Upregulation of the Wnt co-receptor LRP6 promotes hepatocarcinogenesis and enhances cell invasion

Edmund Kwok-Kwan Tung, Betty Yin-Chi Wong, Tai-On Yau, Irene Oi-Lin Ng, Edmund Kwok-Kwan Tung, Betty Yin-Chi Wong, Tai-On Yau, Irene Oi-Lin Ng

Abstract

Background: Activation of the Wnt/β-catenin signaling pathway plays a crucial role in hepatocellular carcinoma (HCC). Low-density lipoprotein (LDL) receptor-related protein-6 (LRP6) is one of the co-receptors of the Wnt/β-catenin pathway and forms a signaling complex with Wnt ligand and Frizzled receptor to activate downstream signaling. However, the role of LRP6 in hepatocarcinogenesis is unclear. In this study, we examined its expression and roles in human HCC.

Methodology/principal findings: Using real-time quantitative RT-PCR, we found that LRP6 was frequently (45%) overexpressed in human HCCs (P = 0.003). In vitro studies showed that ectopic expression of LRP6 increased the protein level of β-catenin. Moreover, overexpression of the full-length and constitutively active LRP6, respectively, activated the WNT/β-catenin signaling pathway, as shown by the TCF/β-catenin reporter assay. With regard to the effects of LRP6 overexpression in HCC cells, stable overexpression of the constitutively active LRP6 in BEL-7402 HCC cells enhanced cell proliferation, cell migration, and invasion in vitro as well as tumorigenicity in nude mice.

Conclusions/significance: Our findings indicate that overexpression of LRP6 contributes to the hyperactivation of the Wnt/β-catenin signaling pathway in human HCCs and suggest it may play a role in hepatocarcinogenesis.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Overexpression of LRP6 in HCC…
Figure 1. Overexpression of LRP6 in HCC cell lines and human HCCs.
(A) Quantitative real-time PCR and Western blotting. Both transcripts and protein of LRP6 were expressed in all seven HCC cell lines. (B) In human HCCs, the transcript level of LRP6 was frequently (45%) up-regulated as compared with their corresponding non-tumorous livers (P = 0.003). (C) Western blot analysis. LRP6 protein level was determined in 28 HCC pairs and was found to be overexpressed in 9 (32%) cases. The 9 cases with LRP6 protein overexpression in the tumors are shown. (D) Immunohistochemical analysis. LRP6 protein was found to be overexpressed in the HCC as compared with the corresponding non-tumorous liver. High power magnification of the tumor showed strong positive staining of LRP6 protein in the cytoplasm (white arrows) and also the membranes (black short arrows) of the tumor cells.
Figure 2. Ectopic expression of LRP6 activated…
Figure 2. Ectopic expression of LRP6 activated the Wnt/β-catenin pathway.
(A) Schematic diagram showing the structural domains of Myc-tagged full length form of LRP6 (myc-FL LRP6) and the constitutively active form (myc-CA LRP6). (B) Western blotting. Myc-FL LRP6 and myc-CA LRP6 were transiently overexpressed in BEL-7402 HCC cell line and human embryonic kidney cell HEK293T cells. The protein level of β-catenin was increased in both BEL-7402 HCC cell line and HEK293T cells. (C) TOP/FOP luciferase reporter assay. Expression of myc-FL LRP6 led to an activation of TCF/β-catenin reporter up to ∼40-fold (without Wnt3a treatment) and ∼120-fold (with Wnt3a treatment), respectively, as compared with the vector control. The fold of induction in myc-CA LRP6 cells reached ∼150-fold even without Wnt3a treatment. (D) Similar results were also observed in HEK293T cells.
Figure 3. Constitutively active LRP6 enhanced cell…
Figure 3. Constitutively active LRP6 enhanced cell proliferation in vitro.
(A) LRP6 expressing BEL-7402 stable cells were established using myc-CA LRP6 construct. The protein level of β-catenin was upregulated as compared with the parental BEL-7402 cells. (B) Cell proliferation assay. The numbers of cells of CA LRP6 Clones #1, #3, #8 and #11 on Day 7 were significantly higher than the vector control BEL-7402 cells (P = 0.032,

Figure 4. Constitutively active LRP6 promoted both…

Figure 4. Constitutively active LRP6 promoted both cell migration and invasion.

