BRAFV600E mutation is the most frequent genetic alteration in papillary thyroid cancer (PTC). β-Catenin (Ctnnb1) is a key downstream component of canonical Wnt signaling pathway and is frequently overexpressed in PTC. BRAFV600E-driven tumors have been speculated to rely on Wnt/β-catenin signaling to sustain its growth, although many details remain to be elucidated. In this study, we investigated the role of β-catenin in BrafV600E-driven thyroid cancer in a transgenic mouse model. In BrafV600E mice with wild-type (WT) Ctnnb1 (BVE-Ctnnb1WT or BVE), overexpression of β-catenin was observed in thyroid tumors. In BrafV600E mice with Ctnnb1 knockout (BVE-Ctnnb1null), thyroid tumor growth was slowed with significant reduction in papillary architecture. This was associated with increased expression of genes involved in thyroid hormone synthesis, elevated 124iodine uptake, and serum T4. The survival of BVE-Ctnnb1null mice was increased by more than 50% during 14-month observation. Mechanistically, downregulation of MAPK, PI3K/Akt, and TGFβ pathways and loss of epithelial–mesenchymal transition (EMT) were demonstrated in the BVE-Ctnnb1null tumors. Treatment with dual β-catenin/KDM4A inhibitor PKF118–310 dramatically improved the sensitivity of BVE-Ctnnb1WT tumor cells to BRAFV600E inhibitor PLX4720, resulting in significant growth arrest and apoptosis in vitro, and tumor regression and differentiation in vivo. These findings indicate that β-catenin signaling plays an important role in thyroid cancer growth and resistance to BRAFV600E inhibitors. Simultaneously targeting both Wnt/β-catenin and MAPK signaling pathways may achieve better therapeutic outcome in BRAFV600E inhibitor-resistant and/or radioiodine-refractory thyroid cancer.

Papillary thyroid cancer (PTC) is the most common type of thyroid cancer, accounting for more than 80% of thyroid cancer cases (1). Although PTC has excellent prognosis with less than 3% mortality at 10 years after treatment, more than 25% of patients developed recurrence during long-term follow-up, which remains a major problem for patients with thyroid cancer (2, 3). In some studies, BRAFV600E mutation has been shown to be one of the factors contributing to thyroid cancer recurrence and mortality (4, 5).

BRAFV600E mutation is the most frequent genetic alteration in PTC occurring in about 45% of cases (6, 7). The mutation constitutively activates the RAS–RAF–MEK–ERK MAPK pathway and promotes initiation and growth of PTC. It has been demonstrated that BrafV600E drives oncogenic transformation of thyroid epithelial cells and development of PTC in transgenic mouse models (8, 9).

The Wnt/β-catenin pathway plays an important role in embryonic development and tissue homeostasis (10). Dysregulation of its signaling is associated with many types of disease including cancer (11). β-Catenin is a key downstream component of both cadherin–catenin cell adhesion complex and Wnt signaling pathway (12, 13). The dual functional protein is involved in the regulation of cadherin-mediated cell–cell adhesion and transcriptional coactivation of Wnt target genes such as MYC (c-Myc), JUN (C-Jun), FOSL1 (Fra-1), CCND1 (cyclin D1), and ABCB1 (multidrug resistance protein 1) via TCF/LEF family of DNA-binding proteins (14, 15).

Aberrant β-catenin expression or localization has been reported in patients with PTC and is associated with c-Myc and cyclin D1 overexpression (16, 17). Frequent activating CTNNB1 mutations and β-catenin nuclear localization have also been reported in the early studies of poorly differentiated or anaplastic thyroid carcinoma (ATC; refs. 18, 19). However, recent studies using the next-generation sequencing are unable to demonstrate frequent CTNNB1 mutations in ATC (20). The functional interactions between β-catenin and MAPK, PI3K/AKT, or CREB (cAMP-response element binding protein) signaling pathways have been shown to promote cell proliferation in thyroid follicular cells and cancer cell lines (21, 22). These early studies suggest that RAS- or BRAF-driven PTC may require Wnt/β-catenin pathway to sustain its growth (23). However, it is not clear to what extent the aberrant β-catenin expression contributes to thyroid cancer development and growth in vivo, especially in the presence of BRAFV600E mutation.

In this study, we used a mouse model of BrafV600E-induced PTC to investigate the role of β-catenin in thyroid cancer growth. We found β-catenin knockout lead to significant reduction in tumor growth, increased survival, and sensitivity to BRAFV600E inhibitor PLX4720.

Reagents

PKF118–310 (β-catenin/KDM4A inhibitor, Catalog No. K4394) was obtained from Sigma-Aldrich (24), and PLX4720 (BRAFV600E inhibitor, Catalog No. S1152) was from Selleck Chemicals (25). Antibodies were purchased from Cell Signaling Technology, Inc.: β-catenin (No. 8480), phospho-Erk ½ (No. 4370), phosphor-Akt (No. 4060), E-cadherin (No. 3195), Slug (No. 9585), Snail (No. 3879), and Vimentin (No. 5741).

Animals

The conditional BrafV600E knock-in and β-catenin knockout mice (Ctnnb1null) were created as described previously (26, 27). Thyroid-specific activation of mutant BrafV600E allele was achieved by crossing with TPO-Cre deleter mice (28, 29). TPO-BrafV600E mice with WT Ctnnb1 (BVE-Ctnnb1WT or BVE) developed PTC at about 5 weeks of age and were used as PTC tumor controls. TPO-BrafWT mice with WT Ctnnb1 were used as normal controls. TPO-BrafV600E mice with thyroid-specific Ctnnb1 knockout (BVE-Ctnnb1null) were obtained by several rounds of breeding among LSL-BrafV600E (26), TPO-Cre (29), and floxed Ctnnb1 mice (27). To knockout Ctnnb1 in the thyroid of BVE mice, floxed Ctnnb1 mice were first crossed with LSL–BrafV600E or TPO–Cre mice to generate floxed Ctnnb1+/−::BrafV600E strain or floxed Ctnnb1+/−::TPO–Cre strain. The floxed Ctnnb1+/−::BrafV600E and floxed Ctnnb1+/−::TPO–Cre mice were then bred together to generate TPO–BrafV600E::Ctnnb1null (BVE-Ctnnb1null) mice, which had thyroid-specific activation of BrafV600E and deletion of β-catenin via Cre-mediated recombination in the thyroid. Mice were housed in autoclaved filter-top cages with autoclaved food and water ad libitum, and maintained on a 12-hour light/12-hour dark cycle (6:00 AM to 6:00 PM). Mice of both sexes were used in the study, and similar results were obtained. The study was approved by the Animal Care and Use Committee of King Faisal Specialist Hospital and Research Centre (RACNo. 2190004) and conducted in compliance with the Guide for the Care and Use of Laboratory Animals (National Research Council).

Genotyping of transgenic mice

Genotyping of Cre-mediated recombination of LSL–BrafV600E mutant allele has been described previously (9, 28). Briefly, primers A (5′-AGTCAATCATCCACAGAGACCT-3′) + C (5′-GCCCAGGCTCTTTATGAGAA-3′) detect both WT Braf (466 bp) and Cre-recombined mutant BrafV600E (518 bp) alleles, respectively. Primers B (5′-GCTTGGCTGGACGTAAACTC-3′) + C detect LSL–BrafV600E allele (140 bp). Primers D (5′-GTTCGCAAGAACCTGATGGACA-3′) and E (5′-CTAGAGCCTGTTTTGCACGTTC-3′) detect Cre gene (350 bp). For genotyping of Cre-mediated recombination of floxed Ctnnb1 allele (27), primers 41 (5′-AAGGTAGAGTGATGAAAGTTGTT-3′) + 42 (5′-CACCATGTCCTCTGTCTATTC-3′) detect both WT (221 bp) and floxed (324 bp) Ctnnb1 alleles, respectively; primers 68 (5′-AATCACAGGGACTTCCATACCAG-3′) + 69 (5′-GCCCAGCCTTAGCCCAACT-3′) detect Cre-recombined deleted Ctnnb1 allele (631 bp). Genomic DNA was isolated from mouse tails or tumor tissues. The PCR conditions were as follows: 94°C for 5 minutes followed by 35 cycles of amplification (94°C for 30 seconds, 58°C for 30 seconds, 72°C for 1 minute) with a final extension at 72°C for 10 minutes.

