1.Effect and Mechanism of Atorvastatin on Reversing Drug Resistance in Leukemia by Regulating Glycolysis through PTEN/mTOR Pathway.
Journal of Experimental Hematology 2023;31(1):38-44
OBJECTIVE:
To investigate the influence and mechanism of atorvastatin on glycolysis of adriamycin resistant acute promyelocytic leukemia (APL) cell line HL-60/ADM.
METHODS:
HL-60/ADM cells in logarithmic growth phase were treated with different concentrations of atorvastatin, then the cell proliferation activity was measured by CCK-8 assay, the apoptosis was detected by flow cytometry, the glycolytic activity was checked by glucose consumption test, and the protein expressions of PTEN, p-mTOR, PKM2, HK2, P-gp and MRP1 were detected by Western blot. After transfection of PTEN-siRNA into HL-60/ADM cells, the effects of low expression of PTEN on atorvastatin regulating the behaviors of apoptosis and glycolytic metabolism in HL-60/ADM cells were further detected.
RESULTS:
CCK-8 results showed that atorvastatin could inhibit the proliferation of HL-60/ADM cells in a concentration-dependent and time-dependent manner (r=0.872, r=0.936), and the proliferation activity was inhibited most significantly when treated with 10 μmol/L atorvastatin for 24 h, which was decreased to (32.3±2.18)%. Flow cytometry results showed that atorvastatin induced the apoptosis of HL-60/ADM cells in a concentration-dependent manner (r=0.796), and the apoptosis was induced most notably when treated with 10 μmol/L atorvastatin for 24 h, which reached to (48.78±2.95)%. The results of glucose consumption test showed that atorvastatin significantly inhibited the glycolytic activity of HL-60/ADM cells in a concentration-dependent and time-dependent manner (r=0.915, r=0.748), and this inhibition was most strikingly when treated with 10 μmol/L atorvastatin for 24 h, reducing the relative glucose consumption to (46.53±1.71)%. Western blot indicated that the expressions of p-mTOR, PKM2, HK2, P-gp and MRP1 protein were decreased in a concentration-dependent manner (r=0.737, r=0.695, r=0.829, r=0.781, r=0.632), while the expression of PTEN protein was increased in a concentration-dependent manner (r=0.531), when treated with different concentrations of atorvastatin for 24 h. After PTEN-siRNA transfected into HL-60/ADM cells, it showed that low expression of PTEN had weakened the promoting effect of atorvastatin on apoptosis and inhibitory effect on glycolysis and multidrug resistance.
CONCLUSION
Atorvastatin can inhibit the proliferation, glycolysis, and induce apoptosis of HL-60/ADM cells. It may be related to the mechanism of increasing the expression of PTEN, inhibiting mTOR activation, and decreasing the expressions of PKM2 and HK2, thus reverse drug resistance.
Humans
;
Atorvastatin/pharmacology*
;
PTEN Phosphohydrolase/pharmacology*
;
Sincalide/metabolism*
;
Drug Resistance, Neoplasm/genetics*
;
TOR Serine-Threonine Kinases/metabolism*
;
Leukemia, Promyelocytic, Acute/drug therapy*
;
Doxorubicin/pharmacology*
;
Apoptosis
;
RNA, Small Interfering/pharmacology*
;
Glycolysis
;
Glucose/therapeutic use*
;
Cell Proliferation
2.Read-through circular RNA rt-circ-HS promotes hypoxia inducible factor 1α expression and renal carcinoma cell proliferation, migration and invasiveness.
Yun Yi XU ; Zheng Zheng SU ; Lin Mao ZHENG ; Meng Ni ZHANG ; Jun Ya TAN ; Ya Lan YANG ; Meng Xin ZHANG ; Miao XU ; Ni CHEN ; Xue Qin CHEN ; Qiao ZHOU
Journal of Peking University(Health Sciences) 2023;55(2):217-227
OBJECTIVE:
To identify and characterize read-through RNAs and read-through circular RNAs (rt-circ-HS) derived from transcriptional read-through hypoxia inducible factor 1α (HIF1α) and small nuclear RNA activating complex polypeptide 1 (SNAPC1) the two adjacent genes located on chromosome 14q23, in renal carcinoma cells and renal carcinoma tissues, and to study the effects of rt-circ-HS on biological behavior of renal carcinoma cells and on regulation of HIF1α.
