Open Access

lncRNA‑ATB promotes stemness maintenance in colorectal cancer by regulating transcriptional activity of the β‑catenin pathway

  • Authors:
    • Xiaojin Yang
    • Hanchuan Tao
    • Cheng Wang
    • Weijun Chen
    • Fu Hua
    • Haixin Qian
  • View Affiliations

  • Published online on: February 26, 2020     https://doi.org/10.3892/etm.2020.8558
  • Pages: 3097-3103
  • Copyright: © Yang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Long non‑coding RNA activated by transforming growth factor‑β (ATB) was recently reported to be involved in a wide range of physiological and pathological processes. However, the role of ATB in colorectal cancer (CRC) stemness remains unclear. In the present study, the functional role of ATB in maintaining stemness of CRC was determined using colony formation and sphere formation assays, and xenograft models. Reverse transcription‑quantitative PCR, western blotting and immunohistochemistry were performed to investigate the mechanisms underlying the effects of ATB. Knockdown of ATB impaired colony formation and sphere formation in CRC cells, accompanied by an inhibition of colon tumor growth. Further results suggested that ATB regulated the transcriptional activity of the β‑catenin pathway by inhibiting β‑catenin expression. In addition, the results confirmed the role of β‑catenin in ATB‑mediated regulation of stemness in CRC. Collectively, the results indicated that ATB is a promising therapeutic target for CRC.

Introduction

Colorectal cancer (CRC) is a common clinical cancer that is associated with high morbidity and mortality (1). Gene mutations, inflammatory condition and imbalances in the microbiome are associated with increased risk of CRC development and progression (2-4). Despite advances in radiotherapy, chemotherapy and surgery, severe side effects compromise the CRC patient health (5,6). Thus, there is an urgent interest and demand for identifying novel therapeutic targets and prognosis predictors for CRC.

Long non-coding RNAs (lncRNAs) comprise a class of transcripts that are >200 nucleotides in length but without any protein-coding ability. Previous studies indicated that lncRNAs are involved in various physiological and pathological processes (7,8). lncRNAs exert their biological functions by binding to DNA, RNA and proteins, thereby regulating protein expression at the transcriptional or post-transcriptional level (9,10). Numerous lncRNAs are dysregulated in different types of cancer and are correlated with tumor cell proliferation, apoptosis, migration and stem-like properties (11-13). lncRNA activated by transforming growth factor-β (ATB) has been identified as a novel transforming growth factor-β (TGF-β)-induced lncRNA that stimulates cancer cell proliferation, metastasis, epithelial-mesenchymal transition (EMT) and angiogenesis in lung, bladder, breast and gastric cancer (14-17). In CRC, ATB promoted cancer cell proliferation and was found to be a predictor of poor prognosis in patients (18,19). However, the regulatory role of ATB in the maintenance of CRC cell stemness remains unclear.

The present study aimed to determine the functional role of ATB in maintaining the stemness of CRC cells. Colony formation assays, sphere formation assays and xenograft models were used to determine the functional role of ATB in maintaining CRC stemness. The mechanisms underlying the effects of ATB were additionally investigated by reverse transcription-quantitative PCR (RT-qPCR), western blotting, and immunohistochemical staining. The present study provided evidence for understanding ATB and identifying a potential therapeutic target for CRC.

Materials and methods

Cell culture and treatment

The human colon cancer cell lines HCT116 and HT29 were purchased from the American Type Culture Collection and cultured in DMEM containing 10% FBS (Gibco; Thermo Fisher Scientific, Inc.). Lentivirus-based short hairpin RNAs (shRNAs) targeting ATB (shATB-1 and shATB-2, two different custom shRNA sequences targeting ATB) and shRNA-negative control (shNC) were obtained from Shanghai GenePharma Co., Ltd. HCT116 and HT29 cells were seeded into 6 well plates (2x105 cells per well). After culture for 24 h, 4x106 lentiviral particles were added into the well and puromycin (2 mg/ml) was added into the medium at 24 h post infection. The stably infected cells were collected for further experiments. A specific activator of the β-catenin pathway, CP21R7 (CP21; cat. no. S7954), was purchased from Selleck Chemicals and used to treat HCT116 and HT29 cells for 48 h at 37˚C with a concentration of 2 mM. The equivalent volume of DMSO was added as a control.

