scholarly journals lncRNA RP11-838N2.3 Promoted Cisplatin Resistance in Lung Adenocarcinoma

2020 ◽  
Vol 2020 ◽  
pp. 1-18
Author(s):  
Jie Chen ◽  
Feng Jiang ◽  
Lijuan Hu ◽  
Fan Zhang ◽  
Junjun Wang ◽  
...  

The mechanism of RP11-838N2.3 promoting cisplatin resistance in lung adenocarcinoma (LAD) was unclear. The RP11-838N2.3 expression level in cells and LAD tissues was detected by qPCR. We constructed lentivirus-mediated GV303 overexpression and GV248 shRNA vector targeting RP11-838N2.3, then infected A549 and A549/DDP cell and furtherly analyzed cell biology. High-throughput gene chip analysis showed that RP11-838N2.3 was significantly upregulated in A549/DDP (change fold=66.056595). The qPCR results showed that the expression level of RP11-838N2.3 in A549/DDP cell was significantly higher than that in A549 cells (P<0.05), and the expression level of RP11-838N2.3 in LAD tissues was also significantly higher than that in adjacent tissues (P<0.05). The expression level of RP11-838N2.3 in cisplatin-insensitive LAD tissues was also significantly higher than that in cisplatin-sensitive LAD tissues (P<0.05). Survival analysis showed that OS (overall survival) and DFS (progression-free survival) of high RP11-838N2.3 expression in the cisplatin-sensitive or cisplatin-insensitive LAD group were lower (P<0.001 and P<0.001) than those of low RP11-838N2.3 expression in the cisplatin-sensitive or cisplatin-insensitive LAD group. CCK8 showed that the OD450 value of RP11-838N2.3 overexpression increased significantly at 24 h, 48 h, and 72 h after transfection, while the knockdown of RP11-838N2.3 caused OD450 value at 24 h, 48 h, and 72 h after transfection significantly reduced, under the action of cisplatin that had the same trend (P<0.05). The cell migration showed that the RP11-838N2.3 overexpression increased significantly migration activity and RP11-838N2.3 knockdown inhibited migration activity at 24 h, 48 h, and 72 h after transfection. The same trend was also observed under the action of cisplatin (P<0.05). The cell invasion showed that the invasion rate of RP11-838N2.3 overexpression increased significantly, while the invasion rate of RP11-838N2.3 knockdown decreased significantly, and the same trend was observed under the action of cisplatin (P<0.05). Apoptosis results showed that the apoptosis rate of RP11-838N2.3 overexpressed cells decreased significantly and the apoptosis rate of RP11-838N2.3 knockdown cells increased significantly, and the same trend was also observed under the action of cisplatin (P<0.05). However, the results of cell cycle showed that there was no significant difference in the proportion of cells in each phase of the cell cycle after RP11-838N2.3 overexpression or knockdown (P>0.05).RP11-838N2.3 was significantly upregulated in cisplatin-resistant cell and tissues of LAD. RP11-838N2.3 could enhance the proliferation, migration, and invasion and inhibit apoptosis of LAD cisplatin-resistant cell. So RP11-838N2.3 could enhance the cisplatin resistance of LAD cells and was a resistant lncRNA molecule.

2020 ◽  
Vol 38 (15_suppl) ◽  
pp. e21022-e21022
Author(s):  
Danni Liu ◽  
Tianhao Mu ◽  
Shifu Chen

