Next Article in Journal
Plants as Biofactories for Therapeutic Proteins and Antiviral Compounds to Combat COVID-19
Previous Article in Journal
A Kinetic Stem Cell Counting Analysis of the Specific Effects of Cell Culture Medium Growth Factors on Adipose-Derived Mesenchymal Stem Cells
Previous Article in Special Issue
Proteomic Analysis of Female Synovial Fluid to Identify Novel Biomarkers for Osteoarthritis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

The Relationship between LRP6 and Wnt/β-Catenin Pathway in Colorectal and Esophageal Cancer

1
Department of Molecular Pathology, Division of Health Sciences, Graduate School of Medicine, Osaka University, Yamadaoka 1-7, Suita City 565-0871, Japan
2
Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Yamadaoka 2-2, Suita City 565-0871, Japan
*
Author to whom correspondence should be addressed.
Submission received: 27 January 2023 / Revised: 21 February 2023 / Accepted: 21 February 2023 / Published: 23 February 2023
(This article belongs to the Special Issue Innovative Biomarker and Precision Medicine)

Abstract

:
High expression of low-density lipoprotein receptor-related protein 6 (LRP6), a key component of the Wnt/β-catenin signaling pathway, is reported to be associated with malignant potential in some solid tumors including breast cancer and hepatocellular carcinoma. Few reports, however, have examined its function and clinical significance in colorectal cancers (CRC) demonstrating constitutive activation of Wnt signaling. Here, we compared the expression level and function of LRP6 in CRC with that of esophageal squamous cell carcinoma (ESCC) bearing few Wnt/β-catenin pathway mutations. On immunohistochemical staining, high LRP6 expression was noted in three of 68 cases (4.4%), and high β-catenin in 38 of 67 cases (56.7%) of CRC. High LRP6 expression was found in 21 of 82 cases (25.6%), and high β-catenin expression in 29 of 73 cases (39.7%) of ESCC. In our in vitro studies, LRP6 knockdown hardly changed Wnt signaling activity in CRC cell lines with mutations in Wnt signaling downstream genes. In contrast, in ESCC cell lines without Wnt signaling-related mutations, LRP6 knockdown significantly decreased Wnt signaling activity. LRP6 function may depend on constitutive activation of Wnt signaling.

1. Introduction

In epithelial tissue, the processes of cell proliferation and differentiation are coordinately regulated [1,2]. One of the most pivotal signaling pathways associated with this regulation is the Wingless and INT-1 (Wnt)/β-catenin pathway [3]. β-catenin is a subunit of the cadherin protein complex, whose stabilization is crucial for the activation of Wnt/β-catenin signaling [4]. In the absence of Wnt, β-catenin is phosphorylated by a protein complex consisting of adenomatous polyposis coli (APC), AXIN, glycogen synthase kinase-3β, and casein kinase 1, followed by ubiquitin-mediated degradation of β-catenin in the proteasome [5]. This active degradation mechanism keeps cytoplasmic β-catenin concentrations consistently low [6].
In the presence of Wnt, Wnt ligands bind to the seven-pass transmembrane receptor Frizzled and the low-density lipoprotein receptor-related protein (LRP) 6, inducing LRP6 phosphorylation. The active form of LRP6 then binds by phosphorylation to AXIN, suppressing degradation of β-catenin and releasing it into the cytoplasm [7]. β-catenin accumulates in the cytoplasm and translocates into the nucleus and binds to the TCF/LEF (T-cell factor/lymphoid enhancer factor) transcription factor to promote expression of genes that regulate cell cycle, proliferation, survival, and differentiation [8] (Supplementary Figure S1).
The abnormal activation of Wnt/β-catenin pathway by gene mutation is involved in the pathogenesis of various diseases, especially human cancers [3]. For example, a high incidence of mutations in the β-catenin gene (CTNNB1) has been observed in liver cancer [9], endometrial cancer [10], and colorectal cancer (CRC) [11]. Mutations in APC, a key tumor suppressor gene, have been detected in most CRCs [12], as well as in some other cancers including gastric cancer [13].
LRP6 is an indispensable transmembrane receptor for Wnt and is overexpressed in several solid tumors, including colorectal [14], liver [15], and breast cancers [16], as well as pancreatic adenocarcinomas [17], in association with increased Wnt/β-catenin signaling.
Increased LRP6 phosphorylation involved in nuclear accumulation of β-catenin also has been observed in CRC, with relevance for tumor malignancy and staging [18], along with poor prognosis [19]. In addition, in a study using whole-exome sequencing, functional variants of LRP6 were identified as novel risk factors for early-onset CRC [20]. With increasing evidence that LRP6 is closely associated with tumor development and progression, various antibodies [21], peptides [16], and small molecules [22] have been developed and show anticancer properties by inhibiting LRP6 function directly or indirectly.
Although the aberration of LRP6 is observed in many types of cancers with various gene mutation frequencies in the Wnt/β-catenin pathway, there are no reports showing whether LRP6 could behave differently in cancers based on Wnt/β-catenin pathway genetic abnormalities. For this reason, the purpose of this study was to elucidate the role of LRP6 in CRC and esophageal squamous carcinoma (ESCC), which shows a low frequency of gene mutations in the Wnt/β-catenin pathway [23], and no reports address LRP6 function.

2. Materials and Methods

2.1. Cell Lines and Culture

Human CRC cell lines (DLD-1, with truncated APC and wild-type β-catenin; HCT116, with a Ser45 deletion in one β-catenin allele and one wild-type allele, wild-type APC) and ESCC cell lines (TE-1 and TE-8: wild-type APC; wild-type β-catenin) were purchased from the American Type Culture Collection (ATCC, Manassas, VA, USA). These cell lines were authenticated by morphologic inspection, short tandem repeat profiling, and mycoplasma testing by the ATCC. The authors also performed the mycoplasma testing on a regular basis. Cell lines were maintained in Dulbecco’s modified Eagle’s medium and Roswell Park Memorial Institute 1640 with 10% fetal bovine serum and 100 U/mL penicillin and 100 μg/mL streptomycin in humid 5% CO2 at 37 °C.

2.2. Clinical Tissue Samples

CRC samples were collected from 68 patients and ESCC samples from 82 patients who underwent surgery at Osaka University Hospital (Osaka, Japan) between 2006 and 2010. As positive controls, we used samples from three cases of breast cancer. Written informed consent was obtained from all patients, in accordance with guidelines approved by the Institutional Research Board. This study was conducted under the supervision of the Ethics Board of Osaka University Hospital (No. 15144).

2.3. siRNA Transfection

Small interfering (si)RNA against LRP6 (siRNA-LRP6) and negative control siRNA (NC-siRNA) were purchased from GeneDesign Inc. (Osaka, Japan). Transfection was performed at a final concentration of 30 nM with Lipofectamine 2000 (Thermo Fisher Scientific, Waltham, MA, USA), based on the manufacturer’s protocol.

