted viral infection-induced IRF3 phosphorylation and IFN-I production. In addition, DDX5 knockdown rendered the mice more resistant to viral infection and enhanced antiviral innate immunity in vivo. Thus, DDX5 suppresses IFN-I antiviral innate immune response by interacting with PP2A-Cß to deactivate IRF3. Together, these findings identify a negative role of DDX5 on regulating IFN-I signaling in innate immune responses.


Subject(s)
DEAD-box RNA Helicases/metabolism , Immunity, Innate , Interferon Regulatory Factor-3/metabolism , Interferon Type I/metabolism , Protein Phosphatase 2/metabolism , Animals , Female , Gene Knockdown Techniques , Mice, Inbred C57BL , Phosphorylation , Protein Binding , Up-Regulation/genetics , Virus Diseases/metabolism , Virus Diseases/pathology
4.
J Stroke Cerebrovasc Dis ; 29(10): 105126, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32912499

ABSTRACT

BACKGROUND: Long non-coding RNAs (LncRNAs) have been reported to play important roles in the pathogenesis and development of many diseases, including cerebral ischemia and reperfusion (I/R) injury. In this study, we aimed to investigate the role of LncRNA-Potassium Voltage-Gated Channel Subfamily Q Member 1 opposite strand/antisense transcript 1 (KCNQ1OT1) in cerebral I/R induced neuronal injury, and its underlying mechanisms. METHODS: Primary mouse cerebral cortical neurons treated with oxygen-glucose deprivation and reoxygenation (OGD/R) in vitro and mice subjected to middle cerebral artery occlusion (MCAO) and reperfusion were used to mimic cerebral I/R injury. Small inference RNA (siRNA) was used to knockdown KCNQ1OT1 or microRNA-153-3p (miR-153-3p). Dual-luciferase assay was performed to detect the interaction between KCNQ1OT1 and miR-153-3p and interaction between miR-153-3p and Fork head box O3a (Foxo3). Flow cytometry analysis was performed to detect neuronal apoptosis. qRT-PCR and Western blotting were performed to detect RNA and protein expressions. RESULTS: KCNQ1OT1 and Foxo3 expressions were significantly increased in neurons subjected to I/R injury in vitro and in vivo, and miR-153-3p expression were significantly decreased. Knockdown of KCNQ1OT1 or overexpression of miR-153-3p weakened OGD/R-induced neuronal injury and regulated Foxo3 expressions. Dual-luciferase analysis showed that KCNQ1OT1 directly interacted with miR-153-3p and Foxo3 is a direct target of miR-153-3p. CONCLUSIONS: Our results indicate that LncRNA-KCNQ1OT1 promotes OGD/R-induced neuronal injury at least partially through acting as a competing endogenous RNA (ceRNA) for miR-153-3p to regulate Foxo3a expression, suggesting LncRNA-KCNQ1OT1 as a potential therapeutic target for cerebral I/R injury.


Subject(s)
Cerebral Cortex/metabolism , Forkhead Box Protein O3/metabolism , Infarction, Middle Cerebral Artery/therapy , MicroRNAs/metabolism , Neurons/metabolism , RNA, Long Noncoding/metabolism , Reperfusion Injury/metabolism , Reperfusion/adverse effects , Animals , Cell Hypoxia , Cells, Cultured , Cerebral Cortex/pathology , Forkhead Box Protein O3/genetics , Gene Expression Regulation , Glucose/deficiency , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/pathology , Male , Mice, Inbred C57BL , MicroRNAs/genetics , Neurons/pathology , RNA, Long Noncoding/genetics , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Signal Transduction
5.
Int Immunopharmacol ; 88: 106937, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32890792

