Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
EClinicalMedicine ; 65: 102305, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37965431

ABSTRACT

Background: Glibenclamide alleviates brain edema and improves neurological outcomes in experimental models of stroke. We aimed to assess whether glibenclamide improves functional outcomes in patients with acute ischemic stroke treated with recombinant tissue plasminogen activator (rtPA). Methods: In this randomized, double-blind, placebo-controlled trial, patients with acute ischemic stroke were recruited to eight academic hospitals in China. Patients were eligible if they were aged 18-74 years, presented with a symptomatic anterior circulation occlusion with a deficit on the NIHSS of 4-25, and had been treated with rtPA within 4.5 h of symptom onset. We used web-based randomization (1:1) to allocate eligible participants to the glibenclamide or placebo group, stratified according to endovascular treatment and baseline stroke severity. Glibenclamide or placebo was taken orally or via tube feeding at a loading dose of 1.25 mg within 10 h after symptom onset, followed by 0.625 mg every 8 h for 5 days. The primary outcome was the proportion of patients with good outcomes (modified Rankin Scale of 0-2) at 90 days, assessed in all randomly assigned patients who had been correctly diagnosed and had begun study medication. The study is registered with ClinicalTrials.gov, NCT03284463, and is closed to new participants. Findings: Between January 1, 2018, and May 28, 2022, 305 patients were randomly assigned, of whom 272 (142 received glibenclamide and 130 received placebo) were included in the primary efficacy analysis. 103 (73%) patients in the glibenclamide group and 94 (72%) in the placebo group had a good outcome (adjusted risk difference 0.002, 95% CI -0.098 to 0.103; p = 0.96). 12 (8%) patients allocated to glibenclamide and seven (5%) patients allocated to placebo died from any cause at 90 days (p = 0.35). The number and type of adverse events were similar between the two groups. There were no drug-related adverse events and no drug-related deaths. Interpretation: The addition of glibenclamide to thrombolytic therapy did not increase the proportion of patients who achieved good outcomes after stroke compared with placebo, but it did not lead to any safety concerns. Funding: Southern Medical University and Nanfang Hospital.

2.
Comput Struct Biotechnol J ; 21: 2809-2823, 2023.
Article in English | MEDLINE | ID: mdl-37206617

ABSTRACT

Stroke is the leading cause of death and disability worldwide, with a growing number of incidences in developing countries. However, there are currently few medical therapies for this disease. Emerged as an effective drug discovery strategy, drug repurposing which owns lower cost and shorter time, is able to identify new indications from existing drugs. In this study, we aimed at identifying potential drug candidates for stroke via computationally repurposing approved drugs from Drugbank database. We first developed a drug-target network of approved drugs, employed network-based approach to repurpose these drugs, and altogether identified 185 drug candidates for stroke. To validate the prediction accuracy of our network-based approach, we next systematically searched for previous literature, and found 68 out of 185 drug candidates (36.8 %) exerted therapeutic effects on stroke. We further selected several potential drug candidates with confirmed neuroprotective effects for testing their anti-stroke activity. Six drugs, including cinnarizine, orphenadrine, phenelzine, ketotifen, diclofenac and omeprazole, have exhibited good activity on oxygen-glucose deprivation/reoxygenation (OGD/R) induced BV2 cells. Finally, we showcased the anti-stroke mechanism of actions of cinnarizine and phenelzine via western blot and Olink inflammation panel. Experimental results revealed that they both played anti-stroke effects in the OGD/R induced BV2 cells via inhibiting the expressions of IL-6 and COX-2. In summary, this study provides efficient network-based methodologies for in silico identification of drug candidates toward stroke.

