1.Amyloid precursor protein regulates 5-fluorouracil resistance in human hepatocellular carcinoma cells by inhibiting the mitochondrial apoptotic pathway.
Xiao-Long WU ; Ying CHEN ; Wen-Cui KONG ; Zhong-Quan ZHAO
Journal of Zhejiang University. Science. B 2020;21(3):234-245
		                        		
		                        			
		                        			Hepatocellular carcinoma (HCC) is a malignant tumor with high morbidity and mortality globally. It accounts for the majority of primary liver cancer cases. Amyloid precursor protein (APP), a cell membrane protein, plays a vital role in the pathogenesis of Alzheimer's disease, and has been found to be implicated in tumor growth and metastasis. Therefore, to understand the relationship between APP and 5-fluorouracil (5-FU) resistance in liver cancer, Cell Counting Kit-8, apoptosis and cell cycle assays, western blotting, and reverse transcription-quantitative polymerase chain reaction (qPCR) analysis were performed. The results demonstrated that APP expression in Bel7402-5-FU cells was significantly up-regulated, as compared with that in Bel7402 cells. Through successful construction of APP-silenced (siAPP) and overexpressed (OE) Bel7402 cell lines, data revealed that the Bel7402-APP751-OE cell line was insensitive, while the Bel7402-siAPP cell line was sensitive to 5-FU in comparison to the matched control group. Furthermore, APP overexpression decreased, while APP silencing increased 5-FU-induced apoptosis in Bel7402 cells. Mechanistically, APP overexpression and silencing can regulate the mitochondrial apoptotic pathway and the expression of apoptotic suppressor genes (B-cell lymphoma-2 (Bcl-2) and B-cell lymphoma-extra large (Bcl-xl)). Taken together, these results preliminarily revealed that APP overexpression contributes to the resistance of liver cancer cells to 5-FU, providing a new perspective for drug resistance.
		                        		
		                        		
		                        		
		                        			Amyloid beta-Protein Precursor/physiology*
		                        			;
		                        		
		                        			Apoptosis/drug effects*
		                        			;
		                        		
		                        			Carcinoma, Hepatocellular/drug therapy*
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Drug Resistance, Neoplasm
		                        			;
		                        		
		                        			Fluorouracil/pharmacology*
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Liver Neoplasms/drug therapy*
		                        			;
		                        		
		                        			Mitochondria/physiology*
		                        			;
		                        		
		                        			Proto-Oncogene Proteins c-bcl-2/genetics*
		                        			;
		                        		
		                        			bcl-X Protein/genetics*
		                        			
		                        		
		                        	
2.Cyclooxygenase-2 promotes ovarian cancer cell migration and cisplatin resistance via regulating epithelial mesenchymal transition.
Lin DENG ; Ding-Qing FENG ; Bin LING
Journal of Zhejiang University. Science. B 2020;21(4):315-326
		                        		
		                        			OBJECTIVE:
		                        			Drug-resistance and metastasis are major reasons for the high mortality of ovarian cancer (OC) patients. Cyclooxygenase-2 (COX-2) plays a critical role in OC development. This study was designed to evaluate the effects of COX-2 on migration and cisplatin (cis-dichloro diammine platinum, CDDP) resistance of OC cells and explore its related mechanisms.
		                        		
		                        			METHODS:
		                        			Cell counting kit-8 (CCK-8) assay was used to detect the cytotoxicity effects of celecoxib (CXB) and CDDP on SKOV3 and ES2 cells. The effect of COX-2 on migration was evaluated via the healing test. Western blot and real-time quantitative polymerase chain reaction (qPCR) were used to analyze E-cadherin, vimentin, Snail, and Slug levels.
		                        		
		                        			RESULTS:
		                        			COX-2 promoted drug-resistance and cell migration. CXB inhibited these effects. The combination of CDDP and CXB increased tumor cell sensitivity, reduced the amount of CDDP required, and shortened treatment administration time. COX-2 upregulation increased the expression of Snail and Slug, resulting in E-cadherin expression downregulation and vimentin upregulation.
		                        		
