1.Extracellular vesicle-carried GTF2I from mesenchymal stem cells promotes the expression of tumor-suppressive FAT1 and inhibits stemness maintenance in thyroid carcinoma.
Jie SHAO ; Wenjuan WANG ; Baorui TAO ; Zihao CAI ; Haixia LI ; Jinhong CHEN
Frontiers of Medicine 2023;17(6):1186-1203
		                        		
		                        			
		                        			Through bioinformatics predictions, we identified that GTF2I and FAT1 were downregulated in thyroid carcinoma (TC). Further, Pearson's correlation coefficient revealed a positive correlation between GTF2I expression and FAT1 expression. Therefore, we selected them for this present study, where the effects of bone marrow mesenchymal stem cell-derived EVs (BMSDs-EVs) enriched with GTF2I were evaluated on the epithelial-to-mesenchymal transition (EMT) and stemness maintenance in TC. The under-expression of GTF2I and FAT1 was validated in TC cell lines. Ectopically expressed GTF2I and FAT1 were found to augment malignant phenotypes of TC cells, EMT, and stemness maintenance. Mechanistic studies revealed that GTF2I bound to the promoter region of FAT1 and consequently upregulated its expression. MSC-EVs could shuttle GTF2I into TPC-1 cells, where GTF2I inhibited TC malignant phenotypes, EMT, and stemness maintenance by increasing the expression of FAT1 and facilitating the FAT1-mediated CDK4/FOXM1 downregulation. In vivo experiments confirmed that silencing of GTF2I accelerated tumor growth in nude mice. Taken together, our work suggests that GTF2I transferred by MSC-EVs confer antioncogenic effects through the FAT1/CDK4/FOXM1 axis and may be used as a promising biomarker for TC treatment.
		                        		
		                        		
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Cell Proliferation
		                        			;
		                        		
		                        			Mice, Nude
		                        			;
		                        		
		                        			Epithelial-Mesenchymal Transition
		                        			;
		                        		
		                        			Thyroid Neoplasms/pathology*
		                        			;
		                        		
		                        			Extracellular Vesicles/pathology*
		                        			;
		                        		
		                        			Mesenchymal Stem Cells
		                        			;
		                        		
		                        			Transcription Factors, TFIII/metabolism*
		                        			;
		                        		
		                        			Neoplastic Stem Cells/pathology*
		                        			
		                        		
		                        	
2.The mechanism of S100A7 inducing the migration and invasion in cervical cancer cells.
Tian TIAN ; Zhen HUA ; Yan KONG ; Ling Zhi WANG ; Xiang Yu LIU ; Yi HAN ; Xue Min ZHOU ; Zhu Mei CUI
Chinese Journal of Oncology 2023;45(5):375-381
		                        		
