1.In Silico Screening of Potential Spike Glycoprotein Inhibitors of SARS-CoV-2 with Drug Repurposing Strategy.
Tian-Zi WEI ; Hao WANG ; Xue-Qing WU ; Yi LU ; Sheng-Hui GUAN ; Feng-Quan DONG ; Chen-le DONG ; Gu-Li ZHU ; Yu-Zhou BAO ; Jian ZHANG ; Guan-Yu WANG ; Hai-Ying LI
Chinese journal of integrative medicine 2020;26(9):663-669
		                        		
		                        			OBJECTIVE:
		                        			To select potential molecules that can target viral spike proteins, which may potentially interrupt the interaction between the human angiotension-converting enzyme 2 (ACE2) receptor and viral spike protein by virtual screening.
		                        		
		                        			METHODS:
		                        			The three-dimensional (3D)-coordinate file of the receptor-binding domain (RBD)-ACE2 complex for searching a suitable docking pocket was firstly downloaded and prepared. Secondly, approximately 15,000 molecular candidates were prepared, including US Food and Drug Administration (FDA)-approved drugs from DrugBank and natural compounds from Traditional Chinese Medicine Systems Pharmacology (TCMSP), for the docking process. Then, virtual screening was performed and the binding energy in Autodock Vina was calculated. Finally, the top 20 molecules with high binding energy and their Chinese medicine (CM) herb sources were listed in this paper.
		                        		
		                        			RESULTS:
		                        			It was found that digitoxin, a cardiac glycoside in DrugBank and bisindigotin in TCMSP had the highest docking scores. Interestingly, two of the CM herbs containing the natural compounds that had relatively high binding scores, Forsythiae fructus and Isatidis radix, are components of Lianhua Qingwen (), a CM formula reportedly exerting activity against severe acute respiratory syndrome (SARS)-Cov-2. Moreover, raltegravir, an HIV integrase inhibitor, was found to have a relatively high binding score.
		                        		
		                        			CONCLUSIONS
		                        			A class of compounds, which are from FDA-approved drugs and CM natural compounds, that had high binding energy with RBD of the viral spike protein. Our work provides potential candidates for other researchers to identify inhibitors to prevent SARS-CoV-2 infection, and highlights the importance of CM and integrative application of CM and Western medicine on treating COVID-19.
		                        		
		                        		
		                        		
		                        			China
		                        			;
		                        		
		                        			Computer Simulation
		                        			;
		                        		
		                        			Coronavirus Infections
		                        			;
		                        		
		                        			diagnosis
		                        			;
		                        		
		                        			drug therapy
		                        			;
		                        		
		                        			Drug Repositioning
		                        			;
		                        		
		                        			methods
		                        			;
		                        		
		                        			Drugs, Chinese Herbal
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Glycoproteins
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Imaging, Three-Dimensional
		                        			;
		                        		
		                        			Mass Screening
		                        			;
		                        		
		                        			methods
		                        			;
		                        		
		                        			Molecular Docking Simulation
		                        			;
		                        		
		                        			methods
		                        			;
		                        		
		                        			Pandemics
		                        			;
		                        		
		                        			Peptidyl-Dipeptidase A
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Pneumonia, Viral
		                        			;
		                        		
		                        			diagnosis
		                        			;
		                        		
		                        			drug therapy
		                        			;
		                        		
		                        			Protein Binding
		                        			;
		                        		
		                        			United States
		                        			;
		                        		
		                        			United States Food and Drug Administration
		                        			
		                        		
		                        	
2.Melatonin Augments the Effects of Fluoxetine on Depression-Like Behavior and Hippocampal BDNF-TrkB Signaling.
Kun LI ; Si SHEN ; Yu-Tian JI ; Xu-Yun LI ; Li-San ZHANG ; Xiao-Dong WANG
Neuroscience Bulletin 2018;34(2):303-311
		                        		