2.4. Immunohistochemical Staining

Formalin-fixed, paraffin-embedded tissues were prepared and sectioned into 4 μm slices. Sections were deparaffinized with xylene and then rehydrated in graded alcohols. Immunostaining was performed using antibodies and the VECTASTAIN Elite ABC Kit (Vector Laboratories, Burlingame, CA, USA), according to the manufacturer’s protocol. Anti-LRP6 rabbit polyclonal antibody (PA5-13145) was obtained from Thermo Fisher Scientific, and anti-β-catenin mouse monoclonal antibody (610153) was obtained from BD Transduction Laboratories (Franklin Lakes, NJ, USA). The stain intensity was graded as 0 to 2 (0 = none; 1 = weak; 2 = strong), and the percentage of positive cells was scored as 1 to 3 (1 = 0–49%; 2 = 50–89%; 3 = 90–100%). The two scores were multiplied together to determine a staining score, with possible scores of 0, 1, 2, 3, 4, or 6. Immunostaining for the LRP6 protein was performed in 68 sets of normal colon tissues and cancer tissues and 82 sets of normal esophageal tissues and cancer tissues. The breast cancer tissue served as a positive control for the primary antibody (Supplementary Figure S2A). A serial section in which phosphate-buffered saline was used instead of the primary antibody served as a negative control (Supplementary Figure S2B). Immunostaining for the β-catenin protein was performed using 67 CRC samples and 73 ESCC samples. The 58 and 68 samples of CRC and ESCC were used for the clinicopathological analysis and survival analysis, respectively. For the relapse-free survival analysis, stage IV cases were omitted.

2.5. Western Blot Analysis

The protein samples were subjected to SDS-PAGE and transferred onto polyvinylidene difluoride membranes. Cell lysates and the antibodies against LRP6 (C47E20) and β-actin (ACTB) were obtained from Cell Signaling Technology, and the antibody against β-catenin (610153) from BD Transduction Laboratories. We collected protein lysate after processing by siRNA at 48 and 72 h. After incubation with secondary antibodies, signals were detected with the Pierce Western Blotting Substrate (Thermo Fisher Scientific).

2.6. RNA Isolation and qRT-PCR

Total RNA from cell lines was isolated using TRIzol reagent (Thermo Fisher Scientific) following the manufacturer’s protocol. RNA concentration and purity were assessed with a Nano Drop One spectrophotometer W1F1 (Thermo Fisher Scientific). Total RNA was reverse transcribed using the High-Capacity RNA-to-cDNA kit (Thermo Fisher Scientific). To measure LRP6 expression level, a real-time quantitative polymerase chain reaction (qRT-PCR) was performed using a LightCycler 2.0 Instrument (F Hoffmann-La Roche AG, Basel, Switzerland) with LightCycler TaqMan Master (F Hoffman-La Roche AG). The product numbers of the TaqMan Gene Expression assay were as follows: ACTB ID Hs01060665_g1 and LRP6 ID Hs00999795_m1. Relative expression was quantified using the ΔΔct method.

2.7. TOPFLASH Assay

Cells were seeded onto 96-well plates (DLD-1, TE-8: 4000 cells/well; HCT116, TE-1: 3000 cells/well), and transfection of siRNA was performed using Lipofectamine 2000 (Thermo Fisher Scientific). At 24 h after siRNA transfection, cells were transfected with 100 ng/well of the TOPFLASH luciferase reporter vector (pGL4.49 [luc2P/TCF-LEF RE/Hygro]; Promega, Fitchburg, WI, USA) using Lipofectamine 2000 and treated with 100 ng/mL human recombinant Wnt3a (R&D Systems, Minneapolis, MN, USA). After 24 h, cells were collected with the reporter lysis buffer for luciferase activity measurement using the Dual-Luciferase Reporter Assay System (Promega). Firefly luciferase activity was normalized against Renilla luciferase activity for each transfected well. The transfection efficiency was evaluated with 1 ng/well of the Renilla luciferase reporter vector (pRL-CMV, Promega). We incubated four kinds of cultured cells under unstimulated conditions, collected the lysate, and measured firefly luciferase activity (TOPFLASH), as well as Renilla luciferase activity (Renilla) as endogenous control.

2.8. Statistical Analysis

Data were expressed as means ± standard deviations. Statistical analysis was performed using JMP Pro 14 software (SAS Institute, Cary, NC, USA). Statistical differences were analyzed with the Student’s t-test for continuous variables and the chi-squared test for noncontinuous data. Survival curves were generated using the Kaplan–Meier method and assessed using the log-rank test. A value of p < 0.05 was considered statistically significant.

3. Results

3.1. Immunohistochemistry for LRP6 and β-Catenin in CRC

In normal mucosa of colon tissues, LRP6-positive cells were generally localized at the bottom of the colonic epithelium (Figure 1A, arrow), whereas CRC tissue samples expressed the LRP6 protein mainly in the cytoplasm to varying extents (Figure 1B,C). With regard to staining intensity, most CRC samples showed weak staining for LRP6 (59/68: 86.8%) (Figure 1D,E). Most CRC samples also had a staining score of 2 or 3 (55/68: 80.9%) (Figure 1F).
In contrast to the results for LRP6 staining, many cases showed strong β-catenin expression (strong: 2) at a high incidence (90–100%), mainly in the cytoplasm (Figure 2A–D). As a result, staining scores were relatively higher than for LRP6, and 38 of 67 cases (56.7%) had scores of 4 or 6 points (Figure 2E).

3.2. Immunohistochemistry for LRP6 and β-Catenin in ESCC

In normal squamous epithelium of esophageal tissues, LRP6 expression was found mainly in the parabasal layer (Figure 3A). In ESCC tissue samples, expression of the LRP6 protein was similar to that for CRC, and 21 of 82 cases (25.6%) were classified with a high staining score of 4 or 6 (Figure 3B,C and Supplementary Figure S3). Many samples showed overall cytoplasmic β-catenin staining similar to CRC, but a greater proportion of ESCC vs. CRC cases had weak intensity. We found that 29 out of 73 cases (39.7%) had a staining score of 4 or 6 for β-catenin in ESCC (Figure 3D,E and Supplementary Figure S3).

3.3. Influence of LRP6 on Wnt/β-Catenin Signaling Activity

With the introduction of LRP6-siRNA into HCT116 and TE-8, LRP6 mRNA expression decreased compared with expression in the control group receiving NC-siRNA (Figure 4A). Likewise, protein levels decreased at 48 and 72 h (Figure 4B). Next, we investigated the relation between LRP6 expression and Wnt/β-catenin signaling activity in CRC and ESCC cell lines. Under the unstimulated condition, Wnt/β-catenin signaling activity in TE-1 and TE-8 cells was lower than in CRC cells (Figure 4C).
As shown in Figure 4D, we performed TOPFLASH assay. In DLD-1 cells, which carry the homozygous mutation in APC, we found no response to stimulation with Wnt3a, along with unchanged Wnt/β-catenin signaling under LRP6 suppression (Figure 4E). In contrast, in HCT116 cells, which carry the heterozygous mutation in CTNNB1 encoding β-catenin, Wnt/β-catenin signaling activity significantly increased with Wnt3a stimulation and did not decline with LRP6 suppression through the introduction of LRP6-siRNA (Figure 4F).
In TE-1 and TE-8 cells, which do not carry a mutation in APC or CTNNB1, signaling activity increased under Wnt3a stimulation in both cell lines (p < 0.01), and introduction of LRP6-siRNA significantly dampened this increase (p < 0.01 or 0.05) (Figure 4G,H).