Colony formation assay

A total of 2x103 HCT116 or HT29 cells were seeded into each well of a six-well plate containing 2 ml DMEM supplemented with 10% FBS. A total of 14 days later, plates were collected and washed with PBS three times at room temperature. Next, 4% paraformaldehyde was added to fix the cells for 15 min at room temperature. After washing cells with PBS three times at room temperature, the cells were stained with 2% crystal violet (Beyotime Institute of Biotechnology) for 15 min at room temperature. The colonies in each well were counted under a light microscope (Nikon Corporation) by two experimenters and analyzed. Three independent experiments were performed in each group.

Sphere formation assay

A total of 2x104 HCT116 or HT29 cells were seeded into each well of a six-well plate containing 2 ml serum-free DMEM. Five days later, the images of spheres were captured using an inverted microscope (x40; Nikon Corporation). The number of spheres in each well was counted after dilution for 100 times with the inverted microscope (x40; Nikon Corporation) by two experimenters and analyzed. Three independent experiments were performed in each group.

RNA extraction and RT-qPCR

Total RNA was extracted using TRIzol™ reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. Reverse transcription (RT) was performed on isolated total RNA using a PrimeScript One Step RT-PCR kit (cat. no. RR047A; Takara Bio, Inc.), and qPCR was performed using a SYBR green Real Time PCR kit (Cat. No. RR085A; Takara Bio, Inc.), according to the manufacturer's protocol. The following thermocycling conditions were used for the qPCR: Initial denaturation at 95˚C for 2 min; 40 cycles of 94˚C for 15 sec, 58˚C for 30 sec and 70˚C for 30 sec; extension at 72˚C for 5 min. GAPDH was used as an internal control. The primers were supplied as follow: ATB-F, 5'-TCTGGCTGAGGCTGGTTGAC-3'; ATB-R, 5'-ATCTCTGGGTGCTGGTGAAGG-3'; GAPDH-F, 5'-TCAAGGCTGAGAACGGGAAG-3'; GAPDH-R, 5'-TCGCCCCACTTGATTTTGGA-3'. The relative gene expression was calculated using the 2-ΔΔCq method (20).

Western blotting

Total protein from HCT116 and HT29 cells was extracted using RIPA buffer (Beyotime Institute of Biotechnology) containing 1% protease inhibitor cocktail (EMD Millipore). The protein concentration was determined by BCA kit (Beyotime Institute of Biotechnology). Subsequently, 20 µg of total protein was loaded in each lane of an SDS-PAGE (10% for β-catenin, LEF1 and GSK-3β or 12% for GAPDH). Following electrophoresis, proteins were transferred onto PVDF membranes (EMD Millipore). Membranes were blocked with 5% milk in TBS-Tween-20 buffer for 2 h at room temperature. The membranes were then incubated with primary antibodies supplied by Cell Signaling Technology, Inc. against β-catenin (1:1,000; cat. no. 8480), lymphoid enhancer-binding factor 1 (LEF1; 1:80; cat. no. 76010) and glycogen synthase kinase-3β (GSK-3β; 1:1,000; cat. no. 12456) overnight at 4˚C. Following incubation with HRP conjugated secondary antibody (1:10,000; cat. nos. ZDR 5306 and ZDR 5307; Origene Technologies, Inc.) at 22-25˚C for 2 h, an enhanced chemiluminescence kit (EMD Millipore) was used to detect specific bands. The density of each band was analyzed using ImageJ software (version: 1.52, National Institutes of Health).

Animal studies

Animal studies were performed in accordance with the institutional guidelines of Soochow University. Female BALB/c nude mice (age, 6 weeks; weight, 18-20 g; 5 mice in each group; humidity 40-60%) were purchased from the Model Animal Research Center of Nanjing University and allowed to acclimate for 1 week before use. All of the mice were housed at 25˚C under a 12 h light/dark cycle with ad libitum access to food and water. For xenograft models, 5x106 HT29 cells were injected into the right flank via the subcutaneous vein of mice. At 10 days post-injection, the tumor length and width was measured using a vernier caliper every 5 days. The tumor volume was calculated as tumor length x tumor width2 x0.52(21). At 25 days post-injection, the mice were sacrificed and tumors were collected, weighed and used for histopathological studies.