		                        			
		                        			Depression is a debilitating psychiatric disorder with a huge socioeconomic burden, and its treatment relies on antidepressants including selective serotonin reuptake inhibitors (SSRIs). Recently, the melatonergic system that is closely associated with the serotonergic system has been implicated in the pathophysiology and treatment of depression. However, it remains unknown whether combined treatment with SSRI and melatonin has synergistic antidepressant effects. In this study, we applied a sub-chronic restraint stress paradigm, and evaluated the potential antidepressant effects of combined fluoxetine and melatonin in adult male mice. Sub-chronic restraint stress (6 h/day for 10 days) induced depression-like behavior as shown by deteriorated fur state, increased latency to groom in the splash test, and increased immobility time in the forced-swim test. Repeated administration of either fluoxetine or melatonin at 10 mg/kg during stress exposure failed to prevent depression-like phenotypes. However, combined treatment with fluoxetine and melatonin at the selected dose attenuated stress-induced behavioral abnormalities. Moreover, we found that the antidepressant effects of combined treatment were associated with the normalization of brain-derived neurotrophic factor (BDNF)-tropomyosin receptor kinase B (TrkB) signaling in the hippocampus, but not in the prefrontal cortex. Our findings suggest that combined fluoxetine and melatonin treatment exerts synergistic antidepressant effects possibly by restoring hippocampal BDNF-TrkB signaling.
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Antidepressive Agents
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Behavior, Animal
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			Brain-Derived Neurotrophic Factor
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Depression
		                        			;
		                        		
		                        			Drug Synergism
		                        			;
		                        		
		                        			Drug Therapy, Combination
		                        			;
		                        		
		                        			Fluoxetine
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Hippocampus
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Male
		                        			;
		                        		
		                        			Melatonin
		                        			;
		                        		
		                        			pharmacology
		                        			;
		                        		
		                        			Membrane Glycoproteins
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Mice, Inbred C57BL
		                        			;
		                        		
		                        			Protein-Tyrosine Kinases
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Restraint, Physical
		                        			;
		                        		
		                        			Signal Transduction
		                        			;
		                        		
		                        			drug effects
		                        			
		                        		
		                        	
3.The binding of a monoclonal antibody to the apical region of SCARB2 blocks EV71 infection.
Xuyuan ZHANG ; Pan YANG ; Nan WANG ; Jialong ZHANG ; Jingyun LI ; Hao GUO ; Xiangyun YIN ; Zihe RAO ; Xiangxi WANG ; Liguo ZHANG
Protein & Cell 2017;8(8):590-600
		                        		
		                        			
		                        			Entero virus 71 (EV71) causes hand, foot, and mouth disease (HFMD) and occasionally leads to severe neurological complications and even death. Scavenger receptor class B member 2 (SCARB2) is a functional receptor for EV71, that mediates viral attachment, internalization, and uncoating. However, the exact binding site of EV71 on SCARB2 is unknown. In this study, we generated a monoclonal antibody (mAb) that binds to human but not mouse SCARB2. It is named JL2, and it can effectively inhibit EV71 infection of target cells. Using a set of chimeras of human and mouse SCARB2, we identified that the region containing residues 77-113 of human SCARB2 contributes significantly to JL2 binding. The structure of the SCARB2-JL2 complex revealed that JL2 binds to the apical region of SCARB2 involving α-helices 2, 5, and 14. Our results provide new insights into the potential binding sites for EV71 on SCARB2 and the molecular mechanism of EV71 entry.
		                        		
		                        		
		                        		
		                        			Amino Acid Sequence
		                        			;
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Antibodies, Monoclonal
		                        			;
		                        		
		                        			chemistry
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Binding Sites
		                        			;
		                        		
		                        			Cell Line
		                        			;
		                        		
		                        			Crystallography, X-Ray
		                        			;
		                        		
		                        			Enterovirus A, Human
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			growth & development
		                        			;
		                        		
		                        			immunology
		                        			;
		                        		
		                        			Fibroblasts
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			virology
		                        			;
		                        		
		                        			Gene Expression
		                        			;
		                        		
		                        			HEK293 Cells
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Immunoglobulin Fab Fragments
		                        			;
		                        		
		                        			chemistry
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Lysosome-Associated Membrane Glycoproteins
		                        			;
		                        		
		                        			chemistry
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			immunology
		                        			;
		                        		
		                        			Mice
		                        			;
		                        		
		                        			Models, Molecular
		                        			;
		                        		
		                        			Protein Binding
		                        			;
		                        		
		                        			Protein Conformation, alpha-Helical
		                        			;
		                        		
		                        			Protein Conformation, beta-Strand
		                        			;
		                        		
		                        			Protein Interaction Domains and Motifs
		                        			;
		                        		
