scholarly journals Autophagy-Sirtuin1(SIRT1) Modulated The Endothelial Progenitor Cells (EPCs) Proliferation and Migration Via wnt/β-Catenin/GSK3β Signaling Pathway and Alleviated The Coronary Atherosclerosis (CAS) in Mice

Author(s):  
Yanwei Li ◽  
Wei Cui ◽  
Bing Song ◽  
Xuying Ye ◽  
Zhuqing Li ◽  
...  

Abstract Background and purpose: SIRT1 exerted its link to CAS risk in humans and EPCs presented its reparative role in CAS. In this study, we explored the role of SIRT1 in CAS mice and also its modulation in EPCs.Methods and materials: ApoE-/- mice were fed with high-fat and high-glucose food to establish the CAS animal model with the normally-raised C57BL/6 mice as a healthy control group. 5 ApoE mice were intravenously injected with SIRT1 activator, SRT 2104 and another 5 were injected with inhibitor Nicotinamide in tail. Weight changes were recorded per week. Blood samples were taken from posterior orbital venous plexus and detected by automatic biochemical analyzer for lipid concentrations. Coronary artery tissues were observed. HE staining displayed the pathological condition while Immunohistochemistry (IHC) evaluated the CD34+/VEGFR2+ relative density. In vitro, EPCs were isolated from bone marrow each group and then purified, cultured and verified using immunofluorescence staining (IFS). Thereafter, we examined the modulatory mechanism of SIRT1 in EPCs by RT-PCR, MTT, Western Blot (WB) and colony formation, scratch methods, connecting the wnt/β-catenin/GSK3β signaling pathway.Results:SIRT1 activation negatively regulated the weight and TC, TG and LDL. SIRT1 activator alleviated the lesion area and decreased the CD34+/VEGFR2+ density which was higher in coronary artery tissues in CAS and SIRT1 inhibitor groups. In vitro, SIRT1 activator promoted the bone marrow-derived EPCs proliferation, migration and activated wnt/β-catenin/GSK3β signaling pathway while inhibited the autophagy biomarkers ATG1 and LC3II. Furthermore, inhibition of autophagy led to the upregulation of SIRT1 and increase in cell proliferation, migration and wnt/β-catenin/GSK3β activity. The suppression of the pathway in turn lowered SIRT1 expression in EPCs, attenuated the cell proliferation and migration and promoted autophagy. Conclusion: Taken all the findings together, this research disclosed that SIRT1 activator might perform its protective role in CAS through autophagy inhibition via wnt/β-catenin/GSK3β signaling pathway in EPCs.

2019 ◽  
Author(s):  
Yanli Liu ◽  
Yarong Zheng ◽  
Yekai Zhou ◽  
Yi Liu ◽  
Mengjuan Xie ◽  
...  

Abstract Background: To investigate the expression and significance of mechanistic target of rapamycin complex 1(mTORC1) in diabetic retinopathy(DR), and to find new targets and new methods for the treatment of DR.Methods: A DR rat model was prepared by general feeding combined with intraperitoneal injection of 10% streptozotocin (60 mg/kg). The rats were randomly divided into a control group (NDM group) and diabetes group (DM group).Three months later,the degrees of retinopathy were determined using hematoxylin and eosin staining,and the levels of p-S6, VEGF, and PEDF proteins were detected by immunohistochemistry and western blotting. Human retinal capillary endothelial cells (HRCECs) were cultured in high glucose conditions,then treated with rapamycin or transfected with siTSC1.The protein levels of p-S6 were assessed by western blotting. The 5-ethynyl-2´-deoxyuridine assay was used to detect cell proliferation, and the Transwell assay was used to detect cell migration.Results: A DM rat model was successfully developed. The expressions of p-S6 and VEGF proteins were significantly increased in the DM group (p < 0.05), and the expression of PEDF protein was significantly decreased compared with the control group (p < 0.05). In vitro,the p-S6 protein in high glucose(HG) induced HRCECs was increased compared with the normal control (p < 0.05), and cell proliferation and migration were increased compared with the normal glucose(NG) group (p < 0.05). After transfection with siTSC1 to activate mTORC1,the expression of p-S6 was increased,as well as cell proliferation and migration.In contrast rapamycin decreased p-S6 expression in HG induced HRCECs, as well as decrased proliferation and migration (p < 0.05).Conclusion: The mTORC1 played an important role in DR. After activation, mTORC1 induced expression of the p-S6 protein, regulated the expressions of VEGF and PEDF proteins, and changed the proliferation and migration of endothelial cells.The mTORC1 can therefore be used as a new target,as well as in the treatment of DR.


2018 ◽  
Vol 2018 ◽  
pp. 1-8 ◽  
Author(s):  
He Hu ◽  
Shanshan Li ◽  
Jianqiao Li ◽  
Chao Huang ◽  
Fang Zhou ◽  
...  