3.
Neuroreport ; 34(9): 463-470, 2023 06 07.
Article in English | MEDLINE | ID: mdl-37161987

ABSTRACT

Emerging evidence has proved that inflammatory responses aggravate the pathological progression of Parkinson's disease. This study aimed to identify the role of Interleukin-1 receptor-associated kinase-M (IRAK-M) as an important negative regulator of innate immunity, in the pathological progression of Parkinson's disease. In the present study, a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) injection was administered to prepare the acute and sub-acute Parkinson's disease mouse models. Western blot analysis was utilized to examine the protein expressions of tyrosine hydroxylase and IRAK-M. The mRNA expression levels of IRAK-M, interleukin (IL)-6, IL-ß, and cyclooxygenase-2 were evaluated via using reverse transcription quantitative PCR (RT-qPCR). The expression of tyrosine hydroxylase-positive neurons in corpus striatum and substantia nigra pars compacta (SNc) tissues was detected using immunohistochemistry. The results showed that the protein and mRNA levels of IRAK-M were considerably upregulated in corpus striatum and SNc tissues in the sub-acute Parkinson's disease model. Furthermore, IRAK-M knockout significantly enhanced the MPTP-induced loss of tyrosine hydroxylase-positive fibers in corpus striatum and tyrosine hydroxylase-positive neurons in SNc, and intensified the effect of MPTP on the activation of microglial cells and the expression of inflammatory cytokines. In addition, sub-acute Parkinson's disease mice with IRAK-M deletion exhibited worse motor abilities than those of wild-type littermates. Overall, the present study suggested that IRAK-M reduces dopaminergic neuron damage in sub-acute Parkinson's disease by suppressing inflammation, which may provide a new therapeutic target for Parkinson's disease treatment.


Subject(s)
Dopaminergic Neurons , Parkinson Disease , Animals , Mice , Interleukin-1 Receptor-Associated Kinases , Tyrosine 3-Monooxygenase , Blotting, Western , Disease Models, Animal , Interleukin-6
4.
Mol Med Rep ; 27(4)2023 04.
Article in English | MEDLINE | ID: mdl-36866740

ABSTRACT

Blood­brain barrier (BBB) damage is one of the main causes of poor outcomes and increased mortality rates following cerebral ischemia­reperfusion injury. Apolipoprotein E (ApoE) and its mimetic peptide have been previously reported to exhibit potent neuroprotective properties in various central nervous system disease models. Therefore, the present study aimed to investigate the possible role of the ApoE mimetic peptide COG1410 in cerebral ischemia­reperfusion injury and its potential underlying mechanism. Male SD rats were subjected to 2 h middle cerebral artery occlusion followed by 22 h reperfusion. Evans blue leakage and IgG extravasation assays results revealed that COG1410 treatment significantly reduced BBB permeability. In addition, in situ zymography and western blotting were used to prove that COG1410 was able to downregulate the activities of MMPs and upregulate the expression of occludin in the ischemic brain tissue samples. Subsequently, COG1410 was found to significantly reverse microglia activation while also suppressing inflammatory cytokine production, according to immunofluorescence signal of Iba­1 and CD68 and protein expression of COX­2. Consequently, this neuroprotective mechanism mediated by COG1410 was further tested using the BV2 cell line in vitro, which was exposed to oxygen glucose deprivation followed by reoxygenation. The mechanism of COG1410 was found to be mediated, as least partly, through the activation of triggering receptor expressed on myeloid cells 2. In conclusion, the data suggest that COG1410 can alleviate BBB injury and neuroinflammation following ischemic stroke.


Subject(s)
Blood-Brain Barrier , Ischemic Stroke , Male , Animals , Rats , Rats, Sprague-Dawley , Apolipoproteins E
5.
J Mol Histol ; 52(5): 943-953, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34403009

ABSTRACT

The pathophysiological mechanism of carotid atherosclerosis (CAS) involves endothelial cell dysfunction, vascular smooth muscle cells (VSMCs), and macrophage activation, which ultimately leads to fibrosis of the vessel wall. lncRNA works weightily in the formation of CAS, but the function and mechanism of lncRNA LINC01123 in stable plaque formation are still equivocal. We collected blood samples from 35 CAS patients as well as 33 healthy volunteers. VSMCs treated with oxidized low-density lipoprotein (ox-LDL) were utilized as the CAS cell models. We applied qRT-PCR for detecting LINC01123, miR-1277-5p and KLF5 mRNA expression, CCK-8 method and BrdU test for determining cell proliferation, Transwell test for measuring cell migration, as well as Western blot for assaying KLF5 protein expression. Dual-luciferase reporter experiment was adopted for assessing the interaction between LINC01123 and miR-1277-5p, as well as KLF5 and miR-1277-5p. LINC01123 and KLF5 expression were dramatically up-regulated, while miR-1277-5p expression was down-regulated in CAS patients and ox-LDL-induced CAS cell models. Overexpressed LINC01123 notedly promoted VSMCs migration and proliferation. LINC01123 knockdown repressed cell proliferation and migration. Also, LINC01123 targeted miR-1277-5p and down-regulated its expression, while miR-1277-5p could negatively regulate KLF5 expression. LINC01123 is highly expressed in CAS patients, and promotes cell proliferation and migration via regulating miR-1277-5p/KLF5 axis in ox-LDL-induced VSMCs. It might be involved in the fibrous plaque formation.