		                        			
		                        			Objective: To investigate the mechanism of S100A7 inducing the migration and invasion in cervical cancers. Methods: Tissue samples of 5 cases of cervical squamous cell carcinoma and 3 cases of adenocarcinoma were collected from May 2007 to December 2007 in the Department of Gynecology of the Affiliated Hospital of Qingdao University. Immunohistochemistry was performed to evaluate the expression of S100A7 in cervical carcinoma tissues. S100A7-overexpressing HeLa and C33A cells were established with lentiviral systems as the experimental group. Immunofluorescence assay was performed to observe the cell morphology. Transwell assay was taken to detect the effect of S100A7-overexpression on the migration and invasion of cervical cancer cells. Reverse transcription-quantitative real-time polymerase chain reaction (RT-qPCR) was used to examine the mRNA expressions of E-cadherin, N-cadherin, vimentin and fibronectin. The expression of extracellular S100A7 in conditioned medium of cervical cancer cell was detected by western blot. Conditioned medium was added into Transwell lower compartment to detect cell motility. Exosomes were isolated and extracted from the culture supernatant of cervical cancer cell, the expressions of S100A7, CD81 and TSG101 were detected by western blot. Transwell assay was taken to detect the effect of exosomes on the migration and invasion of cervical cancer cells. Results: S100A7 expression was positively expressed in cervical squamous carcinoma and negative expression in adenocarcinoma. Stable S100A7-overexpressing HeLa and C33A cells were successfully constructed. C33A cells in the experimental group were spindle shaped while those in the control group tended to be polygonal epithelioid cells. The number of S100A7-overexpressed HeLa cells passing through the Transwell membrane assay was increased significantly in migration and invasion assay (152.00±39.22 vs 105.13±15.75, P<0.05; 115.38±34.57 vs 79.50±13.68, P<0.05). RT-qPCR indicated that the mRNA expressions of E-cadherin in S100A7-overexpressed HeLa and C33A cells decreased (P<0.05) while the mRNA expressions of N-cadherin and fibronectin in HeLa cells and fibronectin in C33A cells increased (P<0.05). Western blot showed that extracellular S100A7 was detected in culture supernatant of cervical cancer cells. HeLa cells of the experimental group passing through transwell membrane in migration and invasion assays were increased significantly (192.60±24.41 vs 98.80±47.24, P<0.05; 105.40±27.38 vs 84.50±13.51, P<0.05) when the conditional medium was added into the lower compartment of Transwell. Exosomes from C33A cell culture supernatant were extracted successfully, and S100A7 expression was positive. The number of transmembrane C33A cells incubated with exosomes extracted from cells of the experimental group was increased significantly (251.00±49.82 vs 143.00±30.85, P<0.05; 524.60±52.74 vs 389.00±63.23, P<0.05). Conclusion: S100A7 may promote the migration and invasion of cervical cancer cells by epithelial-mesenchymal transition and exosome secretion.
		                        		
		                        		
		                        		
		                        			Female
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Uterine Cervical Neoplasms/pathology*
		                        			;
		                        		
		                        			HeLa Cells
		                        			;
		                        		
		                        			Fibronectins/metabolism*
		                        			;
		                        		
		                        			Culture Media, Conditioned
		                        			;
		                        		
		                        			Carcinoma, Squamous Cell/metabolism*
		                        			;
		                        		
		                        			Adenocarcinoma
		                        			;
		                        		
		                        			Cadherins/metabolism*
		                        			;
		                        		
		                        			RNA, Messenger/metabolism*
		                        			;
		                        		
		                        			Cell Movement
		                        			;
		                        		
		                        			Epithelial-Mesenchymal Transition/genetics*
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Cell Proliferation
		                        			;
		                        		
		                        			S100 Calcium Binding Protein A7/metabolism*
		                        			
		                        		
		                        	
3.Exosomal miR-485-3p derived from pancreatic ductal epithelial cells inhibits pancreatic cancer metastasis through targeting PAK1.
Mingzhe LI ; Jiaxin ZHOU ; Zhengkui ZHANG ; Jisong LI ; Feng WANG ; Ling MA ; Xiaodong TIAN ; Zebin MAO ; Yinmo YANG
Chinese Medical Journal 2022;135(19):2326-2337
		                        		
		                        			BACKGROUND:
		                        			Cell competition is an important feature in pancreatic cancer (PC) progression, but the underlying mechanism remains elusive. This study aims to explore the role of exosomes derived from normal pancreatic ductal epithelial cells involved in PC progression.
		                        		
		                        			METHODS:
		                        			PC cells and pancreatic stellate cells (PSCs) were treated with exosomes isolated from pancreatic ductal epithelial cells. Cell proliferation was assessed by CCK8 assays. Cell migration and invasion were assessed by Transwell assays. PC and matched adjacent non-tumor tissue specimens were obtained from 46 patients pathologically diagnosed with PC at Peking University First Hospital from 2013 to 2017. Tissue miR-485-3p and p21-activated kinase-1 (PAK1) expression was examined by real-time polymerase chain reaction (RT-PCR), and the relationship of the two was analyzed using Pearman's product-moment correlation. The clinical significance of miR-485-3p was analyzed using the Chi-square test, Wilcoxon rank-sum test, and Fisher exact probability, respectively. The binding of miR-485-3p to PAK1 5'-untranslated region (5'-UTR) was examined by luciferase assay. PC cells were xenografted into nude mice as a PC metastasis model.
		                        		
