1.Advances of the regulatory mechanism of cyclin, cyclin- dependent kinases and related kinase inhibitors in cell cycle progression.
Jianfeng PAN ; Fangzheng SHANG ; Rong MA ; Youjun RONG ; Yanjun ZHANG
Chinese Journal of Biotechnology 2023;39(4):1525-1547
		                        		
		                        			
		                        			Cell cycle plays a crucial role in cell development. Cell cycle progression is mainly regulated by cyclin dependent kinase (CDK), cyclin and endogenous CDK inhibitor (CKI). Among these, CDK is the main cell cycle regulator, binding to cyclin to form the cyclin-CDK complex, which phosphorylates hundreds of substrates and regulates interphase and mitotic progression. Abnormal activity of various cell cycle proteins can cause uncontrolled proliferation of cancer cells, which leads to cancer development. Therefore, understanding the changes in CDK activity, cyclin-CDK assembly and the role of CDK inhibitors will help to understand the underlying regulatory processes in cell cycle progression, as well as provide a basis for the treatment of cancer and disease and the development of CDK inhibitor-based therapeutic agents. This review focuses on the key events of CDK activation or inactivation, and summarizes the regulatory processes of cyclin-CDK at specific times and locations, as well as the progress of research on relevant CDK inhibitor therapeutics in cancer and disease. The review concludes with a brief description of the current challenges of the cell cycle process, with the aim to provide scientific references and new ideas for further research on cell cycle process.
		                        		
		                        		
		                        		
		                        			Cyclin-Dependent Kinases/metabolism*
		                        			;
		                        		
		                        			Cyclins/metabolism*
		                        			;
		                        		
		                        			Protein Serine-Threonine Kinases
		                        			;
		                        		
		                        			Cell Cycle Proteins/metabolism*
		                        			;
		                        		
		                        			Cell Cycle/physiology*
		                        			;
		                        		
		                        			Cyclin-Dependent Kinase 2
		                        			
		                        		
		                        	
2.4E-BP1 counteracts human mesenchymal stem cell senescence via maintaining mitochondrial homeostasis.
Yifang HE ; Qianzhao JI ; Zeming WU ; Yusheng CAI ; Jian YIN ; Yiyuan ZHANG ; Sheng ZHANG ; Xiaoqian LIU ; Weiqi ZHANG ; Guang-Hui LIU ; Si WANG ; Moshi SONG ; Jing QU
Protein & Cell 2023;14(3):202-216
		                        		
		                        			
		                        			Although the mTOR-4E-BP1 signaling pathway is implicated in aging and aging-related disorders, the role of 4E-BP1 in regulating human stem cell homeostasis remains largely unknown. Here, we report that the expression of 4E-BP1 decreases along with the senescence of human mesenchymal stem cells (hMSCs). Genetic inactivation of 4E-BP1 in hMSCs compromises mitochondrial respiration, increases mitochondrial reactive oxygen species (ROS) production, and accelerates cellular senescence. Mechanistically, the absence of 4E-BP1 destabilizes proteins in mitochondrial respiration complexes, especially several key subunits of complex III including UQCRC2. Ectopic expression of 4E-BP1 attenuates mitochondrial abnormalities and alleviates cellular senescence in 4E-BP1-deficient hMSCs as well as in physiologically aged hMSCs. These f indings together demonstrate that 4E-BP1 functions as a geroprotector to mitigate human stem cell senescence and maintain mitochondrial homeostasis, particularly for the mitochondrial respiration complex III, thus providing a new potential target to counteract human stem cell senescence.
		                        		
		                        		
		                        		
		                        			Mesenchymal Stem Cells/physiology*
		                        			;
		                        		
		                        			Cellular Senescence
		                        			;
		                        		
		                        			Homeostasis
		                        			;
		                        		
		                        			Cell Cycle Proteins/metabolism*
		                        			;
		                        		
		                        			Adaptor Proteins, Signal Transducing/metabolism*
		                        			;
		                        		
		                        			Mitochondria/metabolism*
		                        			;
		                        		
		                        			Electron Transport Complex III/metabolism*
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Cells, Cultured
		                        			
		                        		
		                        	