		                        			CONCLUSIONS
		                        			COX-2 promotes cancer cell migration and CDDP resistance and may serve as a potential target for curing OC.
		                        		
		                        		
		                        		
		                        			Celecoxib/pharmacology*
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			Cell Movement
		                        			;
		                        		
		                        			Cisplatin/pharmacology*
		                        			;
		                        		
		                        			Cyclooxygenase 2/physiology*
		                        			;
		                        		
		                        			Drug Resistance, Neoplasm
		                        			;
		                        		
		                        			Epithelial-Mesenchymal Transition
		                        			;
		                        		
		                        			Female
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Ovarian Neoplasms/pathology*
		                        			;
		                        		
		                        			Polymerase Chain Reaction
		                        			
		                        		
		                        	
3.MicroRNAs involved in drug resistance of breast cancer by regulating autophagy.
Nan WEN ; Qing LV ; Zheng-Gui DU
Journal of Zhejiang University. Science. B 2020;21(9):690-702
		                        		
		                        			
		                        			Autophagy is a conserved catabolic process characterized by degradation and recycling of cytosolic components or organelles through a lysosome-dependent pathway. It has a complex and close relationship to drug resistance in breast cancer. MicroRNAs (miRNAs) are small noncoding molecules that can influence numerous cellular processes including autophagy, through the posttranscriptional regulation of gene expression. Autophagy is regulated by many proteins and pathways, some of which in turn have been found to be regulated by miRNAs. These miRNAs may affect the drug resistance of breast cancer. Drug resistance is the main cause of distant recurrence, metastasis and death in breast cancer patients. In this review, we summarize the causative relationship between autophagy and drug resistance of breast cancer. The roles of autophagy-related proteins and pathways and their associated miRNAs in drug resistance of breast cancer are also discussed.
		                        		
		                        		
		                        		
		                        			Autophagy/physiology*
		                        			;
		                        		
		                        			Breast Neoplasms/pathology*
		                        			;
		                        		
		                        			Drug Resistance, Neoplasm
		                        			;
		                        		
		                        			Female
		                        			;
		                        		
		                        			Gene Expression Regulation, Neoplastic
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			MicroRNAs/physiology*
		                        			;
		                        		
		                        			Signal Transduction/physiology*
		                        			
		                        		
		                        	
4.Complex interplay between tumor microenvironment and cancer therapy.
Frontiers of Medicine 2018;12(4):426-439
		                        		
		                        			
		                        			Tumor microenvironment (TME) is comprised of cellular and non-cellular components that exist within and around the tumor mass. The TME is highly dynamic and its importance in different stages of cancer progression has been well recognized. A growing body of evidence suggests that TME also plays pivotal roles in cancer treatment responses. TME is significantly remodeled upon cancer therapies, and such change either enhances the responses or induces drug resistance. Given the importance of TME in tumor progression and therapy resistance, strategies that remodel TME to improve therapeutic responses are under developing. In this review, we provide an overview of the essential components in TME and the remodeling of TME in response to anti-cancer treatments. We also summarize the strategies that aim to enhance therapeutic efficacy by modulating TME.
		                        		
		                        		
		                        		
		                        			Antineoplastic Agents
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Drug Resistance
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Neoplasm Staging
		                        			;
		                        		
		                        			Neoplasms
		                        			;
		                        		
		                        			drug therapy
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			Treatment Outcome
		                        			;
		                        		
		                        			Tumor Microenvironment
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			physiology
		                        			
		                        		
		                        	