		                        			Receptors, Scavenger
		                        			;
		                        		
		                        			chemistry
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			immunology
		                        			;
		                        		
		                        			Receptors, Virus
		                        			;
		                        		
		                        			chemistry
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			immunology
		                        			;
		                        		
		                        			Recombinant Fusion Proteins
		                        			;
		                        		
		                        			chemistry
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			immunology
		                        			;
		                        		
		                        			Sequence Alignment
		                        			;
		                        		
		                        			Sequence Homology, Amino Acid
		                        			;
		                        		
		                        			Sf9 Cells
		                        			;
		                        		
		                        			Spodoptera
		                        			;
		                        		
		                        			Thermodynamics
		                        			
		                        		
		                        	
4.Equol protects PC12 neuronal cells against hypoxia/reoxygenation injury in vitro by reducing reactive oxygen species production.
Wei YU ; Xiuling DENG ; Zhen MA ; Yan WANG
Journal of Southern Medical University 2016;36(1):1-7
OBJECTIVEBoth of gp91(phox) (an isoform of nicotinamide adenine dinucleotide phosphate-reduced oxidases) and Src (a non-receptor protein tyrosine kinase) play a prominent role in mediating hypoxia/reoxygenation injury of neurons. The present study was designed to investigate the neuroprotective effect of equol, a predominant active metabolite of daidzein, against hypoxia/reoxygenation injury in rat pheochromocytoma cell line (PC12) and explore the underlying mechanisms.
METHODSPC12 cells exposed to hypoxia/reoxygenation injury were examined for reactive oxygen species (ROS) using dihydroethidium and 2', 7'-dichlorofluorescein diacetate and analyzed for changes in lactate dehydrogenase (LDH) activity and malondialdehyde (MDA) content. The expression levels of gp91(phox) and phosphorylated Src-Tyr416 (p-Src) were measured using Western blotting.
RESULTSEquol dose-dependently restored the cell viability and decreased LDH activity and MDA content in culture medium of PC12 cells exposed to hypoxia/reoxygenation. Pretreatment of the cells with 10(-5) and 10(-6) mol/L equol inhibited hypoxia/reoxygenation-induced increase of ROS. PC12 cells treated with equol prior to hypoxia/reoxygenation injury showed significant enhancement of the protein levels of gp91(phox) and p-Src.
CONCLUSIONEquol confers neuroprotection against hypoxia/reoxygenation injury in PC12 cells by inhibiting the generation of ROS very likely as a result of down-regulation of gp91(phox) and inhibition of Src phosphorylation.
Animals ; Cell Hypoxia ; Cell Survival ; Down-Regulation ; Equol ; pharmacology ; L-Lactate Dehydrogenase ; metabolism ; Malondialdehyde ; metabolism ; Membrane Glycoproteins ; metabolism ; NADPH Oxidase 2 ; NADPH Oxidases ; metabolism ; Neurons ; drug effects ; metabolism ; Neuroprotective Agents ; pharmacology ; PC12 Cells ; Phosphorylation ; Rats ; Reactive Oxygen Species ; metabolism ; src-Family Kinases ; metabolism
5.Idebenone Maintains Survival of Mutant Myocilin Cells by Inhibiting Apoptosis.
Yue GUAN ; Juan LI ; Tao ZHAN ; Jian-Wen WANG ; Jian-Bo YU ; Lan YANG
Chinese Medical Journal 2016;129(16):2001-2004
		                        		
		                        		
		                        		
		                        			Animals
		                        			;
		                        		
		                        			Apoptosis
		                        			;
		                        		
		                        			drug effects
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			COS Cells
		                        			;
		                        		
		                        			Cercopithecus aethiops
		                        			;
		                        		
		                        			Cytoskeletal Proteins
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Eye Proteins
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Glaucoma, Open-Angle
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Glycoproteins
		                        			;
		                        		
		                        			genetics
		                        			;
		                        		
		                        			metabolism
		                        			;
		                        		
		                        			Humans
		                        			;
		                        		
		                        			Mutation
		                        			;
		                        		
		                        			Ubiquinone
		                        			;
		                        		
		                        			analogs & derivatives
		                        			;
		                        		
		                        			pharmacology
		                        			
		                        		
		                        	