e21022 Background: 2019-nCoV, a novel coronavirus (2019-nCoV), has infected tens of thousands of people in Wuhan, Hubei Province, China. This new coronavirus has led to hundreds of deaths, and especially threatened people with underlying diseases. ACE2 was reported as the 2019-nCoV putative receptor. The expression abundance of ACE2 may cause different disease progressions. Here, we evaluated the expression profile of ACE2 from 283 samples in lung adenocarcinoma . Methods: 283 samples from the public datasets (GEO: GSE31210 and GEO: GSE2109) were grouped by tissue, ethnicity and gender. The data was normalized by the RMA algorithm. Student 's t-test was used to compare the expression of ACE2 in different groups . Results: The expression level of ACE2 of primary lung tumor was significantly higher than normal lung tissue ( p < 0.0001). ACE2 was significantly differential-expressed between Asian and Caucasian lung adenocarcinoma patients ( p < 0.0001). However, no significant difference of the expression of ACE2 was observed by gender ( p = 0.33). Conclusions: ACE2 was highly expressed in primary lung adenocarcinoma , compared with normal lung tissues. The expression level of ACE2 was higher in Asian lung adenocarcinoma patients than Caucasian’s. These conclusions imply that Asian people with lung adenocarcinoma may be more vulnerable to 2019-nCoV.


2018 ◽  
Vol 50 (1) ◽  
pp. 92-107 ◽  
Author(s):  
Qi-ming Shen ◽  
Hao-you Wang ◽  
Shun Xu

Background/Aims: To investigate the clinical significance and functional mechanisms of membrane-associated RING-CH protein 9 (MARCH9) in lung adenocarcinoma (LAC). Methods: Immunohistochemistry staining was performed to explore the expression of MARCH9 in LAC tissues and adjacent normal lung tissues. Patients’ prognosis was evaluated using overall survival. The prognostic role of MARCH9 was tested with univariate and multivariate analyses. To confirm the effect of MARCH9 in cell proliferation and invasion, overexpression of MARCH9 was induced in two LAC cell lines. Cell cycle, apoptosis, migration, invasion, and immunoprecipitation experiments were performed to further explore the signaling pathways involved. Results: Analysis of a series of 143 clinical samples revealed that MARCH9 was down-regulated in tumor tissues compared with normal lung tissues, and this was closely associated with lymph node metastasis (P = 0.004). Univariate and multivariate analyses indicated that MARCH9 was an independent prognostic biomarker for LAC; low MARCH9 expression indicated poor overall survival. Cellular studies with A549 and H1299 cells demonstrated that MARCH9 can attenuate tumor migration and invasion but had little effect on cell cycle or apoptosis. Moreover, an interaction between MARCH9 and ICAM-1 protein was identified, and overexpression of MARCH9 was found to attenuate the oncogenic effect of ICAM-1, suggesting that MARCH9 may inhibit tumor progression by downregulating ICAM-1 signaling. Conclusion: MARCH9 downregulation in LAC tissues correlated with poor clinical outcomes. MARCH9 may serve as a novel biomarker and potential therapeutic target for LAC.


2019 ◽  
Vol 14 (1) ◽  
pp. 454-461 ◽  
Author(s):  
Huamin Zhang ◽  
Haowei Zhang ◽  
Sihui Wang ◽  
Zhihai Ni ◽  
Tiejun Wang

AbstractTargeting 6-phosphogluconate dehydrogenase (6PGD) can inhibit cancer cell proliferation and tumor growth. However, the relationship between 6PGD and cisplatin resistance still needs further study. Cisplatin-sensitive and cisplatin-resistant ovarian cancer OV2008 and C13* lines and lung cancer A549 and A549DDP lines were treated with different concentrations of cisplatin and cell viability was evaluated. We also compared the growth rates and the cell cycle distributions between cisplatin-sensitive and cisplatin-resistant cells. The expression level of 6PGD was detected by immunoblotting. The Chou-Talalay method was used to evaluate the effect of a combination treatment using cisplatin and the small molecule inhibitor 1-Hydroxy-8-methoxy-anthraquinon (S3) that targets 6PGD. The cisplatin-resistant ovarian and lung cancer cell lines grew faster than the cisplatin- sensitive cell lines, with more cells in S and G2 phases in cisplatin-resistant cell lines. The expression level of 6PGD in cisplatin-resistant cell lines was significantly increased compared with cisplatin-sensitive cell lines. Furthermore, treatment of cells with the S3 small molecule inhibitor of 6PGD together with cisplatin could overcome cisplatin resistance. The expression level of 6PGD in cisplatin-resistant cells lines was significantly upregulated, and the resistance to cisplatin of drug-resistant cells lines could be overcome when treated with the small molecule inhibitor S3 that specifically targets 6PGD.