Subject(s)
Cell Movement/genetics , Kruppel-Like Transcription Factors/metabolism , Lipoproteins, LDL/pharmacology , MicroRNAs/genetics , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , RNA, Long Noncoding/metabolism , Signal Transduction , Base Sequence , Carotid Artery Diseases/blood , Carotid Artery Diseases/genetics , Carotid Artery Diseases/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Proliferation/genetics , Female , Humans , Male , MicroRNAs/metabolism , Middle Aged , Myocytes, Smooth Muscle/drug effects , RNA, Long Noncoding/blood , RNA, Long Noncoding/genetics , Signal Transduction/drug effects , Up-Regulation/drug effects , Up-Regulation/genetics
6.
Drug Des Devel Ther ; 15: 2385-2399, 2021.
Article in English | MEDLINE | ID: mdl-34103899

ABSTRACT

PURPOSE: Many researches have investigated the functions of tetramethylpyrazine (TMP) in Alzheimer's disease (AD). This study aimed to discuss the underlying mechanism of TMP in AD mice. METHODS: TMP (200 mg/kg) was administered to 6-month-old APP/PS1 transgenic mice, and behavioral changes and hippocampal nerve injury in AD mice were detected. Apoptosis and autophagy-related protein levels were detected. Changes in gene expression before and after TMP treatment were compared using transcriptome sequencing. The effects of Cullin 4B (CUL4B) overexpression and somatostatin receptor 4 (SSTR4) silencing on AD symptoms and SSTR4 ubiquitination in APP/PS1 mice were observed. SH-SY5Y and PC12 cells were treated with 25 µmol/L Aß25-35 and TMP to observe cell viability, apoptosis, and autophagy. Cell viability and apoptosis were measured again after treatment with proteasome inhibitor MG132 or lysosomal inhibitor 3-mA. RESULTS: TMP treatment improved the behavioral cognition of APP/PS1 mice and improved the neuronal apoptosis and damage in brain tissue. CUL4B was significantly upregulated in APP/PS1 mouse brain tissue, and SSRT4 protein was downregulated, and the levels of CUL4B and SSRT4 were negatively correlated. TMP treatment downregulated CUL4B, inhibited SSRT4 ubiquitination and upregulated SSRT4 protein level in APP/PS1 mouse brain tissue, while CUL4B overexpression or SSRT4 silencing reversed the effect of TMP. TMP and MG132 improved the decreased activity, increased apoptosis and increased SSRT4 protein in SH-SY5Y and PC12 cells treated with Aß25-35, but not 3-mA. CUL4B overexpression promoted the ubiquitination of SSTR4 in cells, which partially reversed the effect of TMP. CONCLUSION: TMP could improve the cognitive ability of AD mice by inhibiting CUL4B expression and the ubiquitination degradation of SSTR, and alleviating neuronal apoptosis and injury. This study may offer a new therapeutic option for AD treatment.


Subject(s)
Alzheimer Disease/drug therapy , Pyrazines/pharmacology , Receptors, Somatostatin/antagonists & inhibitors , Alzheimer Disease/metabolism , Animals , Cognition/drug effects , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Rats , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism , Tumor Cells, Cultured , Ubiquitination/drug effects
7.
BMC Neurol ; 20(1): 239, 2020 Jun 11.
Article in English | MEDLINE | ID: mdl-32527232