6.Ulinastatin inhibits hypoxia-induced phenotype modulation of pulmonary artery smooth muscle cells by up-regulating PPAR-γ expression.
Kun TANG ; Chang LIU ; Lin CHEN ; Jing GAO ; Chao ZHANG
Journal of Southern Medical University 2016;36(10):1322-1327
OBJECTIVETo evaluate the effect of ulinastatin on hypoxia-induced phenotype modulation of pulmonary artery smooth muscle cells (PASMCs) and explore the underlying mechanism.
METHODSCultured PASMCs from SD rats were exposed to normoxic condition, normoxia with ulinastatin treatment, hypoxia, or hypoxia with ulinastatin treatment. After 24 h of exposures, the cells were examined for SM-α-actin and caplonin expressions with immunofluorescence assay and for cell migration with CCK-8 andH-TdR assays. Western blotting was used for detecting the expressions of PPAR-γ in the cells, and PPAR-γ-responsive firefly luciferase reporter was employed for measuring the transcriptional activity of PPAR-γ. The PPAR-γ inhibitor GW9662 was used to explore the mechanism of the inhibitory effect of ulinastatin on hypoxia induced-phenotype modulation of PASMCs by measuring the changes in cell proliferation and migration.
RESULTSUlinastatin obviously enhanced the expressions of SM-α-actin and calponin (P<0.05), inhibited the proliferation and migration (P<0.05), and up-regulated the expression of PPAR-γ in PASMCs exposed to hypoxia (P<0.05). Pretreatment of the cells with GW9662 abolished the effect of ulinastatin on hypoxia-induced phenotype modulation of PASMCs and enhanced the cell proliferation and migration (P<0.05).
CONCLUSIONUlinastatin inhibits hypoxia-induced phenotype modulation of PASMCs from rats possibly by up-regulating the expression of PPAR-γ.
Actins ; metabolism ; Animals ; Calcium-Binding Proteins ; metabolism ; Cell Hypoxia ; Cell Proliferation ; Cells, Cultured ; Glycoproteins ; pharmacology ; Microfilament Proteins ; metabolism ; Myocytes, Smooth Muscle ; cytology ; drug effects ; PPAR gamma ; metabolism ; Phenotype ; Pulmonary Artery ; cytology ; Rats ; Rats, Sprague-Dawley ; Up-Regulation
7.Effects of ulinastatin on immune function of spleen in severely burned rats and its mechanism.
Juncong LI ; Chao HU ; Yongming YAO ; Hongming YANG
Chinese Journal of Burns 2016;32(5):266-271
OBJECTIVETo observe the effects of ulinastatin on immune function of splenic CD4(+) T lymphocytes and CD4(+) CD25(+) regulatory T lymphocytes (Tregs) and content of high mobility group box 1 (HMGB1) in peripheral blood of severely burned rats, and to analyze the possible mechanisms.
METHODSNinety-six male SD rats were divided into sham injury group, burn group, and ulinastatin group according to the random number table, with 32 rats in each group. Rats in sham injury group were sham injured on the back by immersing in 37 ℃ warm water for 12 s. Rats in burn group and ulinastatin group were inflicted with 30% total body surface area full-thickness scald (hereinafter referred to as burn) on the back by immersing in 94 ℃ hot water for 12 s. Immediately after injury, rats in each group were intraperitoneally injected with saline (40 mL/kg), meanwhile rats in ulinastatin group were intraperitoneally injected with ulinastatin (4×10(4) U/kg), once per 12 h, till post injury hour 72. Eight rats of each group were respectively selected on post injury day (PID) 1, 3, 5, and 7 to collect abdominal aortic blood samples. Serum content of HMGB1 was detected by enzyme-linked immunosorbent assay (ELISA). And then, rats of the 3 groups were sacrificed immediately to collect spleens and separate CD4(+) CD25(+) Tregs and CD4(+) T lymphocytes. Flow cytometer was used to detect positive expression rates of cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and forkhead-winged helix transcription factor p3 (Foxp3) in CD4(+) CD25(+) Tregs. Content of IL-10 in culture supernatant of CD4(+) CD25(+) Tregs, and content of interleukin 2 (IL-2), IL-4, and γ interferon (IFN-γ) in culture supernatant of CD4(+) T lymphocytes was detected by ELISA. The proliferative activity of CD4(+) T lymphocytes was determined by microplate reader. The sample number of above-mentioned experiments was 8 at each time point in each group. Data were processed with analysis of variance of factorial design and LSD test.
