1.Interleukin-17 promotes mouse hepatoma cell proliferation by antagonizing interferon-γ.
Jie LI ; Kun YAN ; Yi YANG ; Hua LI ; Zhidong WANG ; Xin XU
Journal of Southern Medical University 2019;39(1):1-5
OBJECTIVE:
To investigate the interaction between interleukin-17 (IL-17) and interferon-γ (IFN-γ) and how their interaction affects the growth of mouse hepatoma Hepa1-6 cells.
METHODS:
Hepa1-6 cells treated with IL-17 and IFN-γ either alone or in combination were examined for changes in cell proliferation using MTT assay and in cell cycle distribution using flow cytometry. Western blotting was used to detect the protein expression levels of proliferating cell nuclear antigen (PCNA), cyclin D1, P21 and P16 and the phosphorylation of p38MAPK, ERK1/2 and Stat1 in the cells.
RESULTS:
Compared with control group, IFN-γ treatment obviously inhibited the growth and proliferation of Hepa1-6 cells, induced cell cycle arrest at G0/G1 phase, reduced the protein expression of PCNA and cyclin D1, and increased the protein expression of P21. IL-17 alone had no effect on the growth of Hepa1-6 cells. In the combined treatment, IL-17 significantly antagonized the effects of IFN-γ. Compared with those treated with IFN-γ alone, the cells with the combined treatment showed significantly decreased G0/G1 cell population, increased the protein expressions of PCNA and cyclin D1, and decreased the protein expression of P21. IL-17 significantly inhibited IFN-γ-induced phosphorylation of p38MAPK and ERK1/2 without affecting the phosphorylation of Stat1.
CONCLUSIONS
IL-17 obviously reverses the antitumor effects of IFN-γ to promote the proliferation of mouse hepatoma cells and accelerate the development of hepatocellular carcinoma.
Animals
;
Carcinoma, Hepatocellular
;
metabolism
;
pathology
;
Cell Cycle
;
Cell Line, Tumor
;
Cell Proliferation
;
drug effects
;
Cyclin D1
;
metabolism
;
Cyclin-Dependent Kinase Inhibitor p21
;
metabolism
;
Interferon-gamma
;
antagonists & inhibitors
;
Interleukin-17
;
pharmacology
;
Liver Neoplasms
;
metabolism
;
pathology
;
Mice
;
Neoplasm Proteins
;
metabolism
;
Proliferating Cell Nuclear Antigen
;
metabolism
2.Silencing of Long Non-Coding RNA MALAT1 Promotes Apoptosis of Glioma Cells.
Jianping XIANG ; Shifeng GUO ; Shuling JIANG ; Yuelong XU ; Jiwei LI ; Li LI ; Jinyu XIANG
Journal of Korean Medical Science 2016;31(5):688-694
The metastasis-associated lung adenocarcinoma transcription 1 (MALAT1) is a highly conserved long non-coding RNA (lncRNA) gene. However, little is known about the pathological role of lncRNA MALAT1 in glioma. In the present study, we explored the expression level of lncRNA MALAT1 in primary glioma tissues as well as in U87 and U251 glioma cell lines. Using qRT-PCR, we found that the expression of lncRNA MALAT1 was significantly increased in glioma tissues compared with that of paracancerous tissues. Meanwhile, the expression of MALAT1 was highly expressed in U98 and U251 cells. In order to explore the function of MALAT1, the expression of MALAT1 was greatly reduced in U87 and U251 cells transfected with siRNA specifically targeting MALAT1. Consequently, cell viability of U87 and U251 cells were drastically decreased after the knockdown of MALAT1. Concomitantly, the apoptosis rate of the two cell lines was dramatically increased. Furthermore, the expression levels of some tumor markers were reduced after the knockdown of MALAT1, such as CCND1 and MYC. In summary, the current study indicated a promoting role of MALAT1 in the development of glioma cell.