Histology and IHC

Histology and IHC staining was performed as described previously (9). Briefly, 4-μm-thick formalin-fixed paraffin-embedded tissue sections were prepared and stained with hematoxylin and eosin (H&E) or with β-catenin antibody (1:100 dilution; Rabbit mAb No. 8480, Cell Signaling Technology, Inc). A DAKO LSAB + kit with horseradish peroxidase (HRP) was used for immunostaining (DAKO). The sections were counterstained with Mayer's hematoxylin.

Thyroid-stimulating hormone (TSH) and thyroid hormone (T4) measurements

Blood was collected by cardiac puncture. Serum TSH and T4 were measured using MILLIPLEX MAP Mouse Pituitary Magnetic Bead Panel (MPTMAG-49K) and Multi-Species Hormone Magnetic Bead Panel (MSHMAG-21K), respectively, following the manufacturer's instructions (EMD Millipore Corporation).

Establishment of thyroid cancer cell lines

Thyroid tumors were collected aseptically from donor mice (BVE and BVE-Ctnnb1null) using blunt dissection, mechanically dissociated by mincing and passing through a 40-μM mesh sterile screen, and suspended in DMEM/F12 growth medium (10% FBS, 100 units/mL penicillin, 100 μg/mL streptomycin) as described previously (28). Cells were cultured in DMEM/F12 growth medium containing 4 mU/mL bovine TSH (Sigma-Aldrich). For establishment of immortalized cell lines, cells were cultured continually for more than 6 months and passaged weekly or biweekly at 1:3 ratio with 0.25% trypsin. BVE-Ctnnb1WT (BVE) and BVE-Ctnnb1null cell lines had about 25 and 20 passages, respectively before they were used for experiments. Thyroid origin of the cell lines was confirmed by genotyping to rule out fibroblast contamination. Genotyping of the cell lines and expression of genes for thyroid hormone synthesis were verified every 6 months when the cells were in culture.

RNA sequencing (RNA-seq) for quantification of differentially expressed genes (DEG)

Total RNA from WT control thyroid tissues and thyroid tumors (BVE and BVE-Ctnnb1null) were isolated by TRI Reagent solution (No. T9424, Sigma-Aldrich). Libraries were constructed using an Illumina TruSeq RNA Library Prep Kit according to the manufacturer's procedure. All sequencing was performed on Illumina Hiseq 4000 with at least 20 million clean reads. The significant DEGs were selected on the basis of the following criteria: Log2-fold change > 2, FDR < 0.001, and P value from difference test <0.01.

PET/CT imaging of thyroid uptake of 124iodine (124I)

Mice at 4 months of age were anesthetized with 5% isoflurane mixed with oxygen, and maintained at 1.5% isoflurane during the scan. Mice were then given 5 MBq (megabecquerel) 124I-labeled NaI solution by tail vein injection. Thyroid 124I uptake from WT control, BVE, and BVE-Ctnnb1null mice was measured at different time intervals (30 minutes, 2 hours, and 24 hours) by nanoScan PET/CT (Mediso Medical Imaging Systems).

Western blot analysis

Cell lysates were obtained by extraction in RIPA buffer (20 mmol/L Tris-HCl, pH 7.4, 150 mmol/L NaCl, 5 mmol/L EDTA, 1% NP-40) containing Pierce's Halt Protease Inhibitor Cocktail (Thermo Fisher Scientific). Protein concentration was determined by Bradford's assay using a Bio-Rad Protein Assay Kit (Bio-Rad). Proteins (20–40 μg) were loaded onto a 12% SDS-PAGE and were transferred to a PVDF membrane. Western blot analysis was performed using antibodies (1:1,000 dilution, Cell Signaling Technology, Inc.) against β-catenin (No. 8480), phospho-Erk ½ (No. 4370), phosphor-Akt (No. 4060), E-Cadherin (No. 3195), Slug (No. 9585), Snail (No. 3879), and Vimentin (No. 5741).

Wound-healing assay

BVE-Ctnnb1WT and BVE-Ctnnb1null cells were seeded in 6-well plates (105 cells/well), respectively, and a linear scratch was created with a sterile pipette tip when the cells reached confluent monolayer. The cells were rinsed three times with medium to remove cellular debris. Cell migration or wound-healing were monitored by microscopy after 16-hour culture.

Flow cytometry analysis for apoptosis

BVE-Ctnnb1WT cells were cultured with different concentrations of PKF118–310, PLX4720, or both for 24 hours. The apoptosis was analyzed by flow cytometer using Vybrant Apoptosis Assay Kit (Molecular Probes).

Colony formation assay

BVE-Ctnnb1WT and BVE-Ctnnb1null cells were plated into 12-well plates (5 × 102 cells/well), respectively and cultured for 14 days in the presence of different concentrations of PLX4720, PKF118–310, or both. Cells were then fixed with methanol for 10 minutes and stained with 0.5% crystal violet dye in methanol:de-ionized water (1:5) for 10 minutes. After three washes with de-ionized water to remove excess crystal violet dye, the crystal violet dye was released from cells by incubation with 1% SDS for 2 hours before optical density (OD)570 nm measurement.

In vivo administration of PKF118–310, PLX4720, or combined PKF118–310 and PLX4720

BVE mice at 2 months of age were equally divided into four groups. Group 1 (n = 5) received an intraperitoneal (i.p.) injection of 2.5 μg PKF118–310 (0.4 mg/kg) dissolved in 50-μL phosphate-buffered saline pH7.4 (PBS), three times per week (day 1, 3, 5) for 2 months; group 2 (n = 5) received a daily i.p. injection of PLX4720 (20 mg/kg) for 2 months. PLX4720 was first dissolved in DMSO, followed by PBS (100 μL), which was then injected into mice; group 3 (n = 5) received an alternate i.p. injection of PKF118–310 (0.4 mg/kg, day 1, 3, 5) and PLX4720 (20 mg/kg, day 2, 4, 6) for 2 months; group 4 (n = 5) received an i.p. injection of vehicle (PBS) only. At the end of 2-month treatment, the mice were sacrificed and thyroids were harvested for histology analysis. Kaplan–Meier analysis of survival were performed in BVE-Ctnnb1WT mice with or without treatment (n = 12 in each group).

Statistical analysis

Student t test (two-tailed) was used to compare two groups and one-way ANOVA was used to compare multiple groups. A P value of 0.05 or less was considered significant.

Reduction of thyroid tumor growth in BVE-ctnnb1null mice

We first examined β-catenin (Ctnnb1) expression in normal thyroids from WT control mice (n = 10, pooled RNA) and BrafV600E-induced PTC (n = 3) by RNA-seq analysis. Consistent with previous findings in human PTC, β-catenin expression was increased by more than threefold in both early and late stages of PTC (Fig. 1A, P < 0.001). This was confirmed by IHC of strong cytoplasmic staining (Fig. 1B-b). To investigate the impact of β-catenin overexpression on BrafV600E-mediated thyroid tumorigenesis and growth, we knocked out Ctnnb1 gene by cross-breeding floxed Ctnnb1−/−::BrafV600E with floxed Ctnnb1−/−::TPO–Cre mice. The Ctnnb1 knockout was confirmed by genotyping (Fig. 1C) and absence of β-catenin staining by IHC (Fig. 1D-b). As shown in Fig. 2A-c and d, BVE-Ctnnb1null tumor growth was slowed and tumor was mainly located in the center of thyroid lobe with reduced papillary architecture, whereas BVE-Ctnnb1WT tumor could be seen occupying almost the entire thyroid lobe with dominant papillary architecture at 4 months of age (Fig. 2A-a and b). The tumor weight was significantly reduced: 0.035 ± 0.004 g in BVE-Ctnnb1null versus 0.063 ± 0.008 g in BVE mice (P < 0.05; Fig. 2C). Similar results were observed in the late stage of BVE-Ctnnb1null tumors (10 months of age): 0.106 ± 0.02 g in the BVE-Ctnnb1null versus 0.207 ± 0.04 g in the BVE mice (P < 0.05; Fig. 2C). The reduction in tumor growth was more apparent morphologically in the late stage of BVE-Ctnnb1null tumors: reduction of papillary tumor volume, more follicular architecture, and many lymphocytes and macrophage infiltration for possible tumor clearance (Fig. 2B-c and d) whereas in the BVE mice, the entire thyroid bed was occupied by papillary tumor with few lymphocytes and macrophage infiltration (Fig. 2B-a and b). The bodyweight of BVE-Ctnnb1null mice was increased by 20% (12.82 ± 0.98g, n = 16 at 4 months of age) as compared with BVE mice (10.28 ± 0.66 g, n = 14, P < 0.05), but was still 50% less than WT control mice (24.58 ± 0.76 g, n = 16). Although thyroid function (serum TSH and T4) was improved in the BVE-Ctnnb1null mice, they still had severe hypothyroidism with high level of serum TSH and low level of T4 (Fig. 2D). Taken together, these data indicate Ctnnb1 deletion reduces tumor growth and promotes differentiation, but it is not sufficient to reverse hypothyroidism caused by BrafV600E mutation.