Immunohistochemical staining and RNA-fluorescence in situ hybridization

The tumor tissues were fixed with 4% paraformaldehyde at 22-25˚C for 48 h and embedded in paraffin. After heating at 65˚C for 2 h, the paraffin-embedded sections (5 µm) of colon tumor tissues were dewaxed with xylene and gradient alcohol and then subjected to antigen retrieval with citrate buffer (OriGene Technologies, Inc.). Following blocking with 5% normal goat serum (Origene, Technologies, Inc.) at 37˚C for 15 min, the sections were incubated with primary antibody against β-catenin (1:200; cat. no. 8480, Cell Signaling Technology, Inc.) at 4˚C overnight. After immunoperoxidase staining with a Streptavidin-Peroxidase kit (OriGene Technologies, Inc.), 3,3'-diamino-benzidine (Fuzhou Maixin Biotechnology Development Co., Ltd.) was used to detect the target protein. The cell nucleus was stained with hematoxylin at 22-25˚C for 3 min (Beyotime Institute of Biotechnology). For RNA-fluorescence in situ hybridization, a custom probe for the specific detection of ATB was synthesized by Guangzhou Ribobio Co., Ltd. The process of sample preparation and hybridization was followed by the use of a fluorescence in situ hybridization kit (cat. no. C10910; Guangzhou Ribobio Co., Ltd). Images of sections were captured by light microscope (x100; model BX51; Olympus Corporation). The β-catenin positive cells and total cells in each image were counted and the percent of positive cells were analyzed.

Statistical analysis

GraphPad Prism version 5.0 software (GraphPad Software, Inc.) was used for statistical analysis. One-way ANOVA followed with Dunnett's test was used to determine statistical significance for more than two groups. All data are presented as the mean ± SEM. P<0.05 indicated a statistically significant difference.

Results

ATB knockdown impairs CRC stemness maintenance in vitro

To investigate the potential role of ATB in CRC, lentivirus-based shRNAs targeting ATB were used to infect HCT116 and HT29 cells. Following puromycin selection, stably-infected cells were collected for determination of ATB expression by RT-qPCR. The results indicated that shATB significantly downregulated ATB expression in both HCT116 (Fig. 1A) and HT29 (Fig. 1B) cells compared with shNC. The stably infected cells were then analyzed using a colony formation assay. Colony formation ability was significantly reduced in both HCT116 (Fig. 1C) and HT29 (Fig. 1D) cells after ATB knockdown. Furthermore, results of the sphere formation assay indicated that ATB knockdown significantly impaired sphere formation in HCT116 (Fig. 1E) and HT29 (Fig. 1F) cells. Collectively, the above results demonstrated the positive role of ATB in CRC stemness maintenance.

ATB promotes colon tumor growth in vivo

Based on the in vitro study results, an in vivo study was conducted to determine the functional role of ATB in colon tumor growth. HT29 cells that were stably transfected with shNC or shATB were injected into the right subcutaneous vein to establish a xenograft model. HT29 tumor tissues were collected for determination of ATB expression by RNA-fluorescence in situ hybridization. A small number of ATB-positive cells was observed in the colon tumor tissues after ATB knockdown (Fig. 2A). Further statistical analysis confirmed significant downregulation of ATB expression in shATB-infected HT29 tumors (Fig. 2A). The results suggested that ATB knockdown in HT29 cells significantly inhibited tumor growth (Fig. 2B), as evidenced by a mean reduction of 76% in tumor volume (shATB-1 and sh-ATB-2 vs. shNC; Fig. 2C) and 68% decrease in tumor weight (shATB-1 and sh-ATB-2 vs. shNC; Fig. 2D). The above results demonstrated the oncogenic role of ATB in CRC.