3.Discovery of small molecule degraders for modulating cell cycle.
Liguo WANG ; Zhouli YANG ; Guangchen LI ; Yongbo LIU ; Chao AI ; Yu RAO
Frontiers of Medicine 2023;17(5):823-854
		                        		
		                        			
		                        			The cell cycle is a complex process that involves DNA replication, protein expression, and cell division. Dysregulation of the cell cycle is associated with various diseases. Cyclin-dependent kinases (CDKs) and their corresponding cyclins are major proteins that regulate the cell cycle. In contrast to inhibition, a new approach called proteolysis-targeting chimeras (PROTACs) and molecular glues can eliminate both enzymatic and scaffold functions of CDKs and cyclins, achieving targeted degradation. The field of PROTACs and molecular glues has developed rapidly in recent years. In this article, we aim to summarize the latest developments of CDKs and cyclin protein degraders. The selectivity, application, validation and the current state of each CDK degrader will be overviewed. Additionally, possible methods are discussed for the development of degraders for CDK members that still lack them. Overall, this article provides a comprehensive summary of the latest advancements in CDK and cyclin protein degraders, which will be helpful for researchers working on this topic.
		                        		
		                        		
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Cell Cycle/physiology*
		                        			;
		                        		
		                        			Cell Division
		                        			;
		                        		
		                        			Cyclin-Dependent Kinases/metabolism*
		                        			;
		                        		
		                        			Cyclins/metabolism*
		                        			
		                        		
		                        	
4.Carfilzomib inhibits the growth of lung adenocarcinoma via upregulation of Gadd45a expression.
Fang YANG ; Wang-Wang LIU ; Hui CHEN ; Jia ZHU ; Ai-Hua HUANG ; Fei ZHOU ; Yi GAN ; Yan-Hua ZHANG ; Li MA
Journal of Zhejiang University. Science. B 2020;21(1):64-76
		                        		
		                        			
		                        			Proteasome inhibitors have shown remarkable success in the treatment of hematologic neoplasm. There has been a lot of attention to applying these drugs for solid tumor treatment. Recent preclinical study has signified the effectiveness on cell proliferation inhibition in lung adenocarcinoma treated by carfilzomib (CFZ), a second generation proteasome inhibitor. However, no insight has been gained regarding the mechanism. In this study, we have systematically investigated the CFZ functions in cell proliferation and growth, cell cycle arrest, and apoptosis in lung adenocarcinoma cells. Flow cytometry experiments showed that CFZ significantly induced G2/M cell cycle arrest and apoptosis in lung adenocarcinoma. MTS and colony formation assays revealed that CFZ substantially inhibited survival of lung adenocarcinoma cells. All results were consistently correlated to the upregulation expression of Gadd45a, which is an important gene in modulating cell cycle arrest and apoptosis in response to physiologic and environmental stresses. Here, upregulation of Gadd45a expression was observed after CFZ treatment. Knocking down Gadd45a expression suppressed G2/M arrest and apoptosis in CFZ-treated cells, and reduced cytotoxicity of this drug. The protein expression analysis has further identified that the AKT/FOXO3a pathway is involved in Gadd45a upregulation after CFZ treatment. These findings unveil a novel mechanism of proteasome inhibitor in anti-solid tumor activity, and shed light on novel preferable therapeutic strategy for lung adenocarcinoma. We believe that Gadd45a expression can be a highly promising candidate predictor in evaluating the efficacy of proteasome inhibitors in solid tumor therapy.
		                        		
		                        		
		                        		
		                        			Adenocarcinoma of Lung/pathology*
		                        			;
		                        		
		                        			Apoptosis/drug effects*
		                        			;
		                        		
		                        			Cell Cycle Checkpoints/drug effects*
		                        			;
		                        		
		                        			Cell Cycle Proteins/genetics*
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Forkhead Box Protein O3/physiology*
		                        			;
		                        		
		                        			Gene Expression Regulation, Neoplastic/drug effects*
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Lung Neoplasms/pathology*
		                        			;
		                        		
		                        			Oligopeptides/pharmacology*
		                        			;
		                        		
		                        			Proto-Oncogene Proteins c-akt/physiology*
		                        			;
		                        		
		                        			Up-Regulation
		                        			
		                        		
		                        	
5.Role of LINC00152 in non-small cell lung cancer.
Journal of Zhejiang University. Science. B 2020;21(3):179-191
		                        		
		                        			
		                        			Non-small cell lung cancer (NSCLC) accounts for about 85% of all lung cancer cases. The pathogenesis of NSCLC involves complex gene networks that include different types of non-coding RNAs, such as long non-coding RNAs (lncRNAs). The role of lncRNAs in NSCLC is gaining an increasing interest as their function is being explored in various human cancers. Recently, a new oncogenic lncRNA, LINC00152 (cytoskeleton regulator RNA (CYTOR)), has been identified in different tumor types. In NSCLC, the high expression of LINC00152 in tumor tissue and peripheral blood samples has been shown to be associated with worse prognoses of NSCLC patients. Overexpression of LINC00152 has been confirmed to promote the proliferation, invasion, and migration of NSCLC cells in vitro, as well as increase tumor growth in vivo. This review discusses the role of LINC00152 in NSCLC.
		                        		
		                        		
		                        		
		                        			Apoptosis
		                        			;
		                        		
		                        			Biomarkers, Tumor/blood*
		                        			;
		                        		
		                        			Carcinoma, Non-Small-Cell Lung/radiotherapy*
		                        			;
		                        		
		                        			Cell Cycle Checkpoints
		                        			;
		                        		
		                        			Computational Biology
		                        			;
		                        		
		                        			Epithelial-Mesenchymal Transition
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Lung Neoplasms/radiotherapy*
		                        			;
		                        		
		                        			Prognosis
		                        			;
		                        		
		                        			RNA, Long Noncoding/physiology*
		                        			;
		                        		
		                        			Radiation Tolerance
		                        			
		                        		
		                        	
6.GSK923295 as a potential antihepatocellular carcinoma agent causing delay on liver regeneration after partial hepatectomy.
Jia-Cheng TANG ; Ke WU ; Xing ZHENG ; Ming XU ; Yi DAI ; Sai-Sai WEI ; Xiu-Jun CAI
Chinese Medical Journal 2019;132(3):311-318
		                        		
		                        			BACKGROUND:
		                        			The clinical trials emerged centromere protein E inhibitor GSK923295 as a promising anticancer drug, but its function in hepatocellular carcinoma (HCC) remain needs to be fully elucidated, especially as chemotherapy after hepatectomy for liver tumors. We aimed to describe anti-HCC activities of GSK923295 and compare its antiproliferative effects on liver regeneration after partial hepatectomy (PH).
		                        		
		                        			METHODS:
		                        			All subjects were randomized to treatment with either vehicle or GSK923295. Antitumor activity of GSK923295 was assessed by xenograft growth assays. The C57BL/6 mice were subjected to 70% PH and the proliferation was calculated by liver coefficient, further confirmed by immunohistochemistry. The proliferation and cell cycle analysis of liver cell AML12 and HCC cells LM3, HUH7, and HepG2 were investigated using the cell counting kit-8 assay and Flow Cytometry. The chromosome misalignment and segregation in AML12 cells were visualized by immunofluorescence.
		                        		
		                        			RESULTS:
		                        			Treatment with GSK923295 induced antiproliferation in HCC cell lines. It also caused delay on HCC tumor growth instead of regression both in a HCC cell line xenograft model and patient-derived tumor xenograft model. With microarray analysis, CENtromere Protein E was gradually increased in mouse liver after PH. Exposure of liver cells to GSK923295 resulted in delay on a cell cycle in mitosis with a phenotype of misaligned chromosomes and chromosomes clustered. In 70% PH mouse model, GSK923295 treatment also remarkably reduced liver regeneration in later stage, in parallel with the mitotic marker phospho-histone H3 elevation.
		                        		
		                        			CONCLUSION
		                        			The anticancer drug GSK923295 causes a significant delay on HCC tumor growth and liver regeneration after PH in later stage.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Antineoplastic Agents
		                        			;
		                        		
		                        			therapeutic use
		                        			;
		                        		
		                        			Blotting, Western
		                        			;
		                        		
		                        			Bridged Bicyclo Compounds, Heterocyclic
		                        			;
		                        		
		                        			therapeutic use
		                        			;
		                        		
		                        			Carcinoma, Hepatocellular
		                        			;
		                        		
		                        			drug therapy
		                        			;
		                        		
		                        			surgery
		                        			;
		                        		
		                        			Cell Cycle
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Cell Proliferation
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Chromosomal Proteins, Non-Histone
		                        			;
		                        		
		                        			antagonists & inhibitors
		                        			;
		                        		
		                        			Electrophoresis, Polyacrylamide Gel
		                        			;
		                        		
		                        			Female
		                        			;
		                        		
		                        			Fluorescent Antibody Technique
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Immunohistochemistry
		                        			;
		                        		
		                        			Liver Neoplasms
		                        			;
		                        		
		                        			drug therapy
		                        			;
		                        		
		                        			surgery
		                        			;
		                        		
		                        			Liver Regeneration
		                        			;
		                        		
		                        			physiology
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Mice, Inbred C57BL
		                        			;
		                        		
		                        			Real-Time Polymerase Chain Reaction
		                        			;
		                        		
		                        			Sarcosine
		                        			;
		                        		
		                        			analogs & derivatives
		                        			;
		                        		
		                        			therapeutic use
		                        			;
		                        		
		                        			Xenograft Model Antitumor Assays
		                        			
		                        		
		                        	
7.Cardiac Hypertrophy is Positively Regulated by MicroRNA‑24 in Rats
Juan GAO ; Min ZHU ; Rui-Feng LIU ; Jian-Shu ZHANG ; Ming XU
Chinese Medical Journal 2018;131(11):1333-1341
BackgroundMicroRNA-24 (miR-24) plays an important role in heart failure by reducing the efficiency of myocardial excitation-contraction coupling. Prolonged cardiac hypertrophy may lead to heart failure, but little is known about the role of miR-24 in cardiac hypertrophy. This study aimed to preliminarily investigate the function of miR-24 and its mechanisms in cardiac hypertrophy.
MethodsTwelve Sprague-Dawley rats with a body weight of 50 ± 5 g were recruited and randomly divided into two groups: a transverse aortic constriction (TAC) group and a sham surgery group. Hypertrophy index was measured and calculated by echocardiography and hematoxylin and eosin staining. TargetScans algorithm-based prediction was used to search for the targets of miR-24, which was subsequently confirmed by a real-time polymerase chain reaction and luciferase assay. Immunofluorescence labeling was used to measure the cell surface area, and H-leucine incorporation was used to detect the synthesis of total protein in neonatal rat cardiac myocytes (NRCMs) with the overexpression of miR-24. In addition, flow cytometry was performed to observe the alteration in the cell cycle. Statistical analysis was carried out with GraphPad Prism v5.0 and SPSS 19.0. A two-sided P < 0.05 was considered as the threshold for significance.
ResultsThe expression of miR-24 was abnormally increased in TAC rat cardiac tissue (t = -2.938, P < 0.05). TargetScans algorithm-based prediction demonstrated that CDKN1B (p27, Kip1), a cell cycle regulator, was a putative target of miR-24, and was confirmed by luciferase assay. The expression of p27 was decreased in TAC rat cardiac tissue (t = 2.896, P < 0.05). The overexpression of miR-24 in NRCMs led to the decreased expression of p27 (t = 4.400, P < 0.01), and decreased G0/G1 arrest in cell cycle and cardiomyocyte hypertrophy.
ConclusionMiR-24 promotes cardiac hypertrophy partly by affecting the cell cycle through down-regulation of p27 expression.
Animals ; Cardiomegaly ; genetics ; pathology ; Cell Cycle ; genetics ; physiology ; Cyclin-Dependent Kinase Inhibitor p27 ; genetics ; metabolism ; Male ; MicroRNAs ; genetics ; Myocardium ; metabolism ; Myocytes, Cardiac ; cytology ; metabolism ; Rats ; Rats, Sprague-Dawley
8.Effects of Nerve Growth Factor on Cardiac Fibroblasts Proliferation, Cell Cycle, Migration, and Myofibroblast Transformation.
Chinese Medical Journal 2018;131(7):813-817
BackgroundRecent research indicates that nerve growth factor (NGF) promotes cardiac repair following myocardial infarction by promoting angiogenesis and cardiomyocyte survival. The purpose of this study was to investigate the effects of NGF on cardiac fibroblasts (CFs) proliferation, cell cycle, migration, and myofibroblast transformation in vitro.
MethodsCFs were obtained from ventricles of neonatal Sprague-Dawley rats and incubated with various concentrations of NGF (0, 0.01, 0.1, 1, 10, and 100 ng/ml; 0 ng/ml was designated as the control group). Cell proliferation and cell cycle of the CFs were measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and flow cytometry (FCM), respectively. A cell scratch wound model and transwell were carried out to observe effects of NGF on migration of CFs after 24 h of culture. Real-time polymerase chain reaction (RT-PCR) and Western blotting were used to measure α-smooth muscle actin (α-SMA) at mRNA and protein levels after CFs were incubated with various concentrations of NGF.
ResultsExpression of α-SMA measured by RT-PCR and Western blotting significantly increased in the 1 and 10 ng/ml NGF groups (P < 0.05). Absorbance values of CFs showed that NGF did not influence the proliferation of CFs (The Avalues were 0.178 ± 0.038, 0.182 ± 0.011, 0.189 ± 0.005, 0.178 ± 0.010, 0.185 ± 0.025, and 0.177 ± 0.033, respectively, in the 0, 0.01, 0.1, 1, 10, and 100 ng/ml NGF groups [P = 0.800, 0.428, 0.981, 0.596, and 0.913, respectively, compared with control group]), and FCM analysis showed that the percentage of CFs in G0/G1, S, and G2/M phases was not changed (P > 0.05). The cell scratch wound model and transwell showed that CFs migration was not significantly different (P > 0.05).
ConclusionNGF induces myofibroblast transformation but does not influence proliferation, cell cycle, or migration of CFs in vitro.
Actins ; metabolism ; Animals ; Cell Cycle ; drug effects ; physiology ; Cell Movement ; drug effects ; physiology ; Cell Proliferation ; physiology ; Cells, Cultured ; Myofibroblasts ; cytology ; drug effects ; Nerve Growth Factor ; metabolism ; pharmacology ; Rats ; Rats, Sprague-Dawley
9.Effect of Upregulated DNA Replication and Sister Chromatid Cohesion 1 Expression on Proliferation and Prognosis in Hepatocellular Carcinoma.
Xing-Wang XIE ; Xue-Yan WANG ; Wei-Jia LIAO ; Ran FEI ; Xu CONG ; Qian CHEN ; Lai WEI ; Hong-Song CHEN ; Yu WANG
Chinese Medical Journal 2018;131(23):2827-2835
		                        		
		                        			Background:
		                        			DNA replication and sister chromatid cohesion 1 (DSCC1) (also called DCC1) is a component of an alternative replication factor C complex that loads proliferating cell nuclear antigen onto DNA during S phase of the cell cycle. It is located at 8q24 and frequently amplified in hepatocellular carcinoma (HCC). However, the role of DSCC1 in the carcinogenesis and progress of HCC has not been fully investigated. Here, we aimed to assert the importance of DSCC1 in the HCC.
		                        		
		                        			Methods:
		                        			In this study, copy number variation data and RNA sequencing data were used to calculate the DNA copy number and mRNA expression of DSCC1 in HCC. Quantitative polymerase chain reaction, Western blotting, and immunohistochemistry analysis were used to determine the mRNA and protein level of DSCC1 in HCC. The Kaplan-Meier analysis and univariate and multivariate Cox regression analysis were used to assess the association of DSCC1 with the overall survival (OS) of HCC patients. Moreover, lentiviral shRNA was used to knockdown DSCC1, and then, colony-forming assay, cell cycle assay, and cell proliferation assay were performed to evaluate the impact of DSCC1 silencing on HCC cell lines.
		                        		
		                        			Results:
		                        			We found that DSCC1 was amplified and highly expressed in HCC tumor tissues than in nontumor tissues. We then found that the overexpression of both mRNA and protein of DSCC1 was linked to the bad prognosis of HCC patients. Astonishingly, the protein level of DSCC1 was an independent prognostic factor for OS (hazard ratio, 1.79; 95% confidence interval, 1.17-2.74; P = 0.007). Furthermore, the clonogenic capacity of DSCC1-amplified HCC cell lines (MHCC-97H, MHCC-97L, and Hep3B) was significantly inhibited by transduction of a lentiviral shRNA that targets DSCC1. We also showed that knockdown of DSCC1 induced G0-G1 cell cycle arrest (increased from 60% to more than 80%) and greatly inhibited the proliferation of HCC cell lines.
		                        		
		                        			Conclusion
		                        			These results suggest that DSCC1 is a putative HCC driver gene that promotes proliferation and is associated with poor prognosis in HCC.
		                        		
		                        		
		                        		
		                        			Blotting, Western
		                        			;
		                        		
		                        			Carcinoma, Hepatocellular
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			Cell Cycle
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			physiology
		                        			;
		                        		
		                        			Cell Cycle Checkpoints
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			physiology
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Cell Proliferation
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			physiology
		                        			;
		                        		
		                        			DNA Replication
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			physiology
		                        			;
		                        		
		                        			Female
		                        			;
		                        		
		                        			Hep G2 Cells
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Immunohistochemistry
		                        			;
		                        		
		                        			Liver Neoplasms
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			Male
		                        			;
		                        		
		                        			Middle Aged
		                        			;
		                        		
		                        			Multivariate Analysis
		                        			;
		                        		
		                        			Proportional Hazards Models
		                        			;
		                        		
		                        			Real-Time Polymerase Chain Reaction
		                        			
		                        		
		                        	
10.Restoration of Brain Acid Soluble Protein 1 Inhibits Proliferation and Migration of Thyroid Cancer Cells.
Run-Sheng GUO ; Yue YU ; Jun CHEN ; Yue-Yu CHEN ; Na SHEN ; Ming QIU
Chinese Medical Journal 2016;129(12):1439-1446
BACKGROUNDBrain acid soluble protein 1 (BASP1) is identified as a novel potential tumor suppressor in several cancers. However, its role in thyroid cancer has not been investigated yet. In the present study, the antitumor activities of BASP1 against the growth and migration of thyroid cancer cells were evaluated.
METHODSBASP1 expression in thyroid cancer tissues and normal tissues were examined by immunohistochemical staining and the association between its expression and prognosis was analyzed. pcDNA-BASP1 carrying full length of BASP1 cDNA was constructed to restore the expression of BASP1 in thyroid cancer cell lines (BHT-101 and KMH-2). The cell proliferation in vitro and in vivo was evaluated by WST-1 assay and xenograft tumor models, respectively. Cell cycle distribution after transfection was analyzed using flow cytometry. Cell apoptosis after transfection was examined by annexin V/propidium iodide assay. The migration was examined using transwell assay.
RESULTSBASP1 expression was abundant in normal tissues while it is significantly decreased in cancer tissues (P = 0.000). pcDNA-BASP1 restored the expression of BASP1 and significantly inhibited the growth of BHT-101 and KMH-2 cells as well as xenograft tumors in nude mice (P = 0.000). pcDNA-BASP1 induced G1 arrest and apoptosis in BHT-101 and KMH-2 cells. In addition, pcDNA-BASP1 significantly inhibited the cell migration.
CONCLUSIONSDownregulation of BASP1 expression may play a role in the tumorigenesis of thyroid cancer. Restoration of BASP1 expression exerted extensive antitumor activities against growth and migration of thyroid cancer cells, which suggested that BASP1 gene might act as a potential therapeutic agent for the treatment of thyroid cancer.
Aged ; Animals ; Apoptosis ; genetics ; physiology ; Calmodulin-Binding Proteins ; genetics ; metabolism ; Cell Cycle ; genetics ; physiology ; Cell Line, Tumor ; Cell Movement ; genetics ; physiology ; Cell Proliferation ; genetics ; physiology ; Cytoskeletal Proteins ; genetics ; metabolism ; Female ; Gene Expression Regulation, Neoplastic ; genetics ; physiology ; Humans ; Male ; Membrane Proteins ; genetics ; metabolism ; Mice ; Mice, Nude ; Middle Aged ; Nerve Tissue Proteins ; genetics ; metabolism ; Repressor Proteins ; genetics ; metabolism ; Thyroid Neoplasms ; metabolism ; pathology ; Xenograft Model Antitumor Assays
            
Result Analysis
Print
Save
E-mail