LGR5 promotes the proliferation of colorectal cancer cells via the Wnt/β-catenin signaling pathway

  • Authors:
    • Yu Lin
    • Tingyu Wu
    • Qianqian Yao
    • Shuming Zi
    • Long Cui
    • Ming Yang
    • Jinming Li
  • View Affiliations

  • Published online on: April 23, 2015     https://doi.org/10.3892/ol.2015.3144
  • Pages: 2859-2863
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Leucine-rich repeat-containing G-protein coupled receptor 5 (LGR5) is an established cancer stem cell marker and is a target gene of the Wnt/β‑catenin signaling pathway, a critical pathway in the process of tumor initiation and growth. In the present study, the mRNA expression levels of LGR5, adenomatous polyposis coli (APC) and β‑catenin were detected in 20 colorectal cancer (CRC) tissues and matched healthy mucosa samples using reverse transcription‑quantitative polymerase chain reaction. HT-29 CRC cell line was treated with siRNA-Lgr5; the APC, β-catenin and LGR5 RNA expressions were detected and cell viability was measured using a CCK8 assay. The results revealed that LGR5 was significantly overexpressed in CRC tissue compared with healthy mucosa (P<0.05). Furthermore, knockdown of LGR5 by small interfering RNA decreased the expression of APC and β‑catenin in HT29 colon cancer cells as well as inhibited the proliferation of HT29 cells. These findings demonstrated that LGR5 expression is critical for the promotion of neoplastic CRC cell proliferation, indicating that LGR5 may be a novel therapeutic target for CRC.

Introduction

Colorectal cancer (CRC) is one of the most common causes of cancer-associated mortality, accounting for >600,000 mortalities per year, worldwide (1). Tumor recurrence and metastasis to distant organs are the predominant contributing factors to the high mortality and poor survival rates associated with this disease (2). Recently, a number of studies have indicated that only a subpopulation of tumor cells, termed cancer stem cells (CSC), are capable of regenerating the tumor (35). In addition, CSCs may be involved in therapeutic resistance, tumor relapse and metastasis (3). Thus, the emergence of the CSC theory may have significant implications in cancer therapy. CSCs are considered to originate from mutant wild-type stem cells (6). In 2007, Barker et al (7) identified that leucine-rich repeat-containing G protein-coupled receptor 5 (LGR5) expression was restricted to cells in the crypt base of the small and large intestines; in addition, LGR5 was considered to be a stem cell marker. A subsequent study proposed that intestinal epithelial tumors may originate from LGR5-positive stem cells (8).

LGR5, also known as HG38 and G protein-coupled receptor 49 (9,10), is a target gene of the Wnt/β-catenin signaling pathway (11), acting as receptor for the Wnt/β-catenin signaling agonist R-spondin (12,13). This signaling pathway has a critical role in normal development and the maintenance of adult stem cells as well as in tumor pathogenesis and growth (14,15). In healthy mucosa tissue, β-catenin is maintained at low cytoplasmic levels due to degradation regulated by a destruction complex composed of glycogen synthase kinase 3, Axin, adenomatous polyposis coli (APC) and other factors (16). In the progression of the majority of cases of CRC, the Wnt signaling pathway is activated early via truncations of APC and, less frequently, mutations of β-catenin (17). These mutations inhibit the activity of the destruction complex, resulting in the accumulation and nuclear translocation of β-catenin, ultimately resulting in transcriptional activation of target genes (18,19). Nuclear β-catenin is involved in two processes that are essential for embryonic development: Epithelial-mesenchymal transition and stem cell formation (20). Accumulating data indicates that aberrant nuclear expression of β-catenin may confer these two traits to tumor cells, therefore driving malignant tumor progression (21,22).

It is generally accepted that upregulation of LGR5 is associated with activated Wnt/β-catenin signaling, resulting in the overexpression of LGR5 in various types of cancer, including hepatocellular carcinoma, ovarian cancer and CRC (23,24). However, the underlying mechanisms for the role of LGR5 in carcinogenesis and intracellular signaling are poorly understood. Previous studies have identified that LGR5 and its homologs function as receptors of the R-spondin family of stem cell factors in order to enhance Wnt/β-catenin signaling (25). Furthermore, knockdown of LGR5 induced cell death in adenoma and carcinoma cells (26); in addition, LGR5-positive stem cell fractions were capable of forming tumors via activation of the Wnt/β-catenin signaling pathway (8). However, alternative studies propose that loss of LGR5 does not affect the proliferation or migration of intestinal cells (27). The aims of the present study were to further clarify the association between Lgr5, β-catenin and APC in the Wnt/β-catenin signaling pathway and to identify a novel method for the treatment of colorectal cancer.