		                        			RESULTS:
		                        			Exosomes from pancreatic ductal epithelial cells suppressed PC cell migration and invasion as well as the secretion and migration of PSCs. MiR-485-3p was enriched in the exosomes of pancreatic ductal epithelial cells but deficient in those of PC cells and PSCs, in accordance with the lower level in PSCs and PC cells than that in pancreatic ductal cells. And the mature miR-485-3p could be delivered into these cells by the exosomes secreted by normal pancreatic duct cells, to inhibit PC cell migration and invasion. Clinical data analysis showed that miR-485-3p was significantly decreased in PC tissues (P < 0.05) and was negatively associated with lymphovascular invasion (P = 0.044). As a direct target of miR-485-3p, PAK1 was found to exert an inhibitory effect on PC cells, and there was a significantly negative correlation between the expression levels of miR-485-3p and PAK1 (r = -0.6525, P < 0.0001) in PC tissues. Moreover, miR-485-3p could suppress PC metastasis in vivo by targeting p21-activated kinase-1.
		                        		
		                        			CONCLUSIONS
		                        			Exosomal miR-485-3p delivered by normal pancreatic ductal epithelial cells into PC cells inhibits PC metastasis by directly targeting PAK1. The restoration of miR-485-3p by exosomes or some other vehicle might be a novel approach for PC treatment.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			MicroRNAs/metabolism*
		                        			;
		                        		
		                        			Mice, Nude
		                        			;
		                        		
		                        			p21-Activated Kinases/metabolism*
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Pancreatic Neoplasms/genetics*
		                        			;
		                        		
		                        			Epithelial Cells/metabolism*
		                        			;
		                        		
		                        			Pancreatic Ducts/pathology*
		                        			;
		                        		
		                        			Cell Proliferation
		                        			;
		                        		
		                        			Cell Movement
		                        			;
		                        		
		                        			Gene Expression Regulation, Neoplastic
		                        			
		                        		
		                        	
4.Cancer cells corrupt normal epithelial cells through miR-let-7c-rich small extracellular vesicle-mediated downregulation of p53/PTEN.
Weilian LIANG ; Yang CHEN ; Hanzhe LIU ; Hui ZHAO ; Tingting LUO ; Hokeung TANG ; Xiaocheng ZHOU ; Erhui JIANG ; Zhe SHAO ; Ke LIU ; Zhengjun SHANG
International Journal of Oral Science 2022;14(1):36-36
		                        		
		                        			
		                        			Tumor volume increases continuously in the advanced stage, and aside from the self-renewal of tumor cells, whether the oncogenic transformation of surrounding normal cells is involved in this process is currently unclear. Here, we show that oral squamous cell carcinoma (OSCC)-derived small extracellular vesicles (sEVs) promote the proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) of normal epithelial cells but delay their apoptosis. In addition, nuclear-cytoplasmic invaginations and multiple nucleoli are observed in sEV-treated normal cells, both of which are typical characteristics of premalignant lesions of OSCC. Mechanistically, miR-let-7c in OSCC-derived sEVs is transferred to normal epithelial cells, leading to the transcriptional inhibition of p53 and inactivation of the p53/PTEN pathway. In summary, we demonstrate that OSCC-derived sEVs promote the precancerous transformation of normal epithelial cells, in which the miR-let-7c/p53/PTEN pathway plays an important role. Our findings reveal that cancer cells can corrupt normal epithelial cells through sEVs, which provides new insight into the progression of OSCC.
		                        		