RESULTS(1) Compared with that in sham injury group, serum content of HMGB1 of rats in burn group was significantly increased from PID 1 to 7 (with P values below 0.01). Compared with that in burn group, serum content of HMGB1 of rats in ulinastatin group was significantly decreased from PID 1 to 7 (with P values below 0.01). (2) Compared with those in sham injury group, the positive expression rates of CTLA-4 and Foxp3 in CD4(+) CD25(+) Tregs and content of IL-10 in culture supernatant of CD4(+) CD25(+) Tregs of rats in burn group were significantly increased from PID 1 to 7 (with P values below 0.01), peaking on PID 3 [(65±10)%, (76±10)%, and (28.2±4.4) pg/mL respectively]. These 3 indexes of rats in sham injury group on PID 3 were (45±7)%, (46±7)%, and (11.2±2.3) pg/mL respectively. Compared with those in burn group, the positive expression rates of CTLA-4 and Foxp3 in CD4(+) CD25(+) Tregs and content of IL-10 in culture supernatant of CD4(+) CD25(+) Tregs of rats in ulinastatin group were significantly decreased from PID 1 to 7 (P<0.05 or P<0.01), reaching the nadir on PID 7 [(43±6)%], PID 1 [(50±8)%], and PID 7 [(12.4±3.4) pg/mL] respectively. These 3 indexes of rats in burn group on PID 7, 1, and 7 were (58±8)%, (71±9)%, and (19.7±2.8) pg/mL respectively. (3) Compared with those in sham injury group, the content of IL-2 and IFN-γ in culture supernatant of CD4(+) T lymphocytes of rats was significantly decreased, while the content of IL-4 in culture supernatant of CD4(+) T lymphocytes of rats was significantly increased in burn group from PID 1 to 7, with P values below 0.01. Compared with that in burn group, the content of IL-2 and IFN-γ in culture supernatant of CD4(+) T lymphocytes of rats was significantly increased, while the content of IL-4 in culture supernatant of CD4(+) T lymphocytes of rats was significantly decreased in ulinastatin group from PID 1 to 7, P<0.05 or P<0.01. (4) Compared with that in sham injury group, the proliferative activity of CD4(+) T lymphocytes of rats in burn group was significantly decreased from PID 1 to 7 (with P values below 0.01). Compared with that in burn group, the proliferative activity of CD4(+) T lymphocytes of rats in ulinastatin group was significantly increased from PID 1 to 7 (with P values below 0.01).
CONCLUSIONSUlinastatin can weaken the immunosuppressive function mediated by splenic CD4(+) CD25(+) Tregs in severely burned rats, and improve proliferative function and secretory function of splenic CD4(+) T lymphocytes, which may be attributed to the inhibiting effect of ulinastatin on the release of HMGB1 in large amount.
Animals ; Burns ; drug therapy ; CTLA-4 Antigen ; metabolism ; Enzyme-Linked Immunosorbent Assay ; Flow Cytometry ; Forkhead Transcription Factors ; metabolism ; Glycoproteins ; pharmacology ; HMGB1 Protein ; blood ; Interferon-gamma ; metabolism ; Interleukin-10 ; metabolism ; Interleukin-2 ; metabolism ; Interleukin-4 ; metabolism ; Male ; Random Allocation ; Rats ; Rats, Sprague-Dawley ; Spleen ; drug effects ; T-Lymphocytes, Regulatory ; cytology ; drug effects
8.Salidroside protects PC12 cells from HO-induced apoptosis via suppressing NOX2-ROS-MAPKs signaling pathway.
Zhi-Lin QI ; Yin-Hua LIU ; Shi-Mei QI ; Lie-Feng LING ; Zun-Yong FENG ; Qiang LI
Journal of Southern Medical University 2016;37(2):178-183