ATB inhibits the expression and activation of β-catenin

To determine the mechanisms underlying stemness maintenance in CRC by ATB, stably-transfected HCT116 and HT29 cells were collected for further molecular determination. Expression levels of the downstream targets of the Wnt signaling pathway were measured, and the results indicated that ATB knockdown significantly inhibited the expression of G1/S-specific cyclin-D1, transcription factor-1, c-Myc, matrix metalloproteinase 7, and twist-related protein (Twist) both in HCT116 and HT29 cells compared with shNC (Fig. 3A). Western blotting was performed to determine the expression levels of β-catenin, LEF1, and GSK-3β in CRC cells. Significant downregulation of β-catenin expression was observed in both ATB-knockdown HCT116 and HT29 cells compared with shNC (Fig. 3B). However, ATB knockdown did not significantly affect the expression of LEF1 and GSK-3β in CRC cells (Fig. 3B). Reduced levels of β-catenin-positive cells were found in shATB-infected HT29 tumor tissues compared with shNC-infected HT29 tumor tissues (Fig. 3C). Taken together, the results demonstrated the regulatory role of ATB on β-catenin expression in CRC.

β-catenin plays a crucial role in ATB-mediated stemness maintenance in CRC

To determine whether β-catenin was important for regulating stemness in CRC by ATB, HT29 cells were treated with CP21, a specific activator of the β-catenin pathway. ATB knockdown significantly inhibited the colony formation ability of HT29 cells (Fig. 4A and B). However, CP21 treatment inhibited the decrease of colony formation mediated by shATB in HT29 cells (Fig. 4A and B). Collectively, the results confirmed that β-catenin was required for ATB-mediated maintenance of stemness in CRC.

Discussion

Considering their extensive biological functions, lncRNAs are involved in a wide range of physiological and pathological processes (22). In the present study, ATB knockdown impaired colony and sphere formation of CRC cells and significantly inhibited colon tumor growth. Further results suggested that ATB regulated the transcriptional activity of the β-catenin pathway by inhibiting β-catenin expression. In addition, the results confirmed that β-catenin was necessary for ATB-mediated regulation of stemness in CRC.

ATB is dysregulated in various types of cancer, including CRC (19), lung cancer (23), and liver cancer (24). ATB acts as an oncogene, and its dysregulation is commonly associated with poor prognosis of patients (25). In CRC, ATB is upregulated in colon cancer tissues and correlated with clinical cancer stage (19). In addition, ATB is highly expressed in lung cancer tissues and associated with tumor size and metastasis (23). A functional study indicated that ATB promoted proliferation, migration and invasion in osteosarcoma cells (26). Knockdown of ATB in endometrial cancer cells was found to impair cell viability by inducing caspase-3-related tumor apoptosis and G1/S arrest (27). In addition, ATB was responsible for the proliferation and apoptosis of CRC cells (18). The results demonstrated that ATB knockdown impaired stemness maintenance of CRC cells, indicated by the inhibition of colony formation and sphere formation, and colon tumor growth inhibition. The findings expand the current understanding of ATB function.

Recent studies demonstrated that ATB acts as a competing RNA by binding with microRNA (miR)-494 in lung cancer cells (28), miR-141-3p in gastric cancer cells (29), miR-590-5p in melanoma (30), and miR-200 family members in diverse types of cancer (18,26,31,32). ATB regulates transcriptional coactivator YAP1 (YAP1), NF-κB, p38 mitogen-activated protein kinase (MAPK), Twist, and TGF-β in lung cancer, glioma, breast cancer and malignant melanoma (23,30,33,34). In hepatic fibrosis, ATB knockdown downregulated β-catenin expression by upregulating the expression of endogenous miR-200a (35). In the present study, the results suggested that ATB regulated the transcriptional activity of the β-catenin pathway by inhibiting β-catenin expression. Further results confirmed the role of β-catenin during ATB-mediated regulation of stemness in CRC. miR-200a, which is involved in regulating CRC cell proliferation and β-catenin expression (36,37), could serve as the direct target of ATB in regulating stemness maintenance. However, further studies are required to verify the above results.

The present study demonstrated the role of ATB in maintaining stemness of CRC by regulating β-catenin expression. However, further studies are required to investigate the direct target of ATB for regulating β-catenin expression in CRC. Collectively, the results indicated that ATB is a promising therapeutic target for CRC.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

XY and HT were involved in the acquisition of the data and drafting the manuscript. CW, WC and FH were involved in the analysis and interpretation of the data. HQ was involved in the conception and design of the present study. All authors read and approved the final manuscript.