3.4. Relationship between the Expression of LRP6 or β-Catenin Protein and Prognosis in CRC or ESCC

To explore the relationship of LRP6 expression and β-catenin protein with clinicopathological correlation and prognosis in CRC and ESCC, we divided the cases with clinical follow-up data by high LRP6 expression (staining score >3) and low LRP6 expression (staining score < 2). Similarly, we performed analyses with two other groups, one with high β-catenin expression (staining score > 4) and one with low β-catenin expression (staining score < 3). The clinicopathological status of 58 CRC patients and 68 ESCC patients, which was stratified by LRP6 and β-catenin expression level, are shown in Table 1 and Table 2. High expression of LRP6 in CRC was significantly associated with lymphatic invasion (p = 0.035, Table 1). Kaplan–Meier survival curves indicated no significant difference between either set of compared groups in relapse-free survival (RFS) or overall survival (OS) (Supplementary Figures S4 and S5).

4. Discussion

4.1. The Function of LRP6 May Differ Depending on Genetic Abnormalities in the Wnt/β-Catenin Pathway

In CRC, gene mutations in proteins downstream of the Wnt/β-catenin signaling pathway, such as APC (67%), CTNNB1 (6%), and AXIN2 (5%), are often observed [24]. Liver cancer predominantly has mutations in CTNNB1 (25%) and AXIN1 (8%) genes [24]. On the other hand, breast [25] and pancreatic cancer [26] cases rarely have mutations in these genes. Although overexpression of LRP6 is observed in these types of cancer [27], it is not known whether the function of LRP6 differs depending on genetic abnormalities in the Wnt/β-catenin pathway.
We performed an LRP6 knockdown in CRC and ESCC cells to investigate how LRP6 expression affects Wnt/β-catenin signaling. In the luciferase reporter assay using the TOPFLASH plasmid to evaluate Wnt/β-catenin signaling, we found almost no change under Wnt3a stimulation or LRP6 knockdown in DLD-1 cells carrying a homozygous APC mutation. One possible explanation is that the signal was activated in DLD-1 because of an APC mutation affecting the downstream signaling in the Wnt/β-catenin pathway, so that the upstream factor LRP6 did not significantly affect this activity. To confirm whether LRP6 knockdown only affects the Wnt/β-catenin pathway when it is not constitutively active, an experiment with overexpression of APC protein in DLD-1 to reduce Wnt/-catenin pathway activation may be helpful. Because APC is a huge protein, the center third of APC (cAPC), which contains both β-catenin and AXIN binding domain and is sufficient to promote degradation of β-catenin protein [28], may be useful for this experiment. HCT116 cells, which carry a heterozygous mutation (one wild-type allele and one mutant allele with inactivation of SER45) in CTNNB1, responded to stimulation by Wnt3a but not to LRP6 knockdown. The wild-type allele in HCT116 provided susceptibility to Wnt3a, but the mutant SER45 allele likely caused accumulation of β-catenin. These results suggest that LRP6 is not important for the activation of Wnt/β-catenin signaling in CRC, which harbors genetic abnormalities in this pathway. There are several controversial studies showing whether LRP6 expression contributes to Wnt/β-catenin signal activation in CRC with APC and CTNNB1 mutations. Raisch et al. demonstrated that LRP6 did not affect tumorigenesis in APCMin/+ mice, and they also showed that LRP6 knockdown did not affect the colony formation activity and cell growth in CRC cells with APC or CTNNB1 mutations, suggesting that LRP6 is dispensable for tumorigenesis induced by an aberrant Wnt/β-catenin pathway [29]. Chen et al. showed that LRP6 knockout in CRC cells with APC mutations did not alter the activity of the Wnt/β-catenin pathway [30]. On the other hand, Yao et al. reported that LRP6 overexpression in CRC cells activated the Wnt/β-catenin pathway [19]. Saito-Diaz et al. showed that LRP6 is required for activation of the Wnt/β-catenin pathway in CRC cells with APC mutations, but not in CRC cells with CTNNB1 mutations [31], and Cabel et al. confirmed this result with a single cell analysis [32]. Our results support the former studies.
Unlike DLD-1 and HCT116 cells, the TE-1 and TE-8 ESCC cell lines without mutations in APC and CTNNB1 show no enhanced signal activity downstream of the Wnt/β-catenin pathway. Therefore, they responded to Wnt3a stimulation and showed a decrease in this response under LRP6-siRNA exposure, suggesting that LRP6 is important for regulating the Wnt/β-catenin pathway in ESCC. Our results support previous findings that LRP6 is associated with cell migration, invasion, and epithelial-to-mesenchymal transition in ESCC cell lines [33].
Taken together, our results suggest that the function of LRP6 may differ depending on genetic abnormalities in the Wnt/β-catenin pathway (Supplementary Figure S6).

4.2. The Expression of LRP6 and β-Catenin in CRC and ESCC

LRP6 is a membrane protein associated with the Wnt/β-catenin signaling pathway (Supplementary Figure S1) and an important mediator of the intestinal stem cell niche [29]. It contributes to maintenance of the intestinal crypt structure and transmits signals downstream by binding to Wnt ligands [34]. Our immunostaining showed that in normal colonic epithelial tissue, LRP6-positive cells were observed at the crypts of the glandular duct that are sites of active cell division, with increased Wnt/β-catenin signaling. In normal esophageal tissue, LRP6-positive cells are mainly located in the parabasal layer; consistent with this layer is the squamous epithelium proliferative zone where stem cells are present [35].
In cancer, many CRC samples demonstrate a high positivity rate for β-catenin and strong staining intensity, which is consistent with previous reports. Although the staining intensity for LRP6 was not as strong in many CRC samples, the positivity rate was high. This result supports the idea that LRP6 may not be important for the activation of the Wnt/β-catenin pathway in CRC with genetic abnormalities such as APC or CTNNB1 mutations.
In contrast, for β-catenin staining scores in ESCC samples that were lower than in CRC samples and related to LRP6, the positivity rate did not differ significantly compared with CRC samples, but the number of ESCC samples with strong intensity tended to increase. This result suggests that increased expression of LRP6 is associated with the activation of the Wnt/β-catenin pathway in ESCC.
These results are consistent with our in vitro data and further support the idea that LRP6 functions differentially in cancers depending on the presence or absence of genetic abnormalities in the Wnt/β-catenin pathway.