		                        		
		                        		
		                        			Carcinoma, Squamous Cell/pathology*
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Cell Movement
		                        			;
		                        		
		                        			Cell Proliferation
		                        			;
		                        		
		                        			Cell Transformation, Neoplastic
		                        			;
		                        		
		                        			Down-Regulation
		                        			;
		                        		
		                        			Epithelial Cells/metabolism*
		                        			;
		                        		
		                        			Extracellular Vesicles/pathology*
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			MicroRNAs/metabolism*
		                        			;
		                        		
		                        			Mouth Neoplasms/pathology*
		                        			;
		                        		
		                        			PTEN Phosphohydrolase/metabolism*
		                        			;
		                        		
		                        			Tumor Suppressor Protein p53/metabolism*
		                        			
		                        		
		                        	
5.Mesenchymal stem cell therapy for acute respiratory distress syndrome: from basic to clinics.
Protein & Cell 2020;11(10):707-722
		                        		
		                        			
		                        			The 2019 novel coronavirus disease (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has occurred in China and around the world. SARS-CoV-2-infected patients with severe pneumonia rapidly develop acute respiratory distress syndrome (ARDS) and die of multiple organ failure. Despite advances in supportive care approaches, ARDS is still associated with high mortality and morbidity. Mesenchymal stem cell (MSC)-based therapy may be an potential alternative strategy for treating ARDS by targeting the various pathophysiological events of ARDS. By releasing a variety of paracrine factors and extracellular vesicles, MSC can exert anti-inflammatory, anti-apoptotic, anti-microbial, and pro-angiogenic effects, promote bacterial and alveolar fluid clearance, disrupt the pulmonary endothelial and epithelial cell damage, eventually avoiding the lung and distal organ injuries to rescue patients with ARDS. An increasing number of experimental animal studies and early clinical studies verify the safety and efficacy of MSC therapy in ARDS. Since low cell engraftment and survival in lung limit MSC therapeutic potentials, several strategies have been developed to enhance their engraftment in the lung and their intrinsic, therapeutic properties. Here, we provide a comprehensive review of the mechanisms and optimization of MSC therapy in ARDS and highlighted the potentials and possible barriers of MSC therapy for COVID-19 patients with ARDS.
		                        		
		                        		
		                        		
		                        			Adoptive Transfer
		                        			;
		                        		
		                        			Alveolar Epithelial Cells
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Apoptosis
		                        			;
		                        		
		                        			Betacoronavirus
		                        			;
		                        		
		                        			Body Fluids
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			CD4-Positive T-Lymphocytes
		                        			;
		                        		
		                        			immunology
		                        			;
		                        		
		                        			Clinical Trials as Topic
		                        			;
		                        		
		                        			Coinfection
		                        			;
		                        		
		                        			prevention & control
		                        			;
		                        		
		                        			therapy
		                        			;
		                        		
		                        			Coronavirus Infections
		                        			;
		                        		
		                        			complications
		                        			;
		                        		
		                        			immunology
		                        			;
		                        		
		                        			Disease Models, Animal
		                        			;
		                        		
		                        			Endothelial Cells
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			Extracorporeal Membrane Oxygenation
		                        			;
		                        		
		                        			Genetic Therapy
		                        			;
		                        		
		                        			methods
		                        			;
		                        		
		                        			Genetic Vectors
		                        			;
		                        		
		                        			administration & dosage
		                        			;
		                        		
		                        			therapeutic use
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Immunity, Innate
		                        			;
		                        		
		                        			Inflammation Mediators
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Lung
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			physiopathology
		                        			;
		                        		
		                        			Mesenchymal Stem Cell Transplantation
		                        			;
		                        		
		                        			methods
		                        			;
		                        		
		                        			Mesenchymal Stem Cells
		                        			;
		                        		
		                        			physiology
		                        			;
		                        		
		                        			Multiple Organ Failure
		                        			;
		                        		
		                        			etiology
		                        			;
		                        		
		                        			prevention & control
		                        			;
		                        		
		                        			Pandemics
		                        			;
		                        		
		                        			Pneumonia, Viral
		                        			;
		                        		
		                        			complications
		                        			;
		                        		
		                        			immunology
		                        			;
		                        		
		                        			Respiratory Distress Syndrome, Adult
		                        			;
		                        		
		                        			immunology
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			therapy
		                        			;
		                        		