Purpose. Recent research has provided novel insight into the function of fibromodulin (FMOD) in wound healing and angiogenesis. The role of FMOD in initiation of proliferative vitreoretinopathy (PVR) has not been studied. This study investigated the effect of FMOD on human retinal pigment epithelial (RPE) cell, which plays an essential role in the progression of PVR, and the possible mechanisms. Methods. Small interfering (si) RNA-based gene transfer technology was used to decrease FMOD expression and to study its effects on RPEs in vitro. Cell Counting Kit-8 assays, transwells, and flow cytometry analysis were used to measure cell proliferation, migration, cell cycle, and apoptosis. Western blot was used to measure expression of vascular endothelial growth factor (VEGF), VEGF receptor 2 (VEGFR2), extracellular signal-related kinase 1/2 (ERK1/2), and phosphoinositide 3 kinase (PI3K/AKT). Results. After transfection of RPEs with a FMOD-specific siRNA, cell proliferation and migration were inhibited to the percentage of 65% ± 5% and 39% ± 10%, respectively, compared to the control group. Depletion of FMOD induced cell cycle arrest and apoptosis in RPE cells. Downregulation of VEGF, VEGFR2, and phosphorylated AKT (p-AKT) were detected in transfected RPEs. Conclusion. Depletion of FMOD selectively downregulated the expression of VEGF and VEGFR2 and inhibited the signaling pathway of AKT phosphorylation, which consequently inhibited the proliferation and migration of RPE Cell.


2020 ◽  
Vol 64 (4) ◽  
Author(s):  
Hu Li ◽  
Zheng Zeng ◽  
Xiang Yang ◽  
Ye Chen ◽  
Lei He ◽  
...  

Ovarian cancer (OC) is one of the most prevalent and deadly types of gynecological malignancy. Since current treatments are not effective against OC, it is imperative to develop novel potential therapeutic targets for managing OC. In this study, we aimed to uncover the underlying molecular mechanism of long non-coding RNA (lncRNA) GClnc1 related to p53 signaling pathway in OC. The expression of lncRNA H19 GClnc1 was markedly higher in OC samples than the related normal tissues. Next, we found that lncRNA GClnc1 inhibited p53. In addition, the lncRNA GClnc1 overexpression promoted the cell proliferation and migration in vitro. Subsequently, p53 silencing obligated the effect of lncRNA GCln1 knock down on cell proliferation and migration. To sum up, LncRNA GClnc1 contributes to the progression of OC by regulating p53 signaling pathway. Meanwhile, our findings also suggested that lncRNA GClnc1 may serve as a novel therapeutic target for OC patients.


Author(s):  
Jian Zhang ◽  
ZhenFeng Shi ◽  
JinXing Huang ◽  
XiaoGuang Zou

This study aimed to investigate the pivotal role of cystatin B (CSTB) in the development of gastric cancer and to explore its possible regulatory mechanism. Human gastric cancer SGC-7901 cells as a model in vitro were transfected with plasmid PCDNA3.1-CSTB and siRNA-CSTB using Lipofectamine 2000. Quantitative real-time PCR (qRT-PCR) and Western blotting were performed to determine the relative expression of CSTB and PI3K/Akt/mTOR pathway-related protein. Moreover, MTT assay, Transwell assay, and flow cytometry were used to assess cell proliferation, migration, and apoptosis, respectively. The results showed that CSTB was significantly downregulated in SGC-7901 cells compared with gastric epithelial cells. CSTB was successfully overexpressed and suppressed after cells were transfected with pc-CSTB and si-CSTB, respectively. Moreover, cell viability and migration were significantly decreased after being transfected with pc-CSTB when compared with the control group, while being obviously increased after transfection with si-CSTB. However, cell apoptosis was significantly induced after being transfected with pc-CSTB, while being obviously suppressed after transfection with si-CSTB. Besides, the expression levels of p-PI3K, p-Akt, and p-mTOR proteins were all significantly decreased in the pc-CSTB transfection group when compared with the control group, while being increased in the si-CSTB transfection group. Our findings suggest that CSTB downregulation may promote the development of gastric cancer by affecting cell proliferation and migration, and the PI3K/Akt/mTOR signaling pathway was activated in this process. CSTB may serve as a potential therapeutic target for gastric cancer.


2021 ◽  
Vol 22 (6) ◽  
pp. 3046
Author(s):  
Ming-Huei Chou ◽  
Hui-Ching Chuang ◽  
Yu-Tsai Lin ◽  
Ming-Hsien Tsai ◽  
Ying-Hsien Kao ◽  
...  