Patients and methods

Patients and specimens

Specimens were collected from 20 patients with CRC who underwent surgical resection at the Department of Colorectal Surgery of Xin Hua Hospital Affiliated to Shanghai Jiaotong University School of Medicine (Shanghai, China) between November 2010 and May 2013. Tumor and paired healthy adjacent colorectal mucosa tissue samples were collected from each patient. All samples were obtained from the surgically resected material, immediately frozen in liquid nitrogen (Novobio Scientific, Shanghai, China) and stored at −80°C. The current study was approved by the Ethics Committee of Shanghai Jiaotong University School of Medicine and all samples were obtained following receipt of written informed consent from all patients.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

RNA extraction was performed using TRIzol reagent (Invitrogen Life Technologies, Carlsbad, CA, USA), according to the manufacturer's instructions. Complementary DNA was then synthesized using SuperScript III Reverse Transcriptase (Invitrogen Life Technologies). Subsequently, qPCR was performed on a CFX96TM Real-Time System (Bio-Rad Laboratories, Inc., Hercules, CA, USA) using the following primers (Invitrogen Life Technologies): APC, F 5′-GCTCCAAGCCCAACCTTAA-3′ and R 5′-GTTTTCGCCATCCACCAG-3′; β-catenin, F 5′-CATTCAGCAGAAGGTCCGA-3′ and R 5′-CTGGAAAACGCCATCACC-3′; and LGR5, F 5′-GTGGCAGCAAGTATGGCG-3′ and R 5′-AGCAAAGGGAATTGAGCAAG-3′. Fold induction values were calculated using the cycle threshold (Ct) method (2−ΔΔCt) (28). All experiments were performed in triplicate and independently repeated a minimum of three times.

Small interfering RNA (siRNA)

To knockdown LGR5, short hairpin RNA (shRNA) of the human LGR5 lentivirus gene transfer vector was constructed (Novobio Scientific). This gene transfer vector encoded the RNA sequence for green fluorescent protein (GFP). The following LGR5 siRNA sequence was used to target nucleotides: 5′-GTCTGCAATCAGTTACCTA-3′. Titer was measured by detecting GFP-positive HEK293T cells (Novobio Scientific) using fluorescence microscopy (IX51 microscope, Olympus Corporation, Tokyo, Japan), with the recombinant LGR5-targeting siRNA lentivirus prepared and titered to a concentration of 2.5×109 transfection units/ml. A scramble siRNA (Novobio Scientific) was used as a negative control (NC).

Detection of cell proliferation

NC-siRNA- and LGR5-siRNA-transfected cells were seeded into 96-well plates at a density of 4×103 cells/well and incubated for 24 h. On days 1, 3, 5 and 7, 10 µl Cell Counting Kit 8 (CCK8) solution (Dojindo Molecular Technologies, Inc., Shanghai, China) was added to each well. Color intensity was measured using an RT-2100C microplate reader (Rayto Life and Analytical Sciences Co.,Ltd., Nanshen, China) an absorbance of 450 nm to obtain cell growth curves. All experiments were performed in triplicate and repeated independently three times.

Statistical analysis

SAS software (version 8.5; SAS Institute, Inc., Cary, NC, USA) was used for all statistical analyses. Kruskal-Wallis non-parametric tests were performed to analyze differences in LGR5, APC and β-catenin mRNA expression between CRC and corresponding healthy mucosal tissues. In addition, a paired t-test was used to compare differences between the LGR5 knockdown group and negative control group. P<0.05 was considered to indicate a statistically significant difference between values.

Results

LGR5 and β-catenin expression is elevated and APC expression is reduced in CRC tissues

RNA was extracted from 20 CRC and adjacent healthy tissues samples, then subjected to RT-qPCR to determine the mRNA expression profiles of LGR5, β-catenin and APC. As demonstrated in Fig. 1, there were significant differences in LGR5, β-catenin and APC mRNA expression levels between the CRC and healthy colorectal mucosa samples (P=0.0484, 0.0032 and 0.0006, respectively). As shown in Table I, LGR5, β-catenin and APC expression in the CRC samples were divided by their expression in the matched healthy mucosa samples to obtain the tumor/normal healthy tissue expression (T/N) ratio. A T/N ratio of >1 indicated increased expression in CRC. Of the 20 CRC samples investigated, 14 (70%) exhibited elevated levels of LGR5 expression and 15 (75%) demonstrated elevated β-catenin expression compared with their corresponding healthy mucosa samples, with mean T/N ratios of 3.57 and 1.78, respectively. By contrast, APC mRNA expression was decreased in 17 (85%) CRC samples with a mean T/N ratio of 0.87.

Table I.

T/N ratio of LGR5, β-catenin and APC mRNA expression.

Table I.

T/N ratio of LGR5, β-catenin and APC mRNA expression.

CaseLGR5β-cateninAPC
  12.736.370.58
  21.250.890.25
  30.761.653.32
  41.691.100.15
  53.101.160.46
  62.612.580.38
  72.030.981.07
  81.091.260.20
  920.470.990.32
100.771.990.47
115.531.860.31
120.011.077.06
130.032.860.28
140.191.610.55
159.640.860.49
1611.232.730.61
174.030.660.48
180.291.960.34
191.381.460.06
202.561.500.11

[i] Values are presented as the T/N ratio. T/N, tumor/normal healthy tissue expression; LGR5, leucine-rich repeat-containing G-protein coupled receptor 5; APC, adenomatous polyposis coli.

siRNA-mediated knockdown of LGR5 inhibits the expression of APC and β-catenin

To investigate the functional relevance of LGR5 expression in CRC cell lines, the expression of LGR5 was knocked down in the HT-29 CRC cell line. Specific siRNA was constructed for LGR5 and its ability to knock down LGR5 mRNA was evaluated. RT-qPCR identified that the treatment of HT-29 cells with LGR5-siRNA resulted in a significant 52% decrease in LGR5 mRNA expression (P=0.0003) compared with the empty vector NC cells (Fig. 2A), indicating that the depletion of LGR5 using the siRNA method was effective. As illustrated in Fig. 2B and C, knockdown of LGR5 significantly decreased the expression of APC and β-catenin mRNA by 29% (P=0.0003) and 30% (P=0.001), respectively, compared with cells transfected with NC-siRNA at 48 h post-transfection. As APC is known to antagonize the transcriptional activity of β-catenin by promoting its nuclear export and its proteasomal destruction in the cytoplasm, decreasing the expression of APC may enhance Wnt/β-catenin signaling (1719). These results indicated that knockdown of LGR5 may inhibit the expression of β-catenin as well as promote β-catenin accumulation and nuclear translocation by downregulating APC.

Knockdown of LGR5 inhibits CRC cell proliferation

To investigate the effect of LGR5 on CRC cells viability, a viability curve of LGR5-knockdown HT29 cells was constructed by performing a CCK8 assay. As indicated in Fig. 3, HT29 cell growth was significantly inhibited following LGR5 knockdown compared with the growth of control group cells (P<0.001). This therefore indicated that downregulation of LGR5 expression using siRNA significantly inhibited the growth of HT-29 cells.

Discussion

LGR5, which has been established as a stem cell marker in the small intestine and colon, has also been identified as a downstream target gene of the Wnt signaling pathway (7). The current study demonstrated that LGR5 was significantly upregulated in CRC compared with healthy mucosa, which is comparable with the results of a number of previous studies (24,29,30). The Wnt signaling pathway is comprised of a vast number of proteins, including APC and β-catenin, two proteins that are critical in CRC tumorigenesis (18). Therefore, the present study aimed to evaluate the association between LGR5, APC and β-catenin expression and CRC, as well as identify the role of LGR5 in Wnt signaling.

The mRNA expression levels of LGR5, APC and β-catenin were detected in 20 CRC tissue samples and their corresponding healthy mucosa samples. The results demonstrated significant differences in LGR5, β-catenin and APC mRNA expression between CRC and healthy colorectal mucosa, indicating that CRC may be associated with aberrant activation of the Wnt/β-catenin signaling pathway. In CRC, mutations in β-catenin, Axin and certain signaling pathways may result in the accumulation of β-catenin and enhance Wnt signaling activation (31). However, APC mutations, which result in aberrant activation of the Wnt signaling pathway, occur most frequently in CRCs (17).