OBJECTIVETo investigate the molecular mechanism by which salidroside protects PC12 cells from HO-induced apoptosis.
METHODSPC12 cells cultured in DMEM supplemented with 10% horse serum and 5% fetal bovine serum were pretreated with different doses of salidroside for 2 h and then stimulated with HOfor different lengths of time. The expression levels of PARP and caspase 3 and the phosphorylation of p38, ERK and JNK were determined with Western blotting. The cell nuclear morphology was observed after DAPI staining. The production of ROS was detected using a ROS detection kit, and the levels of gp91and p47in the membrane and cytoplasm were detected by membrane-cytoplasm separation experiment; the binding between gp91and p47was assayed by coimmunoprecipitation experiment.
RESULTSSalidroside dose-dependently suppressed cell apoptosis, lowered phosphorylation levels of p38, ERK and JNK, inhibited the production of ROS, reduced the binding between gp91and p47, and inhibited the activity of NOX2 in PC12 cells exposed to HO.
CONCLUSIONSalidroside protects PC12 cells from HO-induced apoptosis at least partly by suppressing NOX2-ROS-MAPKs signaling pathway.
Animals ; Apoptosis ; Caspase 3 ; metabolism ; Glucosides ; pharmacology ; Hydrogen Peroxide ; MAP Kinase Signaling System ; drug effects ; Membrane Glycoproteins ; metabolism ; NADPH Oxidase 2 ; NADPH Oxidases ; metabolism ; Neuroprotective Agents ; pharmacology ; PC12 Cells ; Phenols ; pharmacology ; Phosphorylation ; Rats ; Reactive Oxygen Species ; metabolism
9.Reversal of stemness in multidrug-resistant hepatocellular carcinoma cells by SIS3.
Wei YAN ; Ting WEN ; Suqiong LIN ; Zhongcai LIU ; Wenchao YANG ; Guoyang WU ; Email: WUGUOYANG_MAIL@ALIYUN.COM.
Chinese Journal of Oncology 2015;37(10):731-735
OBJECTIVETo investigate whether SIS3, a specific inhibitor of Smad3 phosphorylation, can reverse the stemness of multidrug-resistant(MDR) hepatocellular carcinoma cells.
METHODSMDR HCC Huh7.5.1/ADM cell lines were developed by exposing parental cells to stepwise increasing concentrations of ADM. CCK-8 assay was used to determine the cellular sensitivity of various anticancer drugs. Flow cytometry (FCM) was used to analyze the expression level of cancer stem cell marker CD133. Clone formation assay and mouse subcutaneous xenograft tumors were used to investigate the tumorigenicity in vitro and in vivo. Western blotting (WB) was used to analyze the changes of expressions of CD133, Smad3, Bcl-2, Bax and p-Smad3 in different conditions.
RESULTSADM treatment of HCC cells in vitro resulted in a development of subline, Huh7.5.1/ADM cells, with CSC phenotypes: stable MDR phenotype (besides ADMc Huh7.5.1/ADM cells were also more resistant to some other anticancer drugs including VCR, MMC and CTX ) (IC50: 0.215 ± 0.018 vs. 0.123 ± 0.004, 0.145 ± 0.009 vs. 0.014 ± 0.002, 1.021 ± 0.119 vs. 0.071 ± 0.006, 27.007 ± 1.606 vs. 1.919 ± 0.032) (unit: µg/ml) (P<0.05). Huh7.5.1/ADM cells enriched the cancer stem-like cell fraction (CD133-positive subpopulation) (76.06 ± 2.948% vs. 25.38 ± 4.349%) (P<0.05), had stronger tumorigenicity in vivo and colony formation ability, and activated the Smad3 activity. Inhibition of Smad3 activity by SIS3 decreased stemness of the Huh7.5.1/ADM cells: CD133-positive subpopulation (48.49 ± 2.304% vs. 76.06 ± 2.948%) (P<0.05); ADM IC50: (0.112 ± 0.019 vs. 0.215 ± 0.018), VCR IC50 (0.065 ± 0.013 vs. 0.145±0.009), MMC IC₅₀ (0.749 ± 0.121 vs. 1.021 ± 0.119), CTX IC50 (10.576 ± 1.248 vs. 27.007 ± 1.606) (unit: µg/ml) (P<0.05), and decreased tumorigenicity and colony formation ability.
CONCLUSIONSIS3 as a specific inhibitor of Smad3 signal is involved in the stemness of multidrug resistant hepatocellular carcinoma cells.