ABSTRACT

BACKGROUND: Thrombolysis with recombinant tissue plasminogen activator (rtPA) improves outcome for patients with acute ischemic stroke (AIS), but many of them still have substantial disability. Glibenclamide (US adopted name, glyburide), a long-acting sulfonylurea, shows promising result in treating AIS from both preclinical and clinical studies. This study investigates the safety and efficacy of glibenclamide combined with rtPA in treating AIS patients. METHODS: This is a prospective, randomized, double-blind, placebo-controlled, multicenter trial with an estimated sample size of 306 cases, starting in January 2018. Patients aged 18 to 74 years, presented with a symptomatic anterior circulation occlusion with a deficit on the NIHSS of 4 to 25 points and treated with intravenous rtPA within the first 4.5 h of their clinical onsets, are eligible for participation in this study. The target time from the onset of symptoms to receive the study drug is of 10 h. Subjects are randomized 1: 1 to receive glibenclamide or placebo with a loading dose of 1.25 mg, followed by 0.625 mg every 8 h for total 5 days. The primary efficacy endpoint is 90-day good outcome, measured as modified Rankin Scale of 0 to 2. Safety outcomes are all-cause 30-day mortality and early neurological deterioration, with a focus on cardiac- and glucose-related serious adverse events. DISCUSSION: This study will provide valuable information about the safety and efficacy of oral glibenclamide for AIS patients treated with rtPA. This would bring benefits to a large number of patients if the agent is proved to be effective. TRIAL REGISTRATION: The trial was registered on September 14th 2017 at www.clinicaltrials.gov having identifier NCT03284463. Registration was performed before recruitment was initiated.


Subject(s)
Glyburide/therapeutic use , Randomized Controlled Trials as Topic , Stroke/drug therapy , Tissue Plasminogen Activator/therapeutic use , Adolescent , Adult , Aged , Brain Ischemia/complications , Constriction, Pathologic , Double-Blind Method , Female , Fibrinolytic Agents/therapeutic use , Humans , Male , Middle Aged , Multicenter Studies as Topic , Prospective Studies , Stroke/etiology , Treatment Outcome , Young Adult
8.
IUBMB Life ; 71(10): 1515-1521, 2019 10.
Article in English | MEDLINE | ID: mdl-31290617

ABSTRACT

Mitochondrial dysfunction has been associated with the pathogenesis of a variety of neurodegenerative diseases. Sitagliptin is a dipeptidyl-peptidase-4 (DPP-4) inhibitor that has been approved for the treatment of type 2 diabetes (T2DM). In the current study, we report that sitagliptin increased the expression of PGC-1α, NRF1, and TFAM in human SH-SY5Y neuronal cells. Notably, our data indicate that sitagliptin promoted mitochondrial biogenesis by increasing the amount of mtDNA, the levels of mitochondria-related genes such as TOMM20, TOMM40, TIMM9, NDUFS3, ATP5C1, and the expression of oxidative phosphorylation subunits complex I and complex IV. Additionally, we found that sitagliptin induced a "gain of mitochondrial function" in SH-SY5Y cells by increasing the mitochondrial respiratory rate and adenosine triphosphate (ATP) production. Significantly, our results demonstrate that sitagliptin activated the transcriptional factor CREB by inducing its phosphorylation at Ser133. Inhibition of CREB using its specific inhibitor H89 abolished the effects of sitagliptin on the expression of PGC-1α, NRF1, and TFAM, as well as an increase in mtDNA amount and ATP production. These findings suggest that sitagliptin could become a potential agent for the treatment of neurological disorders.


Subject(s)
DNA-Binding Proteins/genetics , Mitochondrial Proteins/genetics , Nervous System Diseases/genetics , Nuclear Respiratory Factor 1/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Sitagliptin Phosphate/pharmacology , Transcription Factors/genetics , Adenosine Triphosphate/genetics , Animals , Cell Line , Cyclic AMP Response Element-Binding Protein/genetics , Diabetes Mellitus, Type 2/drug therapy , Dipeptidyl Peptidase 4/genetics , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Humans , Isoquinolines/pharmacology , Mitochondria/drug effects , Mitochondria/genetics , Nervous System Diseases/drug therapy , Nervous System Diseases/pathology , Organelle Biogenesis , Sulfonamides/pharmacology
9.
Inflammation ; 41(6): 2129-2135, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30116933

ABSTRACT

Arctiin is a lignin isolated from Arctium lappa which has been known to have anti-viral and anti-inflammatory effects. The aim of this study is to explore the protective effect of arctiin on lipopolysaccharide (LPS)-induced inflammatory responses in acute lung injury (ALI) model of mice. Male BALB/c mice were pretreated with commercial arctiin (10, 20, and 40 mg/kg) 1 h prior to LPS challenge. Twelve hours later, airway inflammation was assessed. We assessed the effects of arctiin on the LPS-induced production of TNF-α, IL-6, and IL-1ß in the bronchoalveolar lavage fluid (BALF). The lung wet-to-dry weight ratio, myeloperoxidase (MPO) activity, and inflammatory signaling pathway were also detected. Our results showed that arctiin not only significantly ameliorated LPS-stimulated lung histopathological changes but also reduced the lung MPO activity. Arctiin also dramatically decreased the production of TNF-α, IL-1ß, and IL-6 in the BALF. In addition, arctiin significantly inhibited LPS-induced PI3K/Akt phosphorylation as well as NF-κB activation. In conclusion, our results suggested that arctiin protected against LPS-induced ALI through inhibiting PI3K/AKT/NF-κB signaling pathway.