2021 ◽  
Author(s):  
Yu Wang ◽  
Yunxia Zhao ◽  
Xiangwei Zhang ◽  
Yuanzhu Jiang ◽  
Wei Ma ◽  
...  

Abstract Background: We aimed at investigation of the effect and the underlying neurotrophin signaling pathway of the upstream transcription factor 1 (USF1) in lung adenocarcinoma (LUAD).Methods: The Cancer Genome Atlas (TCGA) database was used to analyze USF1 expression data and to extract patients’ clinical records. Immunohistochemical assay and Western blotting (WB) were used to determine the expression levels of USF1 in LUAD. The neurotrophin signaling pathway was analyzed by bioinformatic analysis while the expression of all related proteins was determined by WB. In addition cellular viability, proliferation, migration and invasion potential were investigated by the CCK-8, colony formation, wound healing and transwell. Meanwhile, the effect of USF1 in LUAD progression was investigated in a mouse model. The link between USF1 and UGT1A3 (UDP Glucuronosyltransferase Family 1 Member A3) was studied by the dual-luciferase reporter assay. Results: We have detected a high expression level of USF1 in LUAD, which was associated with advanced tumor stage, nodal metastasis, and poor patient’s survival rate. The knockdown of USF1 inhibited cellular viability, proliferation, migration and invasion. Meanwhile, USF1 knockdown inhibited tumor growth in a mouse model. Besides, USF1 targeted UGT1A3, which was proven by the fact that the USF1 knockdown decreased the expression level of UGT1A3, whereas the upregulated expression of UGT1A3 increased cellular viability and proliferation. We have proved that the neurotrophin signaling pathway in LUAD was activated by USF1 and UGT1A3. The expression of the related proteins was also inhibited by the USF1 knockdown, while the overexpression of IRAK increased cancer cells’ migration and invasion potential.Conclusion: USF1 was highly expressed in LUAD and promoted LUAD progression by regulating the neurotrophin signaling pathway. These findings provide a new theoretical data that could serve as a good foundation for the treatment of LUAD.


2018 ◽  
Vol 19 (10) ◽  
pp. 2986 ◽  
Author(s):  
Samana Batool ◽  
Thomson Joseph ◽  
Mushraf Hussain ◽  
Miza Vuai ◽  
Kavish. Khinsar ◽  
...  

Present study aimed to elucidate the anticancer effect and the possible molecular mechanism underlying the action of Latcripin 1 (LP1), from the mushroom Lentinula edodes strain C91-3 against gastric cancer cell lines SGC-7901 and BGC-823. Cell viability was measured by Cell Counting Kit-8 (CCK-8); morphological changes were observed by phase contrast microscope; autophagy was determined by transmission electron microscope and fluorescence microscope. Apoptosis and cell cycle were assessed by flow cytometer; wound-healing, transwell migration and invasion assays were performed to investigate the effect of LP1 on gastric cancer cell’s migration and invasion. Herein, we found that LP1 resulted in the induction of autophagy by the formation of autophagosomes and conversion of light chain 3 (LC3I into LC3II. LP1 up-regulated the expression level of autophagy-related gene (Atg7, Atg5, Atg12, Atg14) and Beclin1; increased and decreased the expression level of pro-apoptotic (Bax) and anti-apoptotic (Bcl-2) proteins respectively, along with the activation of Caspase-3. At lower-doses, LP1 have shown to arrest cells in the S phase of the cell cycle and decreased the expression level of matrix metalloproteinase MMP-2 and MMP-9. In addition, it has also been shown to regulate the phosphorylation of one of the most hampered gastric cancer pathway, that is, protein kinase B/mammalian target of rapamycin (Akt/mTOR) channel and resulted in cell death. These findings suggested LP1 as a potential natural anti-cancer agent, for exploring the gastric cancer therapies and as a contender for further in vitro and in vivo investigations.