Ethics approval and consent to participate

This animal study was approved by the Ethics Committee of Soochow University and complied with their guidelines.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018.PubMed/NCBI View Article : Google Scholar

2 

Dai L, Liu Y, Cheng L, Wang H, Lin Y, Shi G, Dong Z, Li J, Fan P, Wang Q, et al: SARI attenuates colon inflammation by promoting STAT1 degradation in intestinal epithelial cells. Mucosal Immunol. 12:1130–1140. 2019.PubMed/NCBI View Article : Google Scholar

3 

Brenner H, Kloor M and Pox CP: Colorectal cancer. Lancet. 383:1490–1502. 2014.PubMed/NCBI View Article : Google Scholar

4 

Humphries A and Wright NA: Colonic crypt organization and tumorigenesis. Nat Rev Cancer. 8:415–424. 2008.PubMed/NCBI View Article : Google Scholar

5 

Brungs D, Aghmesheh M, de Souza P, Carolan M, Clingan P, Rose J and Ranson M: Safety and efficacy of oxaliplatin doublet adjuvant chemotherapy in elderly patients with stage III colon cancer. Clin Colorectal Cancer. 17:e549–e555. 2018.PubMed/NCBI View Article : Google Scholar

6 

Li J, Li XL, Yuan Y and Zhang SZ: Disputes and exploration of neoadjuvant and adjuvant therapy for colon cancer. Zhonghua Wei Chang Wai Ke Za Zhi. 22:329–335. 2019.(In Chinese). PubMed/NCBI View Article : Google Scholar

7 

Zheng J, Mao Y, Dong P, Huang Z and Yu F: Long noncoding RNA HOTTIP mediates SRF expression through sponging miR-150 in hepatic stellate cells. J Cell Mol Med. 23:1572–1580. 2019.PubMed/NCBI View Article : Google Scholar

8 

Wu S, Bono J and Tao YX: Long noncoding RNA (lncRNA): A target in neuropathic pain. Expert Opin Ther Targets. 23:15–20. 2019.PubMed/NCBI View Article : Google Scholar

9 

Liu H, Li C, Yang J, Sun Y, Zhang S, Yang J, Yang L, Wang Y and Jiao B: Long noncoding RNA CASC9/miR-519d/STAT3 positive feedback loop facilitate the glioma tumourigenesis. J Cell Mol Med. 22:6338–6344. 2018.PubMed/NCBI View Article : Google Scholar

10 

Yuan SX, Zhang J, Xu QG, Yang Y and Zhou WP: Long noncoding RNA, the methylation of genomic elements and their emerging crosstalk in hepatocellular carcinoma. Cancer Lett. 379:239–244. 2016.PubMed/NCBI View Article : Google Scholar

11 

Dai L, Li J, Dong Z, Liu Y, Chen Y, Chen N, Cheng L, Fang C, Wang H, Ji Y, et al: Temporal expression and functional analysis of long non-coding RNAs in colorectal cancer initiation. J Cell Mol Med. 23:4127–4138. 2019.PubMed/NCBI View Article : Google Scholar

12 

Wu Q, Guo L, Jiang F, Li L, Li Z and Chen F: Analysis of the miRNA-mRNA-lncRNA networks in ER+ and ER-breast cancer cell lines. J Cell Mol Med. 19:2874–2887. 2015.PubMed/NCBI View Article : Google Scholar

13 

Lorenzi L, Avila Cobos F, Decock A, Everaert C, Helsmoortel H, Lefever S, Verboom K, Volders PJ, Speleman F, Vandesompele J and Mestdagh P: Long noncoding RNA expression profiling in cancer: Challenges and opportunities. Genes Chromosomes Cancer. 58:191–199. 2019.PubMed/NCBI View Article : Google Scholar

14 

Chen Y, Wei G, Xia H, Tang Q and Bi F: Long noncoding RNA-ATB promotes cell proliferation, migration and invasion in gastric cancer. Mol Med Rep. 17:1940–1946. 2018.PubMed/NCBI View Article : Google Scholar