		                        			Translational Medical Research
		                        			
		                        		
		                        	
6.Effects of Long Non-coding RNA Plasmacytoma Variant Translocation 1 Gene on Inflammatory Response and Cell Migration in Infected Gastric Epithelial Cell Line.
Xiao-Hui JING ; Ling-Xue LI ; Tao-Tao HAN ; Juan SHI
Acta Academiae Medicinae Sinicae 2020;42(2):228-235
		                        		
		                        			
		                        			To investigate the mechanism of long non-coding RNA plasmacytoma variant translocation 1 (PVT1) in gastric cancer caused by (HP) infection. The expression of PVT1 was detected by quantitative real-time polymerase chain reaction in HP-infected normal gastric epithelial cells GES-1. Gastric cancer cell line SGC-7901 was transfected with PVT1 small interfering RNA and co-cultured with HP,and then the inflammatory cytokines such as tumor necrosis factor-α (TNF-α),interleukin (IL) -1β,IL-6 and IL-8 were detected. After PVT1 was knocked down,the effects of PVT1 on the proliferation and migration of gastric cancer cells were examined by cell scratch assay. RNA-pulldown combined with mass spectrometry was used to detect the protein binding to PVT1,and the result of mass spectrometry was verified by RNA-pulldown combined with Western blot. In HP-infected normal gastric epithelial cells GES-1,quantitative real-time polymerase chain reaction showed that PVT1 was significantly up-regulated (=7.160,=0.019). PVT1 was knocked down in gastric cancer cells,and then infected with HP. The expressions of inflammatory factors including TNF-α (=3.899,=0.011),IL-1β (=14.610,=0.000),and IL-8 (=6.557,=0.001) were significantly inhibited. Although PVT1 knockdown had no significant effect on the proliferation ability of gastric cancer cells,it inhibited the migration of cells. PVT1 might interact with RPS8 protein. PVT1 may act as a pro-inflammatory factor and regulate gastric cancer caused by HP infection.
		                        		
		                        		
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Cell Movement
		                        			;
		                        		
		                        			Cytokines
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Epithelial Cells
		                        			;
		                        		
		                        			cytology
		                        			;
		                        		
		                        			microbiology
		                        			;
		                        		
		                        			Gene Knockdown Techniques
		                        			;
		                        		
		                        			Helicobacter Infections
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			Helicobacter pylori
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Inflammation
		                        			;
		                        		
		                        			RNA, Long Noncoding
		                        			;
		                        		
		                        			genetics
		                        			
		                        		
		                        	
7.Recapitulation of SARS-CoV-2 infection and cholangiocyte damage with human liver ductal organoids.
Bing ZHAO ; Chao NI ; Ran GAO ; Yuyan WANG ; Li YANG ; Jinsong WEI ; Ting LV ; Jianqing LIANG ; Qisheng ZHANG ; Wei XU ; Youhua XIE ; Xiaoyue WANG ; Zhenghong YUAN ; Junbo LIANG ; Rong ZHANG ; Xinhua LIN
Protein & Cell 2020;11(10):771-775
		                        		
		                        		
		                        		
		                        			Betacoronavirus
		                        			;
		                        		
		                        			isolation & purification
		                        			;
		                        		
		                        			pathogenicity
		                        			;
		                        		
		                        			Bile Acids and Salts
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Bile Ducts, Intrahepatic
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			virology
		                        			;
		                        		
		                        			Cell Culture Techniques
		                        			;
		                        		
		                        			Coronavirus Infections
		                        			;
		                        		
		                        			complications
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			Cytokine Release Syndrome
		                        			;
		                        		
		                        			etiology
		                        			;
		                        		
		                        			physiopathology
		                        			;
		                        		
		                        			Cytopathogenic Effect, Viral
		                        			;
		                        		
		                        			Epithelial Cells
		                        			;
		                        		
		                        			enzymology
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			virology
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Hyperbilirubinemia
		                        			;
		                        		
		                        			etiology
		                        			;
		                        		
		                        			Liver
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			Organoids
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			virology
		                        			;
		                        		
		                        			Pandemics
		                        			;
		                        		
		                        			Peptidyl-Dipeptidase A
		                        			;
		                        		
		                        			analysis
		                        			;
		                        		
		                        			Pneumonia, Viral
		                        			;
		                        		
		                        			complications
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			Receptors, Virus
		                        			;
		                        		
		                        			analysis
		                        			;
		                        		
		                        			Serine Endopeptidases
		                        			;
		                        		
		                        			analysis
		                        			;
		                        		
		                        			Viral Load
		                        			
		                        		
		                        	
8.Inhibition of chemotherapy-related breast tumor EMT by application of redox-sensitive siRNA delivery system CSO-ss-SA/siRNA along with doxorubicin treatment.
Xuan LIU ; Xue-Qing ZHOU ; Xu-Wei SHANG ; Li WANG ; Yi LI ; Hong YUAN ; Fu-Qiang HU
Journal of Zhejiang University. Science. B 2020;21(3):218-233
		                        		
		                        			
		                        			Metastasis is one of the main reasons causing death in cancer patients. It was reported that chemotherapy might induce metastasis. In order to uncover the mechanism of chemotherapy-induced metastasis and find solutions to inhibit treatment-induced metastasis, the relationship between epithelial-mesenchymal transition (EMT) and doxorubicin (DOX) treatment was investigated and a redox-sensitive small interfering RNA (siRNA) delivery system was designed. DOX-related reactive oxygen species (ROS) were found to be responsible for the invasiveness of tumor cells in vitro, causing enhanced EMT and cytoskeleton reconstruction regulated by Ras-related C3 botulinum toxin substrate 1 (RAC1). In order to decrease RAC1, a redox-sensitive glycolipid drug delivery system (chitosan-ss-stearylamine conjugate (CSO-ss-SA)) was designed to carry siRNA, forming a gene delivery system (CSO-ss-SA/siRNA) downregulating RAC1. CSO-ss-SA/siRNA exhibited an enhanced redox sensitivity compared to nonresponsive complexes in 10 mmol/L glutathione (GSH) and showed a significant safety. CSO-ss-SA/siRNA could effectively transmit siRNA into tumor cells, reducing the expression of RAC1 protein by 38.2% and decreasing the number of tumor-induced invasion cells by 42.5%. When combined with DOX, CSO-ss-SA/siRNA remarkably inhibited the chemotherapy-induced EMT in vivo and enhanced therapeutic efficiency. The present study indicates that RAC1 protein is a key regulator of chemotherapy-induced EMT and CSO-ss-SA/siRNA silencing RAC1 could efficiently decrease the tumor metastasis risk after chemotherapy.
		                        		
		                        		
		                        		
		                        			Amines/chemistry*
		                        			;
		                        		
		                        			Antineoplastic Agents/adverse effects*
		                        			;
		                        		
		                        			Breast Neoplasms/pathology*
		                        			;
		                        		
		                        			Chitosan/chemistry*
		                        			;
		                        		
		                        			Doxorubicin/adverse effects*
		                        			;
		                        		
		                        			Drug Delivery Systems
		                        			;
		                        		
		                        			Epithelial-Mesenchymal Transition/drug effects*
		                        			;
		                        		
		                        			Female
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			MCF-7 Cells
		                        			;
		                        		
		                        			Neoplasm Metastasis/prevention & control*
		                        			;
		                        		
		                        			Oxidation-Reduction
		                        			;
		                        		
		                        			RNA, Small Interfering/administration & dosage*
		                        			;
		                        		
		                        			Reactive Oxygen Species/metabolism*
		                        			;
		                        		
		                        			rac1 GTP-Binding Protein/physiology*
		                        			
		                        		
		                        	