Figure 1.

β-Catenin expression in BVE-Ctnnb1−WT and BVE-Ctnnb1null thyroid tumors. A, Expression of Ctnnb1 (β-catenin) in different age groups of BVE-Ctnnb1−WT thyroid tumors by RNA-seq analysis. Three tumor samples were used for each group and compared with pooled WT control thyroids from WT mice (n = 10). Data are expressed as mean ± SEM in RPKM. B, IHC staining of β-catenin in WT control thyroid (a) and BVE-Ctnnb1−WT thyroid tumor (b). Increased cytoplasmic staining is noted in the tumor tissue (b). C, Genotyping of BVE-Ctnnb1WT and BVE-Ctnnb1null tumors and tumor-derived cell lines. The PCR products were run on a 1.2% agarose gel. Primers A+C detect 466 bp WT Braf and Cre-recombined 518 bp BrafV600E alleles, respectively; primers B+C detect 140 bp LSL–BrafV600E allele; primers D+E detect 350 bp Cre allele; primers 41+42 detect 221 bp WT and 324 bp floxed Ctnnb1 alleles, respectively; primer 68+69 detect Cre-recombined deleted Ctnnb1 allele (631 bp, deletion of exons 2–6). The LSL–BrafV600E allele (140 bp) and floxed Ctnnb1 allele (324 bp) were not detected in the BVE-Ctnnb1null cell line, indicating complete Cre-mediated recombination driven by TPO promoter. D, IHC staining of β-catenin in Ctnnb1 knockout thyroid tumor (BVE-Ctnnb1null). Strong cytoplasmic staining is noted in the invading papillae of BVE-Ctnnb1−WT tumor (a) and absence of β-catenin staining in the BVE-Ctnnb1null tumor (b).

Figure 1.

β-Catenin expression in BVE-Ctnnb1−WT and BVE-Ctnnb1null thyroid tumors. A, Expression of Ctnnb1 (β-catenin) in different age groups of BVE-Ctnnb1−WT thyroid tumors by RNA-seq analysis. Three tumor samples were used for each group and compared with pooled WT control thyroids from WT mice (n = 10). Data are expressed as mean ± SEM in RPKM. B, IHC staining of β-catenin in WT control thyroid (a) and BVE-Ctnnb1−WT thyroid tumor (b). Increased cytoplasmic staining is noted in the tumor tissue (b). C, Genotyping of BVE-Ctnnb1WT and BVE-Ctnnb1null tumors and tumor-derived cell lines. The PCR products were run on a 1.2% agarose gel. Primers A+C detect 466 bp WT Braf and Cre-recombined 518 bp BrafV600E alleles, respectively; primers B+C detect 140 bp LSL–BrafV600E allele; primers D+E detect 350 bp Cre allele; primers 41+42 detect 221 bp WT and 324 bp floxed Ctnnb1 alleles, respectively; primer 68+69 detect Cre-recombined deleted Ctnnb1 allele (631 bp, deletion of exons 2–6). The LSL–BrafV600E allele (140 bp) and floxed Ctnnb1 allele (324 bp) were not detected in the BVE-Ctnnb1null cell line, indicating complete Cre-mediated recombination driven by TPO promoter. D, IHC staining of β-catenin in Ctnnb1 knockout thyroid tumor (BVE-Ctnnb1null). Strong cytoplasmic staining is noted in the invading papillae of BVE-Ctnnb1−WT tumor (a) and absence of β-catenin staining in the BVE-Ctnnb1null tumor (b).

Close modal
Figure 2.

Morphology of thyroid tumor after Ctnnb1 knockout. A, Histology of thyroid tumors in BVE-Ctnnb1WT and BVE-Ctnnb1null mice at 4 months of age. Tumor growth is slower in the BVE-Ctnnb1null (c and d) than BVE-Ctnnb1WT mice (a and b). H&E staining. B, Histology of thyroid tumors in BVE-Ctnnb1WT and BVE-Ctnnb1null mice at 10 months of age. The entire thyroid is occupied by the tumor with papillary architecture from the BVE-Ctnnb1WT mice (a and b), and characteristic nuclear features such as nuclear grooves, nuclear clearing, irregularity, and overlapping nuclei are indicated by an arrow (b). Follicular architecture in the BVE-Ctnnb1null mice (c and d) and many lymphocyte/macrophage infiltration are observed (indicated by an arrow). C, Thyroid tumor weight in BVE-Ctnnb1WT and BVE-Ctnnb1null mice. Thyroid tumor weight was measured from six mice from each group. D, Thyroid function tests in BVE-Ctnnb1WT and BVE-Ctnnb1null mice. Serum TSH and T4 levels were measured from six mice (5 months old) from each group. Data are presented as mean ± SEM. *, indicates statistically significant.

Figure 2.

Morphology of thyroid tumor after Ctnnb1 knockout. A, Histology of thyroid tumors in BVE-Ctnnb1WT and BVE-Ctnnb1null mice at 4 months of age. Tumor growth is slower in the BVE-Ctnnb1null (c and d) than BVE-Ctnnb1WT mice (a and b). H&E staining. B, Histology of thyroid tumors in BVE-Ctnnb1WT and BVE-Ctnnb1null mice at 10 months of age. The entire thyroid is occupied by the tumor with papillary architecture from the BVE-Ctnnb1WT mice (a and b), and characteristic nuclear features such as nuclear grooves, nuclear clearing, irregularity, and overlapping nuclei are indicated by an arrow (b). Follicular architecture in the BVE-Ctnnb1null mice (c and d) and many lymphocyte/macrophage infiltration are observed (indicated by an arrow). C, Thyroid tumor weight in BVE-Ctnnb1WT and BVE-Ctnnb1null mice. Thyroid tumor weight was measured from six mice from each group. D, Thyroid function tests in BVE-Ctnnb1WT and BVE-Ctnnb1null mice. Serum TSH and T4 levels were measured from six mice (5 months old) from each group. Data are presented as mean ± SEM. *, indicates statistically significant.

Close modal

Expression of thyroid hormone synthesis-related genes in normal thyroid, BVE, and BVE-ctnnb1null thyroid tumors