15 

Xu S, Yi XM, Tang CP, Ge JP, Zhang ZY and Zhou WQ: Long non-coding RNA ATB promotes growth and epithelial-mesenchymal transition and predicts poor prognosis in human prostate carcinoma. Oncol Rep. 36:10–22. 2016.PubMed/NCBI View Article : Google Scholar

16 

Zhai X and Xu W: Long noncoding RNA ATB promotes proliferation, migration, and invasion in bladder cancer by suppressing MicroRNA-126. Oncol Res. 26:1063–1072. 2018.PubMed/NCBI View Article : Google Scholar

17 

Zhang Y, Li J, Jia S, Wang Y, Kang Y and Zhang W: Down-regulation of lncRNA-ATB inhibits epithelial-mesenchymal transition of breast cancer cells by increasing miR-141-3p expression. Biochem Cell Biol. 97:193–200. 2019.PubMed/NCBI View Article : Google Scholar

18 

Gao Z, Zhou H, Wang Y, Chen J and Ou Y: Regulatory effects of lncRNA ATB targeting miR-200c on proliferation and apoptosis of colorectal cancer cells. J Cell Biochem. 121:332–343. 2020.PubMed/NCBI View Article : Google Scholar

19 

Yue B, Qiu S, Zhao S, Liu C, Zhang D, Yu F, Peng Z and Yan D: LncRNA-ATB mediated E-cadherin repression promotes the progression of colon cancer and predicts poor prognosis. J Gastroenterol Hepatol. 31:595–603. 2016.PubMed/NCBI View Article : Google Scholar

20 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001.PubMed/NCBI View Article : Google Scholar

21 

Cheng L, Yang Q, Li C, Dai L, Yang Y, Wang Q, Ding Y, Zhang J, Liu L, Zhang S, et al: DDA1, a novel oncogene, promotes lung cancer progression through regulation of cell cycle. J Cell Mol Med. 21:1532–1544. 2017.PubMed/NCBI View Article : Google Scholar

22 

Li CH and Chen Y: Insight into the role of long noncoding RNA in cancer development and progression. Int Rev Cell Mol Biol. 326:33–65. 2016.PubMed/NCBI View Article : Google Scholar

23 

Wei L, Wu T, He P, Zhang JL and Wu W: LncRNA ATB promotes the proliferation and metastasis of lung cancer via activation of the p38 signaling pathway. Oncol Lett. 16:3907–3912. 2018.PubMed/NCBI View Article : Google Scholar

24 

Lee YR, Kim G, Tak WY, Jang SY, Kweon YO, Park JG, Lee HW, Han YS, Chun JM, Park SY and Hur K: Circulating exosomal noncoding RNAs as prognostic biomarkers in human hepatocellular carcinoma. Int J Cancer. 144:1444–1452. 2019.PubMed/NCBI View Article : Google Scholar

25 

Li J, Li Z, Zheng W, Li X, Wang Z, Cui Y and Jiang X: LncRNA-ATB: An indispensable cancer-related long noncoding RNA. Cell Prolif. 50:2017.doi: 10.1111/cpr.12381. PubMed/NCBI View Article : Google Scholar

26 

Han F, Wang C, Wang Y and Zhang L: Long noncoding RNA ATB promotes osteosarcoma cell proliferation, migration and invasion by suppressing miR-200s. Am J Cancer Res. 7:770–783. 2017.PubMed/NCBI

27 

Zheng X, Liu M, Song Y and Feng C: Long noncoding RNA-ATB impairs the function of tumor suppressor miR-126-mediated signals in endometrial cancer for tumor growth and metastasis. Cancer Biother Radiopharm. 34:47–55. 2019.PubMed/NCBI View Article : Google Scholar

28 

Cao Y, Luo X, Ding X, Cui S and Guo C: LncRNA ATB promotes proliferation and metastasis in A549 cells by down-regulation of microRNA-494. J Cell Biochem. 119:6935–6942. 2018.PubMed/NCBI View Article : Google Scholar

29 

Lei K, Liang X, Gao Y, Xu B, Xu Y, Li Y, Tao Y, Shi W and Liu J: Lnc-ATB contributes to gastric cancer growth through a MiR-141-3p/TGFβ2 feedback loop. Biochem Biophys Res Commun. 484:514–521. 2017.PubMed/NCBI View Article : Google Scholar