5.MiR-145 inhibits drug resistance to Oxaliplatin in colorectal cancer cells through regulating G protein coupled receptor 98.
Qiang FU ; Jing CHENG ; Jindai ZHANG ; Yonglei ZHANG ; Xiaobing CHEN ; Jianguo XIE ; Suxia LUO
Chinese Journal of Gastrointestinal Surgery 2017;20(5):566-570
OBJECTIVETo predict and identify the target gene of miR-145, and to explore the underlying mechanism of the inhibition of miR-145 on drug resistance to Oxaliplatin (L-OHP) in human colorectal cancer cells.
METHODSL-OHP-resistant human colorectal cancer cell line (HCT116/L-OHP) was established in vitro by exposing to increased concentrations of L-OHP in cell culture medium. MiR-145-mimics and its negative control (NC-miRNA) were transfected into HCT116/L-OHP cells using liposome to establish HCT116/L-OHPover-expressing miR-145 and HCT116/L-OHP. The target genes of miR-145 were predicted by bioinformatic analysis, and validated by dual luciferase activity assay. After determination of G protein coupled receptor 98(GPR98) as target gene, corresponding plasmids were constructed and transfected to establish HCT116/L-OHPover-expressing GPR98 and HCT116/L-OHP. HCT116/L-OHP cells over-expressing both GPR98 and miR-145 (HCT116/L-OHP) were acquired through modification of the binding sites of GPR98 cDNA with miR-145. CCK-8 assay was used to assess the proliferation (A value) and sensitivity to L-OHP (the lower the IC50, the stronger the sensitivity) in HCT116/L-OHP cells. Real-time quantitative PCR was used to measure the mRNA expression of miR-145 and GPR98. Western blot was used to examine the protein expression of GPR98 and drug-resistant associated protein, such as P-glycoprotein (gp), multiple drug-resistance protein 1(MRP1), cancer-inhibition gene PTEN.
RESULTSHCT116/L-OHP cell line was successfully established with ICof (42.34±1.05) mg/L and miR-145 mRNA expression of 0.27±0.04, which was higher than (9.81±0.95) mg/L (t=39.784, P=0.000) and lower than 1.00±0.09 (t=13.021, P=0.000) in HCT116 cells. Based on HCT116/L-OHP cells, HCT116/L-OHPcells were established successfully, with relative miR-145 expression of 10.01±1.05, which was higher than 1.06±0.14 in HCT116/L-OHPand 1.00±0.16 in HCT116/L-OHP (F=161.797, P=0.000). GPR98 was identified to be the target gene of miR-145. The relative mRNA and protein expressions of GPR98 in HCT116/L-OHPcells were 8.48±0.46 and 1.71±0.09, respectively, which were higher than those in HCT116/L-OHP(mRNA: 3.65±0.40, protein: 1.21±0.10) and HCT116/L-OHP (mRNA: 3.49±0.35, protein: 1.22±0.08; all P<0.05). The A value was 1.31±0.10, and the relative protein expressions of P-gp and MRP1 were 1.53±0.18 and 1.49±0.20 in HCT116/L-OHPcells, which were higher than those in HCT116/L-OHP (A value: 0.82±0.08, relative protein expression: 1.00±0.06 and 1.21±0.13, all P<0.05). The A value was 0.89±0.08, and the relative protein expressions of P-gp and MRP were 1.02±0.24 and 1.38±0.25 in HCT116/L-OHPcells, which were higher than those in HCT116/L-OHP(A value: 0.20±0.05, relative protein expression: 0.20±0.07, 0.55±0.10, all P<0.05). The relative protein expression of PTEN in HCT116/L-OHPcells was 0.12±0.03, which was lower than 1.25±0.14 in HCT116/L-OHP cells(P<0.05). In addition, relative protein expressions of P-gp and MRP1 were 1.02±0.24 and 1.38±0.25 in HCT116/L-OHPcells, which were higher than those in HCT116/L-OHPcells (0.20±0.07 and 0.55±0.10), while PTEN expression in HCT116/L-OHPcells was lower as compared to HCT116/L-OHPcells (1.41±0.16 vs. 1.98±0.13, P<0.05).
CONCLUSIONMiR-145 inhibits drug resistance to L-OHP of HCT116 cells through suppressing the expression of target gene GPR98.
ATP Binding Cassette Transporter, Sub-Family B ; drug effects ; ATP-Binding Cassette, Sub-Family B, Member 1 ; drug effects ; Cell Line, Tumor ; drug effects ; physiology ; Colorectal Neoplasms ; physiopathology ; Down-Regulation ; drug effects ; genetics ; Drug Resistance, Neoplasm ; drug effects ; genetics ; physiology ; HCT116 Cells ; drug effects ; physiology ; Humans ; In Vitro Techniques ; MicroRNAs ; genetics ; pharmacology ; Multidrug Resistance-Associated Proteins ; drug effects ; Organoplatinum Compounds ; pharmacology ; PTEN Phosphohydrolase ; drug effects ; RNA, Messenger ; Receptors, G-Protein-Coupled ; drug effects ; genetics
6.Research Progress on Drug-resistance of Acute Lymphoblastic Leukemia--Review.
Jing-Jing MA ; Yue CHEN ; Liang YU
Journal of Experimental Hematology 2016;24(1):261-265
		                        		