9.Extracellular signal regulated kinase 5 promotes cell migration, invasion and lung metastasis in a FAK-dependent manner.
Weiwei JIANG ; Fangfang CAI ; Huangru XU ; Yanyan LU ; Jia CHEN ; Jia LIU ; Nini CAO ; Xiangyu ZHANG ; Xiao CHEN ; Qilai HUANG ; Hongqin ZHUANG ; Zi-Chun HUA
Protein & Cell 2020;11(11):825-845
		                        		
		                        			
		                        			This study was designed to evaluate ERK5 expression in lung cancer and malignant melanoma progression and to ascertain the involvement of ERK5 signaling in lung cancer and melanoma. We show that ERK5 expression is abundant in human lung cancer samples, and elevated ERK5 expression in lung cancer was linked to the acquisition of increased metastatic and invasive potential. Importantly, we observed a significant correlation between ERK5 activity and FAK expression and its phosphorylation at the Ser
		                        		
		                        		
		                        		
		                        			A549 Cells
		                        			;
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Cell Movement
		                        			;
		                        		
		                        			Epithelial-Mesenchymal Transition/genetics*
		                        			;
		                        		
		                        			Focal Adhesion Kinase 1/metabolism*
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Lung Neoplasms/pathology*
		                        			;
		                        		
		                        			MAP Kinase Signaling System
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Mitogen-Activated Protein Kinase 7/metabolism*
		                        			;
		                        		
		                        			Neoplasm Invasiveness
		                        			;
		                        		
		                        			Neoplasm Metastasis
		                        			;
		                        		
		                        			Neoplasm Proteins/metabolism*
		                        			
		                        		
		                        	
10.Role of ovarian tumor stem-like cells sorted from human epithelial ovarian cancer SKOV3 cells in vasculogenic mimicry formation.
Jun LIANG ; Huimin XING ; Xiaohua WU ; Lei ZHANG ; Jun ZHAO
Journal of Southern Medical University 2019;39(9):1065-1070
		                        		
		                        			OBJECTIVE:
		                        			To isolate tumor stem-like cells from human epithelial ovarian cancer SKOV3 cells and explore their role in the formation of vascularization mimicry (VM).
		                        		
		                        			METHODS:
		                        			SKOV3 cells were passaged to the 7th generation by suspension culture in serum-free medium, and the percentages of CD133- and CD117-positive cells in the 1st, 3rd, 5th and 7th generations were analyzed using flow cytometry. The proliferative activity of the cells sorted from the 7th generation SKOV3 cells was assessed with colony formation assay. A three-dimensional cell culture model was established to compare the ability of VM formation between the sorted cells and the parental SKOV3 cells. The expression levels of matrix metalloproteinases-2 (MMP-2) and MMP-9 in the two groups were detected using real-time PCR and Western blotting.
		                        		
		                        			RESULTS:
		                        			Some SKOV3 cells formed typical cell spheres with suspension growth in serum-free medium and were passaged to the 7th generation. Flow cytometry revealed that the percentage of CD133-positive cells increased with cell passaging. The cloning efficiency of the sorted cells was significantly higher than that of the parental SKOV3 cells (50.33% 5.33%, < 0.001). The VM formation ability of the sorted cells was stronger than that of the parental SKOV3 cells in the three-dimensional cell culture system. RT-PCR and Western blotting showed that the expression levels of MMP-2 and MMP-9 were significantly higher in the 7th passage cells than in the parental cells ( < 0.05).
		                        		
		                        			CONCLUSIONS
		                        			The sorted cells from SKOV3 cells cultured in serum-free medium exhibit biological properties of tumor stem cells with strong VM formation ability, suggesting their role in VM formation.
		                        		
		                        		
		                        		
		                        			Carcinoma, Ovarian Epithelial
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Cell Movement
		                        			;
		                        		
		                        			Female
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Matrix Metalloproteinase 2
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Matrix Metalloproteinase 9
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Neoplastic Stem Cells
		                        			;
		                        		
		                        			cytology
		                        			;
		                        		
		                        			Neovascularization, Pathologic
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			Ovarian Neoplasms
		                        			;
		                        		
		                        			pathology
		                        			
		                        		
		                        	
            
Result Analysis
Print
Save
E-mail