30 

Mou K, Liu B, Ding M, Mu X, Han D, Zhou Y and Wang LJ: lncRNA-ATB functions as a competing endogenous RNA to promote YAP1 by sponging miR-590-5p in malignant melanoma. Int J Oncol. 53:1094–1104. 2018.PubMed/NCBI View Article : Google Scholar

31 

Li Z, Wu X, Gu L, Shen Q, Luo W, Deng C, Zhou Q, Chen X, Li Y, Lim Z, et al: Long non-coding RNA ATB promotes malignancy of esophageal squamous cell carcinoma by regulating miR-200b/Kindlin-2 axis. Cell Death Dis. 8(e2888)2017.PubMed/NCBI View Article : Google Scholar

32 

He T, Zhou H, Li C, Chen Y, Chen X, Li C, Mao J, Lyu J and Meng QH: Methylglyoxal suppresses human colon cancer cell lines and tumor growth in a mouse model by impairing glycolytic metabolism of cancer cells associated with down-regulation of c-Myc expression. Cancer Biol Ther. 17:955–965. 2016.PubMed/NCBI View Article : Google Scholar

33 

Zhou Y, Zheng X, Lu J, Chen W, Li X and Zhao L: Ginsenoside 20(S)-Rg3 inhibits the warburg effect via modulating DNMT3A/MiR-532-3p/HK2 pathway in ovarian cancer cells. Cell Physiol Biochem. 45:2548–2559. 2018.PubMed/NCBI View Article : Google Scholar

34 

Tang F, Wang H, Chen E, Bian E, Xu Y, Ji X, Yang Z, Hua X, Zhang Y and Zhao B: LncRNA-ATB promotes TGF-β-induced glioma cells invasion through NF-κB and P38/MAPK pathway. J Cell Physiol. 234:23302–23314. 2019.PubMed/NCBI View Article : Google Scholar

35 

Fu N, Zhao SX, Kong LB, Du JH, Ren WG, Han F, Zhang QS, Li WC, Cui P, Wang RQ, et al: LncRNA-ATB/microRNA-200a/β-catenin regulatory axis involved in the progression of HCV-related hepatic fibrosis. Gene. 618:1–7. 2017.PubMed/NCBI View Article : Google Scholar

36 

Santasusagna S, Moreno I, Navarro A, Martinez Rodenas F, Hernández R, Castellano JJ, Muñoz C and Monzo M: Prognostic impact of miR-200 family members in plasma and exosomes from tumor-draining versus peripheral veins of colon cancer patients. Oncology. 95:309–318. 2018.PubMed/NCBI View Article : Google Scholar

37 

Yang W, Ning N and Jin X: The lncRNA H19 promotes cell proliferation by competitively binding to miR-200a and derepressing β-catenin expression in colorectal cancer. Biomed Res Int. 2017(2767484)2017.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

April-2020
Volume 19 Issue 4

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yang X, Tao , Wang C, Chen W, Hua F and Qian H: lncRNA‑ATB promotes stemness maintenance in colorectal cancer by regulating transcriptional activity of the β‑catenin pathway. Exp Ther Med 19: 3097-3103, 2020
APA
Yang, X., Tao, ., Wang, C., Chen, W., Hua, F., & Qian, H. (2020). lncRNA‑ATB promotes stemness maintenance in colorectal cancer by regulating transcriptional activity of the β‑catenin pathway. Experimental and Therapeutic Medicine, 19, 3097-3103. https://doi.org/10.3892/etm.2020.8558
MLA
Yang, X., Tao, ., Wang, C., Chen, W., Hua, F., Qian, H."lncRNA‑ATB promotes stemness maintenance in colorectal cancer by regulating transcriptional activity of the β‑catenin pathway". Experimental and Therapeutic Medicine 19.4 (2020): 3097-3103.
Chicago
Yang, X., Tao, ., Wang, C., Chen, W., Hua, F., Qian, H."lncRNA‑ATB promotes stemness maintenance in colorectal cancer by regulating transcriptional activity of the β‑catenin pathway". Experimental and Therapeutic Medicine 19, no. 4 (2020): 3097-3103. https://doi.org/10.3892/etm.2020.8558