*Apoptosis
;
Biomarkers, Tumor/genetics/metabolism
;
Blotting, Western
;
Cell Line, Tumor
;
Cell Movement
;
Cell Proliferation
;
Cyclin D1/genetics/metabolism
;
Down-Regulation
;
Flow Cytometry
;
Glioma/metabolism/pathology
;
Humans
;
Proto-Oncogene Proteins c-myc/genetics/metabolism
;
*RNA Interference
;
RNA, Long Noncoding/antagonists & inhibitors/genetics/*metabolism
;
RNA, Small Interfering/metabolism
;
Real-Time Polymerase Chain Reaction
3.Knockdown of Bmi1 inhibits bladder cancer cell growth both in vitro and in vivo by blocking cell cycle at G1 phase and inducing apoptosis.
Hong-bo LUO ; Bin LI ; Wei-gang YUAN ; Chuan-rui XU
Journal of Huazhong University of Science and Technology (Medical Sciences) 2015;35(5):730-735
Bmi1 is a member of the polycomb group family of proteins, and it drives the carcinogenesis of various cancers and governs the self-renewal of multiple types of stem cells. However, its role in the initiation and progression of bladder cancer is not clearly known. The present study aimed to investigate the function of Bmi1 in the development of bladder cancer. Bmi1 expression was detected in human bladder cancer tissues and their adjacent normal tissues (n=10) by immunohistochemistry, qRT-PCR and Western blotting, respectively. Bmi1 small interference RNA (siRNA) was synthesized and transfected into human bladder carcinoma cells (EJ) by lipofectamine 2000. The Bmil expression at mRNA and protein levels was measured in EJ cells transfected with Bmil siRNA (0, 80, 160 nmol/L) by qRT-PCR and Western blotting, respectively. Cell viability and Ki67 expression (a marker of cell proliferation) were determined in Bmi1 siRNA-transfected cells by CCK-8 assay and qRT-PCR, respectively. Cell cycle of transfected cells was flow-cytometrically determined. Immunofluorescence and Western blotting were used to detect the expression levels of cell cycle-associated proteins cyclin D1 and cyclin E in the cells. Pro-apoptotic proteins Bax and caspase 3 and anti-apoptotic protein Bcl-2 were detected by Western blotting as well. Additionally, xenograft tumor models were established by inoculation of EJ cells (infected with Bmil shRNA/pLKO.1 lentivirus or not) into nude mice. The tumor volumes were measured every other day for 14 days. The results showed that the Bmil expression was significantly increased in bladder tumor tissues when compared with that in normal tissues (P<0.05). Perturbation of Bmi1 expression by using siRNA could significantly inhibit the proliferation of EJ cells (P<0.05). Bmi1 siRNA-transfected EJ cells were accumulated in G1 phase and the expression levels of cyclin D1 and cyclin E were down-regulated. Bax and caspase-3 expression levels were significantly increased and Bcl-2 levels decreased after Bmi1 knockdown. Tumor volume was conspicuously reduced in mice injected with EJ cells with Bmi1 knockdown. Our findings indicate that Bmi1 is a potential driver oncogene of bladder cancer and it may become a potential treatment target for human bladder cancer.
Animals
;
Apoptosis
;
genetics
;
Carcinogenesis
;
genetics
;
metabolism
;
pathology
;
Carcinoma
;
genetics
;
metabolism
;
pathology
;
therapy
;
Caspase 3
;
genetics
;
metabolism
;
Cell Line, Tumor
;
Cyclin D1
;
antagonists & inhibitors
;
genetics
;
metabolism
;
Cyclin E
;
antagonists & inhibitors
;
genetics
;
metabolism
;
G1 Phase Cell Cycle Checkpoints
;
genetics
;
Gene Expression Regulation, Neoplastic
;
Humans
;
Injections, Intralesional
;
Ki-67 Antigen
;
genetics
;
metabolism
;
Mice
;
Mice, Nude
;
Polycomb Repressive Complex 1
;
antagonists & inhibitors
;
genetics
;
metabolism
;
Proto-Oncogene Proteins c-bcl-2
;
antagonists & inhibitors
;
genetics
;
metabolism
;
RNA, Small Interfering
;
administration & dosage
;
genetics
;
metabolism
;
Signal Transduction
;
Tumor Burden
;
Urinary Bladder
;
metabolism
;
pathology
;
Urinary Bladder Neoplasms
;
genetics
;
metabolism
;
pathology
;
therapy
;
Xenograft Model Antitumor Assays
;
bcl-2-Associated X Protein
;
agonists
;
genetics
;
metabolism
4.Application of temperature sensitive yeast models with definite target in the screening of potential human Pin1 inhibitors.