AC133 Antigen ; Animals ; Antibiotics, Antineoplastic ; pharmacology ; Antigens, CD ; metabolism ; Carcinoma, Hepatocellular ; drug therapy ; metabolism ; pathology ; Doxorubicin ; pharmacology ; Drug Resistance, Neoplasm ; Glycoproteins ; metabolism ; Heterografts ; Humans ; Isoquinolines ; pharmacology ; Liver Neoplasms ; drug therapy ; metabolism ; pathology ; Mice ; Neoplasm Proteins ; metabolism ; Neoplastic Stem Cells ; drug effects ; Peptides ; metabolism ; Proto-Oncogene Proteins c-bcl-2 ; metabolism ; Pyridines ; pharmacology ; Pyrroles ; pharmacology ; Smad3 Protein ; antagonists & inhibitors ; metabolism ; Tumor Stem Cell Assay ; bcl-2-Associated X Protein ; metabolism
10.5-aza-2'-deoxycytidine suppresses the growth of human lung adenocarcinoma cells in nude mouse xenograft models and its effect on methylation status and expression of TFPI-2 gene.
Jiangshui LIANG ; Guilin YIN ; Yongqiang DONG ; Zhongsha MA ; Yuehua XIAO ; Tao JI
Chinese Journal of Oncology 2015;37(7):485-489
OBJECTIVETo investigate the inhibitory effect of classic demethylating drug 5-aza-2'-deoxycytidine (5-Aza-CdR) on the growth of human lung adenocarcinoma cells in nude mouse xenograft models, and to observe its effect on methylation status and expression of TFPI-2 gene in the nude mouse xenograft tissues.
METHODSThe nude mouse xenograft model was established by subcutaneous inoculation of human lung adenocarcinoma A549 cells. According to different doses of 5-Aza-CdR, the tumor-bearing nude mice were randomly divided into experimental groups (0.5 mg/kg group, 1 mg/kg group, 2 mg/kg group) and control group (0 mg/kg group). The tumor growth in the nude mice was observed. The methylation status and the expression of TFPI-2 gene mRNA and protein were detected by methylation specific polymerase chain reaction, real-time fluorescent quantitative polymerase chain reaction and Western blot assay.
RESULTSThe nude mice were euthanized at 28 days after intraperitoneal injection of 5-Aza-CdR. The body weight of tumor-bearing nude mice was (27.12 ± 0.38) g in the 0 mg/kg group, (26.80 ± 0.18) g in the 0.5 mg/kg group, (26.67 ± 0.28) g in the 1 mg/kg group, and (26.50 ± 0.26) g in the 2 mg/kg group, showing no significant difference among them (P > 0.05). The volume of xenograft tumors in the 0 mg/kg group was (709.22 ± 2.87)mm³, (400.67 ± 2.68)mm³ in the 0.5 mg/kg group, (285.71 ± 2.91)mm³ in the 1 mg/kg group, and (230.44 ± 3.15)mm³ in the 2 mg/kg group, showing a significant difference (P < 0.05). There were complete methylation of TFPI-2 gene in the 0 mg/kg group, incomplete methylation in the 0.5 and 1 mg/kg groups, and unmethylation in the 2 mg/kg group. The relative mRNA level in the 0, 0.5, 1, 2 mg/kg groups were 1.00 ± 0.00, 1.67 ± 0.07, 3.40 ± 0.24, and 5.55 ± 0.61, respectively (P < 0.05). The relative expression level of TFPI-2 protein in the 0, 0.5, 1, 2 mg/kg groups was 0.18 ± 0.02, 0.36 ± 0.01, 0.64 ± 0.02, and 0.81 ± 0.20, respectively (P < 0.05).
CONCLUSIONS5-Aza-CdR suppresses the tumor growth of human lung adenocarcinoma cells in nude mouse xenograft models, and induces expression of TFPI-2 gene in the xenograft tumor cells. The mechanism might be that 5-Aza-CdR induces re-expression of demethylated TFPI-2 gene by demethylation, and thus inhibits the growth and proliferation of human lung adenocarcinoma cells.
Adenocarcinoma ; drug therapy ; pathology ; Animals ; Antimetabolites, Antineoplastic ; pharmacology ; Azacitidine ; analogs & derivatives ; pharmacology ; Cell Proliferation ; drug effects ; DNA Methylation ; Disease Models, Animal ; Gene Expression Regulation, Neoplastic ; Glycoproteins ; genetics ; Heterografts ; Humans ; Lung Neoplasms ; drug therapy ; pathology ; Mice ; Mice, Nude ; RNA, Messenger ; metabolism ; Random Allocation
            
Result Analysis
Print
Save
E-mail