In the present study, to understand the effects of LGR5 on APC and β-catenin expression, which are two key components of Wnt signaling, LGR5 expression was silenced in the HT-29 CRC cell line using siRNA. A decrease in APC and β-catenin mRNA expression was observed following knockdown of LGR5. These results indicated that LGR5 may be involved in regulating Wnt/β-catenin signaling via modulation of the expression of APC and β-catenin. The role of APC and β-catenin in CRC tumorigenesis has been well studied. It was reported that >90% of cases of CRC exhibit cytoplasmic accumulation of β-catenin (32). When activated and accumulated in the cytoplasm, β-catenin is transferred to the nucleus, where it activates numerous nuclear transcription factors, such as transcription factor (TCF)/lymphoid enhancer-binding factor, which results in the activation of downstream target molecules. Abnormal expression of these molecules may result in abnormal proliferation and tumorigenesis (33). APC is an important tumor suppressor that downregulates the transcriptional activity of β-catenin by the following three mechanisms: i) Reducing the levels of cytoplasmic β-catenin by binding to Axin; ii) promoting the export of nuclear β-catenin; and iii) sequestering β-catenin, preventing it from binding to TCF (34). The simultaneous decrease in APC and β-catenin expression observed in the present study provided evidence that LGR5 may mediate bidirectional regulation in the Wnt/β-catenin signaling pathway (β-catenin- or APC-directed signaling). Accumulating data has demonstrated that the silencing of LGR5 influences the functional and molecular outcome of CRC cells; for example, previous reports have indicated that knocking down endogenous LGR5 in cultured CRC cell lines reduced their proliferation, migration, growth rates and colony formation capability (24,35,36). However, Walker et al (37) reported that the ablation of LGR5 increased invasion, induced anchorage-independent growth and enhanced tumorigenicity in a xenograft model. Based on these controversial results, the current authors proposed that LGR5 may act as a positive regulator of tumor growth when the β-catenin signaling pathway is predominant and acts as a negative regulator when the APC signaling pathway is predominant.

In the latter experiments of the present study, LGR5 downregulation resulted in the attenuation of HT29 cell proliferation. This data indicated that LGR5 may have a role in the regulation of CRC cell growth and proliferation, which is consistent with the results of previous investigations into basal cell carcinoma (15), Ewing sarcoma (24) and glioma (38). Thus, LGR5 may have the potential to serve as a therapeutic target in patients with CRC. However, future studies treating LGR5 as a therapeutic target should consider the bidirectional regulation of LGR5. Additionally, the current authors proposed that the blocking effect of LGR5 on APC may improve treatment efficiency.

In conclusion, the current results demonstrated that the majority of cases of CRC were associated with abnormal expression of LGR5, β-catenin and APC. Furthermore, knockdown of LGR5 significantly decreased the expression of β-catenin and APC. Due to the critical role of APC and β-catenin in colorectal tumor initiation and growth via the Wnt signaling pathway, LGR5 may be a potential therapeutic target for patients with CRC. However, the role of LGR5 in Wnt/β-catenin signaling requires further investigation.

Acknowledgements

The present study was supported by grants from the Program of Shanghai's Subject Chief Scientist from Shanghai Municipal Health Bureau (no. XBR2011032), the Biomedical Engineering Research Funds of Shanghai Jiaotong University (no. YG2011MS32), the Special Fund for Outstanding Young Teachers of Shanghai Municipal Education Commission (no. JDY10112), the Ministry of Health (no. W2011JZC27) and Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine (no. 11YJ005).

References

1 

Cunningham D, Atkin W, Lenz HJ, Lynch HT, Minsky B, Nordlinger B and Starling N: Colorectal cancer. Lancet. 375:1030–1047. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Weitz J, Koch M, Debus J, Höhler T, Galle PR and Büchler MW: Colorectal cancer. Lancet. 365:153–165. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Visvader JE and Lindeman GJ: Cancer stem cells in solid tumours: Accumulating evidence and unresolved questions. Nat Rev Cancer. 8:755–768. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Odoux C, Fohrer H, Hoppo T, et al: A stochastic model for cancer stem cell origin in metastatic colon cancer. Cancer Res. 68:6932–6941. 2008. View Article : Google Scholar : PubMed/NCBI

5 

Wu XS, Xi HQ and Chen L: Lgr5 is a potential marker of colorectal carcinoma stem cells that correlates with patient survival. World J Surg Oncol. 10:2442012. View Article : Google Scholar : PubMed/NCBI

6 

Lobo NA, Shimono Y, Qian D and Clarke MF: The biology of cancer stem cells. Annu Rev Cell Dev Biol. 23:675–699. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Barker N, van Es JH, Kuipers J, et al: Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature. 449:1003–1007. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Barker N, Ridgway RA, van Es JH, et al: Crypt stem cells as the cells-of-origin of intestinal cancer. Nature. 457:608–611. 2009. View Article : Google Scholar : PubMed/NCBI

9 

McDonald T, Wang R, Bailey W, et al: Identification and cloning of an orphan G protein-coupled receptor of the glycoprotein hormone receptor subfamily. Biochem Biophys Res Commun. 247:266–270. 1998. View Article : Google Scholar : PubMed/NCBI

10 

Hsu SY, Liang SG and Hsueh AJ: Characterization of two LGR genes homologous to gonadotropin and thyrotropin receptors with extracellular leucine-rich repeats and a G protein-coupled, seven-transmembrane region. Mol Endocrinol. 12:1830–1845. 1998. View Article : Google Scholar : PubMed/NCBI

11 

Van der Flier LG, Sabates-Bellver J, Oving I, et al: The intestinal Wnt/TCF signature. Gastroenterology. 132:628–632. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Carmon KS, Gong X, Lin Q, et al: R-spondins function as ligands of the orphan receptors LGR4 and LGR5 to regulate Wnt/beta-catenin signaling. Proc Natl Acad Sci USA. 108:11452–11457. 2011. View Article : Google Scholar : PubMed/NCBI

13 

de Lau W, Barker N, Low TY, et al: Lgr5 homologues associate with Wnt receptors and mediate R-spondin signalling. Nature. 476:293–297. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Early DS, Fontana L and Davidson NO: Translational approaches to addressing complex genetic pathways in colorectal cancer. Transl Res. 151:10–16. 2008. View Article : Google Scholar : PubMed/NCBI

15 

MacDonald BT, Tamai K and He X: Wnt/beta-catenin signaling: Components, mechanisms and diseases. Dev Cell. 17:9–26. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Cadigan KM and Peifer M: Wnt signaling from development to disease: Insights from model systems. Cold Spring Harb Perspect Biol. 1:a0028812009. View Article : Google Scholar : PubMed/NCBI

17 

Schneikert J and Behrens J: The canonical Wnt signalling pathway and its APC partner in colon cancer development. Gut. 56:417–425. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Clevers H and Nusse R: Wnt/β-catenin signaling and disease. Cell. 149:1192–1205. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Metcalfe C and Bienz M: Inhibition of GSK3 by Wnt signalling - two contrasting models. J Cell Sci. 124:3537–3544. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Brabletz T, Hlubek F, Spaderna S, et al: Invasion and metastasis in colorectal cancer: epithelial-mesenchymal transition, mesenchymal: Epithelial transition, stem cells and β-catenin. Cells Tissues Organs. 179:56–65. 2005. View Article : Google Scholar : PubMed/NCBI

21 

Morin PJ and Weeraratna AT: Wnt signaling in human cancer. Cancer Treat Res. 115:169–187. 2003.PubMed/NCBI

22 

Taketo MM: Shutting down Wnt signal-activated cancer. Nat Genet. 36:320–322. 2004. View Article : Google Scholar : PubMed/NCBI

23 

Yamamoto Y, Sakamoto M, Fujii G, et al: Overexpression of orphan G-protein-coupled receptor, Gpr49, in human hepatocellular carcinomas with beta-catenin mutations. Hepatology. 37:528–533. 2003. View Article : Google Scholar : PubMed/NCBI

24 

McClanahan T, Koseoglu S, Smith K, et al: Identification of overexpression of orphan G protein-coupled receptor GPR49 in human colon and ovarian primary tumors. Cancer Biol Ther. 5:419–426. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Carmon KS, Lin Q, Gong X, et al: LGR5 interacts and cointernalizes with Wnt receptors to modulate Wnt/β-catenin signaling. Mol Cell Biol. 32:2054–2064. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Al-Kharusi MR, Smartt HJ, Greenhough A, et al: LGR5 promotes survival in human colorectal adenoma cells and is upregulated by PGE2: Implications for targeting adenoma stem cells with NSAIDs. Carcinogenesis. 34:1150–1157. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Garcia MI, Ghiani M, Lefort A, et al: LGR5 deficiency deregulates Wnt signaling and leads to precocious Paneth cell differentiation in the fetal intestine. Dev Biol. 331:58–67. 2009. View Article : Google Scholar : PubMed/NCBI

28 

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. View Article : Google Scholar : PubMed/NCBI

29 

Uchida H, Yamazaki K, Fukuma M, et al: Overexpression of leucine-rich repeat-containing G protein-coupled receptor 5 in colorectal cancer. Cancer Sci. 101:1731–1737. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Takahashi H, Ishii H, Nishida N, et al: Significance of Lgr5(+ve) cancer stem cells in the colon and rectum. Ann Surg Oncol. 18:1166–1174. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Yang M, Zhong WW and Srivastava N: G protein-coupled lysophosphatidic acid receptors stimulate proliferation of colon cancer cells through the {beta}-catenin pathway. Proc Natl Acad Sci USA. 102:6027–6032. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Hsu HC, Liu YS, Tseng KC, et al: Overexpression of Lgr5 correlates with resistance to 5-FU-based chemotherapy in colorectal cancer. Int J Colorectal Dis. 28:1535–1546. 2013. View Article : Google Scholar : PubMed/NCBI

33 

Hirsch D, Barker N, McNeil N, et al: LGR5 positivity defines stem-like cells in colorectal cancer. Carcinogenesis. 35:849–858. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Walker F, Zhang HH, Odorizzi A and Burgess AW: LGR5 is a negative regulator of tumourigenicity, antagonizes Wnt signalling and regulates cell adhesion in colorectal cancer cell lines. PLoS One. 6:e227332011. View Article : Google Scholar : PubMed/NCBI

35 

Tanese K, Fukuma M, Yamada T, et al: G-protein-coupled receptor GPR49 is up-regulated in basal cell carcinoma and promotes cell proliferation and tumor formation. Am J Pathol. 173:835–843. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Fan XS, Wu HY, Yu HP, et al: Expression of Lgr5 in human colorectal carcinogenesis and its potential correlation with beta-catenin. Int J Colorectal Dis. 25:583–590. 2010. View Article : Google Scholar : PubMed/NCBI

37 

Scannell CA, Pedersen EA, Mosher JT, et al: LGR5 is expressed by Ewing sarcoma and potentiates Wnt/β-catenin signaling. Front Oncol. 3:812013. View Article : Google Scholar : PubMed/NCBI

38 

Wang D, Zhou J, Fan C, et al: Knockdown of LGR5 suppresses the proliferation of glioma cells in vitro and in vivo. Oncol Rep. 31:41–49. 2014.PubMed/NCBI

Related Articles

Journal Cover

June-2015
Volume 9 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lin Y, Wu T, Yao Q, Zi S, Cui L, Yang M and Li J: LGR5 promotes the proliferation of colorectal cancer cells via the Wnt/β-catenin signaling pathway. Oncol Lett 9: 2859-2863, 2015
APA
Lin, Y., Wu, T., Yao, Q., Zi, S., Cui, L., Yang, M., & Li, J. (2015). LGR5 promotes the proliferation of colorectal cancer cells via the Wnt/β-catenin signaling pathway. Oncology Letters, 9, 2859-2863. https://doi.org/10.3892/ol.2015.3144
MLA
Lin, Y., Wu, T., Yao, Q., Zi, S., Cui, L., Yang, M., Li, J."LGR5 promotes the proliferation of colorectal cancer cells via the Wnt/β-catenin signaling pathway". Oncology Letters 9.6 (2015): 2859-2863.
Chicago
Lin, Y., Wu, T., Yao, Q., Zi, S., Cui, L., Yang, M., Li, J."LGR5 promotes the proliferation of colorectal cancer cells via the Wnt/β-catenin signaling pathway". Oncology Letters 9, no. 6 (2015): 2859-2863. https://doi.org/10.3892/ol.2015.3144