Jing ZHANG ; Xiao-Min HAN ; Wen-Hui HU ; Zong-Ru GUO ; Xiao-Bo HE ; Shu-Yi SI
Acta Pharmaceutica Sinica 2014;49(6):854-860
This study is to explore new lead compounds by inhibition of Pin1 for anticancer therapy using temperature sensitive mutants. As Pin1 is conserved from yeast to human, we established a high-throughput screening method for Pin1 inhibitors, which employed yeast assay. This method led to the identification of one potent hits, 8-11. In vitro, 8-11 inhibited purified Pin1 enzyme activity with IC50 of (10.40 +/- 1.68) micromol x L(-1), induced G1 phase arrest and apoptosis, showed inhibitory effects on a series of cancer cell proliferation, reduced Cyclin D1 expression, was defined as reciprocally matched for protein-ligand complex in virtual docking analysis and reduced cell migration ability. In vivo, we could observe reduction of tumor volume after treatment with 8-11 in xenograft mice compared with vehicle DMSO treatment. Altogether, these results provide for the first time the involvement of 8-11 in the anticancer activity against Pin1.
Animals
;
Apoptosis
;
drug effects
;
Cell Proliferation
;
drug effects
;
Cyclin D1
;
metabolism
;
Drug Screening Assays, Antitumor
;
methods
;
G1 Phase
;
High-Throughput Screening Assays
;
methods
;
Humans
;
Mice
;
NIMA-Interacting Peptidylprolyl Isomerase
;
Neoplasms
;
pathology
;
Peptidylprolyl Isomerase
;
antagonists & inhibitors
;
Temperature
;
Xenograft Model Antitumor Assays
;
Yeasts
5.Growth inhibition of combined pathway inhibitors on KRAS mutated non-small cell lung cancer cell line.
Zhan-wen LI ; Zhen-li YANG ; Hai-liang FENG ; Xiao-cui BIAN ; Yan-yan LIU ; Yu-qin LIU
Chinese Journal of Pathology 2013;42(5):330-335
OBJECTIVETo investigate the effect of the selective PI3K inhibitor and MEK inhibitor on KRAS and PTEN co-mutated non-small cell lung cancer cell line NCI-H157 and the relevant mechanisms.
METHODSNCI-H157 was cultured routinely and treated with different concentrations of the two inhibitors. Cell proliferation was detected by MTT cell cycle assay. Based on the MTT results the cells were divided into four groups: the control group, PI3K inhibitor group (GDC-0941, 0.5 and 5.0 µmol/L), combination group I (0.5 µmol/L AZD6244 + 0.5 µmol/L GDC-0941) and combination group II (5.0 µmol/L AZD6244 + 5.0 µmol/L GDC-0941). Colony formation assay was performed to detect colony formation efficiency. The cell cycle and apoptosis were analyzed by flow cytometry. The expression of protein related to apoptosis was tested with Western blot.
RESULTSCell growth was inhibited by the two inhibitors. Combination groups led to stronger cell proliferation inhibition: combination group Ishowed synergistic effect of their actions and combination group II showed an additive effect; in both groups, there were decreased colony number [(77.2 ± 1.54)/well vs (61.50 ± 2.12)/well, P < 0.01] and [(51.00 ± 4.00)/ well vs (22.50 ± 3.53)/well, P < 0.01]; and enhanced apoptotic ratios [(18.30 ± 0.82)% vs (21.32 ± 0.56)%, P < 0.01] and [(27.14 ± 1.58)% vs (42.45 ± 4.42)%, P < 0.01]. In addition, compared to the PI3K inhibitor alone group, the NCI-H157 cells in the combination groups showed increased G0/G1 phase and decreased S phase (P < 0.01). Western blotting showed that the combination groups demonstrated significantly decreased expression of cyclin D1 and cyclin B1, increased p21 and cleaved PARP and decreased bcl-2/bax ratio, compared to the PI3K inhibitor only group.
CONCLUSIONThe combined inhibition of PI3K (AZD6244) and MEK (GDC-0941) has synergistic effects on the proliferation of NCI-H157 cells, but such effects appear to be in a dose-dependent manner.
Apoptosis ; drug effects ; Benzimidazoles ; administration & dosage ; pharmacology ; Carcinoma, Non-Small-Cell Lung ; genetics ; pathology ; Cell Cycle ; drug effects ; Cell Line, Tumor ; Cell Proliferation ; drug effects ; Cyclin B1 ; metabolism ; Cyclin D1 ; metabolism ; Dose-Response Relationship, Drug ; Drug Synergism ; Humans ; Indazoles ; administration & dosage ; pharmacology ; Lung Neoplasms ; genetics ; pathology ; Mitogen-Activated Protein Kinase Kinases ; antagonists & inhibitors ; metabolism ; Mutation ; PTEN Phosphohydrolase ; genetics ; Phosphatidylinositol 3-Kinases ; antagonists & inhibitors ; metabolism ; Poly(ADP-ribose) Polymerases ; metabolism ; Proto-Oncogene Proteins ; genetics ; Proto-Oncogene Proteins c-bcl-2 ; metabolism ; Proto-Oncogene Proteins p21(ras) ; metabolism ; Signal Transduction ; Sulfonamides ; administration & dosage ; pharmacology ; bcl-2-Associated X Protein ; metabolism ; ras Proteins ; genetics
6.Inhibition of survivin expression and mechanisms of reversing drug-resistance of human lung adenocarcinoma cells by siRNA.
Jing-lei LIU ; Yan WANG ; Ji JIANG ; Rui KONG ; Yan-mei YANG ; Hong-fei JI ; Yu-zhi SHI
Chinese Medical Journal 2010;123(20):2901-2907
BACKGROUNDSurvivin, a member of the inhibitor of apoptosis protein (IAP) family, overexpresses in tumor cells and not expresses in terminally differentiated adult tissues. This study aimed to investigate the effects of survivin-specific siRNA on cell proliferation, apoptosis and chemosensitivity to cisplatin in vitro and in vivo and explore the mechanisms about decreasing expression of survivin in reversing cancer cells resistance to chemotherapeutic drug.
METHODSSurvivin-specific siRNA was transfected into A549/DDP cells. The expression of survivin and lung resistance-related protein (LRP) mRNA levels were determined by RT-PCR, chemosensitivity of A549/DDP (cisplatin) cells to cisplatin was determined by MTT assay, and apoptosis and cell cycle were determined by flow cytometry (FCM). The protein expression levels of survivin, LRP, cyclin-D(1), caspase-3 and bcl-2 were determined by Western blotting analyses. The effect of survivin siRNA inhibition on tumor growth was studied in athymic nude mice in vivo.
RESULTSSurvivin-specific siRNA efficiently down-regulated survivin expression. The cell cycle was arrested at G2/M phase, and apoptosis was obviously found. Inhibition of survivin expression could make the IC50 and drug-resistant index of cisplatin decrease, and enhance the cancer cells sensitivity to cisplatin. After transfection by survivin-specific siRNA, expression of LRP and cyclin-D1 were downregulated, caspase-3 expression was upregulated, bcl-2 expression had no obvious change. The animal experiment confirmed knockdown of survivin could inhibit the tumor growth.
CONCLUSIONSSurvivin-specific siRNA can efficiently suppress the expression of survivin, increase apoptosis, inhibit cells proliferation and enhance the chemosensitivity to cisplatin in vitro and in vivo. Suppression of survivin expression helping to reverse drug-resistance may have relationship with downregulation of LRP and upregulation of caspase-3. Anti-tumor strategies based on the inhibition of survivin may be useful in targeting lung adenocarcinomas.
Adenocarcinoma ; drug therapy ; pathology ; Animals ; Apoptosis ; Caspase 3 ; analysis ; Cell Line, Tumor ; Cisplatin ; pharmacology ; Cyclin D1 ; analysis ; Drug Resistance, Neoplasm ; Female ; Humans ; Inhibitor of Apoptosis Proteins ; Lung Neoplasms ; drug therapy ; pathology ; Male ; Mice ; Mice, Inbred BALB C ; Microtubule-Associated Proteins ; antagonists & inhibitors ; genetics ; Proto-Oncogene Proteins c-bcl-2 ; analysis ; RNA, Messenger ; analysis ; RNA, Small Interfering ; genetics ; Vault Ribonucleoprotein Particles ; genetics
7.Influence of DNAzymes against cyclin D1 in tumor cell cycle.