Cell migration and invasion…

Figure 4. Constitutively active LRP6 promoted both cell migration and invasion.
Cell migration and invasion assays were performed using LRP6-stably expressing BEL-7402 cells. (A) Overexpression of myc-CA LRP6 enhanced cell migration in BEL-7402 cells. The numbers of migrated cells in CA LRP6 Clones #3 and #8 were significantly higher than the vector control (P

Figure 5. Constitutively active LRP6 enhanced tumor…

Figure 5. Constitutively active LRP6 enhanced tumor cell growth in vivo .

(A) In vivo nude…

Figure 5. Constitutively active LRP6 enhanced tumor cell growth in vivo.
(A) In vivo nude mice injection assay was performed by injecting myc-CA LRP6 stably expressing and vector control BEL-7402 cells subcutaneously into the flank of the nude mice. (B) The tumor sizes of two myc-CA LRP6 stably expressing tumors were significantly higher as compared with the tumor of vector control (P<0.001). (C) The tumor weight of Clone #3 was higher in myc-CA LRP6 than the control vector (P = 0.002). Another clone (Clone #8) showed a trend of higher tumor weight but the difference did not reach statistical significance (P = 0.165). Error bar = SEM.
Similar articles
Cited by
References
    1. Ferlay J, Shin HR, Bray F, Forman D, Mathers C, et al. Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 2010;127(12):2893–2917. - PubMed
    1. Zender L, Kubicka S. Molecular pathogenesis and targeted therapy of hepatocellular carcinoma. Onkologie. 2008;31:550–555. - PubMed
    1. Wong CM, Ng IO. Molecular pathogenesis of hepatocellular carcinoma. Liver Int. 2008;28:160–174. - PubMed
    1. Takigawa Y, Brown AM. Wnt signaling in liver cancer. Curr Drug Targets. 2008;9:1013–1024. - PMC - PubMed
    1. Llovet JM, Bruix J. Molecular targeted therapies in hepatocellular carcinoma. Hepatology. 2008;48:1312–1327. - PMC - PubMed
Show all 41 references
Publication types
MeSH terms
Substances
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 4. Constitutively active LRP6 promoted both…
Figure 4. Constitutively active LRP6 promoted both cell migration and invasion.
Cell migration and invasion assays were performed using LRP6-stably expressing BEL-7402 cells. (A) Overexpression of myc-CA LRP6 enhanced cell migration in BEL-7402 cells. The numbers of migrated cells in CA LRP6 Clones #3 and #8 were significantly higher than the vector control (P

Figure 5. Constitutively active LRP6 enhanced tumor…

Figure 5. Constitutively active LRP6 enhanced tumor cell growth in vivo .

(A) In vivo nude…

Figure 5. Constitutively active LRP6 enhanced tumor cell growth in vivo.
(A) In vivo nude mice injection assay was performed by injecting myc-CA LRP6 stably expressing and vector control BEL-7402 cells subcutaneously into the flank of the nude mice. (B) The tumor sizes of two myc-CA LRP6 stably expressing tumors were significantly higher as compared with the tumor of vector control (P<0.001). (C) The tumor weight of Clone #3 was higher in myc-CA LRP6 than the control vector (P = 0.002). Another clone (Clone #8) showed a trend of higher tumor weight but the difference did not reach statistical significance (P = 0.165). Error bar = SEM.
Figure 5. Constitutively active LRP6 enhanced tumor…
Figure 5. Constitutively active LRP6 enhanced tumor cell growth in vivo.
(A) In vivo nude mice injection assay was performed by injecting myc-CA LRP6 stably expressing and vector control BEL-7402 cells subcutaneously into the flank of the nude mice. (B) The tumor sizes of two myc-CA LRP6 stably expressing tumors were significantly higher as compared with the tumor of vector control (P<0.001). (C) The tumor weight of Clone #3 was higher in myc-CA LRP6 than the control vector (P = 0.002). Another clone (Clone #8) showed a trend of higher tumor weight but the difference did not reach statistical significance (P = 0.165). Error bar = SEM.