METHODS:
Reverse transcription-polymerase chain reaction (RT-PCR) and Sanger sequencing were used to examine expression of read-through RNAs HIF1α-SNAPC1 and rt-circ-HS in different tumor cells. Tissue microarrays of 437 different types of renal cell carcinoma (RCC) were constructed, and chromogenic in situ hybridization (ISH) was used to investigate expression of rt-circ-HS in different RCC types. Small interference RNA (siRNA) and artificial overexpression plasmids were designed to examine the effects of rt-circ-HS on 786-O and A498 renal carcinoma cell proliferation, migration and invasiveness by cell counting kit 8 (CCK8), EdU incorporation and Transwell cell migration and invasion assays. RT-PCR and Western blot were used to exa-mine expression of HIF1α and SNAPC1 RNA and proteins after interference of rt-circ-HS with siRNA, respectively. The binding of rt-circ-HS with microRNA 539 (miR-539), and miR-539 with HIF1α 3' untranslated region (3' UTR), and the effects of these interactions were investigated by dual luciferase reporter gene assays.
RESULTS:
We discovered a novel 1 144 nt rt-circ-HS, which was derived from read-through RNA HIF1α-SNAPC1 and consisted of HIF1α exon 2-6 and SNAPC1 exon 2-4. Expression of rt-circ-HS was significantly upregulated in 786-O renal carcinoma cells. ISH showed that the overall positive expression rate of rt-circ-HS in RCC tissue samples was 67.5% (295/437), and the expression was different in different types of RCCs. Mechanistically, rt-circ-HS promoted renal carcinoma cell proliferation, migration and invasiveness by functioning as a competitive endogenous inhibitor of miR-539, which we found to be a potent post-transcriptional suppressor of HIF1α, thus promoting expression of HIF1α.
CONCLUSION
The novel rt-circ-HS is highly expressed in different types of RCCs and acts as a competitive endogenous inhibitor of miR-539 to promote expression of its parental gene HIF1α and thus the proliferation, migration and invasion of renal cancer cells.
Humans
;
Carcinoma, Renal Cell/pathology*
;
Cell Proliferation
;
Hypoxia
;
Kidney Neoplasms
;
MicroRNAs/genetics*
;
Neoplasm Invasiveness/genetics*
;
RNA, Circular/metabolism*
;
RNA, Small Interfering
;
Hypoxia-Inducible Factor 1, alpha Subunit/genetics*
3.Effect of SLC7A11 gene downregulation on the gefitinib resistance of lung adenocarcinoma PC9/GR cells and its mechanism.