Dan LI ; Junyi ZHU ; Yayan ZHOU ; Xuanming LIU
Journal of Biomedical Engineering 2009;26(2):374-384
In this study, DNAzymes against cyclin D1 (cyclin D1-DRz) were designed according to the secondary structure of cyclin D1 mRNA which was computed with RNAdraw and Mfold. Cyclin D1-DRz were transfected into tumor cell line u251 and HeLa by oligofectamine. The expression of cyclin D1 was detected by RT-PCR. It was shown that the expression of cyclin D1 gene was suppressed obviously, and the expressions of other cell-cycle related genes such as cyclin E1, cyclin A1 and cyclin B1 were also declined. The cell cycle analysis of tumor cells tansfected with cyclin D1-DRz revealed an arrestment in the G0/G1 phase. In conclusion, the approach is effective and feasible for designing DNAzyme. Cyclin D1-DRz is useful for interfering with the cell cycle procession of tumor cells.
Base Sequence
;
Cell Cycle
;
drug effects
;
Cyclin D1
;
antagonists & inhibitors
;
genetics
;
metabolism
;
DNA, Catalytic
;
genetics
;
metabolism
;
pharmacology
;
HeLa Cells
;
Humans
;
Molecular Sequence Data
;
RNA, Messenger
;
genetics
;
metabolism
;
Transfection
8.DAPT enhances the apoptosis of human tongue carcinoma cells.
Brian E GROTTKAU ; Xi-Rui CHEN ; Claudia C FRIEDRICH ; Xing-Mei YANG ; Wei JING ; Yao WU ; Xiao-Xiao CAI ; Yu-Rong LIU ; Yuan-Ding HUANG ; Yun-Feng LIN
International Journal of Oral Science 2009;1(2):81-89
AIMTo investigate the effect of DAPT (gamma-secretase inhibitor) on the growth of human tongue carcinoma cells and to determine the molecular mechanism to enable the potential application of DAPT to the treatment of tongue carcinoma.
METHODOLOGYHuman tongue carcinoma Tca8113 cells were cultured with DAPT. Cell growth was determined using Indigotic Reduction method. The cell cycle and apoptosis were analyzed by flow cytometry. Real-time PCR and Immuno-Fluorescence (IF) were employed to determine the intracellular expression levels.
RESULTSDAPT inhibited the growth of human tongue carcinoma Tca8113 cells by inducing G0-G1 cell cycle arrest and apoptosis. The mRNA levels of Hairy/Enhancer of Split-1 (Hes-1), a target of Notch activation, were reduced by DAPT in a dose-dependent manner. Coincident with this observation, DAPT induced a dose-dependent promotion of constitutive Caspase-3 in Tca8113 cells.
CONCLUSIONDAPT may have a therapeutic value for human tongue carcinoma. Moreover, the effects of DAPT in tumor inhibition may arise partly via the modulation of Notch-1 and Caspase-3.
Amyloid Precursor Protein Secretases ; antagonists & inhibitors ; Antineoplastic Agents ; administration & dosage ; pharmacology ; Apoptosis ; drug effects ; Basic Helix-Loop-Helix Transcription Factors ; drug effects ; Carcinoma ; pathology ; Caspase 3 ; drug effects ; Cell Line, Tumor ; Cell Membrane ; drug effects ; Cell Nucleus ; drug effects ; Cyclin D1 ; drug effects ; Dipeptides ; administration & dosage ; pharmacology ; Dose-Response Relationship, Drug ; G1 Phase ; drug effects ; Homeodomain Proteins ; drug effects ; Humans ; Receptor, Notch1 ; drug effects ; Repressor Proteins ; drug effects ; Resting Phase, Cell Cycle ; drug effects ; Tongue Neoplasms ; pathology ; Transcription Factor HES-1
9.Tunicamycin enhances TRAIL-induced apoptosis by inhibition of cyclin D1 and the subsequent downregulation of survivin.