To further assess the effect of Ctnnb1 knockout on the differentiation of thyroid follicular cells, we analyzed the expression of genes involved in the thyroid hormone synthesis from WT control thyroids, BVE and BVE-Ctnnb1null tumors by RNA-seq analysis. As shown in Fig. 3A, the expression of genes involved in the thyroid hormone synthesis were significantly downregulated in the BVE tumors (n = 3) as compared with WT control thyroids (n = 10): 80-fold decrease in reads per kilobase per million reads (RPKM) in Tg (529.22 ± 141.41 vs. 42398.8 ± 1499.36), 14-fold decrease in Tpo (47.56 ± 15.83 vs. 691.94 ± 41.08), twofold decrease in Tshr (4.25 ± 1.33 vs. 10.73 ± 6.3), 70-fold decrease in Duox2 (0.22 ± 0.04 vs. 14.08 ± 2.75), 22-fold decrease in Duoxa2 (1.71 ± 0.4 vs. 38.7 ± 9.07), 56-fold decrease in Slc5a5 (4.8 ± 1.39 vs. 269.79 ± 39.51), 72-fold decrease in Slc5a8 (0.51 ± 0.07 vs. 36.25 ± 0.25), 47-fold decrease in Slc26a7 (2.29 ± 0.37 vs. 108.76 ± 63.31), and sixfold decrease in Iyd (42.89 ± 3.74 vs. 241.3 ± 103.12). The deletion of Ctnnb1 gene resulted in upregulation of these genes in the BVE-Ctnnb1null tumors as compared with the BVE tumors. Slc5a5, the sodium-iodide symporter (NIS) for basolateral iodide transport into thyroid follicular cells, was the most upregulated gene with 18-fold increase in its expression (Fig. 3B; Supplementary Fig. S1). However, Ctnnb1 deletion only partially rescued these downregulated genes up to 40% (Fig. 3B). There are four genes involved in the apical iodide efflux into the follicular lumen: Slc26a7 (30–32), Slc26a4 (33), Slc5a8 (34), and Ano1(35). In contrast to Slc26a7 whose expression was downregulated by 47-fold in the BVE tumors, the expression of Slc26a4 and Ano1 was increased by seven- and fourfold, respectively (Fig. 3C), which may compensate the loss of Slc26a7 function and mitigate the deficiency in apical iodide efflux. Finally, we evaluated thyroid 124I uptake by PET-CT scan to see if elevated Slc5a5 expression correlated with increased iodine uptake. As shown in Fig. 3D, 124I uptake in the BVE-Ctnnb1null mice was comparable with the WT mice and modestly higher than the BVE mice at 30 minutes and 2 hours intervals following tail vein injection of 5 MBq 124I-labeled NaI solution. The high concentration of 124I and large tumor size may contribute to the insignificant difference in the early 124I uptake. At 24 hours interval, 124I uptake was about threefold higher in the BVE-Ctnnb1null mice than that in the BVE mice (0.065 ± 0.005 vs. 0.025 ± 0.005 MBq, n = 3), but was still threefold lower than the WT mice (0.223 ± 0.017, n = 3; Fig. 3D). Thus, 124I measurement at 24 hours is more sensitive to detect deficiency in the iodine uptake. These dada confirm that increased Slc5a5 expression results in elevated iodide uptake in the BVE-Ctnnb1null mice.

Figure 3.

Expression of thyroid hormone synthesis-related genes in WT control thyroids, BVE-Ctnnb1WT and BVE-Ctnnb1null thyroid tumors. Tumors (n = 3) were collected at 4 months of age and gene expression was analyzed by RNA-seq. A, Downregulation of thyroid hormone synthesis-related genes in the BVE-Ctnnb1WT thyroid tumors. B, Comparison of thyroid hormone synthesis-related gene expression in BVE-Ctnnb1WT and BVE-Ctnnb1null thyroid tumors. C, Upregulation of Slc26a4 and Ano1 in BVE-Ctnnb1WT thyroid tumors. D,124I PET/CT scan. Five MBq (megabecquerel) 124I-labeled NaI solution was administered to 4-month-old mice (normal, BVE-Ctnnb1WT, or BVE-Ctnnb1null) by tail vain injection. 124I uptake was measured at different time intervals (30 minutes, 2 hours, and 24 hours) by PET/CT scan. The location of 124I uptake by the thyroid was indicated by an arrow. *, indicates statistically significant. Data are expressed as mean ± SEM.

Figure 3.

Expression of thyroid hormone synthesis-related genes in WT control thyroids, BVE-Ctnnb1WT and BVE-Ctnnb1null thyroid tumors. Tumors (n = 3) were collected at 4 months of age and gene expression was analyzed by RNA-seq. A, Downregulation of thyroid hormone synthesis-related genes in the BVE-Ctnnb1WT thyroid tumors. B, Comparison of thyroid hormone synthesis-related gene expression in BVE-Ctnnb1WT and BVE-Ctnnb1null thyroid tumors. C, Upregulation of Slc26a4 and Ano1 in BVE-Ctnnb1WT thyroid tumors. D,124I PET/CT scan. Five MBq (megabecquerel) 124I-labeled NaI solution was administered to 4-month-old mice (normal, BVE-Ctnnb1WT, or BVE-Ctnnb1null) by tail vain injection. 124I uptake was measured at different time intervals (30 minutes, 2 hours, and 24 hours) by PET/CT scan. The location of 124I uptake by the thyroid was indicated by an arrow. *, indicates statistically significant. Data are expressed as mean ± SEM.

Close modal

Reduction in MAPK, PI3K/akt, and TGFβ signaling pathways in BVE-ctnnb1null thyroid tumors

Increased expression of p-Erk, p-Akt, Snail, Slug, and vimentin is associated with thyroid cancer growth and progression (36, 37). Both Snail and Slug are zinc finger transcription factors and downstream targets of TGFβ, which promotes epithelial–mesenchymal transition (EMT) by downregulating the expression of adhesion molecule E-cadherin (38). Therefore, to investigate the mechanisms leading to thyroid tumor growth inhibition in the BVE-Ctnnb1null mice, we studied the expression of p-Erk, p-Akt, Snail (Snail1), Slug (Snail2), vimentin, and E-cadherin in three BVE-Ctnnb1null tumors and two cell lines established from BVE and BVE-Ctnnb1null thyroid tumors by Western blot analysis. As shown in Fig. 4A, the expression levels of p-Erk and p-Akt were decreased in both BVE-Ctnnb1null tumor samples and cell line, suggesting downregulation of MAPK and PI3K/Akt signaling pathways. As shown in Fig. 4B, both Snail and Slug expression levels were reduced as well. As expected, E-cadherin expression was increased and the expression of mesenchymal cell marker vimentin was decreased in both BVE-Ctnnb1null tumor samples and cell line (Fig. 4B). Furthermore, cell migration was also reduced in the BVE-Ctnnb1null cell line (Fig. 4C). The long-term survival of BVE-Ctnnb1null mice was compared with BVE mice (n = 20 in each group). As shown in Fig. 4D, the survival of BVE-Ctnnb1null mice was significantly increased and more than 50% of mice were still alive after 14-month observation (P < 0.0001). These data demonstrate that active β-catenin signaling is required in BRAFV600E-mediated tumor growth.

Figure 4.

Ctnnb1 knockout leads to downregulation of MAPK, PI3K/Akt, TGFβ signaling pathways and reversal of EMT in BVE-Ctnnb1null tumors and cell line. A, Western blot analysis of β-catenin, p-Erk, and p-AKT protein levels in BVE-Ctnnb1null tumor samples and cell line. The phosphorylation of p-Erk and p-AKT was decreased in the BVE-Ctnnb1null tumor samples and cell line. B, Western blot analysis of vimentin, E-cadherin, Slug, and Snail expression in BVE-Ctnnb1null tumor samples and cell line. Vimentin, Slug, and Snail expression levels were reduced whereas E-cadherin expression was increased in the BVE-Ctnnb1null tumor samples and cell line. C, Wound-healing assay. Cell migration was measured by wound-healing assay. BVE-Ctnnb1WT and BVE-Ctnnb1null cells were seeded in 6-well plates (105 cells/well), respectively, and a linear scratch was created when the cells reached confluent monolayer. The cells were further cultured for 16 hours to observe wound-healing or cell migration. The wound-healing or cell migration is reduced in the BVE-Ctnnb1null tumor cells. D, Kaplan–Meier analysis of survival between BVE-Ctnnb1WT and BVE-Ctnnb1null mice (n = 20 in each group).

Figure 4.

Ctnnb1 knockout leads to downregulation of MAPK, PI3K/Akt, TGFβ signaling pathways and reversal of EMT in BVE-Ctnnb1null tumors and cell line. A, Western blot analysis of β-catenin, p-Erk, and p-AKT protein levels in BVE-Ctnnb1null tumor samples and cell line. The phosphorylation of p-Erk and p-AKT was decreased in the BVE-Ctnnb1null tumor samples and cell line. B, Western blot analysis of vimentin, E-cadherin, Slug, and Snail expression in BVE-Ctnnb1null tumor samples and cell line. Vimentin, Slug, and Snail expression levels were reduced whereas E-cadherin expression was increased in the BVE-Ctnnb1null tumor samples and cell line. C, Wound-healing assay. Cell migration was measured by wound-healing assay. BVE-Ctnnb1WT and BVE-Ctnnb1null cells were seeded in 6-well plates (105 cells/well), respectively, and a linear scratch was created when the cells reached confluent monolayer. The cells were further cultured for 16 hours to observe wound-healing or cell migration. The wound-healing or cell migration is reduced in the BVE-Ctnnb1null tumor cells. D, Kaplan–Meier analysis of survival between BVE-Ctnnb1WT and BVE-Ctnnb1null mice (n = 20 in each group).