Subject(s)
Acute Lung Injury/prevention & control , Furans/pharmacology , Glucosides/pharmacology , Signal Transduction/drug effects , Acute Lung Injury/chemically induced , Animals , Furans/therapeutic use , Glucosides/therapeutic use , Lipopolysaccharides , Male , Mice , Mice, Inbred BALB C , NF-kappa B/drug effects , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism
10.
Cell Physiol Biochem ; 44(2): 671-681, 2017.
Article in English | MEDLINE | ID: mdl-29169175

ABSTRACT

BACKGROUND/AIMS: Prior studies have shown that bufalin inhibits cellular proliferation and induces apoptosis in various human cancers. MicroRNA-203 (miR-203) has been shown to function as an important regulator of tumor progression at various stages. In this study, we investigated the effect of miR-203 expression and bufalin treatment on glioma cell proliferation and stem cell-like phenotypes. METHODS: We used cell viability assay, colony formation assay, cell apoptosis assay and neurosphere formation assay to dectect the treatment effect of bufalin on U251 and U87 cells. Cells were transfected with the miR-203 mimic without bufalin treatment or cells were transfected with anti-miR-203 under bufalin treatment, the above expreiments were repeated. RT-PCR was employed to quantify miR-203 expression. Western blot was performed to detect the stem cell-like (CSC) markers, OCT4 and SOX2. Luciferase activity assay was used to determine whether the SPARC is the target of miR-203. RESULTS: Bufalin treatment inhibited cell proliferation, colony formation, and CSC phenotypes and increased cell apoptosis and expression of miR-203. Furthermore, overexpression of miR-203 led to similar outcomes as bufalin treatment with respect to the cell viability, colony formation, cell apoptosis and the phenotypes of glioma cells. While anti-miR-203 attenuated the inhibitory effects of bufalin as promoting cell proliferation, colony formation and CSC phenotyes and inhibiting cell apoptosis. In addition, we identified SPARC as a novel target gene of miR-203. CONCLUSIONS: These findings suggest that miR-203 plays an important role in bufalin's ability to inhibit the growth of glioma cells and the development of stem cell-like phenotypes.


Subject(s)
Antineoplastic Agents/toxicity , Bufanolides/toxicity , Cell Proliferation/drug effects , MicroRNAs/metabolism , Up-Regulation/drug effects , 3' Untranslated Regions , Antagomirs/metabolism , Apoptosis/drug effects , Base Sequence , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Glioma/metabolism , Glioma/pathology , Humans , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Mutagenesis , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Osteonectin/chemistry , Osteonectin/genetics , Osteonectin/metabolism , Phenotype , Sequence Alignment
11.
Biochem Biophys Res Commun ; 486(4): 1129-1136, 2017 May 13.
Article in English | MEDLINE | ID: mdl-28389242

ABSTRACT

Glioblastoma stem-like cells (GSCs) are responsible for the initiation and progression of glioblastoma multiforme (GBM), and microRNAs (miRNAs) play an important role in this disease. However, the mechanisms underlying the role of miRNAs in the stemness of GSCs have not been completely elucidated. We previously showed that miR-181a is downregulated in GBM and may predict prognosis in patients with this disease. Here, we demonstrate that the upregulation of miR-181a suppressed GSC formation and inhibited GBM tumorigenesis by targeting the Notch2 oncogene. We found that miR-181a was downregulated in GSCs derived from human glioblastoma U87MG and U373MG cells. The high expression of miR-181a inhibited the levels of stemness-related markers CD133 and BMI1, attenuated sphere proliferation, promoted cell apoptosis, and reduced the tumorigenicity of GSCs. MiR-181a decreased the expression of Notch2 by targeting the 3'-untranslated region of its mRNA. Notch2 overexpression inhibited the effects of miR-181a downregulation on GSCs, and was negatively correlated with miR-181a expression. Moreover, high Notch2 expression together with low miR-181a expression was correlated with a shorter median overall survival for GBM patients. Together, these data show that miR-181a may play an essential role in GSC formation and GBM progression by targeting Notch2, suggesting that Notch2 and miR-181a have potential prognostic value as tumor biomarkers in GBM patients.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Glioblastoma/metabolism , Glioblastoma/mortality , MicroRNAs/metabolism , Receptor, Notch2/metabolism , Adult , Biomarkers, Tumor/metabolism , Cell Line, Tumor , China/epidemiology , Humans , Male , Middle Aged , Prognosis , Reproducibility of Results , Sensitivity and Specificity , Statistics as Topic , Survival Rate
12.
Int J Oncol ; 49(1): 217-24, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27176932