Hai Yan ZHANG ; Zhen Xian DU ; Bao Qin LIU ; Yan Yan GAO ; Xin MENG ; Yifu GUAN ; Wei Wei DENG ; Hua Qin WANG
Experimental & Molecular Medicine 2009;41(5):362-369
TNF-related apoptosis-inducing ligand (TRAIL) has been proposed as a promising cancer therapy that preferentially induces apoptosis in cancer cells, but not most normal tissues. However, many cancers are resistant to TRAIL by mechanisms that are poorly understood. In this study, we showed that tunicamycin, a naturally occurring antibiotic, was a potent enhancer of TRAIL-induced apoptosis through downregulation of survivin. The tunicamycin-mediated sensitization to TRAIL was efficiently reduced by forced expression of survivin, suggesting that the sensitization was mediated at least in part through inhibition of survivin expression. Tunicamycin also repressed expression of cyclin D1, a cell cycle regulator commonly overexpressed in thyroid carcinoma. Furthermore, silencing cyclin D1 by RNA interference reduced survivin expression and sensitized thyroid cancer cells to TRAIL; in contrast, forced expression of cyclin D1 attenuated tunicamycin-potentiated TRAIL-induced apoptosis via over-riding downregulation of survivin. Collectively, our results demonstrated that tunicamycin promoted TRAIL-induced apoptosis, at least in part, by inhibiting the expression of cyclin D1 and subsequent survivin. Of note, tunicamycin did not sensitize the differentiated thyroid epithelial cells to TRAIL-induced apoptosis. Thus, combined treatment with tunicamycin and TRAIL may offer an attractive strategy for safely treating resistant thyroid cancers.
Anti-Bacterial Agents/*pharmacology
;
*Apoptosis
;
Cell Line, Tumor
;
Cyclin D1/*antagonists & inhibitors/metabolism
;
*Down-Regulation
;
Humans
;
Microtubule-Associated Proteins/*genetics/metabolism
;
TNF-Related Apoptosis-Inducing Ligand/*metabolism
;
Tunicamycin/*pharmacology
10.Tunicamycin enhances TRAIL-induced apoptosis by inhibition of cyclin D1 and the subsequent downregulation of survivin.
Hai Yan ZHANG ; Zhen Xian DU ; Bao Qin LIU ; Yan Yan GAO ; Xin MENG ; Yifu GUAN ; Wei Wei DENG ; Hua Qin WANG
Experimental & Molecular Medicine 2009;41(5):362-369
TNF-related apoptosis-inducing ligand (TRAIL) has been proposed as a promising cancer therapy that preferentially induces apoptosis in cancer cells, but not most normal tissues. However, many cancers are resistant to TRAIL by mechanisms that are poorly understood. In this study, we showed that tunicamycin, a naturally occurring antibiotic, was a potent enhancer of TRAIL-induced apoptosis through downregulation of survivin. The tunicamycin-mediated sensitization to TRAIL was efficiently reduced by forced expression of survivin, suggesting that the sensitization was mediated at least in part through inhibition of survivin expression. Tunicamycin also repressed expression of cyclin D1, a cell cycle regulator commonly overexpressed in thyroid carcinoma. Furthermore, silencing cyclin D1 by RNA interference reduced survivin expression and sensitized thyroid cancer cells to TRAIL; in contrast, forced expression of cyclin D1 attenuated tunicamycin-potentiated TRAIL-induced apoptosis via over-riding downregulation of survivin. Collectively, our results demonstrated that tunicamycin promoted TRAIL-induced apoptosis, at least in part, by inhibiting the expression of cyclin D1 and subsequent survivin. Of note, tunicamycin did not sensitize the differentiated thyroid epithelial cells to TRAIL-induced apoptosis. Thus, combined treatment with tunicamycin and TRAIL may offer an attractive strategy for safely treating resistant thyroid cancers.
Anti-Bacterial Agents/*pharmacology
;
*Apoptosis
;
Cell Line, Tumor
;
Cyclin D1/*antagonists & inhibitors/metabolism
;
*Down-Regulation
;
Humans
;
Microtubule-Associated Proteins/*genetics/metabolism
;
TNF-Related Apoptosis-Inducing Ligand/*metabolism
;
Tunicamycin/*pharmacology

Result Analysis
Print
Save
E-mail