Close modal

Antitumor activities of dual β-catenin/KDM4A inhibitor PKF118–310 and BRAFV600E inhibitor PLX4720

To evaluate the sensitivity of tumor cells to PKF118–310 and PLX4720, BVE-Ctnnb1WT and BVE-Ctnnb1null cell lines were used for colony formation assay to measure the long-term effect of PKF118–310 and PLX4720 on cell proliferation. The cells were cultured in different concentrations of PKF118–310 or PLX4720 alone or in combination for 14 days. As shown in Fig. 5A, PKF118–310 alone at 0.5 μmol/L resulted in 24% decrease in cell viability of BVE-Ctnnb1WT cells. However, it significantly enhanced antiproliferative effect of PLX4720, resulting in 73% decrease in cell viability (reduction of cell viability to 27%) after combined treatment of 1 μmol/L PLX4720 and 0.5 μmol/L PKF118–310 versus 16% decrease in cell viability (84% cells still viable) for 1 μmol/L PLX4720 alone or 24% decrease in cell viability (76% cells remain viable) for 0.5 μmol/L PKF118–310 alone (P < 0.0001, Fig. 5A). In the BVE-Ctnnb1null cells, 0.5 μmol/L PKF118–310 treatment resulted in 40% cell death whereas 0.25 μmol/L PLX4720 lead to more than 80% cell death (Fig. 5A, P < 0.0001), indicating that BVE-Ctnnb1null cells became more sensitive to PKF118–310 and PLX4720 inhibitors. Next, we investigated the combined treatment on apoptosis of BVE-Ctnnb1WT cells. As shown in Fig. 5B, apoptosis was found in more than 70% of cells after combined treatment with 2 μmol/L PLX4720 and 1 μmol/L PKF118–310 for 24 hours.

Figure 5.

In vitro effects of β-catenin inhibitor PKF118–310 and BRAFV600E inhibitor PLX4720 on BVE-Ctnnb1WT and BVE-Ctnnb1null tumor cell growth. A, Colony formation assay. Cells were seeded to 12-well plates (5 × 102 cells/well for BVE-Ctnnb1WT and 1.2 × 103 cells/well for BVE-Ctnnb1null) and cultured in the presence of different concentrations of PLX4720, PKF118–310, or both for 14 days. Colony number was quantified by optical density (OD) measurement at 570 nm after straining with crystal violet dye. Cell viability (colony number) is expressed as percentage of vehicle control. *, Statistically significant. B, Annexin V staining of apoptotic cells after treatment of BVE-Ctnnb1WT cells with PLX4720, PKF118–310, or both for 24 hours. Significant apoptosis is observed at the concentration of 2 μmol/L PLX4720 and 1 μmol/L PKF118–310.

Figure 5.

In vitro effects of β-catenin inhibitor PKF118–310 and BRAFV600E inhibitor PLX4720 on BVE-Ctnnb1WT and BVE-Ctnnb1null tumor cell growth. A, Colony formation assay. Cells were seeded to 12-well plates (5 × 102 cells/well for BVE-Ctnnb1WT and 1.2 × 103 cells/well for BVE-Ctnnb1null) and cultured in the presence of different concentrations of PLX4720, PKF118–310, or both for 14 days. Colony number was quantified by optical density (OD) measurement at 570 nm after straining with crystal violet dye. Cell viability (colony number) is expressed as percentage of vehicle control. *, Statistically significant. B, Annexin V staining of apoptotic cells after treatment of BVE-Ctnnb1WT cells with PLX4720, PKF118–310, or both for 24 hours. Significant apoptosis is observed at the concentration of 2 μmol/L PLX4720 and 1 μmol/L PKF118–310.

Close modal

Finally, we investigated PKF118–310, PLX4720, or combined treatment on thyroid tumor growth and differentiation in the BVE mice. As shown in Fig. 6A-b and c, thyroid tumor growth was slowed following PKF118–310 treatment and the effect was similar to PLX4720 treatment: reduction in tumor volume with many lymphocyte and macrophage infiltration. A further reduction in tumor volume and increased differentiation into follicular architecture were demonstrated after combined treatment of PKF118–310 and PLX4720 (Fig. 6A-d), which was comparable to BVE-Ctnnb1null mice treated with PLX4720 (Fig. 6A-f). As shown in Fig. 6B, significant reduction in tumor load was observed in the PLX4720 (0.049 ± 0.004 g, P < 0.05) and PLX4720 + PKF118–310 (0.032 ± 0.005 g, P < 0.01) groups as compared with the BVE control (0.068 ± 0.007 g). Although there was reduction in tumor volume in the PKF118–310 group (0.054 ± 0.008 g), it was not statistically significant, probably due to small sample size. As shown in Fig. 6C and D, the survival of BVE mice was significantly increased following treatment of PKF118–310 (P < 0.05), or PLX4720 + PKF118–310 (P < 0.01). However, the difference in survival between PKF118–310 and PKF118–310 + PLX4720 groups was modest and not statistically significant (P = 0.33). This is probably due to short follow-up duration and small number of animals in each group. Significant difference may be demonstrated if longer follow-up was observed and more animals were used.

Figure 6.

In vivo effects of PKF118–310 and PLX4720 on BVE tumor growth. A, BVE mice were subject to PLX4720 or PKF118–310 or combined PKF118–310 and PLX4720 treatment for 2 months. Thyroid histology by H&E staining: (a) BVE mice without treatment; (b) BVE mice after PLX4720 treatment; (c) BVE mice after PKF118–310 treatment; (d) BVE mice after PKF118–310 + PLX4720 treatment; (e) BVE-Ctnnb1null mice without treatment; (f) BVE-Ctnnb1null mice after PLX4720 treatment. Tumor growth is slowed after treatment (4×) with many lymphocyte and macrophage infiltration (indicated by arrows, 20×). Re-appearance of thyroid follicles and regression of papillary architecture are noted after treatment especially with combined PKF118–310 and PLX4720 treatment in the BVE mice or BVE-Ctnnb1null mice after PLX4720 treatment. B, Thyroid tumor weight after treatment. Thyroid tumor weight was measured from each group (n = 5) and data were presented as mean ± SEM. C, Kaplan–Meier analysis of BVE-Ctnnb1WT mice survival after PKF118–310 treatment (n = 12). D, Kaplan–Meier analysis of BVE-Ctnnb1WT mice survival after combined PKF118–310 and PLX4720 treatment (n = 12).

Figure 6.

In vivo effects of PKF118–310 and PLX4720 on BVE tumor growth. A, BVE mice were subject to PLX4720 or PKF118–310 or combined PKF118–310 and PLX4720 treatment for 2 months. Thyroid histology by H&E staining: (a) BVE mice without treatment; (b) BVE mice after PLX4720 treatment; (c) BVE mice after PKF118–310 treatment; (d) BVE mice after PKF118–310 + PLX4720 treatment; (e) BVE-Ctnnb1null mice without treatment; (f) BVE-Ctnnb1null mice after PLX4720 treatment. Tumor growth is slowed after treatment (4×) with many lymphocyte and macrophage infiltration (indicated by arrows, 20×). Re-appearance of thyroid follicles and regression of papillary architecture are noted after treatment especially with combined PKF118–310 and PLX4720 treatment in the BVE mice or BVE-Ctnnb1null mice after PLX4720 treatment. B, Thyroid tumor weight after treatment. Thyroid tumor weight was measured from each group (n = 5) and data were presented as mean ± SEM. C, Kaplan–Meier analysis of BVE-Ctnnb1WT mice survival after PKF118–310 treatment (n = 12). D, Kaplan–Meier analysis of BVE-Ctnnb1WT mice survival after combined PKF118–310 and PLX4720 treatment (n = 12).

Close modal

In this study, we have shown that β-catenin cooperates with oncogenic BrafV600E to drive thyroid cancer growth. The oncogenic potential of BRAFV600E is significantly reduced after Ctnnb1 knockout, resulting in tumor growth inhibition, increased sensitivity to BRAFV600E inhibitor, elevated iodide uptake, and improved thyroid function. The therapeutic effect of BRAFV600E inhibitor PLX4720 is further enhanced when dual β-catenin/KDM4A inhibitor PKF118–310 is used together.