Patients with advanced head and neck squamous cell carcinoma (HNSCC) usually show a dismal prognosis. It is this worthwhile to develop new, effective therapeutic regimens for these patients, such as molecular targeted therapy, which is promising as an alternative or combination treatment for HNSCC. The mammalian target of rapamycin (mTOR) pathway, which plays an important role in the carcinogenesis of HNSCC, is the most frequently activated, and is thus worthy of further investigation. In this study, two human HNSCC cell lines, FaDu and SAS, were evaluated for cell growth with trypan blue staining and tumor growth using an orthotopic xenograft model. The immunohistochemical expression of mTOR in the subcutaneous xenograft model and the inhibitory effects of docetaxel on the growth and state of activation of the PI3K/mTOR pathway were also evaluated and examined by colony formation and Western blot, respectively. Cell proliferation and migration were measured by water-soluble tetrazolium salt (WST-1) and OrisTM cell migration assay, respectively. Furthermore, the effects of rapamycin and BEZ235, a phosphatidylinositol 3-kinases (PI3K) and mTOR inhibitor in combination with docetaxel or CCL20 were evaluated in the FaDu and SAS cells. The results showed that the expression of mTOR was significantly higher in the SAS and FaDu xenograft models than in the control. Docetaxel treatment significantly suppressed HNSCC cell proliferation and migration in vitro via the PI3K/mTOR/CCL-20 signaling pathway. Additionally, when administered in a dose-dependent fashion, mTOR inhibitors inhibited the growth and migration of the HNSCC cells. This combination was synergistic with docetaxel, resulting in almost complete cell growth and migration arrest. In conclusion, docetaxel significantly inhibited HNSCC cell proliferation and migration in vitro via the PI3K/mTOR/CCL-20 signaling pathway. The synergistic and additive activity of mTOR inhibitors combined with docetaxel shows potential as a new treatment strategy for HNSCC.


2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Jiong Ma ◽  
Chunxia Zhou ◽  
Xuejun Chen

Abstract Background Hedgehog (Hh) signaling pathway, which is essential for cell proliferation and differentiation, is noted to be aberrantly activated in tumor from increasing studies in recent years. MicroRNAs (miRNAs) as an important non-coding RNA in cells have been proven to possess a regulatory role specific to the Hh signaling pathway. Here, in vitro and in vivo cellular/molecular experiments were adopted to clarify the regulatory mechanism linking miR-636 to the Hh signaling pathway in ovarian cancer (OVC). Methods Protein–protein interaction analysis was performed to identify the hub gene in the Hh pathway. TargetScan database was used to predict the potential upstream regulators for Gli2. qRT-PCR was performed to test the expression of miR-636, while Western blot was conducted to detect the expression of proteins related to the Hh pathway and epithelial-mesenchymal transition (EMT). For cell functional experiments, HO-8910PM OVC cell line was used. MTT assay and wound healing assay were used to measure the effect of miR-636 on cell proliferation and migration. Flow cytometry was carried out to examine the effect of miR-636 on cell cycle, and Western blot was used to identify the change in expression of Hh and EMT-related proteins. Dual-luciferase reporter gene assay was implemented to detect the targeting relationship between miR-636 and Gli2. Xenotransplantation models were established for in vivo examination. Results Gli2 was identified as the hub gene of the Hh pathway and it was validated to be regulated by miR-636 based on the data from TargetScan and GEO databases. In vitro experiments discovered that miR-636 was significantly lowly expressed in OVC cell lines, and overexpressing miR-636 significantly inhibited HO-8910PM cell proliferation, migration and induced cell cycle arrest in G0/G1 phase, while the inhibition of miR-636 caused opposite results. Dual-luciferase reporter gene assay revealed that Gli2 was the target gene of miR-636 in OVC. Besides, overexpressed miR-636 decreased protein expression of Gli2, and affected the expression of proteins related to the Hh signaling pathway and EMT. Rescue experiments verified that overexpression of Gli2 reversed the inhibitory effect of miR-636 on HO-8910PM cell proliferation and migration, and attenuated the blocking effect of miR-636 on cell cycle. The xenotransplantation experiment suggested that miR-636 inhibited cell growth of OVC by decreasing Gli2 expression. Besides, overexpressing Gli2 potentiated the EMT process of OVC cells via decreasing E-cadherin protein expression and increasing Vimentin protein expression, and it reversed the inhibitory effect of miR-636 on OVC cell proliferation in vivo. Conclusion miR-636 mediates the activation of the Hh pathway via binding to Gli2, thus inhibiting EMT, suppressing cell proliferation and migration of OVC. Trial registration: The experimental protocol was established, according to the ethical guidelines of the Helsinki Declaration and was approved by the Human Ethics Committee of The Second Affiliated hospital of Zhejiang University School of Medicine (IR2019001235). Written informed consent was obtained from individual or guardian participants.


Sign in / Sign up

Export Citation Format

Share Document