ABSTRACT

OBJECTIVE: Ischemic stroke is one of the leading causes of death globally, and inflammation is considered as a vital contributor to the pathophysiology of ischemic stroke. Recently, microRNA-421-3p-derived macrophages is found to promote motor function recovery in spinal cord injury. Here, we explored whether microRNA-421-3p is involved in inflammation responses during cerebral ischemia/reperfusion (I/R) injury and its molecular mechanism. METHODS: An in vivo experimental animal model of intraluminal middle cerebral artery occlusion/reperfusion (MCAO/R) and in vitro model of microglial subjected to oxygen-glucose deprivation and reoxygenation (OGD/R) were used. The effects of microRNA-421-3p on cerebral I/R injury and its underlying mechanism were detected by quantitative real-time PCR, western blotting, immunofluorescence staining, RNA immunoprecipitation, flow cytometry, luciferase reporter assay, and bioinformatics analysis. RESULTS: We find that microRNA-421-3p is significantly decreased in cerebral I/R injury in vitro and in vivo. Furthermore, overexpression of microRNA-421-3p evidently suppresses pro-inflammatory factor expressions and inhibits NF-κB p65 protein expression and nuclear translocation in BV2 microglia cells treated with OGD/R. However, microRNA-421-3p neither promotes p65 mRNA expression, nor affects p65 mRNA or protein stability. Moreover, we find the m6A 'reader' protein YTH domain family protein 1 (YTHDF1) is the specific target of microRNA-421-3p, and YTHDF1 specifically binds to the m6a site of p65 mRNA to promote its translation. CONCLUSION: microRNA-421-3p prevents inflammatory response in cerebral ischemia/reperfusion injury through targeting YTHDF1 to inhibit p65 mRNA translation. These findings provide novel insights into understanding the molecular pathogenesis of cerebral I/R injury.


Subject(s)
Infarction, Middle Cerebral Artery/genetics , MicroRNAs , RNA-Binding Proteins/genetics , Reperfusion Injury/genetics , Transcription Factor RelA/genetics , Animals , Cell Line , Cytokines/genetics , Infarction, Middle Cerebral Artery/metabolism , Inflammation/genetics , Inflammation/metabolism , Male , Methyltransferases , Mice, Inbred C57BL , Protein Biosynthesis , RNA-Binding Proteins/metabolism , Reperfusion Injury/metabolism , Transcription Factor RelA/metabolism
6.
Cell Transplant ; 29: 963689720946020, 2020.
Article in English | MEDLINE | ID: mdl-32749163

ABSTRACT

Astragalus membranaceus (Ast) and ligustrazine (Lig) have a protective effect on lower hemorrhagic transformation induced by pharmaceutical thrombolysis. The cerebral ischemia rat model was induced with autologous blood clot injections. A combination of Ast and Lig, or a protein kinase C delta (PKCδ) inhibitor-rottlerin, or a combination of Ast, Lig, and rottlerin was administered immediately after recombinant tissue plasminogen activator injection. The cerebral infarct area, neurological deficits, cerebral hemorrhage status, neuronal damage and tight junctions' changes in cerebral vessels, and the messenger RNA and protein levels of PKCδ, myristoylated alanine-rich C kinase substrate (Marcks), and matrix metallopeptidase 9 (MMP9) were determined after 3 h and 24 h of thrombolysis. The ultrastructure of the neuronal damage and tight junctions was examined under a transmission electron microscope. The expression levels of PKCδ, Marcks, and MMP9 were assessed by immunohistochemistry, western blot, and quantitative real-time polymerase chain reaction . Administration of Ast and Lig not only significantly decreased neurological deficit scores, infarct volumes, and cerebral hemorrhage but also inhibited the disruption due to neuronal dysfunction and the tight junction integrity in the cerebral vessel. Treatment with a combination of Ast and Lig effectively protected ischemia-induced microhemorrhage transformation through PKCδ/Marcks pathway suppression.


Subject(s)
Astragalus propinquus/chemistry , Cerebral Hemorrhage/drug therapy , Pyrazines/therapeutic use , Vasodilator Agents/therapeutic use , Animals , Brain Ischemia , Cerebral Infarction , Male , Rats , Rats, Sprague-Dawley
7.
Int J Immunopathol Pharmacol ; 33: 2058738419869055, 2019.
Article in English | MEDLINE | ID: mdl-31409163