2020 ◽  
Vol 15 (1) ◽  
pp. 848-859
Author(s):  
Wei Wei ◽  
Liefeng Ji ◽  
Wanli Duan ◽  
Jiang Zhu

AbstractCircular RNA sterile alpha motif domain containing 4A (circSAMD4A) was found to be differentially expressed in osteosarcoma and contributed to the tumorigenesis of osteosarcoma. However, the role of circSAMD4A in doxorubicin (DXR) resistance of osteosarcoma is yet to be elucidated. Levels of circSAMD4A, microRNA (miR)-218-5p and Krüppel-like factor 8 (KLF8) were detected using quantitative reverse transcription-polymerase chain reaction. Western blot was applied to detect the protein levels of KLF8, cyclin D1 and p21. Cell viability, cell cycle, migration and invasion were analyzed using Cell Counting Kit-8 assay, flow cytometry and transwell assay, respectively. The interaction between miR-218-5p and circSAMD4A or KLF8 was verified using dual-luciferase reporter assay or RNA immunoprecipitation assay. In vivo experiments were performed using murine xenograft models. CircSAMD4A and KLF8 were elevated in osteosarcoma, and knockdown of circSAMD4A or KLF8 sensitized osteosarcoma cells to DXR by mediating resistant cell viability, migration and invasion inhibition, and cell cycle arrest in vitro. miR-218-5p was decreased in osteosarcoma, and miR-218-5p inhibition enhanced DXR resistance. Besides, miR-218-5p was found to bind to circSAMD4A or KLF8, and subsequent rescue experiments indicated that miR-218-5p inhibition reversed the inhibitory effects of circSAMD4A silencing on DXR resistance, and silencing miR-218-5p enhanced DXR resistance by targeting KLF8 in osteosarcoma cells. Moreover, circSAMD4A could indirectly regulate KLF8 via miR-218-5p. Additionally, circSAMD4A knockdown enhanced the cytotoxicity of DXR in osteosarcoma in vivo via regulating miR-218-5p and KLF8. In all, circSAMD4A enhanced cell DXR resistance in osteosarcoma by regulating the miR-218-5p/KLF8 axis, suggesting a novel therapeutic target for therapy-resistant osteosarcoma.


2020 ◽  
Vol 19 ◽  
pp. 153303382095700
Author(s):  
Dong-Wei Wang ◽  
Hai-Zheng Zheng ◽  
Na Cha ◽  
Xiao-Jie Zhang ◽  
Min Zheng ◽  
...  

AHNAK nucleoprotein 2 (AHNAK2) has been emerged as a crucial protein for neuroblast differentiation and cell migration, thereby involving in the development of various cancers. However, the specific molecular mechanism of AHNAK2 in lung adenocarcinoma is inconclusive. By accessing to the Oncomine dataset and GEPIA website, a higher expression level of AHNAK2 was observed in lung adenocarcinoma tissue samples. Overall survival (OS) curve plotted by Kaplan-Meier method showed that up-regulation of AHNAK2 was related with poor prognosis of lung adenocarcinoma patients. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) analysis and western blot were conducted to examine the expression level of genes in lung adenocarcinoma cells. Through functional in vitro experiments, cell proliferation, migration and invasion were all suppressed after AHNAK2 knockdown using Cell counting kit-8 (CCK-8) assay, wound-healing and transwell analysis. Reduction of AHNAK2 decreased the apoptosis rate using flow cytometry analysis. Moreover, the key markers of MAPK pathway, p-MEK, p-ERK and p-P90RSK were decreased due to the transfection of si-AHNAK2 in A549 cells. U0126, a MEK inhibitor, showed the similar effects on MAPK-related protein levels with si-AHNAK2. To sum up, AHNAK2 is significantly increased in lung adenocarcinoma and plays a carcinogenic role by activating the MAPK signaling pathway, providing a novel insight and raising possibility for lung adenocarcinoma treatment.