The poor response to radioiodine ablation therapy after thyroidectomy is a major cause for thyroid cancer recurrence and poor prognosis (39). The loss of radioiodine avidity is due to aberrant silencing of iodide-metabolizing genes such as SLC5A5, TSHR, TPO, and TG in thyroid cancer cells. Mutant BRAFV600E inhibits these genes by constitutive activation of MAPK and TGFβ pathways (40–42). Suppression of MAPK pathway could partially restore expression of iodide-metabolizing genes in BRAFV600E mutant thyroid cancer cells (43). In this study, we have shown that β-catenin is involved in tumor growth and downregulation of iodide-metabolizing genes in BrafV600E-induced thyroid cancer. β-catenin ablation partially reverses aberrant silencing of iodide-metabolizing genes. The effect is probably indirect and mediated through the reduction of p-Erk activation since its activation is significantly reduced following β-catenin ablation. Our data suggest that constitutive BRAFV600E activation depends on active β-catenin signaling. These novel findings may offer a new therapeutic approach of combined β-catenin and BRAFV600E inhibitors in the treatment of iodine-refractory thyroid cancer.

Thyroid Iodide efflux to the follicular lumen via apical membrane for thyroid hormone synthesis is less well known and may require several genes such as SLC26A4 (Pendrin; ref. 44), ANO1 (Anoctamin 1; ref. 35), SLC5A8 (34), and SLC26A7 (30, 31). In our murine model, the expression of Slc5a8 and Slc26a7 is down-regulated as a result of BrafV600E mutation. Interestingly, mRNA levels of Slc26a4 and Ano1 are increased, which may compensate for the functional loss of Slc5a8 and Slc26a7. However, SLC26A7 may be the most important gene in the iodide efflux since its loss-of-function mutations result in severe hypothyroidism which cannot be rescued by normal SLC5A8, ANO1, and SLC26A4 genes (30, 32). It is well-known that SLC5A5 (NIS) is involved in iodide transport into the thyroid follicular cells. Our data have shown that increased Slc5a5 expression leads to elevated iodide uptake in the BVE-Ctnnb1null mice. Iodotyrosine deiodinase (IYD) catalyzes iodide recycling and promotes iodide retention in the thyroid follicular cells. Given that its expression is reduced in both BVE (about 5% of WT control) and BVE-Ctnnb1null tumors (about 12% of WT control) (Fig. 3A and B), reduced iodide recycling may also contribute to decreased iodide uptake in the BVE and to a lesser extent BVE-Ctnnb1null mice. This may explain significant reduction in the iodide uptake in both BVE and BVE-Ctnnb1null tumors as compared with normal thyroid at 24 hours: 124I being excreted instead of recycled back into the thyroid.

The Wnt/β-catenin signaling pathway is frequently dysregulated in cancer. Extensive crosstalk exits between Wnt/β-catenin and MAPK signaling in cancer (45). The outcome between their interactions depend on the specific cellular context. For example, in melanoma, Wnt/β-catenin signaling may act as a tumor suppressor to promote programmed cell death: BRAFV600E downregulates Wnt/β-catenin signaling cascade. Activation of Wnt/β-catenin signaling synergizes BRAFV600E inhibitor PLX4720 to reduce melanoma growth and increase apoptosis via Wnt-mediated reduction of AXIN1 (46). In contrast, Wnt/β-catenin signaling functions as an oncogene in colorectal cancer: its activation leads to increased MAPK signaling through Ras stabilization and drives malignant transformation (47). In the BrafV600E murine thyroid cancer, β-catenin expression is upregulated and its knockout results in downregulation of multiple signaling pathways such as MAPK, PI3K/Akt, and TGFβ, indicating a positive crosstalk between Wnt/β-catenin and these signaling pathways. Furthermore, active Wnt/β-catenin signaling is required for BrafV600E-mediated tumor growth. Sastre-Perona and colleagues have demonstrated activation of β-catenin signaling by HrasG12V but not BrafV600E in PCCl3 rat thyroid follicular cells (22). In our study, BrafV600E is involved in the activation of β-catenin. The use of different model systems (in vitro rat thyroid follicular cells vs. in vivo BrafV600E-driven thyroid cancer) might contribute to the variation. Interestingly, Damsky and colleagues have reported that β-catenin is a central mediator of BrafV600E-driven melanoma metastasis and regulates both MAPK and PI3K/Akt signaling (48).

Resistance to BRAFV600E inhibitors after prolonged treatment is a major challenge in targeted cancer therapy. Activation of Wnt/β-catenin pathway is one of the mechanisms of resistance. Combined inhibition of both Wnt/β-catenin and MAPK signaling pathways has shown synergistic antitumor effects in BRAFV600E-mutant colorectal cancer cell lines (49). This study also demonstrates Wnt/β-catenin pathway plays an important role in conferring resistance to BRAFV600E inhibitor in BrafV600E-mutant thyroid cancer. The resistance could be reduced by the dual β-catenin/KDM4A inhibitor PKF118–310, which was initially identified as an inhibitor of Tcf/β-catenin signaling by a high-throughput screening of natural compounds (24, 50). PKF118–310 interrupts the interaction between β-catenin and Tcf/Lef transcription factors and has been shown to induce apoptosis, cell-cycle arrest, and suppress in vivo tumor growth in several xenograft models (51–53). PKF118–310 has recently been described as an inhibitor of histone lysine demethylase 4A (KDM4A) in epigenetic histone regulation (54). KDM4A is known to function as an oncogene when it is overexpressed (55, 56). A twofold increase in Kdm4a expression was found in both BVE and BVE-Ctnnb1null tumors and cell lines. Ctnnb1 ablation did not affect its expression in the BVE-Ctnnb1null tumors (Supplementary Fig. S2). It is thus possible that Kdm4a overexpression may be involved in the tumor growth of both BVE and BVE-Ctnnb1null mice. The in vitro effects of PKF118–310 on BVE-Ctnnb1null cells (inhibition of cell proliferation) support KDM4A involvement in thyroid cancer growth. The reduction of tumor volume in vivo in the BVE mice may be through its dual inhibition of Tcf/β-catenin signaling and KDM4A-mediate epigenetic modifications. Given its dual inhibition functions and few KDM4-selective inhibitors available, this tool drug may be a good candidate for further development into a cancer therapeutic drug.

In summary, we have demonstrated Wnt/β-catenin signaling pathway plays an important role in thyroid cancer growth and differentiation. Dual β-catenin/KDM4A inhibitor PKF118–310 significantly increases the antitumor activity of BRAFV600E inhibitor PLX4720 leading to prolonged survival of BrafV600E-mutant thyroid cancer. Targeting both MAPK and Wnt/β-catenin pathways may have significant therapeutic benefit for BRAFV600E inhibitor-resistant and/or radioiodine-refractory thyroid cancer.

No disclosures were reported.

M. Zou: Conceptualization, data curation, formal analysis, validation, investigation, visualization, methodology, writing–original draft, writing–review and editing. H.A. BinEssa: Formal analysis, validation, investigation, visualization, methodology, writing–review and editing. Y.H. Al-Malki: Data curation, investigation, methodology, writing–review and editing. S. Al-Yahya: Data curation, investigation, methodology. M. Al-Alwan: Data curation, investigation, methodology, writing–review and editing. I. Al-Jammaz: Data curation, investigation, methodology. K.S.A. Khabar: Data curation, investigation, writing–review and editing. F. Almohanna: Data curation, investigation, project administration. A.M. Assiri: Resources, data curation, project administration. B.F. Meyer: Resources, supervision, investigation. A.S. Alzahrani: Conceptualization, resources, supervision, writing–review and editing. F.A. Al-Mohanna: Conceptualization, resources, investigation, writing–review and editing. Y. Shi: Conceptualization, supervision, funding acquisition, validation, investigation, methodology, writing–original draft, writing–review and editing.