		                        			
		                        			Acute lymphoblastic leukemia (ALL) is a malignant clonal disease, its treatment methods include chemotherapy, hematopoietic stem cell transplantation, immunotherapy and molecular targeted therapy. Clinically, ALL patients need to get complete remission through chemotherapy, and then choose the other treatment according to the patient's condition. But the drug resistance has been a biggest obstacle in treatment of ALL. There are many research reports about drug-resistant of ALL at present. In this review, the classic drug resistance mechanisms, such as membrane transporter, gene modifications and some newly finding mechanisms including such as bone marrow microenvironment and Micro RNA and so on are summarized.
		                        		
		                        		
		                        		
		                        			Bone Marrow
		                        			;
		                        		
		                        			physiology
		                        			;
		                        		
		                        			Cellular Microenvironment
		                        			;
		                        		
		                        			Drug Resistance, Neoplasm
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Membrane Transport Proteins
		                        			;
		                        		
		                        			physiology
		                        			;
		                        		
		                        			MicroRNAs
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			Precursor Cell Lymphoblastic Leukemia-Lymphoma
		                        			;
		                        		
		                        			drug therapy
		                        			
		                        		
		                        	
7.Aberrant DNA methylation and its targeted therapy in acute myeloid leukemia.
Xueying LI ; Lixia ZHU ; Xiujin YE
Journal of Zhejiang University. Medical sciences 2016;45(4):387-394
		                        		
		                        			
		                        			The occurrence and development of acute myeloid leukemia (AML) is not only related to gene mutations, but also influenced by abnormal epigenetic regulation, in which DNA methylation is one of the most important mechanisms. Abnormal DNA methylation may lead to the activation of oncogene and the inactivation of tumor suppressor gene, resulting in the occurrence of leukemia. The mutations of DNA methylation enzymes associated with AML may have certain characteristics. The AML with recurrent cytogenetic abnormalities is also related to abnormal methylation. Some fusion genes can alter DNA methylation status to participate in the pathogenesis of leukemia. In addition, chemotherapy drug resistance in patients with AML is associated with the change of gene methylation status. Considering the reversibility of the epigenetic modification, targeted methylation therapy has become a hotspot of AML research.
		                        		
		                        		
		                        		
		                        			DNA Methylation
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			physiology
		                        			;
		                        		
		                        			DNA Modification Methylases
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			physiology
		                        			;
		                        		
		                        			Drug Resistance, Neoplasm
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			Epigenesis, Genetic
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			physiology
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Leukemia, Myeloid, Acute
		                        			;
		                        		
		                        			etiology
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			pathology
		                        			;
		                        		
		                        			Mutation
		                        			;
		                        		
		                        			genetics
		                        			
		                        		
		                        	