4.3. Clinical Significance of LRP6 Expression in CRC and ESCC

By analyzing the relevance of LRP6 expression to prognosis and clinicopathological parameters, we found a significant correlation between high LRP expression and lymphatic invasion in CRC, suggesting that LRP6 is associated with the invasive activity of CRCs. This is consistent with the studies that show LRP6 is associated with metastasis and poor prognosis in multiple types of cancers such as breast cancer [36], liver cancer [37], and oral squamous cell carcinoma [38]. In addition, Yao et al. reported that LRP6 promoted the migration of CRC cells through regulation of cytoskeleton dynamics [19]. However, as described above, this group showed that LRP6 overexpression activated the Wnt/β-catenin pathway even in CRC cells with APC or CTNNB1 mutations. Therefore, further studies will be needed to reveal the underlying mechanism.
Although high LRP expression was significantly correlated with lymphatic invasion in CRC, Kaplan–Meier curves showed no significant difference in OS or RFS between groups with high vs. low LRP6 expression in CRC. This discrepancy may be because we analyzed only small number of samples for survival analysis in this study. Therefore, further investigation using a higher sample number will be required.

4.4. The Potential of LRP6 as a Therapeutic Target

Small molecule inhibitor, monoclonal antibody and modified peptides, which target the Wnt/β-catenin pathway have been developed and have proceeded to clinical trial [39]. The target of these drugs are various proteins in the Wnt/β-catenin pathway [40], such as ligands (e.g., WNT5a) [41], receptors and coreceptors (e.g., Fzd) [42], intracellular components (e.g., β-catenin) [43], and transcription cofactors (e.g., CBP) [44]. With regards to LRP6, Niclosamide [45], Salinomycin [46], and Rottlerin [47] are known to suppress LRP6 expression.
Our results suggest that LRP6 inhibition and other Wnt/β-catenin inhibitors may be effective for ESCC in case of LRP6 overexpression and upregulated Wnt/β-catenin pathway. Meanwhile, LRP6 inhibition may not be effective against CRC with genetic abnormalities in the Wnt/β-catenin pathway.
However, it is reported that LRP6 affects not only the Wnt/β-catenin pathway, but also other pathways such as the noncanonical Wnt pathway [48], Wnt-dependent stabilization of proteins (Wnt/STOP) pathway [49], G protein-coupled receptor (GPCR) pathway [50], and Hippo pathway [51]. These pathways are related to tumorigenesis and malignant phenotype of CRC. For example, overexpression of Wnt11, which activates the noncanonical Wnt pathway promotes the proliferation, invasion, and migration of CRC cell lines [52]. Some GPCRs such as S1PR3, S1PR5, and AT1R are associated with tumorigenesis, proliferation, invasion, and migration of CRC cells [53]. Cho et al. reported that upregulation of Hippo pathway genes is related to poor prognosis in patients with CRC [54]. Therefore, LRP6 may be involved in the malignant phenotype of CRC cells via these pathways.
If LRP6 overexpression is associated with the malignant phenotype of CRC independent of the Wnt/β-catenin pathway, it is possible that the combined inhibition of LRP6 with an intracellular component or transcription cofactors of the Wnt/β-catenin pathway may have additive or synergistic effects. Further studies will be needed to investigate this possibility.
Nucleic acid medicine, including small interference RNA (siRNA) and microRNA (miRNA), is considered a next-generation cancer therapy [55]. We previously showed that microRNA-487b (miR-487b) decreased LRP6 expression by directly binding to the 3′-untranslational region (UTR) of LRP6 mRNA, and miR-487b treatment suppressed the proliferation and invasion of CRC cells with APC or CTNNB1 mutations [56]. We developed an improved in vivo drug delivery system, which is named as inorganic nanoparticle device (iNaD), and we showed the effective antitumor effect of miRNA encapsuled into iNaD [57]. Therefore, the combinational treatment of iNaD-miR-487b and Wnt inhibitors (e.g., β-catenin inhibitor, CBP inhibitor) may be a new therapeutic strategy for CRC with genetic abnormalities in the Wnt/β-catenin pathway.

5. Conclusions

Our findings indicate that LRP6 could behave differently in different cancers based on genetic abnormalities related to the Wnt/β-catenin pathway. Further studies are needed to elucidate the function of LRP6 in cancer and characterize its potential as a therapeutic target.

Supplementary Materials

The following supporting information can be downloaded at: https://0-www-mdpi-com.brum.beds.ac.uk/article/10.3390/life13030615/s1, Figure S1: Schematic illustration of Wnt/b-catenin pathway and its association with LRP6. Figure S2: Positive and Negative control of LRP6 immunostaining. (A) Positive control: LRP6 is expressed in a breast cancer. (B) Negative control: Phosphate-buffered saline was used instead of the primary antibody. Figure S3: LRP6 and β-catenin expression in ESCC tissues. (A, B) Classification of the positivity of LRP6 expression in ESCC. (C, D) Classification of LRP6 expression intensity in ESCC. (E, F) Classification of the positivity of β-catenin expression in ESCC. (G, H) Classification of β-catenin expression intensity in ESCC. Figure S4: Survival analysis in CRC tissues. (A) Overall survival of CRC patients with LRP6 high or low expression. (B) Relapse-free survival of CRC patients with LRP6 high or low expression. (C) Overall survival of CRC patients with β-catenin high or low expression. (D) Relapse-free survival of CRC patients with β-catenin high or low expression. Figure S5: Survival analysis in ESCC tissues. (A) Overall survival of ESCC patients with LRP6 high or low expression. (B) Relapse-free survival of ESCC patients with LRP6 high or low expression. (C) Overall survival of ESCC patients with β-catenin high or low expression. (D) Relapse-free survival of ESCC patients with β-catenin high or low expression. Figure S6: Schematic illustration of the relationship between Wnt/b-catenin pathway and LRP6 in CRC and ESCC.

Author Contributions

Conceptualization, A.S., M.M. and H.Y. (Hirofumi Yamamoto); methodology, M.M., K.O. and H.Y. (Hirofumi Yamamoto); validation, K.O., N.N., M.W., H.Y. (Hiroyuki Yamamoto) and Y.Y.; formal analysis, M.W., H.Y. (Hiroyuki Yamamoto), X.W., S.S. and Y.Y.; investigation, A.S., K.O., N.N., M.W., H.Y. (Hiroyuki Yamamoto) and S.K.; resources, X.W., Y.Y. and H.Y. (Hirofumi Yamamoto); data curation, K.O., N.N. and Y.Y.; writing—original draft preparation, A.S., M.M., Y.Y. and H.Y. (Hirofumi Yamamoto); writing—review and editing, M.M., Y.Y. and H.Y. (Hirofumi Yamamoto); visualization, N.N. and Y.Y.; supervision, H.Y. (Hirofumi Yamamoto); project administration, H.Y. (Hirofumi Yamamoto); funding acquisition, X.W., A.S. and H.Y. (Hirofumi Yamamoto). All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by Grant-in-Aid for JSPS KAKENHI C (No. 16K07174) to Xin Wu.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki, and approved by the Institutional Review Board of Osaka University (Approval No. 15144).