ABSTRACT

To investigate the expression and clinical significance of miR-181a and its target genes in glioblastoma multiforme (GBM), the expression levels of miR-181a and three target genes in human normal brain tissues and GBM were analyzed in silico using gene microarray, gene ontology, KEGG pathway and hierarchical clustering analysis followed by validation with quantitative RT-PCR. Our results show that miR-181a is down-regulated in GBM patients. The three target genes, ANGPT2, ARHGAP18 and LAMC1, are negatively correlated with the expression of miR-181a. Moreover, high expression of ANGPT2 or LAMC1 together with large size of GBM is correlated with a shorter median overall survival. In conclusion, our results showed that miR-181a and it targets ANGPT2 and LAMC1 might be predictors of prognosis in GBM patients.


Subject(s)
Angiopoietin-2/biosynthesis , GTPase-Activating Proteins/biosynthesis , Glioblastoma/genetics , Laminin/biosynthesis , MicroRNAs/biosynthesis , Adult , Aged , Angiopoietin-2/genetics , Cell Line, Tumor , Disease-Free Survival , Female , GTPase-Activating Proteins/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Humans , Kaplan-Meier Estimate , Laminin/genetics , Male , MicroRNAs/genetics , Middle Aged , Prognosis
13.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 32(4): 462-7, 2015 Aug.
Article in Chinese | MEDLINE | ID: mdl-26252085

ABSTRACT

OBJECTIVE: To investigate the effect of curcumin on oligomer formation and mitochondrial ATP-sensitive potassium channels (mitoKATP) induced by overexpression or mutation of α-synuclein. METHODS: Recombinant plasmids α-synuclein-pEGFP-A53T and α-synuclein-pEGFP-WT were transfected into PC12 cells by lipofectamin method, and intervened by application of curcumin (20 µmol/L) and 5-hydroxydecanoate (5-HD). Oligomer formation in the cultured cells was identified by Western blotting and Dot blotting. Cytotoxicity and apoptosis of the PC12 cells were measured by lactate dehydrogenase (LDH) and JC-1 assays. mitoKATP were identified by Western blotting and whole cell patch clamp. RESULTS: Curcumin has significantly reduced the oligomer formation induced by overexpression or mutation of α-synuclein in the cultured cells. LDH has decreased by 36.3% and 23.5%, and red/green fluorescence ratio of JC-1 was increased respectively by 48.46% and 50.33% after application of curcumin (P<0.05). Protein expression of Kir6.2 has decreased and mitoKATP channel current has significantly increased (P<0.05). CONCLUSION: Curcumin can inhibit α-synuclein gene overexpression or mutation induced α-synuclein oligomers formation. It may block apoptosis induced by wild-type overexpression or mutation of α-synuclein. By stabilizing mitochondrial membrane potential. Opening of mitoKATP channel may have been the initiating protective mechanism of apoptosis induced by wild-type overexpression or mutation of α-synuclein. Curcumin may antagonize above cytotoxicity through further opening the mitoKATP channel.