Yun Long JIA ; Yan ZHAO ; Shu Man ZHEN ; Zi Shuo CHENG ; Bo Yang ZHENG ; Yue Ping LIU ; Li Hua LIU
Chinese Journal of Oncology 2023;45(9):779-786
Objective: To screen the key genes involved in gefitinib resistance of lung adenocarcinoma PC9/GR cells which harbored 19 exon mutation of epidermal growth factor receptor (EGFR) gene, and discuss the effect and mechanism of downregulation of solute carrier family 7 member 11 (SLC7A11) on the gefitinib resistance of PC9/GR cells. Methods: RNA microarray was conducted to detect the gene expressions in PC9 and PC9/GR cells. The differently expressed genes were screened by using limma package of R language and analyzed by Kyoto encyclopedia of genes and genomes (KEGG) pathway enrichment analysis. Western blotting was performed to determine the expression of SLC7A11 protein in PC9 and PC9/GR cells. PC9/GR cells were infected with lentivirus plasmid containing short hairpin RNA (shRNA) targeting SLC7A11 or negative control shRNA (sh-NC), respectively. Real-time quantitative polymerase chain reaction (RT-qPCR) was performed to evaluate the efficacy of shRNA on the expression of SLC7A11 mRNA. Cell counting kit-8 (CCK-8) assay was conducted to determine the suppressing effect of gefitinib on PC9/GR cells. Mito-Tracker Red CMXRos probe and malondialdehyde (MDA) assay kit were used to evaluate gefitinib-induced ferroptosis in PC9/GR cells. Immunohistochemistry (IHC) was conducted to detect the expression of SLC7A11 protein in the tumor tissues of advanced stage lung adenocarcinoma patients harboring 19 exon mutation of EGFR gene. Thirty-six advanced stage lung adenocarcinoma patients who received EGFR-tyrosihe kinase inhibitor(TKI) as first-line treatment in Fourth Hospital of Hebei Medical Unviersity were enrolled. Kaplan-Meier survival curve was drawn to analyze the correlation between SLC7A11 expression and progression-free survival (PFS) of the patients. Results: RNA array demonstrated that 2 888 genes were differently expressed between PC9 and PC9/GR cells. KEGG analysis showed that ferroptosis-related gene was one of the most enriched region of the differently expressed genes between PC9 and PC9/GR cells. These ferroptosis-related gene cohort contained 13 genes, among which SLC7A11 exhibited the most significant difference. Western blotting showed that the expression of SLC7A11 protein in PC9/GR cells was significantly higher than that in PC9 cells (0.76±0.03 vs. 0.19±0.02, P<0.001). The 50% inhibiting concentration (IC(50)) of gefitinib was 35.08 μmol/L and 64.01 μmol/L for sh-SLC7A11 and sh-NC group PC9/GR cells, respectively. PC9/GR cells in sh-SLC7A11 group exhibited significantly lower density of mitochondria fluorescence after gefitinib treatment, compared to the sh-NC group (213.77±26.50 vs. 47.88±4.55, P<0.001). In addition, PC9/GR cells in sh-SLC7A11 group exhibited significantly higher MDA after gefitinib treatment, compared to the sh-NC group [(15.43±1.60) μmol/mg vs. (82.18±7.77) μmol/mg, P<0.001]. The PFS of the patients with low expression of SLC7A11 (n=18) was significantly longer than the patients with high expression of SLC7A11 (n=18, 16.77 months vs. 9.14 months, P<0.001). Conclusion: Downregulation of SLC7A11 could increase the sensitivity of PC9/GR cells to gefitinib by promoting ferroptosis.
Humans
;
Gefitinib/therapeutic use*
;
Antineoplastic Agents/therapeutic use*
;
Lung Neoplasms/pathology*
;
Down-Regulation
;
Quinazolines/therapeutic use*
;
Drug Resistance, Neoplasm/genetics*
;
ErbB Receptors/metabolism*
;
Adenocarcinoma of Lung
;
Protein Kinase Inhibitors/therapeutic use*
;
RNA, Small Interfering/genetics*
;
Cell Line, Tumor
;
Amino Acid Transport System y+/metabolism*
4.Effect of SLC7A11 gene downregulation on the gefitinib resistance of lung adenocarcinoma PC9/GR cells and its mechanism.
Yun Long JIA ; Yan ZHAO ; Shu Man ZHEN ; Zi Shuo CHENG ; Bo Yang ZHENG ; Yue Ping LIU ; Li Hua LIU
Chinese Journal of Oncology 2023;45(9):779-786
Objective: To screen the key genes involved in gefitinib resistance of lung adenocarcinoma PC9/GR cells which harbored 19 exon mutation of epidermal growth factor receptor (EGFR) gene, and discuss the effect and mechanism of downregulation of solute carrier family 7 member 11 (SLC7A11) on the gefitinib resistance of PC9/GR cells. Methods: RNA microarray was conducted to detect the gene expressions in PC9 and PC9/GR cells. The differently expressed genes were screened by using limma package of R language and analyzed by Kyoto encyclopedia of genes and genomes (KEGG) pathway enrichment analysis. Western blotting was performed to determine the expression of SLC7A11 protein in PC9 and PC9/GR cells. PC9/GR cells were infected with lentivirus plasmid containing short hairpin RNA (shRNA) targeting SLC7A11 or negative control shRNA (sh-NC), respectively. Real-time quantitative polymerase chain reaction (RT-qPCR) was performed to evaluate the efficacy of shRNA on the expression of SLC7A11 mRNA. Cell counting kit-8 (CCK-8) assay was conducted to determine the suppressing effect of gefitinib on PC9/GR cells. Mito-Tracker Red CMXRos probe and malondialdehyde (MDA) assay kit were used to evaluate gefitinib-induced ferroptosis in PC9/GR cells. Immunohistochemistry (IHC) was conducted to detect the expression of SLC7A11 protein in the tumor tissues of advanced stage lung adenocarcinoma patients harboring 19 exon mutation of EGFR gene. Thirty-six advanced stage lung adenocarcinoma patients who received EGFR-tyrosihe kinase inhibitor(TKI) as first-line treatment in Fourth Hospital of Hebei Medical Unviersity were enrolled. Kaplan-Meier survival curve was drawn to analyze the correlation between SLC7A11 expression and progression-free survival (PFS) of the patients. Results: RNA array demonstrated that 2 888 genes were differently expressed between PC9 and PC9/GR cells. KEGG analysis showed that ferroptosis-related gene was one of the most enriched region of the differently expressed genes between PC9 and PC9/GR cells. These ferroptosis-related gene cohort contained 13 genes, among which SLC7A11 exhibited the most significant difference. Western blotting showed that the expression of SLC7A11 protein in PC9/GR cells was significantly higher than that in PC9 cells (0.76±0.03 vs. 0.19±0.02, P<0.001). The 50% inhibiting concentration (IC(50)) of gefitinib was 35.08 μmol/L and 64.01 μmol/L for sh-SLC7A11 and sh-NC group PC9/GR cells, respectively. PC9/GR cells in sh-SLC7A11 group exhibited significantly lower density of mitochondria fluorescence after gefitinib treatment, compared to the sh-NC group (213.77±26.50 vs. 47.88±4.55, P<0.001). In addition, PC9/GR cells in sh-SLC7A11 group exhibited significantly higher MDA after gefitinib treatment, compared to the sh-NC group [(15.43±1.60) μmol/mg vs. (82.18±7.77) μmol/mg, P<0.001]. The PFS of the patients with low expression of SLC7A11 (n=18) was significantly longer than the patients with high expression of SLC7A11 (n=18, 16.77 months vs. 9.14 months, P<0.001). Conclusion: Downregulation of SLC7A11 could increase the sensitivity of PC9/GR cells to gefitinib by promoting ferroptosis.
Humans
;
Gefitinib/therapeutic use*
;
Antineoplastic Agents/therapeutic use*
;
Lung Neoplasms/pathology*
;
Down-Regulation
;
Quinazolines/therapeutic use*
;
Drug Resistance, Neoplasm/genetics*
;
ErbB Receptors/metabolism*
;
Adenocarcinoma of Lung
;
Protein Kinase Inhibitors/therapeutic use*
;
RNA, Small Interfering/genetics*
;
Cell Line, Tumor
;
Amino Acid Transport System y+/metabolism*
5.MiR-125b-5 suppresses ovarian cancer cell migration and invasion by targeted downregulation of CD147.
Zhen HUANG ; Hao Ming SHEN ; Hong Yu DENG ; Li Sha SUN ; Bin QÜ
Journal of Southern Medical University 2022;42(9):1389-1396
OBJECTIVE:
To investigate whether miR-125b-5p regulates biological behaviors of ovarian cancer cells by targeted regulation of CD147 expression.
METHODS:
RT-qPCR was used to detect the expression of miR-125b-5p and CD147 mRNA in ovarian cancer tissues and cancer cell lines. SKOV3 cells transfected with miR-125b-5p mimic and HO8910 cells transfected with miR-125b-5p inhibitor were examined for changes in proliferation, migration and invasion using CCK-8 assay, colonyforming assay and Transwell assay. Starbase was used to predict the potential binding sites between miR-125b-5p and CD147, and double luciferase reporter gene assay was used to verify the targeting relationship. In SKOV3 cells, the effects of cotransfection with miR-125b-5p mimic and pcDNA3.1-CD147 (or pcDNA3.1) plasmid on cell proliferation, migration and invasion were assessed with CCK-8 assay and Transwell assay.
RESULTS:
The expression of miR-125b-5p was significantly lowered and that of CD147 was increased in both ovarian cancer tissues and ovarian cancer cell lines (P < 0.05). Overexpression of miR-125b-5p in SKOV3 cells resulted in significantly suppressed cell proliferation, migration and invasion, while downregulation of miR-125b-5p in HO8910 cells promoted cell proliferation, migration and invasion. Bioinformatic analysis predicted that miR-125b-5p binds to CD147, which was confirmed by luciferase reporter gene assay. RT-qPCR and Western blotting showed that miR-125b-5p negatively regulated CD147 expression (P < 0.05). In SKOV3 cells, the inhibitory effects of miR-125b-5p mimic on cell proliferation, invasion and migration were significantly attenuated by co-transfection of the cells with pcDNA3.1-CD147 plasmid.
CONCLUSION
miR-125b-5p inhibits the migration and invasion of ovarian cancer cells by negatively regulating the expression of CD147.
Basigin
;
Carcinoma, Ovarian Epithelial
;
Cell Line, Tumor
;
Cell Movement/genetics*
;
Cell Proliferation/genetics*
;
Down-Regulation
;
Female
;
Gene Expression Regulation, Neoplastic
;
Humans
;
MicroRNAs/metabolism*
;
Neoplasm Invasiveness/genetics*
;
Ovarian Neoplasms/genetics*
;
RNA, Messenger/genetics*
;
Sincalide/metabolism*
6.Nur77 promotes invasion and migration of gastric cancer cells through the NF-κB/IL-6 pathway.
Wei LI ; Yong Kang SHI ; Yu Hua GUO ; Sheng Wang TIAN
Journal of Southern Medical University 2022;42(9):1410-1417
OBJECTIVE:
To analyze the association of Nur77 with overall survival of gastric cancer patients and investigate the role of Nur77 in invasion and migration of gastric cancer cells.
METHODS:
Oncomine database was used to analyze the expression of Nur77 in gastric cancer and gastric mucosa tissues, and the distribution characteristics of Nur77 protein between gastric cancer and normal tissues were compared using Human Protein Atlas. GEPIA2 was used to analyze the relationship of Nur77 expression and the patients' survival. The expression of Nur77 in gastric cancer cell lines GES-1, AGS and MKN-45 were detected by Western blotting. The regulatory interactions between IL-6 and Nur77 were verified by transfecting the cells with specific Nur-77 siRNA and Nur-77-overexpressing plasmid. The changes in migration ability of the cells following Nur-77 knockdown were assessed with scratch assay. The effect of Nur-77 overexpression or IL-6 knockdown, or their combination, on migration and invasion of the gastric cancer cells were examined using Transwell assay. The effect of Nur77 expression level on NF-κB/IL-6 pathway activation was analyzed using Western blotting.
RESULTS:
Oncomine database showed that gastric cancer tissues expressed a significantly higher level of Nur77 mRNA than normal tissues (P < 0.05). Nur77 expression was detected mostly in the nucleus, and a high Nur77 expression was associated with a poor survival outcome of the patients (P < 0.05). In gastric cancer cells, the high expression of Nur77 participated in the regulation of IL-6. Nur77 silencing significantly lowered the migration ability of the cells (P < 0.05), and IL-6 silencing significantly attenuated the enhanced migration caused by Nur77 overexpression (P < 0.05). Nur77 participates in the activation of NF-κB/IL-6 signaling pathway by regulating the expression of p-p65, p65, p-Stat3 and Stat3.