2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Jiali Fu ◽  
Jingjing Pan ◽  
Xiang Yang ◽  
Yan Zhang ◽  
Fanggui Shao ◽  
...  

Abstract Aim This study aimed to explore the mechanism of LncRNA urothelial carcinoma-associated 1 (UCA1) promoting cisplatin resistance in lung adenocarcinoma (LUAD). Method The UCA1 expression level in LUAD cell lines was detected by reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR). We overexpressed UCA1 in A549 cells and downregulated UCA1 in A549/DDP cells by the lentivirus‑mediated technique. Subsequently, in vitro, and in vivo functional experiments were performed to investigate the functional roles of UCA1 in the growth and metastasis of LUAD cell lines. Furthermore, RNA pulldown, mass spectrometry, and RNA immunoprecipitation technique were performed to analyze various downstream target factors regulated by UCA1. Results The results revealed a higher UCA1 expression level in A549/DDP cells and LUAD tissues than in A549 cells and adjacent cancer tissues. UCA1 expression was significantly associated with distant metastasis, clinical stage, and survival time of patients with LUAD. UCA1 overexpression significantly increased the proliferation, invasion, clone formation, and cisplatin resistance ability and enhanced the expression levels of proliferating cell nuclear antigen and excision repair cross-complementing gene 1 in A549 cells. However, these trends were mostly reversed after the knockdown of UCA1 in A549/DDP cells. Tumorigenic assays in nude mice showed that UCA1 knockdown significantly inhibited tumor growth and reduced cisplatin resistance. Enolase 1 was the RNA-binding protein (RBP) of UCA1. Conclusion Based on the results, we concluded that UCA1 promoted LUAD progression and cisplatin resistance and hence could be a potential diagnostic marker and therapeutic target in patients with LUAD.


2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Ru-nan Zhang ◽  
Dong-mei Wu ◽  
Li-ping Wu ◽  
Guo-wei Gao

Abstract Background Emerging studies have shown that long noncoding RNAs (lncRNAs) predominantly function in the carcinogenesis of multiple developing human tumors. The current study aimed to investigate the underlying mechanisms of LINC00337 in lung adenocarcinoma. Methods We analyzed TCGA and GTEx datasets and chose LINC00337 as the research object. Cell proliferation, cell apoptosis, cell cycle, migration, and invasion were detected in the gain and loss experiments of LINC00337 both in vitro and in vivo. Moreover, RNA pull-down, luciferase reporter assays, western blotting analysis, and rescue experiments were performed to investigate the underlying molecular mechanisms of LINC00337 function. Results LINC00337 expression was remarkably upregulated in lung adenocarcinoma. In addition, LINC00337 knockdown was shown to repress cell migration, invasion, and proliferation, as well as the cell cycle, and gear up apoptosis in lung adenocarcinoma in vitro and in vivo. With respect to the mechanism, LINC00337 knockdown boosted miR-1285-3p expression and then restrained YTHDF1 expression post-transcriptionally. Crucially, both miR-1285-3p decrement and YTHDF1 overexpression successfully reversed the influence on cell proliferation, migration, invasion, and apoptosis caused by LINC00337 shRNA. Conclusions These results suggest that LINC00337 acts as an oncogenic lncRNA, targeting miR-1285-3p and regulating YTHDF1 expression, to promote the progression of lung adenocarcinoma.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4717-4717
Author(s):  
Zonghong Shao ◽  
Xifeng Dong ◽  
Rong Fu

Objective To compare the response of GPI-AP negative or positive bone marrow mononuelear cells(BMMNCs) from PNH patients to DAG in vitro and explore the related mechanism. Methods Seventeen PNH patients as well as fourteen normal controls were enrolled. CD59-/CD59+ cells were sorted by magnetic activated cell-sorting system. Then the cells were incubated in IMDM medium containing several hemapoitic growth factors with DAG or G-CSF for 48h in vitro. The cell cycle kinetics and apoptosis of these cells were detected by flow cytometry(FCM). The expressions of CD114 on CD34+CD59- and CD34+CD59+ bone marrow cells(BMC) after incubated with G-CSF were measured by FCM. And another 14 PNH peripheral blood samples were obtained, the expression of CD44/CD49d on CD59- and CD59+ cells were analyzed by FCM respectively. The mRNA of CD114 and CD44/CD49d was also tested in 22 PNH patients vs 14 controls and CD59- vs CD59+ cells from 14 PNH patients by Q-PCR. Results After incubated with DAG for 48h in vitro, the death rate and apoptosis rate for GPI-AP negative and positive cells(CD59-/CD59+ BMMNCs cells): for CD59- BMMNCs, compared with control group, the death rate of DAG group increased (27.29±22.04% vs 19.10±20.93%), apoptosis rate also increased(10.55±12.34% vs 7.2±6.76%), there was no significant difference for them; for CD59+ BMMNCs, compared with control, the death rate of cells from DAG group increased significantly (31.89±26.75% vs 12.83±18.92%)(P<0.05), whereas there was no significant difference for the apoptosis rate (9.66±7.96% vs 6.31±1.32%); for the CD59- and CD59+ BMMNCs, the death rate was significant higher than apoptosis rate respectively(P<0.05). For cell cycle kinetics, there was no significant difference between the two kinds of BMMNCs. As to the percentage of CD114, compared with control group, it increased significantly in CD34+CD59+ BMMNCs from G-CSF group (48.12±41.20% vs 12.84±15.32%) (P<0.05), whereas there was no significant difference for CD34+CD59- BMMNCs (41.76±44.62% vs 26.79±41.62%). And the variation of CD114 for CD34+CD59+ BMMNCs was higher than that for CD34+CD59- BMMNCs(33.97±36.03% vs 14.88±27.02%)(P<0.05). The expression of CD44/CD49d protein: the expression of CD44 for CD59+was higher than that for CD59- cells(97.66±4.21% vs 93.46±9.52%, P<0.05); and there was no significant difference for CD49d expression in CD59- and CD59+ cells(38.46±27.37% vs 43.79±24.77%). The mRNA expressions of CD114, CD44 and CD49d in 22 PNH patients compared with 15 control and CD59- cells compared with CD59+ cells from 14 PNH patients : for CD114, its mRNA expression was higer for CD59+ cells compared with that for CD59- cells(2.78±2.52 vs 1.69±2.34, P<0.05), but there was no significant difference for CD114 in PNH patients and controls; for CD44, the significant difference exited for PNH patients compared with controls and CD59- cells compared with CD59+ cells(1.73±2.20 vs 3.80±3.87, P<0.05; 0.82±0.75 vs 2.38±2.42, P<0.05); for CD49d, no significant difference exited for PNH patients compared with controls and CD59- cells compared with CD59+ cells(2.83±2.62 vs 2.56±3.04; 1.74±2.60 vs 1.94±3.02). Conclusions In vitro, effect of DAG was similar on CD59- and CD59+ BMMNCs, the style of death was necrosis not apotosis and the cell cycle was not influenced by DAG. The variation of CD114 for CD34+CD59- after G-CSF stimulation was less than that in CD34+CD59+ cells, and mRNA of CD114 was lower in CD59- cells compared with CD59+ cells, which may indicating the mechanism for the remission of PNH patients after DAG chemotherapy. The protein and mRNA of CD44 was lower in PNH patients and CD59- cells compared with control and CD59+ cells respectively, which may explain the inferior growth of PNH cells, because they can not fully use the BM microenvironment. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document