ABSTRACT

The purpose of the study was to evaluate the effect of Astragalus membranaceus extract and ligustrazine combination on ameliorating inflammation in cerebral ischemic rats that have undergone thrombolysis. Astragalus membranaceus and ligustrazine per se, or a combination of A. membranaceus and ligustrazine was administered by intraperitoneal injection immediately after surgery and sham surgery. After the induction of thrombolysis, the neurological function was measured and cerebral lesion volume was determined. The regulatory T cells in the spleen were measured by flow cytometry. To explore the protective effects of the combination drug on the neurological function and inflammation, the expression of transcription factor Foxp3 and cytokines, including transforming growth factor beta 1, interleukin 10, interleukin 4, interleukin 1 beta, interferon gamma, interleukin 17, in damaged brain was examined using reverse transcription polymerase chain reaction, Western blot, and enzyme-linked immunosorbent assay. The cerebral lesion volume was markedly reduced in the combination drug-treated rats compared to the rats treated with either A. membranaceus or ligustrazine alone (P < 0.05). The neurological function, regulatory T cells, transcription factor Foxp3, transforming growth factor beta 1, interleukin 10, and interleukin 4 were markedly elevated in the rats treated with combination drugs (P < 0.05). The expression of interleukin 1 beta, interferon gamma, and interleukin 17 was reduced in the rats treated with combination drug therapy (P < 0.05). Treatment with a combination of A. membranaceus and ligustrazine can ameliorate inflammation after thrombolysis and regulate the related cytokines by elevating the expression of endogenous regulatory T cells.


Subject(s)
Astragalus propinquus/chemistry , Brain Ischemia/drug therapy , Cerebral Infarction/drug therapy , Fibrinolytic Agents/pharmacology , Inflammation/drug therapy , Plant Extracts/pharmacology , Pyrazines/pharmacology , Animals , Brain Ischemia/metabolism , Cerebral Infarction/metabolism , Cytokines/metabolism , Disease Models, Animal , Drug Therapy, Combination/methods , Inflammation/metabolism , Male , Rats , Rats, Wistar , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism
8.
Neuropharmacology ; 158: 107682, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31278927

ABSTRACT

Yin-Yang 1 (YY1) has been identified as playing critical roles in multiple diseases. However, little is known regarding its roles and mechanisms in cerebral ischemia/reperfusion (I/R) injury. This study is aimed to explore the roles of YY1 in regulating neuronal apoptosis in cerebral I/R injury and its underlying mechanisms. Primary mouse cerebral cortical neurons were isolated and subjected to OGD/R to mimic cerebral I/R injury in vitro. The roles of YY1 on OGD/R-induced neuronal injury were investigated by performing western blotting, quantitative real-time polymerase chain reaction, TUNEL, RNA-binding protein immunoprecipitation, chromatin immunoprecipitation, chromatin isolation by RNA purification assay, glucose uptake assay, lactate production assay, and extracellular acidification rate assay. YY1-binding long non-coding RNAs (LncRNAs) in neurons subjected to OGD/R were identified by RIP and RNA sequencing. The roles of YY1 on cerebral I/R in vivo were detected by assessing neuronbehaviour, infarct size, and neuronal apoptosis. We found that YY1 expression is downregulated, and LncRNA GAS5 is upregulated in neurons subjected to OGD/R. OGD/R treatment promotes YY1 interacting with GAS5 in neurons, and YY1 negatively regulates GAS5 expression by binding to GAS5 promoter to repress its transcription. Besides, YY1 and GAS5 bind to the same region of PFKFB3 promoter to promote PFKFB3 expression and strengthen neuronal glycolysis, resulting in aggravating OGD/R-induced neuronal apoptosis. Knockdown of YY1 or GAS5 protects against I/R-induced ischemic brain damage and improves overall neurological functions in vivo. Overall, YY1 interacts with LncRNA GAS5 to promote PFKFB3 transcription to enhance neuronal glycolysis, resulting in aggravating cerebral I/R injury.


Subject(s)
Brain Ischemia/genetics , Glucose/metabolism , Glycolysis/genetics , Neurons/metabolism , Phosphofructokinase-2/genetics , RNA, Long Noncoding/genetics , Reperfusion Injury/genetics , YY1 Transcription Factor/genetics , Animals , Apoptosis/genetics , Brain Ischemia/metabolism , Cerebral Cortex/cytology , Chromatin Immunoprecipitation , Immunoprecipitation , In Situ Nick-End Labeling , Male , Mice , Primary Cell Culture , RNA, Long Noncoding/metabolism , Real-Time Polymerase Chain Reaction , Reperfusion Injury/metabolism , Up-Regulation , YY1 Transcription Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL