scholarly journals Targeting EZH2 Promotes Chemosensitivity of BCL-2 Inhibitor through PIK3IP1-PI3K/AKT Axis in Acute Myeloid Leukemia

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 3345-3345
Author(s):  
Chan Yang ◽  
Jie Zi ◽  
Chunhua Song ◽  
Zheng Ge

Abstract Introduction Enhancer of zeste homolog 2 (EZH2) is the catalytic subunit of the polycomb repressive complex 2 (PRC2), which plays critical roles in transcription repressions.[1] PRC2 is required for acute Myeloid Leukemia (AML) cell survival, and EZH2 is overexpressed and related to worse prognosis in AML, therefore, EZH2 may involve in leukemogenesis through inhibition of the tumor suppressor genes in AML. [2, 3] BCL-2, one of the key anti-apoptosis proteins is dysregulated in AML. Venetoclax (Ven, ABT-199), a BCL-2 selective inhibitor has anti-tumor activity in AML but its overall response rate of monotherapy was unsatisfied owing to the clinical resistance to the inhibitor. [4] In this study, we examined the effect of targeting EZH2 on chemosensitivity of BCL-2 inhibitor in AML and the underlying mechanisms. Methods Cell Counting Kit-8 assay was used for cell proliferation and cytotoxicity in U937 and MV-4-11 AML cells treated with vehicle control, EZH2 inhibitor (DZNeP), Ven and Combination (Combo, Ven+DZNeP) for 48 hours. Synergistic effect was analyzed with Calcusyn. RNA-seq was performed with total RNA isolated from U937 cells treated with 2μM DZNeP, 7.5μM Ven or vehicle for 48 hours. Apoptosis was measured by cell staining with Annexin V+propidium iodide (PI) following flow cytometry analysis. EZH2 mRNA level was examined by qPCR in 39 newly-diagnosed AML patients from February 1, 2016 to February 28, 2019 at our institute with an approval of the Ethics Committee. Level of the apoptotic effectors was detected by western blot. GEPIA (Gene Expression Profiling Interactive Analysis) and R2 genomics analysis and visualization application were utilized for survival analysis. Results Both Ven and DZNeP had a dose-dependent and time-dependent effect on cell proliferation arrest in U937 and MV4-11 cells. DZNeP significantly sensitized the effect of Ven on cell proliferation arrest compared to single drug only (P<0.001) (Fig.1a). CalcuSyn analysis showed the synergistic effect of the Ven+DZNeP on cell proliferation arrest (Fig.1b). Total apoptosis rate increased significantly in the Ven +DZNeP group in U937(32.04%±2.83) and MV-4-11 (25.73%±0.34) cells compared to single drug controls (Fig.1c). Also, level of the apoptotic effectors, PARP, Cleaved caspase-3 and Cleaved caspase-9 were significantly increased in Ven+DZNeP group compared to single drug and vehicle control (Fig.1d). Moreover, 1727 significantly regulated genes were identified by RNA-seq in U937 cells upon Ven treatment compared to vehicle control, and 1376 upon DZNeP treatment, in which 333 were overlapped (104 genes changed in the same direction but 228 changed in the opposite) (Fig 2a). The overlapped regulated genes upon the two drugs treatment are mainly involved in PIK3/AKT/mTOR, G1/S transition of mitotic cell cycle, apoptosis, et al. PIK3AP1, PIK3C2B and PIK3R3, the activators of PI3K signaling are up-regulated upon Ven treatment; and PI3KIP1, the suppressor of PI3K/AKT/mTOR signaling pathway is down-regulated by Ven but up-regulated by DZNeP. qPCR data showed that Ven+DZNeP significantly upregulated PIK3IP1 and down-regulated the expression of PI3K activators in the cells (Fig 2b). It is reported that the PI3K/AKT/mTOR signaling pathway is related to the clinical resistance of Ven in AML patients. Thus, our data not only showed the synergistic effect of Ven+DZNeP but also revealed a model that targeting EZH2 by DZNeP might conquer the Ven-induced bypass-effect through upregulation of PIK3IP1 to repression of PI3K/AKT signaling pathway. In addition, EZH2 mRNA level was quantified in 39 newly diagnosis patients with AML and 20 health control, and data showed EZH2 is increased in AML (p<0.05). Metadata analysis support our results, and EZH2 overexpression is associated with short overall survival (p<0.05) (Fig 2c). Database analysis assured that the expression of PIK3IP1 in patients with AML is lower than normal control. High PIK3IP1 showed a trend to a better outcome, which further supports that PIK3IP is a tumor suppressor in AML (Fig 2d). Conclusions Our data demonstrate the DZNeP sensitizes the effect of Ven in AML. Our results also reveal a novel mechanism that accounts for the synergistic effect of the two drugs and the fact that DZNeP may increase the chemosensitivity of BCL-2 inhibitor through PIK3IP1-PI3K/AKT axis in AML. Our findings suggest the potential combined therapy of Ven+DZNeP for AML. Figure 1 Figure 1. Disclosures No relevant conflicts of interest to declare.

2020 ◽  
Vol 40 (2) ◽  
Author(s):  
Yanting Zhang ◽  
Jie Bai ◽  
Wangli Si ◽  
Shanshan Yuan ◽  
Yijun Li ◽  
...  

Abstract As a zinc transporter, SLC39A7 (zip7) is vital in intestinal epithelial self-renewal, and recent studies suggested that SLC39A7 was related to cancer progression. Whereas, little is known about the role of SLC39A7 in gastric cancer (GC). In the present study, qRT-PCR analysis demonstrated that SLC39A7 mRNA level was increased in both GC tissues and cell lines. Overexpressing SLC39A7 boosted cell proliferation and migration, while inhibited apoptosis in GC. It was also found that si-SLC39A7 suppressed Akt/mTOR pathway and activation of Akt/mTOR pathway reversed the effects of si-SLC39A7 on GC development. Through prediction website, we found that SLC39A7 was directly regulated by miR-139-5p. miR-139-5p mimic had adverse effects on SLC39A7 expression and influence in the GC cell proliferation, migration and apoptosis by Akt/mTOR signaling pathway, while miR-139-5p inhibitor showed opposite effects. To conclude, our studies showed that SLC39A7 was negatively regulated by miR-139-5p. Besides, SLC39A7 positively regulated GC development through Akt/mTOR signaling pathway. These results indicate that SLC39A7 may be a candidate target gene for GC treatment.


2021 ◽  
Vol 20 ◽  
pp. 153303382199007
Author(s):  
Wenlin Liu ◽  
Jiandong Zhan ◽  
Rong Zhong ◽  
Rui Li ◽  
Xiaoli Sheng ◽  
...  

Background: Laryngeal cancer is one of the most common malignant tumors among head and neck cancers. Accumulating studies have indicated that long noncoding RNAs (lncRNAs) play an important role in laryngeal cancer occurrence and progression, however, the functional roles and relative regulatory mechanisms of lncRNA growth arrest-specific transcript 5 (GAS5) in laryngeal cancer progression remain unclear. Methods: The expression of lncRNA GAS5 in both laryngeal cancer tissues and cell lines was evaluated using quantitative reverse transcription-polymerase chain reaction (RT-qPCR) assay. The relationships between lncRNA GAS5 expression and clinical parameters were also analyzed. To determine the biological function of lncRNA GAS5, a lncRNA GAS5-specific plasmid was first transfected into laryngeal cancer cells using lentiviral technology. Cell counting kit-8 assay, flow cytometry, and Transwell assays were used to detect in vitro cell proliferation, apoptosis, cycle distribution, and metastasis abilities, respectively. Furthermore, in vivo cell growth experiments were also performed using nude mice. Additionally, western blotting was performed to identify the underlying regulatory mechanism. Results: In the current study, lncRNA GAS5 was downregulated in laryngeal cancer tissues and its low expression was closely associated with poor tumor differentiation, advanced TNM stage, lymph node metastasis, and shorter overall survival time. In addition, lncRNA GAS5 upregulation significantly inhibited laryngeal cancer cell proliferation both in vitro and in vivo. Moreover, in response to lncRNA GAS5 overexpression, more laryngeal cancer cells were arrested at the G2/M stage, accompanied by increased cell apoptosis rates and suppressed migration and invasion capacities. Mechanistically, our data showed that the overexpression of lncRNA GAS5 significantly regulated the PI3K/AKT/mTOR signaling pathway. Conclusion: LncRNA GAS5 might act as a suppressor gene during laryngeal cancer development, as it suppressed cell proliferation and metastasis by regulating the PI3K/AKT/mTOR signaling pathway; thus, lncRNA GAS5 is a promising therapeutic biomarker for the treatment of laryngeal cancer.


Tumor Biology ◽  
2017 ◽  
Vol 39 (5) ◽  
pp. 101042831769756 ◽  
Author(s):  
Hui Shi ◽  
Jin Pu ◽  
Xiao-Li Zhou ◽  
Yun-Ye Ning ◽  
Chong Bai

This study aimed to investigate the effects of long non-coding RNA ROR (regulator of reprogramming) on cisplatin (DDP) resistance in patients with non-small-cell lung cancer by regulating PI3K/Akt/mTOR signaling pathway. Human cisplatin-resistant A549/DDP cell lines were selected and divided into control group, negative control group, si-ROR group, ROR over-expression group, Wortmannin group, and ROR over-expression + Wortmannin group. MTT assay was used to determine the optimum inhibitory concentration of DDP. Quantitative real-time polymerase chain reaction and western blotting were applied to detect expressions of long non-coding RNA ROR, PI3K, Akt, and mTOR. Colony-forming assay, scratch test, Transwell assay, and flow cytometry were conducted to detect cell proliferation, migration, invasion, and apoptosis, respectively. Tumor-formation assay was performed to detect the growth of transplanted tumors. Long non-coding RNA ROR expression was high in human A549/DDP cell lines. Compared with the control and negative control groups, the mRNA and protein expressions of PI3K, Akt, mTOR, and bcl-2 decreased, whereas the mRNA and protein expression of bax and the sensitivity of cells to DDP significantly increased. Cell proliferation, migration, and invasion abilities decreased in the si-ROR and Wortmannin groups. In comparison with control and negative control groups, the mRNA and protein expressions of PI3K, Akt, mTOR, and bcl-2 increased, whereas the mRNA and protein expressions of bax decreased, the sensitivity of cells to DDP significantly increased, and cell proliferation, migration, and invasion abilities decreased in the ROR over-expression group. For nude mice in tumor-formation assay, compared with control and negative control groups, the tumor weight was found to be lighter (1.03 ± 0.15) g, the protein expressions of PI3K, Akt, mTOR, and bcl-2 decreased, and the protein expression of bax increased in the si-ROR group. Long non-coding RNA ROR may affect the sensitivity of lung adenocarcinoma cells to DDP by targeting PI3K/Akt/mTOR signaling pathway.


2018 ◽  
Vol 32 ◽  
pp. 205873841881434 ◽  
Author(s):  
Genglong Zhu ◽  
Xialei Liu ◽  
Haijing Li ◽  
Yang Yan ◽  
Xiaopeng Hong ◽  
...  

Liver cancer is one of the most common and lethal cancers in human digestive system, which kills more than half a million people every year worldwide. This study aimed to investigate the effects of kaempferol, a flavonoid compound isolated from vegetables and fruits, on hepatic cancer HepG2 cell proliferation, migration, invasion, and apoptosis, as well as microRNA-21 (miR-21) expression. Cell viability was detected using cell counting kit-8 (CCK-8) assay. Cell proliferation was measured using 5-bromo-2′-deoxyuridine (BrdU) incorporation assay. Cell apoptosis was assessed using Guava Nexin assay. Cell migration and invasion were determined using two-chamber migration (invasion) assay. Cell transfection was used to change the expression of miR-21. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) was performed to analyze the expressions of miR-21 and phosphatase and tensin homologue (PTEN). Expression of key proteins involved in proliferation, apoptosis, migration, invasion, and phosphatidylinositol 3-kinase/protein kinase 3/mammalian target of rapamycin (PI3K/AKT/mTOR) pathway were evaluated using western blotting. Results showed that kaempferol significantly inhibited HepG2 cell proliferation, migration, and invasion, and induced cell apoptosis. Kaempferol remarkably reduce the expression of miR-21 in HepG2 cells. Overexpression of miR-21 obviously reversed the effects of kaempferol on HepG2 cell proliferation, migration, invasion, and apoptosis. Moreover, miR-21 negatively regulated the expression of PTEN in HepG2 cells. Kaempferol enhanced the expression of PTEN and inactivated PI3K/AKT/mTOR signaling pathway in HepG2 cells. In conclusion, kaempferol inhibited proliferation, migration, and invasion of HepG2 cells by down-regulating miR-21 and up-regulating PTEN, as well as inactivating PI3K/AKT/mTOR signaling pathway.


2018 ◽  
Vol 42 (9) ◽  
pp. 1192-1199 ◽  
Author(s):  
Liyin Li ◽  
Yanhua Qi ◽  
Xiaobo Ma ◽  
Guosheng Xiong ◽  
Lijun Wang ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document