8.Autophagy in 5-Fluorouracil Therapy in Gastrointestinal Cancer: Trends and Challenges.
Jia-Cheng TANG ; Yi-Li FENG ; Xiao LIANG ; Xiu-Jun CAI
Chinese Medical Journal 2016;129(4):456-463
OBJECTIVE5-Fluorouracil (5-FU)-based combination therapies are standard treatments for gastrointestinal cancer, where the modulation of autophagy is becoming increasingly important in offering effective treatment for patients in clinical practice. This review focuses on the role of autophagy in 5-FU-induced tumor suppression and cancer therapy in the digestive system.
DATA SOURCESAll articles published in English from 1996 to date those assess the synergistic effect of autophagy and 5-FU in gastrointestinal cancer therapy were identified through a systematic online search by use of PubMed. The search terms were "autophagy" and "5-FU" and ("colorectal cancer" or "hepatocellular carcinoma" or "pancreatic adenocarcinoma" or "esophageal cancer" or "gallbladder carcinoma" or "gastric cancer").
STUDY SELECTIONCritical reviews on relevant aspects and original articles reporting in vitro and/or in vivo results regarding the efficiency of autophagy and 5-FU in gastrointestinal cancer therapy were reviewed, analyzed, and summarized. The exclusion criteria for the articles were as follows: (1) new materials (e.g., nanomaterial)-induced autophagy; (2) clinical and experimental studies on diagnostic and/or prognostic biomarkers in digestive system cancers; and (3) immunogenic cell death for anticancer chemotherapy.
RESULTSMost cell and animal experiments showed inhibition of autophagy by either pharmacological approaches or via genetic silencing of autophagy regulatory gene, resulting in a promotion of 5-FU-induced cancer cells death. Meanwhile, autophagy also plays a pro-death role and may mediate cell death in certain cancer cells where apoptosis is defective or difficult to induce. The dual role of autophagy complicates the use of autophagy inhibitor or inducer in cancer chemotherapy and generates inconsistency to an extent in clinic trials.
CONCLUSIONAutophagy might be a therapeutic target that sensitizes the 5-FU treatment in gastrointestinal cancer.
Antimetabolites, Antineoplastic ; therapeutic use ; Autophagy ; physiology ; Drug Resistance, Neoplasm ; Fluorouracil ; therapeutic use ; Gastrointestinal Neoplasms ; drug therapy ; pathology ; Humans
9.Effects of methylseleninic acid on cisplatin-resistant ovarian cancer cells (SKOV3/DDP) and the mechanisms.
Ying TAN ; Qing FENG ; Xin SUN ; Min XUE ; Ning JIANG ; Xinliang DENG
Journal of Central South University(Medical Sciences) 2016;41(12):1305-1311
		                        		
		                        			
		                        			To investigate the reversal effect of methylseleninic acid on cisplatin (DDP)-resistant ovarian cancer cells and the underlying mechanisms.
 Methods: SKOV3/DDP cells were incubated with cisplatin at different concentrations for 48 h, then the proliferation rate of SKOV3/DDP cells was detected by MTT assays, and the expression of β-catenin in SKOV3/DDP cells was examined by Western blot. The inhibitory effect of methyl-seleninic acid (MSA) combined with DDP at different concentrations on SKOV3/DDP cells was assayed by MTT method. Western blot was used to detect the expression of β-catenin protein in the cells.
 Results: The inhibitory rate for proliferation in DDP-treated SKOV3/DDP cells with different concentrations is lower than that in the SKOV3 cells (P<0.05); β-catenin expression in SKOV3/DDP cells was significantly higher than that in the SKOV3 cells (P<0.05). The inhibitory rate for proliferation in SKOV3/DDP cells with different concentrations of MSA was increased with the increase in concentration (P<0.05). The inhibitory rate for proliferation in SKOV3/DDP cells with 2 or 6 μmol/L MSA plus cisplatin was lower than that in cisplatin alone group (P<0.05). β-catenin expression in SKOV3 /DDP cells with 2 or 6 μmol/L MSA plus cisplatin was higher than that in the cisplatin alone group (P<0.05).
 Conclusion: MSA can reverse cisplatin resistance on SKOV3 / DDP cells, which may be related to the decrease in β-catenin expression.
		                        		
		                        		
		                        		
		                        			Antineoplastic Agents
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Carcinoma
		                        			;
		                        		
		                        			physiopathology
		                        			;
		                        		
		                        			Cell Line, Tumor
		                        			;
		                        		
		                        			physiology
		                        			;
		                        		
		                        			Cell Proliferation
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Cisplatin
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Drug Resistance, Neoplasm
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			Female
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Organoselenium Compounds
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Ovarian Neoplasms
		                        			;
		                        		
		                        			physiopathology
		                        			;
		                        		
		                        			beta Catenin
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			metabolism
		                        			
		                        		
		                        	
10.Influence of HMGB1/MAPK/m-TOR signaling pathway on cell autophagy and chemotherapy resistance in K562 cells.
Liying LIU ; Fei GAO ; Yanqiong YE ; Zhiheng CHEN ; Yunpeng DAI ; Ping ZHAO ; Guotao GUAN ; Mingyi ZHAO
Journal of Central South University(Medical Sciences) 2016;41(10):1016-1023
		                        		
		                        			
		                        			To observe the effect of high-mobility group box 1 (HMGB1) on autophagy and chemotherapy resistance in human leukemiacell line (K562) cells, and to explore the underlying mechanisms.
 Methods: The K562 cells were cultured in vitro and divided into 6 groups: a chemotherapeutic group, a chemotherapeutic control group, a HMGB1 preconditioning group, a HMGB1 preconditioning control group, a HMGB1 siRNA group and a siRNA control group. The chemotherapeutic group was further divided into a vincristine (VCR) group, an etoposide (VP-16) group, a cytosine arabinoside (Ara-C) group, a adriamycin (ADM) group and a arsenic trioxide (As2O3) group. The cell activity was evaluated by cell counting kit-8. The protein levels of HMGB1, microtubule-associate protein1light chain3 (LC3), AMP-activated protein kinase (AMPK) and mammalian target of rapamycin (m-TOR) were determined by Western blotting. The level of serum HMGB1 was evaluated by enzyme-linked immunosorbent assay (ELISA). The autophagy was examined by monodansylcadaverine staining and observed under transmission electron microscopy.
 Results: Compared with the control group, the cell activity was significantly decreased and the level of serum HMGB1 was significantly increased in the chemotherapeutic (VCR, VP-16, Ara-C, ADM and As2O3) groups (all P<0.05). Compared with the control group, the cell activity and the level of serum HMGB1 were significantly increased in the HMGB1 preconditioning group (both P<0.05). Compared with the siRNA control group, the cell activity and the level of serum HMGB1 were significantly decreased in the HMGB1 siRNA group (both P<0.05). Compared with the control group, the expression of LC3-II and the formation of autophagic bodies were increased in the HMGB1 preconditioning group (both P<0.05), the p-AMPK expression was increased and p-mTOR expression was decreased (both P<0.05).
 Conclusion: HMGB1 can increase the autophagy and promote chemotherapy resistance through the pathway of AMPK/m-TOR in K562 cells.
		                        		
		                        		
		                        		
		                        			AMP-Activated Protein Kinases
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			physiology
		                        			;
		                        		
		                        			Arsenic Trioxide
		                        			;
		                        		
		                        			Arsenicals
		                        			;
		                        		
		                        			Autophagy
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			Cytarabine
		                        			;
		                        		
		                        			Doxorubicin
		                        			;
		                        		
		                        			Drug Resistance, Neoplasm
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			physiology
		                        			;
		                        		
		                        			Etoposide
		                        			;
		                        		
		                        			HMGB1 Protein
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			physiology
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			K562 Cells
		                        			;
		                        		
		                        			physiology
		                        			;
		                        		
		                        			Microtubule-Associated Proteins
		                        			;
		                        		
		                        			Oxides
		                        			;
		                        		
		                        			RNA, Small Interfering
		                        			;
		                        		
		                        			Signal Transduction
		                        			;
		                        		
		                        			TOR Serine-Threonine Kinases
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			physiology
		                        			;
		                        		
		                        			Vincristine
		                        			
		                        		
		                        	
            
Result Analysis
Print
Save
E-mail