Informed Consent Statement

Written informed consent was obtained from all patients.

Data Availability Statement

Not applicable.

Acknowledgments

We thank Y. Doki and H. Eguchi, as well as other colleagues in Dept. Gastroenterological Surgery, Osaka University for cooperation in the use of clinical tissue samples.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. van der Flier, L.G.; Clevers, H. Stem cells, self-renewal, and differentiation in the intestinal epithelium. Annu. Rev. Physiol. 2009, 71, 241–260. [Google Scholar] [CrossRef]
  2. Jacobs, I.J.; Ku, W.Y.; Que, J. Genetic and cellular mechanisms regulating anterior foregut and esophageal development. Dev. Biol. 2012, 369, 54–64. [Google Scholar] [CrossRef] [Green Version]
  3. Klaus, A.; Birchmeier, W. Wnt signalling and its impact on development and cancer. Nat. Rev. Cancer 2008, 8, 387–398. [Google Scholar] [CrossRef]
  4. Valenta, T.; Hausmann, G.; Basler, K. The many faces and functions of β-catenin. EMBO J. 2012, 31, 2714–2736. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. MacDonald, B.T.; Tamai, K.; He, X. Wnt/beta-catenin signaling: Components, mechanisms, and diseases. Dev. Cell. 2009, 17, 9–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Park, H.B.; Kim, J.W.; Baek, K.H. Regulation of Wnt Signaling through Ubiquitination and Deubiquitination in Cancers. Int. J. Mol. Sci. 2020, 21, 3904. [Google Scholar] [CrossRef] [PubMed]
  7. Zeng, X.; Huang, H.; Tamai, K.; Zhang, X.; Harada, Y.; Yokota, C.; Almeida, K.; Wang, J.; Doble, B.; Woodgett, J.; et al. Initiation of Wnt signaling: Control of Wnt coreceptor Lrp6 phosphorylation/activation via frizzled, dishevelled and axin functions. Development 2008, 135, 367–375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Lecarpentier, Y.; Schussler, O.; Hébert, J.L.; Vallée, A. Multiple Targets of the Canonical WNT/β-Catenin Signaling in Cancers. Front. Oncol. 2019, 9, 1248. [Google Scholar] [CrossRef] [PubMed]
  9. Zucman-Rossi, J.; Benhamouche, S.; Godard, C.; Boyault, S.; Grimber, G.; Balabaud, C.; Cunha, A.S.; Bioulac-Sage, P.; Perret, C. Differential effects of inactivated Axin1 and activated beta-catenin mutations in human hepatocellular carcinomas. Oncogene 2007, 26, 774–780. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. McConechy, M.K.; Ding, J.; Senz, J.; Yang, W.; Melnyk, N.; Tone, A.A.; Prentice, L.M.; Wiegand, K.C.; McAlpine, J.N.; Shah, S.P.; et al. Ovarian and endometrial endometrioid carcinomas have distinct CTNNB1 and PTEN mutation profiles. Mod. Pathol. 2014, 27, 128–134. [Google Scholar] [CrossRef] [Green Version]
  11. Kim, S.; Jeong, S. Mutation Hotspots in the β-Catenin Gene: Lessons from the Human Cancer Genome Databases. Mol. Cells 2019, 42, 8–16. [Google Scholar] [CrossRef] [PubMed]
  12. Rowan, A.J.; Lamlum, H.; Ilyas, M.; Wheeler, J.; Straub, J.; Papadopoulou, A.; Bicknell, D.; Bodmer, W.F.; Tomlinson, I.P. APC mutations in sporadic colorectal tumors: A mutational “hotspot” and interdependence of the “two hits”. Proc. Natl. Acad. Sci. USA 2000, 97, 3352–3357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Li-Chang, H.H.; Kasaian, K.; Ng, Y.; Lum, A.; Kong, E.; Lim, H.; Jones, S.J.; Huntsman, D.G.; Schaeffer, D.F.; Yip, S. Retrospective review using targeted deep sequencing reveals mutational differences between gastroesophageal junction and gastric carcinomas. BMC Cancer 2015, 15, 32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Rismani, E.; Fazeli, M.S.; Mahmoodzadeh, H.; Movassagh, A.; Azami, S.; Karimipoor, M.; Teimoori-Toolabi, L. Pattern of LRP6 gene expression in tumoral tissues of colorectal cancer. Cancer Biomark. 2017, 19, 151–159. [Google Scholar] [CrossRef] [PubMed]
  15. Tung, E.K.; Wong, B.Y.; Yau, T.O.; Ng, I.O. Upregulation of the Wnt co-receptor LRP6 promotes hepatocarcinogenesis and enhances cell invasion. PLoS ONE 2012, 7, e36565. [Google Scholar] [CrossRef] [Green Version]
  16. Liu, C.C.; 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. USA 2010, 107, 5136–5141. [Google Scholar] [CrossRef] [Green Version]
  17. Garg, B.; Giri, B.; Majumder, K.; Dudeja, V.; Banerjee, S.; Saluja, A. Modulation of post-translational modifications in β-catenin and LRP6 inhibits Wnt signaling pathway in pancreatic cancer. Cancer Lett. 2017, 388, 64–72. [Google Scholar] [CrossRef]
  18. Lemieux, E.; Agnol, S.; Eaudry, K.; Arrier, J.; Rivard, N. Oncogenic KRAS signalling promotes the Wnt/β-catenin pathway through LRP6 in colorectal cancer. Oncogene 2015, 34, 4914–4927. [Google Scholar] [CrossRef]
  19. Yao, Q.; An, Y.; Hou, W.; Cao, Y.N.; Yao, M.F.; Ma, N.N.; Hou, L.; Zhang, H.; Liu, H.J.; Zhang, B. LRP6 promotes invasion and metastasis of colorectal cancer through cytoskeleton dynamics. Oncotarget 2017, 8, 109632–109645. [Google Scholar] [CrossRef]
  20. de Voer, R.M.; Hahn, M.M.; Weren, R.D.; Mensenkamp, A.R.; Gilissen, C.; van Zelst-Stams, W.A.; Spruijt, L.; Kets, C.M.; Zhang, J.; Venselaar, H.; et al. Identification of Novel Candidate Genes for Early-Onset Colorectal Cancer Susceptibility. PLoS Genet. 2016, 12, e1005880. [Google Scholar] [CrossRef]