Subject(s)
Curcumin/pharmacology , KATP Channels/metabolism , Mitochondria/drug effects , Mutation , Parkinson Disease/metabolism , alpha-Synuclein/genetics , Animals , Apoptosis/drug effects , Cell Line , Humans , KATP Channels/chemistry , KATP Channels/genetics , Mitochondria/genetics , Mitochondria/metabolism , Mutation/drug effects , PC12 Cells , Parkinson Disease/drug therapy , Parkinson Disease/genetics , Parkinson Disease/physiopathology , Rats
14.
Mol Med Rep ; 12(2): 3081-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25955795

ABSTRACT

Alzheimer's disease (AD), characterized by ß-amyloid deposition and neurodegeneration, is the most common cause of dementia worldwide. Emerging evidence suggests that ectopic expression of micro (mi)RNAs is involved in the pathogenesis of AD. There is increasing evidence that miRNAs expressed in the brain are involved in neuronal development, survival and apoptosis. The expression of ß-site amyloid precursor protein-cleaving enzyme 1 (BACE1) is regulated by dysregulated miRNAs in the brain. The present study determined the expression levels of the miRNA-29 (miR-29) family in peripheral blood samples of patients with AD and demonstrated a marked decrease in the expression of miR-29c compared with age-matched controls. In addition, a significant increase in the expression of BACE1 was observed in the peripheral blood of patients with AD. Correlation analysis revealed that the expression of miR-29c was negatively correlated with the protein expression of BACE1 in the peripheral blood samples from patients with AD. The present study also investigated the role of miR-29 on hippocampal neurons in vitro and in vivo. The results demonstrated that the upregulation of miR-29c promoted learning and memory behaviors in SAMP8 mice, at least partially, by increasing the activity of protein kinase A/cAMP response element-binding protein, involved in neuroprotection. This evidence suggested that miR-29c may be a promising potential therapeutic target against AD.


Subject(s)
Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Peptides/genetics , Aspartic Acid Endopeptidases/genetics , MicroRNAs/genetics , Neurons/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/metabolism , CREB-Binding Protein/genetics , CREB-Binding Protein/metabolism , Case-Control Studies , Cell Proliferation , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Female , Gene Expression Regulation , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Maze Learning , Mice , Mice, Transgenic , MicroRNAs/metabolism , Neurons/pathology , Primary Cell Culture , Signal Transduction
15.
Mol Med Rep ; 12(1): 1435-42, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25815896

ABSTRACT

Alzheimer's disease (AD), the most common form of dementia in the aged population, presents an increasing clinical challenge in terms of diagnosis and treatment. Neurodegeneration is one of the hallmarks of AD, which consequently induces cognitive impairment. Brain-derived neurotrophic factor (BDNF), a neuroprotective factor, has been implicated in neuronal survival and proliferation. The epigenetic mechanism of BDNF methylation may be responsible for the reduced expression of BDNF in patients with AD. DNA methyltransferase may contribute to the methylation of BDNF, which is involved in neuroprotection in AD. In addition, epigenetic modifications, including a combination of microRNAs (miRNAs/miRs) and DNA methylation, have been suggested as regulatory mechanisms in the control of neuronal survival. In the present study, the expression of miR-29c was determined in the cerebrospinal fluid (CSF) of patients with AD and of healthy control individuals. A marked decrease in the expression of miR-29c was observed in the AD group compared with the normal control group, accompanied by a decreased in the expression of BDNF. Additionally, a significant increase in the expression of DNA methyltransferase 3 (DNMT3) was observed in the CSF from the patients with AD. Correlation analysis revealed that the expression of miR-29c was positively correlated with BDNF and negatively correlated with DNMT3 protein in the CSF of patients with AD. In addition, the regulatory association between miR-29c, DNMT3 and BDNF were also examined in vitro. It was demonstrated that miR-29c directly targeted DNMT3 and contributed to neuronal proliferation by regulating the expression of BDNF, at least partially, through enhancing the activity of the tyrosine receptor kinase B/extracellular signal-regulated kinase signaling pathway. In conclusion, the present study suggested that miR-29c may be a promising potential therapeutic target in the treatment of AD.


Subject(s)
Alzheimer Disease/genetics , Brain-Derived Neurotrophic Factor/biosynthesis , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , MicroRNAs/genetics , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/pathology , Animals , Apoptosis/genetics , Azacitidine/administration & dosage , Brain-Derived Neurotrophic Factor/cerebrospinal fluid , Cell Proliferation/genetics , Cell Survival/genetics , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/cerebrospinal fluid , DNA Methylation/genetics , DNA Methyltransferase 3A , Gene Expression Regulation , Humans , MicroRNAs/cerebrospinal fluid , Neurons/metabolism , Neurons/pathology , Primary Cell Culture , Rats , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...