CONCLUSION
A high Nur77 expression is strongly correlated with a poor prognosis of gastric cancer patients. Nur77 promotes the invasion and migration of gastric cancer cells possibly by regulating the NF-κB/IL-6 signaling pathway.
Cell Line, Tumor
;
Cell Movement
;
Gene Expression Regulation, Neoplastic
;
Humans
;
Interleukin-6/metabolism*
;
NF-kappa B/metabolism*
;
Neoplasm Invasiveness/genetics*
;
Nuclear Receptor Subfamily 4, Group A, Member 1
;
RNA, Messenger/metabolism*
;
RNA, Small Interfering/genetics*
;
Stomach Neoplasms/genetics*
7.Expression of miR-4324 and its targeted gene Talin2 in breast cancer.
Peng GAO ; Hai Tao ZHU ; Wen Hao PEI ; Pei Hai XU ; Yong Xing DING
Journal of Southern Medical University 2022;42(10):1517-1525
OBJECTIVE:
To investigate the regulatory effect of miR-4324 on ankyrin 2(Talin2) expression and biological behaviors of breast cancer cells and the clinical implications of changes in miR-4324 and Talin2 expressions in breast cancer.
METHODS:
In breast cancer and adjacent tissues, the expressions of Talin2 and miR-4324 were examined with immunohistochemistry and qRT-PCR, respectively and the association of Talin2 expression levels with the prognosis and clinicopathological features of breast cancer patients was analyzed.The human breast cancer cell line SKBR-3 was transfected with miR-4324 mimic, miR-4324 inhibitor, si-Talin2, or both miR-4324 inhibitor and si-Talin2, and the changes in biological behaviors of the cells were examined; the cellular expression of Talin2at the mRNA and protein levels were detected with qRT-PCR and Western blotting.Dual luciferase reporter gene assay was used to verify the targeting relationship between miR-4324 and Talin2.The effect of miR-4324-mediated regulation of Talin2 on SKBR-3 cell migration was assessed using Transwell assays.
RESULTS:
Talin2 expression was significantly higher in breast cancer tissues than in the adjacent tissues, and its expression level was correlated with lymph node metastasis and high HER-2 expression in breast cancer (P < 0.05) but not with the patient's age, clinical stage, histological grade or expressions of estrogen and progesterone receptors (P >0.05).The expression of miR-4324 was significantly reduced in breast cancer tissues as compared with the adjacent tissues (P < 0.01).In SKBR-3 cells, transfection with miR-4324 mimics significantly inhibited proliferation, migration and invasion (P < 0.05) and promoted apoptosis (P < 0.01) of the cells.Dual luciferase reporter gene assay confirmed that cotransfection with miR-4324 mimics significantly reduced luciferase activity of Talin2-3'-UTR WT reporter plasmid (P < 0.05).Transfection of the cells with miR-4324 mimics significantly reduced mRNA and protein expressions of Talin2(P < 0.05).Transwell migration assay showed that the migration ability of SKBR-3 cells was significantly enhanced after transfection with miR-4324 inhibitor (P < 0.01), lowered after transfection with si-Talin2(P < 0.01), and maintained at the intermediate level after co-transfection with miR-4324 inhibitor+si-Talin2 group (P < 0.05).
CONCLUSIONS
High expression of Talin2 is associated with lymph node metastasis and HER-2 overexpression in breast cancer patients.Down-regulation of miR-4324 inhibits the proliferation, invasion and migration and induces apoptosis of breast cancer cells, and the inhibitory effect of miR-4324 knockdown on breast cancer cell migration is mediated probably by targeted inhibition of Talin2 expression.