  21. Fenderico, N.; van Scherpenzeel, R.C.; Goldflam, M.; Proverbio, D.; Jordens, I.; Kralj, T.; Stryeck, S.; Bass, T.Z.; Hermans, G.; Ullman, C.; et al. Anti-LRP5/6 VHHs promote differentiation of Wnt-hypersensitive intestinal stem cells. Nat. Commun. 2019, 10, 365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Arensman, M.D.; Nguyen, P.; Kershaw, K.M.; Lay, A.R.; Ostertag-Hill, C.A.; Sherman, M.H.; Downes, M.; Liddle, C.; Evans, R.M.; Dawson, D.W. Calcipotriol Targets LRP6 to Inhibit Wnt Signaling in Pancreatic Cancer. Mol. Cancer Res. 2015, 13, 1509–1519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Ninomiya, I.; Endo, Y.; Fushida, S.; Sasagawa, T.; Miyashita, T.; Fujimura, T.; Nishimura, G.; Tani, T.; Hashimoto, T.; Yagi, M.; et al. Alteration of beta-catenin expression in esophageal squamous-cell carcinoma. Int. J. Cancer 2000, 85, 757–761. [Google Scholar] [CrossRef]
  24. Bugter, J.M.; Fenderico, N.; Maurice, M.M. Mutations and mechanisms of WNT pathway tumour suppressors in cancer. Nat. Rev. Cancer 2021, 21, 5–21. [Google Scholar] [CrossRef] [PubMed]
  25. Brown, A.M. Wnt signaling in breast cancer: Have we come full circle? Breast. Cancer Res. 2001, 3, 351–355. [Google Scholar] [CrossRef] [Green Version]
  26. Gerdes, B.; Ramaswamy, A.; Simon, B.; Pietsch, T.; Bastian, D.; Kersting, M.; Moll, R.; Bartsch, D. Analysis of beta-catenin gene mutations in pancreatic tumors. Digestion 1999, 60, 544–548. [Google Scholar] [CrossRef]
  27. Raisch, J.; Côté-Biron, A.; Rivard, N. A Role for the WNT Co-Receptor LRP6 in Pathogenesis and Therapy of Epithelial Cancers. Cancers 2019, 11, 1162. [Google Scholar] [CrossRef] [Green Version]
  28. Shih, I.M.; Yu, J.; He, T.C.; Vogelstein, B.; Kinzler, K.W. The b-catenin binding domain of adenomatous polyposis coli is sufficient for tumor suppression. Cancer Res. 2000, 60, 1671–1676. [Google Scholar]
  29. Raisch, J.; Côté-Biron, A.; Langlois, M.J.; Leblanc, C.; Rivard, N. Unveiling the Roles of Low-Density Lipoprotein Receptor-Related Protein 6 in Intestinal Homeostasis, Regeneration and Oncogenesis. Cells 2021, 10, 1792. [Google Scholar] [CrossRef]
  30. Chen, M.; He, X. APC Deficiency Leads to β-Catenin Stabilization and Signaling Independent of LRP5/6. Dev. Cell 2019, 49, 825–826. [Google Scholar] [CrossRef]
  31. Saito-Diaz, K.; Benchabane, H.; Tiwari, A.; Tian, A.; Li, B.; Thompson, J.J.; Hyde, A.S.; Sawyer, L.M.; Jodoin, J.N.; Santos, E.; et al. APC Inhibits Ligand-Independent Wnt Signaling by the Clathrin Endocytic Pathway. Dev. Cell 2018, 44, 566–581. [Google Scholar] [CrossRef] [Green Version]
  32. Cabel, C.R.; Alizadeh, E.; Robbins, D.J.; Ahmed, Y.; Lee, E.; Thorne, C.A. Single-Cell Analyses Confirm the Critical Role of LRP6 for Wnt Signaling in APC-Deficient Cells. Dev. Cell 2019, 49, 827–828. [Google Scholar] [CrossRef]
  33. Guan, H.; Liu, J.; Lv, P.; Zhou, L.; Zhang, J.; Cao, W. MicroRNA-590 inhibits migration, invasion and epithelial-to-mesenchymal transition of esophageal squamous cell carcinoma by targeting low-density lipoprotein receptor-related protein 6. Oncol. Rep. 2020, 44, 1385–1392. [Google Scholar] [CrossRef]
  34. Farin, H.F.; Jordens, I.; Mosa, M.H.; Basak, O.; Korving, J.; Tauriello, D.V.; de Punder, K.; Angers, S.; Peters, P.J.; Maurice, M.M.; et al. Visualization of a short-range Wnt gradient in the intestinal stem-cell niche. Nature 2016, 530, 340–343. [Google Scholar] [CrossRef]
  35. Ingravallo, G.; Dall’Olmo, L.; Segat, D.; Fassan, M.; Mescoli, C.; Dazzo, E.; Castoro, C.; Polimeno, L.; Rizzetto, C.; Baroni, M.D.; et al. CDX2 hox gene product in a rat model of esophageal cancer. J. Exp. Clin. Cancer Res. 2009, 28, 108. [Google Scholar] [CrossRef] [Green Version]
  36. Zhang, Y.; Shu, C.; Maimaiti, Y.; Wang, S.; Lu, C.; Zhou, J. LRP6 as a biomarker of poor prognosis of breast cancer. Gland Surg. 2021, 10, 2414–2427. [Google Scholar] [CrossRef]
  37. Jia, Q.; Bu, Y.; Wang, Z.; Chen, B.; Zhang, Q.; Yu, S.; Liu, Q. Maintenance of stemness is associated with the interation of LRP6 and heparin-binding protein CCN2 autocrined by hepatocellular carcinoma. J. Exp. Clin. Cancer Res. 2017, 36, 117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Yuan, Y.; Xie, X.; Jiang, Y.; Wei, Z.; Wang, P.; Chen, F.; Li, X.; Sun, C.; Zhao, H.; Zeng, X.; et al. LRP6 is identified as a potential prognostic marker for oral squamous cell carcinoma via MALDI-IMS. Cell Death Dis. 2017, 8, e3035. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Yu, F.; Yu, C.; Li, F.; Zuo, Y.; Wang, Y.; Yao, L.; Wu, C.; Wang, C.; Ye, L. Wnt/β-catenin signaling in cancers and targeted therapies. Signal Transduct. Target. Ther. 2021, 6, 307. [Google Scholar] [CrossRef] [PubMed]
  40. Wang, Z.; Zhao, T.; Zhang, S.; Wang, J.; Chen, Y.; Zhao, H.; Yang, Y.; Shi, S.; Chen, Q.; Liu, K. The Wnt signaling pathway in tumorigenesis, pharmacological targets, and drug development for cancer therapy. Biomark. Res. 2021, 9, 68. [Google Scholar] [CrossRef] [PubMed]
  41. Säfholm, A.; Tuomela, J.; Rosenkvist, J.; Dejmek, J.; Härkönen, P.; Andersson, T. The Wnt-5a-derived hexapeptide Foxy-5 inhibits breast cancer metastasis in vivo by targeting cell motility. Clin. Cancer Res. 2008, 14, 6556–6563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Gurney, A.; Axelrod, F.; Bond, C.J.; Cain, J.; Chartier, C.; Donigan, L.; Fischer, M.; Chaudhari, A.; Ji, M.; Kapoun, A.M.; et al. Wnt pathway inhibition via the targeting of Frizzled receptors results in decreased growth and tumorigenicity of human tumors. Proc. Natl. Acad. Sci. USA 2012, 109, 11717–11722. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Lepourcelet, M.; Chen, Y.N.; France, D.S.; Wang, H.; Crews, P.; Petersen, F.; Bruseo, C.; Wood, A.W.; Shivdasani, R.A. Small-molecule antagonists of the oncogenic Tcf/beta-catenin protein complex. Cancer Cell 2004, 5, 91–102. [Google Scholar] [CrossRef] [Green Version]