We would like to thank Ms. Roua A. Al-Rijjal, Ms. Anwar F. Al-Enezi, and Mr. Wilfredo Antiquera for excellent technical support; Drs. Shioko Kimura, Catrin Pritchard, and Rolf Kemler for generous gifts of TPO-Cre, LSL-BrafV600E, and floxed Ctnnb1 mice, respectively; and Mr. Cong Li and Kai Huang from BGI for bioinformatics service. This study was supported by KACST grant 13-MED1765-20 to Y. Shi.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Mao
Y
,
Xing
M
. 
Recent incidences and differential trends of thyroid cancer in the USA
.
Endocr Relat Cancer
2016
;
23
:
313
22
.
2.
Grogan
RH
,
Kaplan
SP
,
Cao
H
,
Weiss
RE
,
Degroot
LJ
,
Simon
CA
, et al
A study of recurrence and death from papillary thyroid cancer with 27 years of median follow-up
.
Surgery
2013
;
154
:
1436
46
;
discussion 46–7
.
3.
Dong
W
,
Horiuchi
K
,
Tokumitsu
H
,
Sakamoto
A
,
Noguchi
E
,
Ueda
Y
, et al
Time-varying pattern of mortality and recurrence from papillary thyroid cancer: lessons from a long-term follow-up
.
Thyroid
2019
;
29
:
802
8
.
4.
Xing
M
,
Alzahrani
AS
,
Carson
KA
,
Shong
YK
,
Kim
TY
,
Viola
D
, et al
Association between BRAF V600E mutation and recurrence of papillary thyroid cancer
.
J Clin Oncol
2015
;
33
:
42
50
.
5.
Xing
M
,
Alzahrani
AS
,
Carson
KA
,
Viola
D
,
Elisei
R
,
Bendlova
B
, et al
Association between BRAF V600E mutation and mortality in patients with papillary thyroid cancer
.
JAMA
2013
;
309
:
1493
501
.
6.
Xing
M
. 
BRAF mutation in thyroid cancer
.
Endocr Relat Cancer
2005
;
12
:
245
62
.
7.
Xing
M
. 
Molecular pathogenesis and mechanisms of thyroid cancer
.
Nat Rev Cancer
2013
;
13
:
184
99
.
8.
Knauf
JA
,
Ma
X
,
Smith
EP
,
Zhang
L
,
Mitsutake
N
,
Liao
XH
, et al
Targeted expression of BRAFV600E in thyroid cells of transgenic mice results in papillary thyroid cancers that undergo dedifferentiation
.
Cancer Res
2005
;
65
:
4238
45
.
9.
Zou
M
,
Baitei
EY
,
BinEssa
HA
,
Al-Mohanna
FA
,
Parhar
RS
,
St-Arnaud
R
, et al
Cyp24a1 attenuation limits progression of Braf(V600E) -induced papillary thyroid cancer cells and sensitizes them to BRAF(V600E) inhibitor PLX4720
.
Cancer Res
2017
;
77
:
2161
72
.
10.
MacDonald
BT
,
Tamai
K
,
He
X
. 
Wnt/beta-catenin signaling: components, mechanisms, and diseases
.
Dev Cell
2009
;
17
:
9
26
.
11.
Clevers
H
. 
Wnt/beta-catenin signaling in development and disease
.
Cell
2006
;
127
:
469
80
.
12.
Valenta
T
,
Hausmann
G
,
Basler
K
. 
The many faces and functions of beta-catenin
.
Embo J
2012
;
31
:
2714
36
.
13.
Moon
RT
,
Bowerman
B
,
Boutros
M
,
Perrimon
N
. 
The promise and perils of Wnt signaling through beta-catenin
.
Science
2002
;
296
:
1644
6
.
14.
Kim
JH
,
Park
SY
,
Jun
Y
,
Kim
JY
,
Nam
JS
. 
Roles of Wnt target genes in the journey of cancer stem cells
.
Int J Mol Sci
2017
;
18
:
1604
.
15.
Lien
WH
,
Fuchs
E
. 
Wnt some lose some: transcriptional governance of stem cells by Wnt/beta-catenin signaling
.
Genes Dev
2014
;
28
:
1517
32
.
16.
Ishigaki
K
,
Namba
H
,
Nakashima
M
,
Nakayama
T
,
Mitsutake
N
,
Hayashi
T
, et al
Aberrant localization of beta-catenin correlates with overexpression of its target gene in human papillary thyroid cancer
.
J Clin Endocrinol Metab
2002
;
87
:
3433
40
.
17.
Rezk
S
,
Brynes
RK
,
Nelson
V
,
Thein
M
,
Patwardhan
N
,
Fischer
A
, et al
beta-Catenin expression in thyroid follicular lesions: potential role in nuclear envelope changes in papillary carcinomas
.
Endocr Pathol
2004
;
15
:
329
37
.
18.
Garcia-Rostan
G
,
Tallini
G
,
Herrero
A
,
D'Aquila
TG
,
Carcangiu
ML
,
Rimm
DL
. 
Frequent mutation and nuclear localization of beta-catenin in anaplastic thyroid carcinoma
.
Cancer Res
1999
;
59
:
1811
5
.
19.
Garcia-Rostan
G
,
Camp
RL
,
Herrero
A
,
Carcangiu
ML
,
Rimm
DL
,
Tallini
G
. 
Beta-catenin dysregulation in thyroid neoplasms: down-regulation, aberrant nuclear expression, and CTNNB1 exon 3 mutations are markers for aggressive tumor phenotypes and poor prognosis
.
Am J Pathol
2001
;
158
:
987
96
.
20.
Landa
I
,
Ibrahimpasic
T
,
Boucai
L
,
Sinha
R
,
Knauf
JA
,
Shah
RH
, et al
Genomic and transcriptomic hallmarks of poorly differentiated and anaplastic thyroid cancers
.
J Clin Invest
2016
;
126
:
1052
66
.
21.
Castellone
MD
,
De Falco
V
,
Rao
DM
,
Bellelli
R
,
Muthu
M
,
Basolo
F
, et al
The beta-catenin axis integrates multiple signals downstream from RET/papillary thyroid carcinoma leading to cell proliferation
.
Cancer Res
2009
;
69
:
1867
76
.
22.
Sastre-Perona
A
,
Riesco-Eizaguirre
G
,
Zaballos
MA
,
Santisteban
P
. 
beta-catenin signaling is required for RAS-driven thyroid cancer through PI3K activation
.
Oncotarget
2016
;
7
:
49435
49
.
23.
Ely
KA
,
Bischoff
LA
,
Weiss
VL
. 
Wnt Signaling in Thyroid Homeostasis and Carcinogenesis
.
Genes
2018
;
9
:
204
.
24.
Lepourcelet
M
,
Chen
YN
,
France
DS
,
Wang
H
,
Crews
P
,
Petersen
F
, et al
Small-molecule antagonists of the oncogenic Tcf/beta-catenin protein complex
.
Cancer Cell
2004
;
5
:
91
102
.
25.
Tsai
J
,
Lee
JT
,
Wang
W
,
Zhang
J
,
Cho
H
,
Mamo
S
, et al
Discovery of a selective inhibitor of oncogenic B-Raf kinase with potent antimelanoma activity
.
Proc Natl Acad Sci U S A
2008
;
105
:
3041
6
.
26.
Mercer
K
,
Giblett
S
,
Green
S
,
Lloyd
D
,
DaRocha Dias
S
,
Plumb
M
, et al
Expression of endogenous oncogenic V600EB-raf induces proliferation and developmental defects in mice and transformation of primary fibroblasts
.
Cancer Res
2005
;
65
:
11493
500
.
27.
Brault
V
,
Moore
R
,
Kutsch
S
,
Ishibashi
M
,
Rowitch
DH
,
McMahon
AP
, et al
Inactivation of the beta-catenin gene by Wnt1-Cre-mediated deletion results in dramatic brain malformation and failure of craniofacial development
.
Development
2001
;
128
:
1253
64
.
28.
Zou
M
,
Baitei
EY
,
Al-Rijjal
RA
,
Parhar
RS
,
Al-Mohanna
FA
,
Kimura
S
, et al
TSH overcomes Braf(V600E)-induced senescence to promote tumor progression via downregulation of p53 expression in papillary thyroid cancer
.
Oncogene
2016
;
35
:
1909
18
.
29.
Kusakabe
T
,
Kawaguchi
A
,
Kawaguchi
R
,
Feigenbaum
L
,
Kimura
S
. 
Thyrocyte-specific expression of Cre recombinase in transgenic mice
.
Genesis
2004
;
39
:
212
6
.