Female
;
Humans
;
Breast Neoplasms/pathology*
;
Cell Line, Tumor
;
Cell Movement/genetics*
;
Cell Proliferation/genetics*
;
Gene Expression Regulation, Neoplastic
;
Luciferases/genetics*
;
Lymphatic Metastasis
;
MicroRNAs/metabolism*
;
Neoplasm Invasiveness/genetics*
;
RNA, Messenger
8.KIF2C: a novel link between Wnt/β-catenin and mTORC1 signaling in the pathogenesis of hepatocellular carcinoma.
Shi WEI ; Miaomiao DAI ; Chi ZHANG ; Kai TENG ; Fengwei WANG ; Hongbo LI ; Weipeng SUN ; Zihao FENG ; Tiebang KANG ; Xinyuan GUAN ; Ruihua XU ; Muyan CAI ; Dan XIE
Protein & Cell 2021;12(10):788-809
Hepatocellular carcinoma (HCC) is the most common primary liver malignancy and is the fourth-leading cause of cancer-related deaths worldwide. HCC is refractory to many standard cancer treatments and the prognosis is often poor, highlighting a pressing need to identify biomarkers of aggressiveness and potential targets for future treatments. Kinesin family member 2C (KIF2C) is reported to be highly expressed in several human tumors. Nevertheless, the molecular mechanisms underlying the role of KIF2C in tumor development and progression have not been investigated. In this study, we found that KIF2C expression was significantly upregulated in HCC, and that KIF2C up-regulation was associated with a poor prognosis. Utilizing both gain and loss of function assays, we showed that KIF2C promoted HCC cell proliferation, migration, invasion, and metastasis both in vitro and in vivo. Mechanistically, we identified TBC1D7 as a binding partner of KIF2C, and this interaction disrupts the formation of the TSC complex, resulting in the enhancement of mammalian target of rapamycin complex1 (mTORC1) signal transduction. Additionally, we found that KIF2C is a direct target of the Wnt/β-catenin pathway, and acts as a key factor in mediating the crosstalk between Wnt/β-catenin and mTORC1 signaling. Thus, the results of our study establish a link between Wnt/β-catenin and mTORC1 signaling, which highlights the potential of KIF2C as a therapeutic target for the treatment of HCC.
Adult
;
Aged
;
Animals
;
Carcinoma, Hepatocellular/pathology*
;
Cell Line, Tumor
;
Cell Movement
;
Cell Proliferation
;
Epithelial-Mesenchymal Transition/genetics*
;
Female
;
Gene Expression Regulation, Neoplastic
;
Humans
;
Intracellular Signaling Peptides and Proteins/metabolism*
;
Kinesins/metabolism*
;
Liver Neoplasms/pathology*
;
Male
;
Mice
;
Mice, Inbred BALB C
;
Middle Aged
;
Neoplasm Staging
;
Prognosis
;
Protein Binding
;
RNA, Small Interfering/metabolism*
;
Survival Analysis
;
Tumor Burden
;
Wnt Signaling Pathway
;
Xenograft Model Antitumor Assays
;
beta Catenin/metabolism*
9.Inhibition of chemotherapy-related breast tumor EMT by application of redox-sensitive siRNA delivery system CSO-ss-SA/siRNA along with doxorubicin treatment.
Xuan LIU ; Xue-Qing ZHOU ; Xu-Wei SHANG ; Li WANG ; Yi LI ; Hong YUAN ; Fu-Qiang HU
Journal of Zhejiang University. Science. B 2020;21(3):218-233
Metastasis is one of the main reasons causing death in cancer patients. It was reported that chemotherapy might induce metastasis. In order to uncover the mechanism of chemotherapy-induced metastasis and find solutions to inhibit treatment-induced metastasis, the relationship between epithelial-mesenchymal transition (EMT) and doxorubicin (DOX) treatment was investigated and a redox-sensitive small interfering RNA (siRNA) delivery system was designed. DOX-related reactive oxygen species (ROS) were found to be responsible for the invasiveness of tumor cells in vitro, causing enhanced EMT and cytoskeleton reconstruction regulated by Ras-related C3 botulinum toxin substrate 1 (RAC1). In order to decrease RAC1, a redox-sensitive glycolipid drug delivery system (chitosan-ss-stearylamine conjugate (CSO-ss-SA)) was designed to carry siRNA, forming a gene delivery system (CSO-ss-SA/siRNA) downregulating RAC1. CSO-ss-SA/siRNA exhibited an enhanced redox sensitivity compared to nonresponsive complexes in 10 mmol/L glutathione (GSH) and showed a significant safety. CSO-ss-SA/siRNA could effectively transmit siRNA into tumor cells, reducing the expression of RAC1 protein by 38.2% and decreasing the number of tumor-induced invasion cells by 42.5%. When combined with DOX, CSO-ss-SA/siRNA remarkably inhibited the chemotherapy-induced EMT in vivo and enhanced therapeutic efficiency. The present study indicates that RAC1 protein is a key regulator of chemotherapy-induced EMT and CSO-ss-SA/siRNA silencing RAC1 could efficiently decrease the tumor metastasis risk after chemotherapy.
Amines/chemistry*
;
Antineoplastic Agents/adverse effects*
;
Breast Neoplasms/pathology*
;
Chitosan/chemistry*
;
Doxorubicin/adverse effects*
;
Drug Delivery Systems
;
Epithelial-Mesenchymal Transition/drug effects*
;
Female
;
Humans
;
MCF-7 Cells
;
Neoplasm Metastasis/prevention & control*
;
Oxidation-Reduction
;
RNA, Small Interfering/administration & dosage*
;
Reactive Oxygen Species/metabolism*
;
rac1 GTP-Binding Protein/physiology*
10.Mig- gene silencing inhibits vasculogenic mimicry formation and invasion of glioma U251 cells by suppressing MEK/ERK signaling.
Fan WANG ; Fenglong CHEN ; Weipeng HU ; Yi ZHANG
Journal of Southern Medical University 2019;39(5):566-571
OBJECTIVE:
To investigate the inhibitory effects of silencing migration-inducing gene-7 (Mig-7) on vasculogenic mimicry formation, migration and invasion of human glioma cells and whether MEK/ERK signaling pathway mediates these effects.
METHODS:
Human glioma U251 cells were infected by lentiviral vectors carrying a small interfering RNA targeting Mig-7 gene (sh-Mig-) or a negative control shRNA (sh-NC), and real-time quantitative PCR was used to detect the expression level of Mig- mRNA in the cells. Three-dimensional culture and Transwell chamber invasion assay were used to observe the effect of Mig- gene silencing on vasculogenic mimicry formation and invasion ability of the U251 cells. Western blotting was performed to detect the changes in the protein expression levels of MEK/ERK in the infected cells.
RESULTS:
We successfully obtained a U251 cell line with stable low expression of Mig- gene using RNA interference technique. Compared with the cells infected with sh-NC lentivirus and the non- infected cells, U251 cells infected with the lentiviral vector carrying sh-Mig- showed significantly decreased expression level of Mig- ( < 0.01) with obviously lowered vasculogenic mimicry formation and invasion abilities ( < 0.05). Mig- silencing also significantly lowered the expressions of MEK and ERK proteins in U251 cells ( < 0.05).
CONCLUSIONS
Silencing of Mig-7 gene inhibits vasculogenic mimicry formation and invasion of U251 cells possibly by suppressing MEK/ERK signaling, suggesting the important role of Mig-7 gene in vasculogenic mimicry formation and invasion of human glioma cells.
Cell Line, Tumor
;
Cell Movement
;
Cell Proliferation
;
Gene Silencing
;
Glioma
;
genetics
;
pathology
;
Humans
;
Neoplasm Proteins
;
metabolism
;
RNA, Small Interfering
;
Signal Transduction

Result Analysis
Print
Save
E-mail