  44. Emami, K.H.; Nguyen, C.; Ma, H.; Kim, D.H.; Jeong, K.W.; Eguchi, M.; Moon, R.T.; Teo, J.L.; Kim, H.Y.; Moon, S.H.; et al. A small molecule inhibitor of beta-catenin/CREB-binding protein transcription. Proc. Natl. Acad. Sci. USA 2004, 101, 12682–12687. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Lu, W.; Lin, C.; Roberts, M.J.; Waud, W.R.; Piazza, G.A.; Li, Y. Niclosamide suppresses cancer cell growth by inducing Wnt co-receptor LRP6 degradation and inhibiting the Wnt/β-catenin pathway. PLoS ONE 2011, 6, e29290. [Google Scholar] [CrossRef]
  46. Lu, W.; Li, Y. Salinomycin suppresses LRP6 expression and inhibits both Wnt/β-catenin and mTORC1 signaling in breast and prostate cancer cells. J. Cell. Biochem. 2014, 115, 1799–1807. [Google Scholar] [CrossRef] [Green Version]
  47. Lu, W.; Lin, C.; Li, Y. Rottlerin induces Wnt co-receptor LRP6 degradation and suppresses both Wnt/β-catenin and mTORC1 signaling in prostate and breast cancer cells. Cell Signal. 2014, 26, 1303–1309. [Google Scholar] [CrossRef] [Green Version]
  48. Bryja, V.; Andersson, E.R.; Schambony, A.; Esner, M.; Bryjová, L.; Biris, K.K.; Hall, A.C.; Kraft, B.; Cajanek, L.; Yamaguchi, T.P.; et al. The extracellular domain of Lrp5/6 inhibits noncanonical Wnt signaling in vivo. Mol. Biol. Cell 2009, 20, 924–936. [Google Scholar] [CrossRef] [Green Version]
  49. Acebron, S.P.; Niehrs, C. β-Catenin-Independent Roles of Wnt/LRP6 Signaling. Trends Cell Biol. 2016, 26, 956–967. [Google Scholar] [CrossRef]
  50. Wan, M.; Li, J.; Herbst, K.; Zhang, J.; Yu, B.; Wu, X.; Qiu, T.; Lei, W.; Lindvall, C.; Williams, B.O.; et al. LRP6 mediates cAMP generation by G protein-coupled receptors through regulating the membrane targeting of Gα(s). Sci. Signal. 2011, 4, ra15. [Google Scholar] [CrossRef] [Green Version]
  51. Azzolin, L.; Panciera, T.; Soligo, S.; Enzo, E.; Bicciato, S.; Dupont, S.; Bresolin, S.; Frasson, C.; Basso, G.; Guzzardo, V.; et al. YAP/TAZ incorporation in the β-catenin destruction complex orchestrates the Wnt response. Cell 2014, 158, 157–170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Nishioka, M.; Ueno, K.; Hazama, S.; Okada, T.; Sakai, K.; Suehiro, Y.; Okayama, N.; Hirata, H.; Oka, M.; Imai, K.; et al. Possible involvement of Wnt11 in colorectal cancer progression. Mol. Carcinog. 2013, 52, 207–217. [Google Scholar] [CrossRef]
  53. Zeng, Z.; Ma, C.; Chen, K.; Jiang, M.; Vasu, R.; Liu, R.; Zhao, Y.; Zhang, H. Roles of G Protein-Coupled Receptors (GPCRs) in Gastrointestinal Cancers: Focus on Sphingosine 1-Shosphate Receptors, Angiotensin II Receptors, and Estrogen-Related GPCRs. Cells 2021, 10, 2988. [Google Scholar] [CrossRef] [PubMed]
  54. Cho, S.Y.; Gwak, J.W.; Shin, Y.C.; Moon, D.; Ahn, J.; Sol, H.W.; Kim, S.; Kim, G.; Shin, H.M.; Lee, K.H.; et al. Expression of Hippo pathway genes and their clinical significance in colon adenocarcinoma. Oncol. Lett. 2017, 13, 4055–4076. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Damase, T.R.; Sukhovershin, R.; Boada, C.; Taraballi, F.; Pettigrew, R.I.; Cooke, J.P. The Limitless Future of RNA Therapeutics. Front. Bioeng. Biotechnol. 2021, 9, 628137. [Google Scholar] [CrossRef]
  56. Hata, T.; Mokutani, Y.; Takahashi, H.; Inoue, A.; Munakata, K.; Nagata, K.; Haraguchi, N.; Nishimura, J.; Hata, T.; Matsuda, C.; et al. Identification of microRNA-487b as a negative regulator of liver metastasis by regulation of KRAS in colorectal cancer. Int. J. Oncol. 2017, 50, 487–496. [Google Scholar] [CrossRef] [Green Version]
  57. Wu, X.; Yokoyama, Y.; Takahashi, H.; Kouda, S.; Yamamoto, H.; Wang, J.; Morimoto, Y.; Minami, K.; Hata, T.; Shamma, A.; et al. Improved In Vivo Delivery of Small RNA Based on the Calcium Phosphate Method. J. Pers. Med. 2021, 11, 1160. [Google Scholar] [CrossRef]
Figure 1. LRP6 expression in normal colon and CRC tissues. (A) LRP6 expression in normal colon mucosa. (B,C) Classification of the positivity of LRP6 expression in CRC. (D,E) Classification of LRP6 expression intensity in CRC. (F) Distribution of the staining score, calculated by multiplying the intensity and positivity scores.
Figure 1. LRP6 expression in normal colon and CRC tissues. (A) LRP6 expression in normal colon mucosa. (B,C) Classification of the positivity of LRP6 expression in CRC. (D,E) Classification of LRP6 expression intensity in CRC. (F) Distribution of the staining score, calculated by multiplying the intensity and positivity scores.
Life 13 00615 g001
Figure 2. β-catenin expression in CRC tissues. (A,B) Classification of the positivity of β-catenin expression in CRC. (C,D) Classification of β-catenin expression intensity in CRC. (E) Distribution of the staining score, calculated by multiplying the intensity and positivity scores.
Figure 2. β-catenin expression in CRC tissues. (A,B) Classification of the positivity of β-catenin expression in CRC. (C,D) Classification of β-catenin expression intensity in CRC. (E) Distribution of the staining score, calculated by multiplying the intensity and positivity scores.
Life 13 00615 g002
Figure 3. LRP6 and β-catenin expression in normal esophageal and ESCC tissues. (A) LRP6 expression in normal esophageal tissues. (B,C) Distribution of the LRP6 staining score, calculated by multiplying the intensity and positivity scores. (D,E) Distribution of the β-catenin staining score, calculated by multiplying the intensity and positivity scores.