30.
Zou
M
,
Alzahrani
AS
,
Al-Odaib
A
,
Alqahtani
MA
,
Babiker
O
,
Al-Rijjal
RA
, et al
Molecular analysis of congenital hypothyroidism in Saudi Arabia: SLC26A7 mutation is a novel defect in thyroid dyshormonogenesis
.
J Clin Endocrinol Metab
2018
;
103
:
1889
98
.
31.
Ishii
J
,
Suzuki
A
,
Kimura
T
,
Tateyama
M
,
Tanaka
T
,
Yazawa
T
, et al
Congenital goitrous hypothyroidism is caused by dysfunction of the iodide transporter SLC26A7
.
Commun Biol
2019
;
2
:
270
.
32.
Cangul
H
,
Liao
XH
,
Schoenmakers
E
,
Kero
J
,
Barone
S
,
Srichomkwun
P
, et al
Homozygous loss-of-function mutations in SLC26A7 cause goitrous congenital hypothyroidism
.
JCI Insight
2018
;
3
.
99631
.
33.
Bizhanova
A
,
Kopp
P
. 
Minireview: the sodium-iodide symporter NIS and pendrin in iodide homeostasis of the thyroid
.
Endocrinology
2009
;
150
:
1084
90
.
34.
Rodriguez
AM
,
Perron
B
,
Lacroix
L
,
Caillou
B
,
Leblanc
G
,
Schlumberger
M
, et al
Identification and characterization of a putative human iodide transporter located at the apical membrane of thyrocytes
.
J Clin Endocrinol Metab
2002
;
87
:
3500
3
.
35.
Twyffels
L
,
Strickaert
A
,
Virreira
M
,
Massart
C
,
Van Sande
J
,
Wauquier
C
, et al
Anoctamin-1/TMEM16A is the major apical iodide channel of the thyrocyte
.
Am J Physiol Cell Physiol
2014
;
307
:
C1102
12
.
36.
Knauf
JA
,
Sartor
MA
,
Medvedovic
M
,
Lundsmith
E
,
Ryder
M
,
Salzano
M
, et al
Progression of BRAF-induced thyroid cancer is associated with epithelial-mesenchymal transition requiring concomitant MAP kinase and TGFbeta signaling
.
Oncogene
2011
;
30
:
3153
62
.
37.
Liu
Z
,
Hou
P
,
Ji
M
,
Guan
H
,
Studeman
K
,
Jensen
K
, et al
Highly prevalent genetic alterations in receptor tyrosine kinases and phosphatidylinositol 3-kinase/akt and mitogen-activated protein kinase pathways in anaplastic and follicular thyroid cancers
.
J Clin Endocrinol Metab
2008
;
93
:
3106
16
.
38.
Hardy
RG
,
Vicente-Duenas
C
,
Gonzalez-Herrero
I
,
Anderson
C
,
Flores
T
,
Hughes
S
, et al
Snail family transcription factors are implicated in thyroid carcinogenesis
.
Am J Pathol
2007
;
171
:
1037
46
.
39.
Aashiq
M
,
Silverman
DA
,
Na'ara
S
,
Takahashi
H
,
Amit
M
. 
Radioiodine-refractory thyroid cancer: molecular basis of redifferentiation therapies, management, and novel therapies
.
Cancers
2019
;
11
:
1382
.
40.
Durante
C
,
Puxeddu
E
,
Ferretti
E
,
Morisi
R
,
Moretti
S
,
Bruno
R
, et al
BRAF mutations in papillary thyroid carcinomas inhibit genes involved in iodine metabolism
.
J Clin Endocrinol Metab
2007
;
92
:
2840
3
.
41.
Romei
C
,
Ciampi
R
,
Faviana
P
,
Agate
L
,
Molinaro
E
,
Bottici
V
, et al
BRAFV600E mutation, but not RET/PTC rearrangements, is correlated with a lower expression of both thyroperoxidase and sodium iodide symporter genes in papillary thyroid cancer
.
Endocr Relat Cancer
2008
;
15
:
511
20
.
42.
Riesco-Eizaguirre
G
,
Rodriguez
I
,
De la Vieja
A
,
Costamagna
E
,
Carrasco
N
,
Nistal
M
, et al
The BRAFV600E oncogene induces transforming growth factor beta secretion leading to sodium iodide symporter repression and increased malignancy in thyroid cancer
.
Cancer Res
2009
;
69
:
8317
25
.
43.
Liu
D
,
Hu
S
,
Hou
P
,
Jiang
D
,
Condouris
S
,
Xing
M
. 
Suppression of BRAF/MEK/MAP kinase pathway restores expression of iodide-metabolizing genes in thyroid cells expressing the V600E BRAF mutant
.
Clin Cancer Res
2007
;
13
:
1341
9
.
44.
Gillam
MP
,
Sidhaye
AR
,
Lee
EJ
,
Rutishauser
J
,
Stephan
CW
,
Kopp
P
. 
Functional characterization of pendrin in a polarized cell system. Evidence for pendrin-mediated apical iodide efflux
.
J Biol Chem
2004
;
279
:
13004
10
.
45.
Guardavaccaro
D
,
Clevers
H
. 
Wnt/beta-catenin and MAPK signaling: allies and enemies in different battlefields
.
Sci Signal
2012
;
5
:
pe15
.
46.
Biechele
TL
,
Kulikauskas
RM
,
Toroni
RA
,
Lucero
OM
,
Swift
RD
,
James
RG
, et al
Wnt/beta-catenin signaling and AXIN1 regulate apoptosis triggered by inhibition of the mutant kinase BRAFV600E in human melanoma
.
Sci Signal
2012
;
5
:
ra3
.
47.
Jeong
WJ
,
Yoon
J
,
Park
JC
,
Lee
SH
,
Lee
SH
,
Kaduwal
S
, et al
Ras stabilization through aberrant activation of Wnt/beta-catenin signaling promotes intestinal tumorigenesis
.
Sci Signal
2012
;
5
:
ra30
.
48.
Damsky
WE
,
Curley
DP
,
Santhanakrishnan
M
,
Rosenbaum
LE
,
Platt
JT
,
Rothberg
BEG
, et al
beta-catenin signaling controls metastasis in Braf-activated Pten-deficient melanomas
.
Cancer Cell
2011
;
20
:
741
54
.
49.
Chen
G
,
Gao
C
,
Gao
X
,
Zhang
DH
,
Kuan
SF
,
Burns
TF
, et al
Wnt/beta-catenin pathway activation mediates adaptive resistance to BRAF inhibition in colorectal cancer
.
Mol Cancer Ther
2018
;
17
:
806
13
.
50.
Leow
PC
,
Tian
Q
,
Ong
ZY
,
Yang
Z
,
Ee
PL
. 
Antitumor activity of natural compounds, curcumin and PKF118–310, as Wnt/beta-catenin antagonists against human osteosarcoma cells
.
Invest New Drugs
2010
;
28
:
766
82
.
51.
Wei
W
,
Chua
MS
,
Grepper
S
,
So
S
. 
Small molecule antagonists of Tcf4/beta-catenin complex inhibit the growth of HCC cells in vitro and in vivo
.
Int J Cancer
2010
;
126
:
2426
36
.
52.
Hallett
RM
,
Kondratyev
MK
,
Giacomelli
AO
,
Nixon
AM
,
Girgis-Gabardo
A
,
Ilieva
D
, et al
Small molecule antagonists of the Wnt/beta-catenin signaling pathway target breast tumor-initiating cells in a Her2/Neu mouse model of breast cancer
.
PLoS One
2012
;
7
:
e33976
.
53.
Zeng
S
,
Seifert
AM
,
Zhang
JQ
,
Cavnar
MJ
,
Kim
TS
,
Balachandran
VP
, et al
Wnt/beta-catenin signaling contributes to tumor malignancy and is targetable in gastrointestinal stromal tumor
.
Mol Cancer Ther
2017
;
16
:
1954
66
.
54.
Franci
G
,
Sarno
F
,
Nebbioso
A
,
Altucci
L
. 
Identification and characterization of PKF118–310 as a KDM4A inhibitor
.
Epigenetics
2017
;
12
:
198
205
.
55.
Cloos
PA
,
Christensen
J
,
Agger
K
,
Maiolica
A
,
Rappsilber
J
,
Antal
T
, et al
The putative oncogene GASC1 demethylates tri- and dimethylated lysine 9 on histone H3
.
Nature
2006
;
442
:
307
11
.
56.
Lee
DH
,
Kim
GW
,
Jeon
YH
,
Yoo
J
,
Lee
SW
,
Kwon
SH
. 
Advances in histone demethylase KDM4 as cancer therapeutic targets
.
Faseb J
2020
;
34
:
3461
84
.