References

    1. Ferlay J, Shin HR, Bray F, Forman D, Mathers C, et al. Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 2010;127(12):2893–2917.
    1. Zender L, Kubicka S. Molecular pathogenesis and targeted therapy of hepatocellular carcinoma. Onkologie. 2008;31:550–555.
    1. Wong CM, Ng IO. Molecular pathogenesis of hepatocellular carcinoma. Liver Int. 2008;28:160–174.
    1. Takigawa Y, Brown AM. Wnt signaling in liver cancer. Curr Drug Targets. 2008;9:1013–1024.
    1. Llovet JM, Bruix J. Molecular targeted therapies in hepatocellular carcinoma. Hepatology. 2008;48:1312–1327.
    1. Kim YD, Park CH, Kim HS, Choi SK, Rew JS, et al. Genetic alterations of Wnt signaling pathway-associated genes in hepatocellular carcinoma. J Gastroenterol Hepatol. 2008;23:110–118.
    1. Tommasi S, Pinto R, Pilato B, Paradiso A. Molecular pathways and related target therapies in liver carcinoma. Curr Pharm Des. 2007;13:3279–3287.
    1. Teufel A, Staib F, Kanzler S, Weinmann A, Schulze-Bergkamen H, et al. Genetics of hepatocellular carcinoma. World J Gastroenterol. 2007;13:2271–2282.
    1. Clevers H. Wnt/beta-catenin signaling in development and disease. Cell. 2006;127:469–480.
    1. MacDonald BT, Tamai K, He X. Wnt/beta-catenin signaling: components, mechanisms, and diseases. Dev Cell. 2009;17:9–26.
    1. Wong CM, Fan ST, Ng IO. beta-Catenin mutation and overexpression in hepatocellular carcinoma: clinicopathologic and prognostic significance. Cancer. 2001;92:136–145.
    1. Li Y, Lu W, He X, Schwartz AL, Bu G. LRP6 expression promotes cancer cell proliferation and tumorigenesis by altering beta-catenin subcellular distribution. Oncogene. 2004;23:9129–9135.
    1. Liu CC, Prior J, Piwnica-Worms D, Bu G. LRP6 overexpression defines a class of breast cancer subtype and is a target for therapy. Proc Natl Acad Sci U S A. 2010;107:5136–5141.
    1. Zhang J, Li Y, Liu Q, Lu W, Bu G. Wnt signaling activation and mammary gland hyperplasia in MMTV-LRP6 transgenic mice: implication for breast cancer tumorigenesis. Oncogene. 2010;29:539–549.
    1. Brennan K, Gonzalez-Sancho JM, Castelo-Soccio LA, Howe LR, Brown AM. Truncated mutants of the putative Wnt receptor LRP6/Arrow can stabilize beta-catenin independently of Frizzled proteins. Oncogene. 2004;23:4873–4884.
    1. Hu L, Wen JM, Sham JS, Wang W, Xie D, et al. Establishment of cell lines from a primary hepatocellular carcinoma and its metastatis. Cancer Genet Cytogenet. 2004;148:80–84.
    1. Chan DW, Chan CY, Yam JW, Ching YP, Ng IO. Prickle-1 negatively regulates Wnt/beta-catenin pathway by promoting Dishevelled ubiquitination/degradation in liver cancer. Gastroenterology. 2006;131:1218–1227.
    1. Moon RT, Kohn AD, De Ferrari GV, Kaykas A. WNT and beta-catenin signalling: diseases and therapies. Nat Rev Genet. 2004;5:691–701.
    1. Park JY, Park WS, Nam SW, Kim SY, Lee SH, et al. Mutations of beta-catenin and AXIN I genes are a late event in human hepatocellular carcinogenesis. Liver Int. 2005;25:70–76.
    1. Ishizaki Y, Ikeda S, Fujimori M, Shimizu Y, Kurihara T, et al. Immunohistochemical analysis and mutational analyses of beta-catenin, Axin family and APC genes in hepatocellular carcinomas. Int J Oncol. 2004;24:1077–1083.
    1. Devereux TR, Stern MC, Flake GP, Yu MC, Zhang ZQ, et al. CTNNB1 mutations and beta-catenin protein accumulation in human hepatocellular carcinomas associated with high exposure to aflatoxin B1. Mol Carcinog. 2001;31:68–73.
    1. Satoh S, Daigo Y, Furukawa Y, Kato T, Miwa N, et al. AXIN1 mutations in hepatocellular carcinomas, and growth suppression in cancer cells by virus-mediated transfer of AXIN1. Nat Genet. 2000;24:245–250.
    1. Huang H, Fujii H, Sankila A, Mahler-Araujo BM, Matsuda M, et al. Beta-catenin mutations are frequent in human hepatocellular carcinomas associated with hepatitis C virus infection. Am J Pathol. 1999;155:1795–1801.
    1. Hsu HC, Jeng YM, Mao TL, Chu JS, Lai PL, et al. Beta-catenin mutations are associated with a subset of low-stage hepatocellular carcinoma negative for hepatitis B virus and with favorable prognosis. Am J Pathol. 2000;157:763–770.
    1. Mi K, Johnson GV. Role of the intracellular domains of LRP5 and LRP6 in activating the Wnt canonical pathway. J Cell Biochem. 2005;95:328–338.
    1. Beagle B, Johnson GV. Differential modulation of TCF/LEF-1 activity by the soluble LRP6-ICD. PLoS ONE. 2010;5:e11821.
    1. Pinson KI, Brennan J, Monkley S, Avery BJ, Skarnes WC. An LDL-receptor-related protein mediates Wnt signalling in mice. Nature. 2000;407:535–538.
    1. Tamai K, Semenov M, Kato Y, Spokony R, Liu C, et al. LDL-receptor-related proteins in Wnt signal transduction. Nature. 2000;407:530–535.
    1. Wehrli M, Dougan ST, Caldwell K, O'Keefe L, Schwartz S, et al. arrow encodes an LDL-receptor-related protein essential for Wingless signalling. Nature. 2000;407:527–530.
    1. Merle P, Kim M, Herrmann M, Gupte A, Lefrancois L, et al. Oncogenic role of the frizzled-7/beta-catenin pathway in hepatocellular carcinoma. J Hepatol. 2005;43:854–862.
    1. Merle P, de la Monte S, Kim M, Herrmann M, Tanaka S, et al. Functional consequences of frizzled-7 receptor overexpression in human hepatocellular carcinoma. Gastroenterology. 2004;127:1110–1122.
    1. Kim M, Lee HC, Tsedensodnom O, Hartley R, Lim YS, et al. Functional interaction between Wnt3 and Frizzled-7 leads to activation of the Wnt/beta-catenin signaling pathway in hepatocellular carcinoma cells. J Hepatol. 2008;48:780–791.
    1. Bengochea A, de Souza MM, Lefrancois L, Le Roux E, Galy O, et al. Common dysregulation of Wnt/Frizzled receptor elements in human hepatocellular carcinoma. Br J Cancer. 2008;99:143–150.
    1. Zhang Z, Schittenhelm J, Guo K, Buhring HJ, Trautmann K, et al. Upregulation of frizzled 9 in astrocytomas. Neuropathol Appl Neurobiol. 2006;32:615–624.
    1. Kirikoshi H, Sekihara H, Katoh M. Expression profiles of 10 members of Frizzled gene family in human gastric cancer. Int J Oncol. 2001;19:767–771.
    1. Janssens N, Andries L, Janicot M, Perera T, Bakker A. Alteration of frizzled expression in renal cell carcinoma. Tumour Biol. 2004;25:161–171.
    1. Li Y, Bu G. LRP5/6 in Wnt signaling and tumorigenesis. Future Oncol. 2005;1:673–681.
    1. Hoang BH, Kubo T, Healey JH, Sowers R, Mazza B, et al. Expression of LDL receptor-related protein 5 (LRP5) as a novel marker for disease progression in high-grade osteosarcoma. Int J Cancer. 2004;109:106–111.
    1. Guo Y, Rubin EM, Xie J, Zi X, Hoang BH. Dominant negative LRP5 decreases tumorigenicity and metastasis of osteosarcoma in an animal model. Clin Orthop Relat Res. 2008;466:2039–2045.
    1. Bjorklund P, Svedlund J, Olsson AK, Akerstrom G, Westin G. The internally truncated LRP5 receptor presents a therapeutic target in breast cancer. PLoS ONE. 2009;4:e4243.
    1. Bjorklund P, Akerstrom G, Westin G. An LRP5 receptor with internal deletion in hyperparathyroid tumors with implications for deregulated WNT/beta-catenin signaling. PLoS Med. 2007;4:e328.

Source: PubMed

3
Suscribir