Figure 3. LRP6 and β-catenin expression in normal esophageal and ESCC tissues. (A) LRP6 expression in normal esophageal tissues. (B,C) Distribution of the LRP6 staining score, calculated by multiplying the intensity and positivity scores. (D,E) Distribution of the β-catenin staining score, calculated by multiplying the intensity and positivity scores.
Life 13 00615 g003
Figure 4. Effects of LRP6 on Wnt/β-catenin signaling activity. (A) Expression of LRP6 mRNA in CRC and ESCC cell lines, as measured by qRT-PCR. We used ACTB as endogenous control. * p < 0.01. (B) The protein level of LRP6 in CRC and ESCC cell lines. We used ACTB as endogenous control. (C) Wnt/β-catenin signaling activity under unstimulated conditions in CRC and ESCC cell lines. (D) Experimental plan for investigating signaling activity by adding LRP6-siRNA or Wnt3a to the CRC and ESCC cell lines. (EH) Wnt/β-catenin signaling activity using CRC and ESCC cell lines. We used Renilla as endogenous control. * p < 0.05, ** p < 0.01; n.s.: not significant.
Figure 4. Effects of LRP6 on Wnt/β-catenin signaling activity. (A) Expression of LRP6 mRNA in CRC and ESCC cell lines, as measured by qRT-PCR. We used ACTB as endogenous control. * p < 0.01. (B) The protein level of LRP6 in CRC and ESCC cell lines. We used ACTB as endogenous control. (C) Wnt/β-catenin signaling activity under unstimulated conditions in CRC and ESCC cell lines. (D) Experimental plan for investigating signaling activity by adding LRP6-siRNA or Wnt3a to the CRC and ESCC cell lines. (EH) Wnt/β-catenin signaling activity using CRC and ESCC cell lines. We used Renilla as endogenous control. * p < 0.05, ** p < 0.01; n.s.: not significant.
Life 13 00615 g004
Table 1. Comparison of clinicopathological status stratified by LRP6 or β-catenin expression in CRC tissue samples.
Table 1. Comparison of clinicopathological status stratified by LRP6 or β-catenin expression in CRC tissue samples.
Characteristic LRP6 High (n = 29)LRP6 Low (n = 29)p Value
AgeAverage ± SD64.6 ± 11.7266.1 ± 9.520.591
SexMale17200.412
Female129
Lymphatic invasionPositive1790.035
Negative1220
Venous invasionPositive620.253
Negative2327
Lymph node metastasisPositive630.253
Negative2326
Distant metastasisPositive310.612
Negative2628
Degree of differentiation *Well/Mod28281.000
Poor/Muc11
Tumor locationColon14120.598
Rectum1517
Depth of invasion **~mp23220.753
ss~67
Characteristic β-Catenin High (n = 24)β-Catenin Low (n = 34)p Value
AgeAverage ± SD63.2 ± 9.1666.9 ± 11.410.192
SexMale14230.467
Female1011
Lymphatic invasionPositive12140.506
Negative1220
Venous invasionPositive530.255
Negative1931
Lymph node metastasisPositive451.000
Negative2029
Distant metastasisPositive310.297
Negative2133
Degree of differentiation *Well/Mod23331.000
Poor/Muc11
Tumor locationColon10160.684
Rectum1418
Depth of invasion **~mp18270.692
ss~67
* Well: well differentiated adenocarcinoma, Mod: moderately differentiated adenocarcinoma, Poor: poorly differentiated adenocarcinoma, Muc: mucinous carcinoma. ** mp: muscularis propria, ss: subserosa.
Table 2. Comparison of clinicopathological status stratified by LRP6 or β-catenin expression in ESCC tissue samples.
Table 2. Comparison of clinicopathological status stratified by LRP6 or β-catenin expression in ESCC tissue samples.
Characteristic LRP6 High (n = 49)LRP6 Low (n = 19)p Value
AgeAverage ± SD70.0 ± 8.5469.6 ± 8.730.8675
SexMale43130.0606
Female66
T stageT1, T230150.2537
T3, T4194
Lymphatic invasionPositive3490.1803
Negative148
Venous invasionPositive950.2949
Negative3911
Lymph node metastasisPositive3090.300
Negative1910
Distant metastasisPositive201.000
Negative4718
Degree of differentiation *Well/Mod38170.3251
Poor112
Tumor size (max length)mm, Average ± SD47.1 ± 25.8640.5 ± 25.980.4187
Characteristic β-Catenin High (n = 26)β-Catenin Low (n = 42)p Value
AgeAverage ± SD70.7 ± 7.9569.4 ± 8.920.537
SexMale20360.355
Female66
T stageT1, T216290.524
T3, T41013
Lymphatic invasionPositive18250.249
Negative616
Venous invasionPositive770.215
Negative1634
Lymph node metastasisPositive16230.583
Negative1019
Distant metastasisPositive111.000
Negative2441
Degree of differentiation *Well/Mod19360.198
Poor76
Tumor size (max length)mm, Average ± SD51.5 ± 16.5241.9 ± 29.430.133
* Well: well differentiated adenocarcinoma, Mod: moderately differentiated adenocarcinoma, Poor: Poorly differentiated adenocarcinoma.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Shishido, A.; Miyo, M.; Oishi, K.; Nishiyama, N.; Wu, M.; Yamamoto, H.; Kouda, S.; Wu, X.; Shibata, S.; Yokoyama, Y.; et al. The Relationship between LRP6 and Wnt/β-Catenin Pathway in Colorectal and Esophageal Cancer. Life 2023, 13, 615. https://0-doi-org.brum.beds.ac.uk/10.3390/life13030615

AMA Style

Shishido A, Miyo M, Oishi K, Nishiyama N, Wu M, Yamamoto H, Kouda S, Wu X, Shibata S, Yokoyama Y, et al. The Relationship between LRP6 and Wnt/β-Catenin Pathway in Colorectal and Esophageal Cancer. Life. 2023; 13(3):615. https://0-doi-org.brum.beds.ac.uk/10.3390/life13030615

Chicago/Turabian Style

Shishido, Akemi, Masaaki Miyo, Kazuki Oishi, Natsumi Nishiyama, Meiqiao Wu, Hiroyuki Yamamoto, Shihori Kouda, Xin Wu, Satoshi Shibata, Yuhki Yokoyama, and et al. 2023. "The Relationship between LRP6 and Wnt/β-Catenin Pathway in Colorectal and Esophageal Cancer" Life 13, no. 3: 615. https://0-doi-org.brum.beds.ac.uk/10.3390/life13030615

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop