Open Access

Cadherin‑16 inhibits thyroid carcinoma cell proliferation and invasion

  • Authors:
    • Xiaolin Yang
    • Yukun Li
    • Geling Liu
    • Weina Zha
    • Ying Liu
  • View Affiliations

  • Published online on: March 15, 2022     https://doi.org/10.3892/ol.2022.13265
  • Article Number: 145
  • Copyright: © Yang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Cadherin‑16 (CDH16), a member of the cadherin family of adhesion molecules, serves an important role in the formation and maintenance of the thyroid follicular lumen. Decreased expression of CDH16 has been reported to be associated with tumor stage in papillary thyroid cancer (PTC); however, previous analyses have been limited and the biological role of CDH16 in different subtypes of TC is unknown. To investigate the role of CDH16 in the occurrence and development of TC, bioinformatic analysis of three TC subtypes (PTC, follicular cell‑derived TC and anaplastic TC) was performed using an extended data set from the Gene Expression Omnibus database, with additional confirmation using data from The Cancer Genome Atlas, as well as biopsies from 35 patients with PTC and TC or follicular cell lines. According to the dataset analysis, CDH16 was downregulated in PTC and follicular cell‑derived and anaplastic TC; the downregulation in PTC was independent of DNA copy number variation. Furthermore, low expression levels of CDH16 were significantly correlated with tumor size, lymph node metastasis status and disease stage in 35 patients with PTC. Gene Set Enrichment Analysis suggested that CDH16 participated in DNA replication and cell adhesion pathways. To evaluate CDH16 activity, CDH16 was overexpressed in TC‑derived BCPAP cells. CDH16 overexpression inhibited cell proliferation, migration and invasion and induced apoptosis by downregulating proteins associated with DNA replication and cell adhesion. These results support the identification of CDH16 as a valuable target for TC prognosis and therapy and, to the best of our knowledge, represent the first direct demonstration of its mechanistic role in TC.

Introduction

Thyroid carcinoma (TC) is the most common endocrine malignancy (1), with increasing incidence worldwide (24). Epidemiological data from China show that the age standardized incidence rate of thyroid cancer increased from 3.21/105 in 2005 to 9.61/105 in 2015, with an annual increasing incidence rate of 12.4% (5). There are four primary types of TC according to the cellular derivation and state of differentiation (6): Papillary thyroid cancer (PTC), follicular thyroid cancer (FTC) and anaplastic thyroid cancer (ATC) arise from follicular cells of the thyroid, while medullary thyroid cancer (MTC) arises from parafollicular C cells of the thyroid. Although PTC and FTC, the most common forms of TC, usually have good prognoses (7,8), ~10% of patients suffer disease recurrence and metastasis following treatment (9). Furthermore, MTC and ATC have worse prognoses due to their aggressive behavior (10), with a 1-year survival rate of 5–30% for ATC (11). A major challenge in treatment of TC is failure to respond to radiotherapy and chemotherapy (8,12). Therefore, it is urgent to identify new biomarkers and therapeutic targets.

Cadherins (CDHs) are members of a family of homozygous and Ca2+-dependent cell adhesion glycoproteins, which perform essential roles in embryonic development, normal cell function and tissue integrity preservation by mediating cell-cell adhesion (13). Abnormal expression of CDHs has been reported in carcinogenesis and is associated with tumor initiation and progression (14). Notably, CDH16 has been implicated in differentiation of the kidney (15,16) and thyroid (1719). Furthermore, decreased CDH16 has been observed in renal cell carcinoma (16) and studies suggest that CDH16 may be downregulated in TC (20,21). In one study (21), downregulation of CDH16 was shown to be correlated with unfavorable clinicopathological features of PTC and was inversely associated with expression of cancer-associated genes; however, the analysis did not distinguish between TC subtypes and did not consider the potential contribution of gene copy number variation (CNV). Furthermore, bioinformatic analysis was limited to a single cohort in The Cancer Genome Atlas (TCGA) database and direct expression analysis was limited to a single 16 patient cohort. Though the results of the previous studies (20,21) are consistent with a tumor suppressor role of CDH16, the biological activity of CDH16 in TC has not been directly demonstrated and its role in TC remains hypothetical.

In the present study, the expression and CNV of CDH16 in TC was investigated. Functional assays were also performed to probe its role and molecular mechanism in TC. These studies provide a molecular basis for the role of CDH16 in TC, as well as confirming the potential of CDH16 as a target for TC prognosis and treatment.

Materials and methods

Database analysis

mRNA expression and DNA CN of CDH16 in TC were investigated using the Oncomine 4.5 database (oncomine.org), an integrated data mining platform for collecting, analyzing and delivering cancer transcriptome data (22). The present study analysis focused on four TC studies, including He (accession no. GSE3467), Vasko (accession no. GSE6004), Giordano (accession no. GSE27155) and TCGA thyroid (2325). The CDH16 mRNA expression and DNA CN were assessed between TC and normal thyroid tissue. P<0.05 was considered to indicate a statistically significant difference.

LinkedOmics database (linkedomics.org/login.php), a publicly available portal analyzing multi-omics data from 32 types of cancer in TCGA (26), was used to visualize genes that were co-expressed with CDH16 and perform KEGG pathway analysis of these genes by Gene Set Enrichment Analysis (GSEA).

Tissue specimens and cell culture

A total of 35 paired PTC surgical and corresponding adjacent normal thyroid specimens were obtained from Tangshan Gongren Hospital and Tangshan Renmin Hospital between January 2017 and November 2021. The inclusion criteria were: i) histologically confirmed PTC; ii) voluntary participation in the research; iii) no preoperative radiotherapy, chemotherapy or other treatment for TC. The exclusion criteria were: i) presence of tumor other than TC; ii) heart, lung, liver, kidney or hematopoietic system disease; iii) pregnant or lactating. Patient characteristics are presented in Table I. The samples were stored in liquid nitrogen for reverse transcription-quantitative (RT-q)PCR or fixed in 4% paraformaldehyde solution for 48 h at room temperature and embedded in paraffin for immunohistochemistry (IHC). The present study was approved by the Ethical Committee of Tangshan Gongren Hospital (Hebei, China; approval no. GRYY-LL-2020-104) and all patients provided written informed consent.

Table I.

Association of CDH16 protein expression with clinicopathological features of 35 patients with papillary thyroid cancer.

Table I.

Association of CDH16 protein expression with clinicopathological features of 35 patients with papillary thyroid cancer.

CDH16 expression

Characteristicn+-%P-value
Sex
  Male104660.00.471a
  Female25141144.0
Age, years
  <552011945.00.625
  ≥55157853.3
Tumor size, cm
  ≥21111090.90.001
  <22417729.2
Lymph node metastasis
  Present1851372.20.004
  Absent1713423.5
Disease stage
  I, II28171139.30.041a
  III, IV71685.7

a P-value calculated by Fisher's exact test. +, CDH16-positive expression; -, CDH16-negative expression.

Human TC BCPAP and TPC1 cells were obtained from American Type Culture Collection and the human thyroid follicular cell line Nthy-ori3-1 was purchased from Shanghai Institute of Cell Biology of the Chinese Academy of Sciences (Shanghai, China). BCPAP, TPC1 and Nthy-ori3-1 cells were cultured in RPMI-1640 (Gibco; Thermo Fisher Scientific, Inc.) containing 10% fetal calf serum (Gibco; Thermo Fisher Scientific, Inc.). All cell lines were cultured in a 5% CO2 humidified incubator at 37°C.

IHC staining

Tissue fixation, paraffin embedding, sectioning and dewaxing were performed as previously described (27). Tissue samples were incubated with mouse anti-CDH16 (1:100; cat. no. ab215769; Abcam) overnight at 4°C. Subsequently, sections were incubated with horseradish peroxidase (HRP)-conjugated goat anti-mouse IgG secondary antibody (1:2,000; cat. no. ab205719; Abcam) for 30 min at 37°C. The slides were examined by two blinded senior pathologists from Tangshan Gongren Hospital. Cells within five randomly selected fields of view were counted under a light microscope (magnification, ×20) and those with brown or yellow staining on the cell membrane were considered CDH16-positive. The percentage of positive tumor cells was calculated. A threshold of >25% positively stained tumor cells was used to define CDH16 positivity.

RT-qPCR

RNA was isolated from thyroid tissue samples or cells using TRIzol® (Invitrogen; Thermo Fisher Scientific, Inc.). RNA was reverse transcribed into cDNA using cDNA Synthesis kit (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. The prepared cDNA was subjected to RT-qPCR using a SYBR Green PCR Supermix kit (Invitrogen; Thermo Fisher Scientific, Inc.) with the Rotor Gene-3000 instrument (Corbett Research Ltd.). Reactions were conducted in a 20 µl volume with 1 µl cDNA according to the manufacturer's protocol of the SYBR Green PCR Supermix kit. Primer sequences for CDH16 were forward, 5′-CCCTGAGTTCATCACTTCCC-3′ and reverse, 5′-AGAGTCTGGCTCCCAATCC-3′. Primer sequences for GAPDH were forward, 5′- GAAAGCCTGCCGGTGACTAA-3′ and reverse, 5′-AGGAAAAGCATCACCCGGAG-3′. The PCR protocol was as follows: Initial denaturation at 95°C for 2 min, followed by 45 cycles of 95°C for 15 sec and 60°C for 30 sec. Relative expression was calculated using the 2−ΔΔCq method (28) with GAPDH as a reference gene for normalization.

Western blot analysis

Protein was isolated from cells using RIPA buffer (Beyotime Institute of Biotechnology) with protease inhibitor phenylmethylsulfonyl fluoride (1:200; Beyotime Institute of Biotechnology). Total protein concentration was determined using a BCA kit (Beyotime Institute of Biotechnology), according to the manufacturer's protocol. Aliquots of 25 µg protein were electrophoresed on 10% polyacrylamide gels and transferred onto polyvinylidene fluoride membranes. Membranes were blocked in 5% non-fat milk in TBS containing 0.05% Tween-20 (TBST) for 2 h at room temperature. The membranes were incubated overnight at 4°C with primary antibodies against CDH16 (1:1,000, cat. no. 15107-1-AP, ProteinTech Group, Inc.), DNA polymerase (POL)D1 (1:1,000, cat. no. 15646-1-AP, ProteinTech Group, Inc), minichromosome maintenance (MCM)6 (1:2,000, cat. no. 13347-2-AP; ProteinTech Group, Inc), claudin (CLDN)1 (1:1,500, cat. no. 13050-1-AP, ProteinTech Group, Inc), intercellular adhesion molecule (ICAM)1 (1:2,000, cat. no. 60299-1-Ig, ProteinTech Group, Inc), syndecan (SDC)4 (1:1,000, cat. no. 11820-1-AP, ProteinTech Group, Inc) or β-actin (1:1,000, cat. no. TA-09; OriGene Technologies, Inc.). The membranes were washed with PBS three times and subsequently probed with anti-rabbit (cat. no. ZB-2301) or anti-mouse horseradish peroxidase-linked secondary antibody (both 1:2500; cat. no. ZB-2305) for 60 min at room temperature and visualized with ECL reagent (cat. no. sc-2048; all OriGene Technologies, Inc.). The protein bands were scanned and quantified by Tanon Gis software v4.2 (Tanon Science and Technology Co., Ltd.). Expression of β-actin served as the loading control.

Gene transfection

A full length CDH16 cDNA clone was chemically synthesized by Sino Biological, Inc. and ligated into pCMV3 vector (Sino Biological, Inc.). Empty pCMV3 vector was used as the negative control. Untransfected cells were used as the blank control group. To introduce the vector, BCPAP cells were seeded in 6-well plates at a density of 5×105 cells per well. After incubation at 37°C overnight, the degree of cell fusion was 50–70%. The cells were transiently transfected with plasmid carrying CDH16 or negative control plasmid (both 4 µg/well) using TransIntro EL (TransGen Biotech Co., Ltd.) according to the manufacturer's instructions. The effect of pCMV3-CDH16 on CDH16 mRNA and protein expression was analyzed by RT-qPCR and western blotting, as aforementioned, at 48 h post-transfection.

MTT analysis

Cells (1×104 per well) were seeded in 96-well plates. At 0, 24, 48, 72, and 96 h post-transfection, cell viability was determined by adding 10 µl 5 mg/ml MTT solution (Sigma-Aldrich; Merck KGaA) to each well and incubating the samples for 4 h at 37°C. The cell culture medium was removed and 150 µl DMSO (Sigma-Aldrich; Merck KGaA) was added to dissolve the formazan crystals. The absorbance at 490 nm was measured using a microplate reader (Bio-Rad Laboratories, Inc.). The experiment was repeated three times.

Flow cytometry analysis

Effect of CDH16 on BCPAP cell apoptosis was determined by flow cytometry. Cells (5×105 per well) were seeded in 6-well plates. At 48 h post-transfection, the cells were trypsinized using 0.25% trypsin (Beijing Solarbio Science & Technology Co., Ltd.) at room temperature for 1 min and collected by centrifugation at 800 × g for 5 min at room temperature. The cells were incubated with 0.5 ml binding buffer and 1.0 µl Annexin V-FITC (Merck KGaA) at room temperature for 15 min and resuspended in fresh 0.5 ml binding buffer containing 5 µl PI. Early apoptosis was measured using a Beckman Coulter FC500 flow cytometer (Beckman Coulter, Inc.) and MXP 2.2 software (Beckman Coulter, Inc.).

Scratch test

Effect of CDH16 on BCPAP cell migration was determined by scratch test. The cells were inoculated on 6-well plates at a density of 5×105 cells/well and incubated in RPMI-1640 (Gibco) containing 10% FBS (Gibco) at 37°C for 24 h. When confluency reached 80%, the medium was replaced with serum-free RPMI-1640 and the cells were starved for 24 h. A scratch was then drawn with a 200-µl plastic pipette tip and the plates were rinsed with PBS to remove any cells suspended in culture medium after scratching. The gap distance of each scratch wound was photographed at 0 and 48 h after scratching using an light microscope (Olympus Corporation IX71; ×40 magnification) and scratch area was measured with ImageJ v1.50 (National Institutes of Health). Wound healing was calculated as a percentage.

Transwell invasion assay

Pre-coated Matrigel chambers (BD Biosciences) were used for the invasion assays according to the manufacturer's instructions. After transfection of pCMV3-CDH16 plasmid and empty pCMV3 plasmid at 37°C for 48 h, the cells were collected. A total of 1×104 BCPAP cells were seeded into the upper chamber of the Transwell apparatus in serum-free RPMI-1640, while RPMI-1640 supplemented with 10% FBS was added to the lower chamber. After incubation at 37°C for 24 h, cells adhering to the lower surface were fixed with 100% methanol for 20 min at room temperature and stained with 5% Giemsa solution for 20 min at room temperature. The number of infiltrated cells was calculated under a light microscope (Olympus Corporation IX71) on five randomly selected fields (×100 magnification).

Statistical analysis

All experiments were performed ≥3 times. Statistical analysis was performed using SPSS 17.0 software (SPSS, Inc.). The χ2 or Fisher's exact test was used to analyze differences in CDH16 levels between PTC and normal thyroid tissue in IHC staining. Data are presented as the mean ± standard deviation. The paired t-test was used to compare CDH16 mRNA expression between 35 paired thyroid tissue samples. Pearson correlation test was used in correlation analysis. Single factor analysis of variance (one-way ANOVA) followed by post hoc Fisher's least significant difference test was used to compare >2 samples. P<0.05 was considered to indicate a statistically significant difference.

Results

CDH16 is downregulated in human TC tissue and cell lines independent of CNV

To verify that CDH16 is downregulated in TC, CDH16 transcript levels were evaluated in three independent TC studies from GEO. Data from all three studies in the Oncomine database showed that mRNA expression of CDH16 was significantly lower in PTC than in normal thyroid tissue (P<0.05; Fig. 1A-C). CDH16 expression was also significantly lower in FTC (P<0.05; Fig. 1D) and ATC (P<0.05; Fig. 1E) than in normal thyroid tissue. Notably, CDH16 ranked in the top 5% of all genes downregulated in each of these studies and the fold difference for PTC was >2. To determine whether decreased expression of CDH16 in PTC may be accounted for by DNA CNV, a dataset from TCGA that has previously been shown to have lower CDH16 expression in PTC (21) compared with matched normal tissue was used. The DNA CN of CDH16 showed no significant difference in blood, normal thyroid or TC tissue (P>0.05; Fig. 1F). Collectively, these findings indicated that CDH16 expression was decreased in TC in three PTC datasets as well as FTC and ATC, and also demonstrated that lower expression of CDH16 in TC was not accounted the result of CNV.

To support these findings, RT-qPCR and IHC were performed for 35 paired thyroid tissue samples collected from patients with PTC. CDH16 mRNA expression was significantly decreased in PTC compared with normal tissue (P<0.05; Fig. 2A). Furthermore, IHC showed that CDH16 was localized to the cell membrane of PTC cells. Of 35 PTC tissue samples, 17 (48.6%) stained negative for CDH16, while only two (5.7%) of the normal thyroid tissue samples stained negative for CDH16 (P<0.05; Fig. 2B). Therefore, decreased expression of CDH16 in PTC was observed at both the mRNA and protein level.

Next, to determine whether downregulation of CDH16 was correlated with the outcome of PTC, the association between CDH16 expression and clinicopathological characteristics of 35 patients with PTC was assessed. CDH16 protein expression was associated with tumor size, lymph node metastasis and pathological stage of PTC but there was no significant association with sex or age (Table I).

To confirm the downregulation of CDH16 in TC, CDH16 levels in TC-derived cell lines (BCPAP and TPC1) and the human thyroid follicular cell line Nthy-ori3-1 were measured. CDH16 expression was lower in TC cell lines, particularly BCPAP cells, both at the mRNA and protein level (P<0.05; Fig. 2C and D). These results were consistent with the aforementioned downregulation of CDH16 in patients with advanced PTC and support the use of BCPAP cells as a model for evaluating CDH16 biological activity.

CDH16 expression is negatively associated with expression of proliferation and invasion pathway-associated genes

To determine the role of CDH16 in TC, genes co-expressed with CDH16 in TC samples from TCGA were investigated using the LinkedOmics database. Volcano plot suggested that the most highly co-expressed genes were polypeptide N-acetylgalactosaminyltransferase 7 (GALNT7), NGFI-A binding protein 2 (NAB2) and UDP-galactose-4-epimerase (GALE), and the most highly inversely co-expressed genes are trefoil factor 3 (TFF3), odontogenic, ameloblast associated (ODAM) and dipeptidyl peptidase like 6 (DPP6) (P<0.05; Fig. 3A). KEGG pathway analysis by GSEA showed that negatively CDH16-associated genes were enriched in DNA replication and cell adhesion pathways (Fig. 3B; Table II). Correlation between expression levels of POLD1, MCM6, CLDN1, ICAM1 and SDC4 and those of CDH16 in 503 patients with PTC in TCGA was analyzed. The expression of each of these genes was significantly negatively correlated with expression of CDH16 in PTC (r=−0.290, −0.395, −0.594, −0.552, −0.583, respectively; P<0.001; Fig. 4).

Table II.

Significant enrichment of Kyoto Encyclopedia of Genes and Genomes pathways of cadherin 16 in thyroid carcinoma (LinkedOmics).

Table II.

Significant enrichment of Kyoto Encyclopedia of Genes and Genomes pathways of cadherin 16 in thyroid carcinoma (LinkedOmics).

DescriptionLeading edge genes, nLeading edge gene
DNA replication24MCM6; MCM2; MCM7; MCM4; MCM5; POLD1; RFC2; MCM3; LIG1; POLE3; POLE; RFC3; POLE2; PCNA; RNASEHIA; POLD3; POLA2; PRIM1; RFC4; POLA1; FEN1; RNASEH1; RPA2; PRIM2
Cell adhesion molecules54CLDN1; SDC4; ICAM1; CDH3; NRCAM; CD276; CLDN16; SDC3; CLDN10; CLDN7; CD58; CLDN4; ITGB8; CDH4; PTPRF; NLGN2; ALCAM; CLDN9; VTCN1; CNTNAP1; F11R; MAG; ITGAM; HLA-8; CDH15; CLDN2; ITGB7; HLA-DQB1; ITGA9; HLA-DRA; HLA-DQA1; HLA-G; CD22; HLA-DOB; HLA-A; HLA-C; HLA-DRB1; ITGB1; HLA-DOA; SELL; SELPLG; CD86; ITGB2; CD274; HLA-DPA1; NCAM2; HLA-DRB5; CD40; PTPRM;ICOSLG; HLA-DQA2; HLA-DPB1; SPN; CLDN3
Overexpression of CDH16 inhibits proliferation, migration and invasion of BCPAP cells in vitro

To verify the function of CDH16 in TC cells, BCPAP cells were transfected with CDH16 expression vector. RT-qPCR and western blot assay verified higher mRNA and protein CDH16 levels in CDH16 overexpression compared with negative control BCPAP cells (P<0.05; Fig. 5A and B). To evaluate the effect of CDH16 on proliferation, MTT assay was performed. The proliferation of BCPAP cells was significantly decreased in overexpression compared with negative control cells (P<0.05; Fig. 5C).

To determine whether CDH16 expression affects tumor-associated functions, we the effect of CDH16 overexpression on cell apoptosis, migration and invasion was assessed. Compared with negative control, the CDH16 overexpression group exhibited a significantly higher apoptosis rate, as evaluated by flow cytometry with Annexin V and PI staining (P<0.05; Fig. 5D and E). Cell scratch test indicated that overexpression of CDH16 significantly inhibited migration of BCPAP cells (P<0.05; Fig. 6A and B). Furthermore, Transwell invasion assay indicated that overexpression of CDH16 significantly inhibited invasion capacity of BCPAP cells (P<0.05; Fig. 6C and D). Overall, these results demonstrated that CDH16 promoted apoptosis and inhibited TC cell proliferation, migration and invasion in vitro, which is consistent with the pattern of expression in patients with PTC.

Overexpression of CDH16 downregulates POLD1, MCM6, CLDN1, ICAM1 and SDC4 expression in BCPAP cells in vitro

To verify that CDH16 inhibited cell proliferation, migration and invasion via DNA replication and cell adhesion molecular pathways, western blot assay was used to detect protein expression levels. Following overexpression of CDH16 in TC cells, expression of POLD1, MCM6, CLDN1, ICAM1 and SDC4 was significantly downregulated (53.42±6.38, 37.67±14.27, 42.17±3.36, 37.04±9.34 and 47.83±9.23% decrease, respectively; P<0.05; Fig. 7) compared with negative control. These data indicated that CDH16 may regulate proliferation, migration and invasion of TC cells via DNA replication and cell adhesion pathways.

Discussion

The human genome encodes 115 members of the CDH superfamily and the expression of these family members is tissue-specific (29). CDH1/E-CDH, one of the most classical CDHs, is primarily expressed in epithelial tissue and has been characterized as a tumor suppressor involved in epithelial-mesenchymal transition (3033). Expression of CDH16 was first observed in the rabbit kidney (34) and has since been shown to be expressed in mouse and human thyroid (17,20). CDH16 co-localizes with CDH1 in thyroid follicular cells and serves a key role in thyroid cell polarity acquisition and follicle formation (18,20). Calì et al (20) demonstrated that CDH16 promotes intercellular adhesion to a similar extent as CDH1 and loss of CDH16 precedes loss of CDH1 in TC. CDH1 expression decreases in FTC and has been suggested as a marker for prognosis of TC (35,36). To the best of our knowledge, however, the mechanism of CDH16 in TC has not previously been determined.

The present study characterized CDH16 expression in different forms of TC. The results demonstrated that CDH16 mRNA levels were significantly downregulated in PTC, FTC and ATC compared with normal thyroid tissue. Furthermore, CDH16 ranked within the top 5% of downregulated genes, with the highest rank (<2%) for PTC. These data demonstrated that CDH16 downregulation of expression was a common feature of TC. CDH16 protein expression in TC was correlated with tumor size, disease stage and nodal metastasis in an independent cohort of 35 patients. Consistent with the present results, Li et al (21) demonstrated an association between CDH16 downregulation and lymph node metastasis using TCGA cohorts. These results confirm previous findings suggesting that CDH16 may serve as a biomarker for PTC diagnosis and prognosis (20,21). CNV affects gene expression and is a key pathogenic factor in types of cancer, such as ovarian cancer (37). However, in the present study, no significant CN loss of CDH16 was detected in TC. These results suggested that CDH16 may be a potential diagnostic and prognostic marker for TC and that downregulation of CDH16 was independent of CNV.

CDH16 serves as a tumor suppressor in TC (20,21) but the specific pathogenesis remains largely unclear. Here, CDH16 was co-expressed with genes involved in DNA replication and cell adhesion molecule pathways. To verify the role of CDH16 in TC, CDH16 was overexpressed in BCPAP cells and MTT, flow cytometry, cell scratch and Transwell invasion assays were performed to determine whether overexpression of CDH16 affected proliferation, apoptosis, migration and invasion. The results demonstrated that overexpression of CDH16 resulted in inhibition of proliferation, migration and invasion of TC cells and increased apoptosis.

KEGG analysis demonstrated that CDH16 expression was negatively associated with expression of DNA replication genes. DNA replication is a key event for cell proliferation that is separated into three stages: Initiation, extension and termination (38). At the beginning of replication, the double strands of the DNA helix unwind under the action of helicase (38). Then, using each parent chain as a template and four deoxynucleotides in the surrounding environment as raw materials, a chain complementary to the parent chain is synthesized under the action of DNA polymerase according to complementary base pairing (39). The accuracy of DNA replication in eukaryotes requires proteins such as helicase and DNA POL (40,41). MCM protein complex, which consists of six highly conserved proteins (MCM2-7), initiates DNA replication and unwinding via its replicative helicase activity (42). MCM2-7 proteins are present in proliferating cells (43) and overexpression of MCM2, MCM4 and MCM6 is associated with tumorigenesis (4446). DNA POL α, δ, and ε are key mediators of DNA replication in eukaryotes (41); their mutation or abnormal expression affects the occurrence, development and invasion of human colorectal cancer, stomach adenocarcinoma and pancreatic adenocarcinoma (4749). POLD1 encodes DNA POL δ (50); an increase in its protein expression or activity has been demonstrated to be associated with tumorigenesis in colorectal carcinoma and endometrial carcinoma (5153). In addition, it is associated with the invasive ability of cancer cells. For example, Sanefuji et al (54) reported that POLD1 is a key indicator of the activity and invasion of hepatoma cells and its expression is associated with the degree of vascular invasion of cancer cells. Sigurdson et al (55) found that increased POLD1 expression increases the risk of breast cancer. The present study showed that expression of MCM6 and POLD1 in TC was significantly negatively correlated with CDH16 by analyzing TCGA thyroid cancer data and that expression of MCM6 and POLD1 was significantly downregulated and cell proliferation inhibited after overexpression of CDH16 in BCPAP cells. This suggested that CDH16 inhibited proliferation of TC cells via downregulation of proteins associated with DNA replication.

KEGG analysis showed that CDH16 was associated with the cell adhesion molecular pathway. Adhesion molecules are divided into five groups: Integrins, selectins, CDHs, hyaladherin and immunoglobulin superfamily members (56). Cell adhesion molecules regulate tumor invasion and metastasis (57). By binding to surface adhesion molecules and extracellular matrix or cell ligands, they activate intracellular signaling pathways and endow tumor cells with metastatic ability (57). CLDNs are cytoskeletal proteins of tight junction between cells that not only regulate paracellular transepithelial/transendothelial transport but are also key for cell proliferation and differentiation (58). The abnormal expression of CLDNs can lead to structural and functional damage of epithelial and endothelial cells, which leads to tumor invasion and metastasis (59). CLDN1 is the most studied CLDN in cancer and its overexpression or loss of expression is observed in different types of cancer (60). CLDN1 expression is low in invasive breast, esophageal and prostate cancer (5961). However, CLDN1 is highly expressed in ovarian, colon and thyroid cancer (60,6264). Furthermore, the mechanism of CLDN1 in promoting tumor invasion may involve activation of matrix metalloproteinase 2 (65). Considering the role of CLDN1 in TC, targeting CLDN1 expression may be a promising treatment for TC (66).

Other cell adhesion molecules identified in the present study included ICAM1 and SDC4. ICAM1 is an immune protein superfamily adhesion molecule that is widely expressed on the surface of monocytes, lymphocytes and vascular endothelial and cancer cells (67). Its primary function is to regulate adhesion between cells and between cells and the extracellular matrix. In addition, it is involved in cell signal transduction, proliferation and migration, immune inflammatory response, vascular growth, tumor invasion and metastasis, as well as other physiological and pathological processes (68). ICAM1 is overexpressed in a number of tumor types, including TC (69,70). The binding of ICAM1 and its ligand lymphocyte-associated antigen 1 (LFA1) inhibits natural killer and cytotoxic T cells in the occurrence and development of malignant tumor, helps tumor cells escape immune surveillance and attack and promotes invasion and metastasis of tumor cells (71). SDC, a family of four transmembrane proteoglycans, has been reported to serve key roles in cell proliferation, migration and differentiation (72). SDC4 is highly expressed in TC compared with normal tissue (73). Chen et al (73) revealed that SDC4 expression levels are upregulated in PTC and SDC4 gene silencing suppresses PTC cell migration and invasion by suppressing activation of the Wnt/β-catenin signaling pathway.

The present study confirmed that expression of CLDN1, ICAM1 and SDC4 was negatively correlated with CDH16 expression in TCGA databases. Furthermore, expression of CLDN1, ICAM1 and SDC4 was verified by western blotting following overexpression of CDH16. The present results demonstrated that CDH16-overexpressing TC cells exhibited significantly downregulated levels of CLDN1, ICAM1 and SDC4, which lead to inhibition of cell migration and invasion. Thus, CDH16 and cell adhesion pathways may serve a role in TC via migration and invasion, which is consistent with results of KEGG analysis. These results are in accordance with bioinformatics analysis results and support the findings of the present study, confirming that CDH16 may be a functional tumor suppressor in TC.

In conclusion, the present study used an extended dataset to demonstrate that CDH16 is downregulated in PTC, FTC and ATC and demonstrated that its expression was significantly correlated with tumor size, lymph node metastasis and disease stage in a cohort of 35 patients with PTC. Furthermore, bioinformatics analysis showed that downregulation of CDH16 may promote TC via DNA replication and cell adhesion pathways. The present study also demonstrated that overexpression of CDH16 inhibited proliferation, migration and invasion and induced apoptosis of BCPAP cells. These findings were demonstrated using human tumor samples and cell lines; however, studies in mice would provide in vivo verification. To the best of our knowledge, the present study is the first to provide direct evidence for the tumor suppressive effect of CDH16 in TC. While the molecular mechanism of CDH16 disruption in TC remains unknown, elucidating the signaling mechanism underlying CDH16 downregulation in TC may inform the design of novel therapeutic strategies to treat TC.

Acknowledgements

The authors would like to thank Professor Jun Li from Tangshan Renmin Hospital (Hebei, China) for collecting tissue specimens.

Funding

The present study was supported by The Science and Technology Program of Hebei (grant no. H2018105066).

Availability of data and materials

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

Authors' contributions

XY, YuL and GL designed the study. XY, WZ and YiL performed the experiments. GL and XY performed data analysis. XY and YuL confirm the authenticity of all the raw data. XY drafted the manuscript. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethical Committee of Tangshan Gongren Hospital (approval no. GRYY-LL-2020-104). All population-related studies were carried out according to the World Medical Association Declaration of Helsinki. All patients provided written informed consent.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Pellegriti G, Frasca F, Regalbuto C, Squatrito S and Vigneri R: Worldwide increasing incidence of thyroid cancer: Update on epidemiology and risk factors. J Cancer Epidemiol. 2013:9652122013. View Article : Google Scholar : PubMed/NCBI

2 

Kim J, Gosnell JE and Roman SA: Geographic influences in the global rise of thyroid cancer. Nat Rev Endocrinol. 16:17–29. 2020. View Article : Google Scholar : PubMed/NCBI

3 

Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM and Matrisian LM: Projecting cancer incidence and deaths to 2030: The unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 74:2913–2921. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Siegel RL, Miller KD and Jemal A: Cancer Statistics, 2017. CA Cancer J Clin. 67:7–30. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Wang J, Yu F, Shang Y, Ping Z and Liu L: Thyroid cancer: Incidence and mortality trends in China, 2005–2015. Endocrine. 68:163–173. 2020. View Article : Google Scholar : PubMed/NCBI

6 

Sherman SI: Thyroid carcinoma. Lancet. 361:501–511. 2003. View Article : Google Scholar : PubMed/NCBI

7 

Burns WR and Zeiger MA: Differentiated thyroid cancer. Semin Oncol. 37:557–566. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Rahmani N, Abbas Hashemi S, Fazli M and Raisian M: Clinical management and outcomes of papillary, follicular and medullary thyroid cancer surgery. Med Glas. 10:164–167. 2013.PubMed/NCBI

9 

Grant CS: Papillary thyroid cancer: Strategies for optimal individualized surgical management. Clin Ther. 36:1117–1126. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Manoj G, Deepika K, Ryoko O, Saket J, Vikas M and Wenwen C: Laminin-5γ-2 (LAMC2) is highly expressed in anaplastic thyroid carcinoma and is associated with tumor progression, migration, and invasion by modulating signaling of EGFR. J Clin Endocrinol Metab. 99:E62–E72. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Wu H, Sun Y, Ye H, Yang S, Lee SL and de las Morenas A: Anaplastic thyroid cancer: Outcome and the mutation/expression profiles of potential targets. Pathol Oncol Res. 21:695–701. 2015. View Article : Google Scholar : PubMed/NCBI

12 

Ernani V, Kumar M, Chen AY and Owonikoko TK: Systemic treatment and management approaches for medullary thyroid cancer. Cancer Treat Rev. 50:89–98. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Hulpiau P and van Roy F: Molecular evolution of the cadherin superfamily. Int J Biochem Cell Biol. 41:349–369. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Casal JI and Bartolomé RA: Beyond N-Cadherin, Relevance of Cadherins 5, 6 and 17 in Cancer Progression and Metastasis. Int J Mol Sci. 20:33732019. View Article : Google Scholar : PubMed/NCBI

15 

Thomson RB, Ward DC, Quaggin SE, Igarashi P, Muckler ZE and Aronson PS: cDNA cloning and chromosomal localization of the human and mouse isoforms of Ksp-cadherin. Genomics. 51:445–451. 1998. View Article : Google Scholar : PubMed/NCBI

16 

Thedieck C, Kuczyk M, Klingel K, Steiert I, Müller CA and Klein G: Expression of Ksp-cadherin during kidney development and in renal cell carcinoma. Br J Cancer. 92:2010–2017. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Calì G, Zannini M, Rubini P, Tacchetti C, D'Andrea B, Affuso A, Wintermantel T, Boussadia O, Terracciano D, Silberschmidt D, et al: Conditional inactivation of the E-cadherin gene in thyroid follicular cells affects gland development but does not impair junction formation. Endocrinology. 148:2737–2746. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Koumarianou P, Goméz-López G and Santisteban P: Pax8 controls thyroid follicular polarity through cadherin-16. J Cell Sci. 130:219–231. 2017.PubMed/NCBI

19 

de Cristofaro T, Di Palma T, Fichera I, Lucci V, Parrillo L, De Felice M and Zannini M: An essential role for Pax8 in the transcriptional regulation of cadherin-16 in thyroid cells. Mol Endocrinol. 26:67–78. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Calì G, Gentile F, Mogavero S, Pallante P, Nitsch R, Ciancia G, Ferraro A, Fusco A and Nitsch L: CDH16/Ksp-cadherin is expressed in the developing thyroid gland and is strongly down-regulated in thyroid carcinomas. Endocrinology. 153:522–534. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Li P, Wu Q, Sun Y, Pan X, Han Y, Ye B, Zhang Y, Dong J and Zheng Z: Downregulation of cdh16 in papillary thyroid cancer and its potential molecular mechanism analysed by qRT-PCR, TCGA and in silico analysis. Cancer Manag Res. 11:10719–10729. 2019. View Article : Google Scholar : PubMed/NCBI

22 

Rhodes DR, Kalyana-Sundaram S, Mahavisno V, Varambally R, Yu J, Briggs BB, Barrette TR, Anstet MJ, Kincead-Beal C, Kulkarni P, et al: Oncomine 3.0: Genes, pathways, and networks in a collection of 18,000 cancer gene expression profiles. Neoplasia. 9:166–180. 2007. View Article : Google Scholar : PubMed/NCBI

23 

He H, Jazdzewski K, Li W, Liyanarachchi S, Nagy R, Volinia S, Calin GA, Liu CG, Franssila K, Suster S, et al: The role of microRNA genes in papillary thyroid carcinoma. Proc Natl Acad Sci USA. 102:19075–19080. 2005. View Article : Google Scholar : PubMed/NCBI

24 

Vasko V, Espinosa AV, Scouten W, He H, Auer H, Liyanarachchi S, Larin A, Savchenko V, Francis GL, de la Chapelle A, et al: Gene expression and functional evidence of epithelial-to-mesenchymal transition in papillary thyroid carcinoma invasion. Proc Natl Acad Sci USA. 104:2803–2808. 2007. View Article : Google Scholar : PubMed/NCBI

25 

Giordano TJ, Au AYM, Kuick R, Thomas DG, Rhodes DR, Wilhelm KG Jr, Vinco M, Misek DE, Sanders D, Zhu Z, et al: Delineation, functional validation, and bioinformatic evaluation of gene expression in thyroid follicular carcinomas with the PAX8-PPARG translocation. Clin Cancer Res. 12:1983–1993. 2006. View Article : Google Scholar : PubMed/NCBI

26 

Vasaikar SV, Straub P, Wang J and Zhang B: LinkedOmics: Analyzing multi-omics data within and across 32 cancer types. Nucleic Acids Res. 46D:D956–D963. 2018. View Article : Google Scholar : PubMed/NCBI

27 

Yang X, Liu G, Li W, Zang L, Li D, Wang Q, Yu F and Xiang X: Silencing of zinc finger protein 703 inhibits medullary thyroid carcinoma cell proliferation in vitro and in vivo. Oncol Lett. 19:943–951. 2020.PubMed/NCBI

28 

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

29 

Suzuki ST: Structural and functional diversity of cadherin superfamily: Are new members of cadherin superfamily involved in signal transduction pathway? J Cell Biochem. 61:531–542. 1996. View Article : Google Scholar : PubMed/NCBI

30 

Vleminckx K, Vakaet L Jr, Mareel M, Fiers W and van Roy F: Genetic manipulation of E-cadherin expression by epithelial tumor cells reveals an invasion suppressor role. Cell. 66:107–119. 1991. View Article : Google Scholar : PubMed/NCBI

31 

Perl AK, Wilgenbus P, Dahl U, Semb H and Christofori G: A causal role for E-cadherin in the transition from adenoma to carcinoma. Nature. 392:190–193. 1998. View Article : Google Scholar : PubMed/NCBI

32 

Shiozaki H, Oka H, Inoue M, Tamura S and Monden M: E-cadherin mediated adhesion system in cancer cells. Cancer. 77 (Suppl):1605–1613. 1996. View Article : Google Scholar : PubMed/NCBI

33 

Pal M, Bhattacharya S, Kalyan G and Hazra S: Cadherin profiling for therapeutic interventions in Epithelial Mesenchymal Transition (EMT) and tumorigenesis. Exp Cell Res. 368:137–146. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Thomson RB, Igarashi P, Biemesderfer D, Kim R, Abu-Alfa A, Soleimani M and Aronson PS: Isolation and cDNA cloning of Ksp-cadherin, a novel kidney-specific member of the cadherin multigene family. J Biol Chem. 270:17594–17601. 1995. View Article : Google Scholar : PubMed/NCBI

35 

Brabant G, Hoang-Vu C, Cetin Y, Dralle H, Scheumann G, Mölne J, Hansson G, Jansson S, Ericson LE and Nilsson M: E-cadherin: A differentiation marker in thyroid malignancies. Cancer Res. 53:4987–4993. 1993.PubMed/NCBI

36 

Mitselou A, Ioachim E, Peschos D, Charalabopoulos K, Michael M, Agnantis NJ and Vougiouklakis T: E-cadherin adhesion molecule and syndecan-1 expression in various thyroid pathologies. Exp Oncol. 29:54–60. 2007.PubMed/NCBI

37 

Reid BM, Permuth JB, Chen YA, Fridley BL, Iversen ES, Chen Z, Jim H, Vierkant RA, Cunningham JM, Barnholtz-Sloan JS, et al: Genome-wide analysis of common copy number variation and epithelial ovarian cancer risk. Cancer Epidemiol Biomarkers Prev. 28:1117–1126. 2019. View Article : Google Scholar : PubMed/NCBI

38 

Fragkos M, Ganier O, Coulombe P and Méchali M: DNA replication origin activation in space and time. Nat Rev Mol Cell Biol. 16:360–374. 2015. View Article : Google Scholar : PubMed/NCBI

39 

Ganai RA and Johansson E: DNA replication-A matter of fidelity. Mol Cell. 62:745–755. 2016. View Article : Google Scholar : PubMed/NCBI

40 

Burgers PMJ and Kunkel TA: Eukaryotic DNA replication fork. Annu Rev Biochem. 86:417–438. 2017. View Article : Google Scholar : PubMed/NCBI

41 

Syväoja J, Suomensaari S, Nishida C, Goldsmith JS, Chui GS, Jain S and Linn S: DNA polymerases alpha, delta, and epsilon: Three distinct enzymes from HeLa cells. Proc Natl Acad Sci USA. 87:6664–6668. 1990. View Article : Google Scholar : PubMed/NCBI

42 

Forsburg SL: Eukaryotic MCM proteins: Beyond replication initiation. Microbiol Mol Biol Rev. 68:109–131. 2004. View Article : Google Scholar : PubMed/NCBI

43 

Stoeber K, Tlsty TD, Happerfield L, Thomas GA, Romanov S, Bobrow L, Williams ED and Williams GH: DNA replication licensing and human cell proliferation. J Cell Sci. 114:2027–2041. 2001. View Article : Google Scholar : PubMed/NCBI

44 

Das M, Prasad SB, Yadav SS, Govardhan HB, Pandey LK, Singh S, Pradhan S and Narayan G: Over expression of minichromosome maintenance genes is clinically correlated to cervical carcinogenesis. PLoS One. 8:e696072013. View Article : Google Scholar : PubMed/NCBI

45 

Kwok HF, Zhang SD, McCrudden CM, Yuen HF, Ting KP, Wen Q, Khoo US and Chan KY: Prognostic significance of minichromosome maintenance proteins in breast cancer. Am J Cancer Res. 5:52–71. 2014.PubMed/NCBI

46 

Issac MSM, Yousef E, Tahir MR and Gaboury LA: MCM2, MCM4, and MCM6 in Breast Cancer: Clinical Utility in Diagnosis and Prognosis. Neoplasia. 21:1015–1035. 2019. View Article : Google Scholar : PubMed/NCBI

47 

Shinbrot E, Henninger EE, Weinhold N, Covington KR, Göksenin AY, Schultz N, Chao H, Doddapaneni H, Muzny DM, Gibbs RA, et al: Exonuclease mutations in DNA polymerase epsilon reveal replication strand specific mutation patterns and human origins of replication. Genome Res. 24:1740–1750. 2014. View Article : Google Scholar : PubMed/NCBI

48 

Rayner E, van Gool IC, Palles C, Kearsey SE, Bosse T, Tomlinson I and Church DN: A panoply of errors: Polymerase proofreading domain mutations in cancer. Nat Rev Cancer. 16:71–81. 2016. View Article : Google Scholar : PubMed/NCBI

49 

Wang Y, Chen Y, Wang C, Yang M, Wang Y, Bao L, Wang JE, Kim B, Chan KY, Xu W, et al: MIF is a 3′ flap nuclease that facilitates DNA replication and promotes tumor growth. Nat Commun. 12:29542021. View Article : Google Scholar : PubMed/NCBI

50 

Preston BD, Albertson TM and Herr AJ: DNA replication fidelity and cancer. Semin Cancer Biol. 20:281–293. 2010. View Article : Google Scholar : PubMed/NCBI

51 

Palles C, Cazier JB, Howarth KM, Domingo E, Jones AM, Broderick P, Kemp Z, Spain SL, Guarino E, Salguero I, et al CORGI Consortium; WGS500 Consortium, : Germline mutations affecting the proofreading domains of POLE and POLD1 predispose to colorectal adenomas and carcinomas. Nat Genet. 45:136–144. 2013. View Article : Google Scholar : PubMed/NCBI

52 

Siraj AK, Parvathareddy SK, Bu R, Iqbal K, Siraj S, Masoodi T, Concepcion RM, Ghazwani LO, AlBadawi I, Al-Dayel F, et al: Germline POLE and POLD1 proofreading domain mutations in endometrial carcinoma from Middle Eastern region. Cancer Cell Int. 19:3342019. View Article : Google Scholar : PubMed/NCBI

53 

Nicolas E, Golemis EA and Arora S: POLD1: Central mediator of DNA replication and repair, and implication in cancer and other pathologies. Gene. 590:128–141. 2016. View Article : Google Scholar : PubMed/NCBI

54 

Sanefuji K, Taketomi A, Iguchi T, Sugimachi K, Ikegami T, Yamashita Y, Gion T, Soejima Y, Shirabe K and Maehara Y: Significance of DNA polymerase delta catalytic subunit p125 induced by mutant p53 in the invasive potential of human hepatocellular carcinoma. Oncology. 79:229–237. 2010. View Article : Google Scholar : PubMed/NCBI

55 

Sigurdson AJ, Hauptmann M, Chatterjee N, Alexander BH, Doody MM, Rutter JL and Struewing JP: Kin-cohort estimates for familial breast cancer risk in relation to variants in DNA base excision repair, BRCA1 interacting and growth factor genes. BMC Cancer. 4:92004. View Article : Google Scholar : PubMed/NCBI

56 

Samanta D and Almo SC: Nectin family of cell-adhesion molecules: Structural and molecular aspects of function and specificity. Cell Mol Life Sci. 72:645–658. 2015. View Article : Google Scholar : PubMed/NCBI

57 

Harjunpää H, Llort Asens M, Guenther C and Fagerholm SC: Cell Adhesion Molecules and Their Roles and Regulation in the Immune and Tumor Microenvironment. Front Immunol. 10:10782019. View Article : Google Scholar : PubMed/NCBI

58 

Krause G, Winkler L, Mueller SL, Haseloff RF, Piontek J and Blasig IE: Structure and function of claudins. Biochim Biophys Acta. 1778:631–645. 2008. View Article : Google Scholar : PubMed/NCBI

59 

Singh AB, Sharma A and Dhawan P: Claudin family of proteins and cancer: An overview. J Oncol. 2010:5419572010. View Article : Google Scholar : PubMed/NCBI

60 

Bhat AA, Syed N, Therachiyil L, Nisar S, Hashem S, Macha MA, Yadav SK, Krishnankutty R, Muralitharan S, Al-Naemi H, et al: Claudin-1, A Double-Edged Sword in Cancer. Int J Mol Sci. 21:5692020. View Article : Google Scholar : PubMed/NCBI

61 

Zhou B, Moodie A, Blanchard AA, Leygue E and Myal Y: Claudin 1 in breast cancer: New insights. J Clin Med. 4:1960–1976. 2015. View Article : Google Scholar : PubMed/NCBI

62 

Németh J, Németh Z, Tátrai P, Péter I, Somorácz A, Szász AM, Kiss A and Schaff Z: High expression of claudin-1 protein in papillary thyroid tumor and its regional lymph node metastasis. Pathol Oncol Res. 16:19–27. 2010. View Article : Google Scholar : PubMed/NCBI

63 

Hucz J, Kowalska M, Jarzab M and Wiench M: Gene expression of metalloproteinase 11, claudin 1 and selected adhesion related genes in papillary thyroid cancer. Endokrynol Pol. 57 (Suppl A):18–25. 2006.(In Polish). PubMed/NCBI

64 

Fluge Ø, Bruland O, Akslen LA, Lillehaug JR and Varhaug JE: Gene expression in poorly differentiated papillary thyroid carcinomas. Thyroid. 16:161–175. 2006. View Article : Google Scholar : PubMed/NCBI

65 

Li J, Chigurupati S, Agarwal R, Mughal MR, Mattson MP, Becker KG, Wood WH III, Zhang Y and Morin PJ: Possible angiogenic roles for claudin-4 in ovarian cancer. Cancer Biol Ther. 8:1806–1814. 2009. View Article : Google Scholar : PubMed/NCBI

66 

Piontek A, Eichner M, Zwanziger D, Beier LS, Protze J, Walther W, Theurer S, Schmid KW, Führer-Sakel D, Piontek J, et al: Targeting claudin-overexpressing thyroid and lung cancer by modified Clostridium perfringens enterotoxin. Mol Oncol. 14:261–276. 2020. View Article : Google Scholar : PubMed/NCBI

67 

Pober JS, Gimbrone MA Jr, Lapierre LA, Mendrick DL, Fiers W, Rothlein R and Springer TA: Overlapping patterns of activation of human endothelial cells by interleukin 1, tumor necrosis factor, and immune interferon. J Immunol. 137:1893–1896. 1986.PubMed/NCBI

68 

Kotteas EA, Boulas P, Gkiozos I, Tsagkouli S, Tsoukalas G and Syrigos KN: The intercellular cell adhesion molecule-1 (ICAM-1) in lung cancer: Implications for disease progression and prognosis. Anticancer Res. 34:4665–4672. 2014.PubMed/NCBI

69 

Buitrago D, Keutgen XM, Crowley M, Filicori F, Aldailami H, Hoda R, Liu YF, Hoda RS, Scognamiglio T, Jin M, et al: Intercellular adhesion molecule-1 (ICAM-1) is upregulated in aggressive papillary thyroid carcinoma. Ann Surg Oncol. 19:973–980. 2012. View Article : Google Scholar : PubMed/NCBI

70 

Min IM, Shevlin E, Vedvyas Y, Zaman M, Wyrwas B, Scognamiglio T, Moore MD, Wang W, Park S, Park S, et al: CAR T Therapy Targeting ICAM-1 Eliminates Advanced Human Thyroid Tumors. Clin Cancer Res. 23:7569–7583. 2017. View Article : Google Scholar : PubMed/NCBI

71 

Gray KD, McCloskey JE, Vedvyas Y, Kalloo OR, Eshaky SE, Yang Y, Shevlin E, Zaman M, Ullmann TM, Liang H, et al: PD1 blockade enhances ICAM1-directed CAR T therapeutic efficacy in advanced thyroid cancer. Clin Cancer Res. 26:6003–6016. 2020. View Article : Google Scholar : PubMed/NCBI

72 

Ohkawara B, Glinka A and Niehrs C: Rspo3 binds syndecan 4 and induces Wnt/PCP signaling via clathrin-mediated endocytosis to promote morphogenesis. Dev Cell. 20:303–314. 2011. View Article : Google Scholar : PubMed/NCBI

73 

Chen LL, Gao GX, Shen FX, Chen X, Gong XH and Wu WJ: SDC4 gene silencing favors human papillary thyroid carcinoma cell apoptosis and inhibits epithelial mesenchymal transition via Wnt/beta-catenin pathway. Mol Cells. 41:853–867. 2018.PubMed/NCBI

Related Articles

Journal Cover

May-2022
Volume 23 Issue 5

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yang X, Li Y, Liu G, Zha W and Liu Y: Cadherin‑16 inhibits thyroid carcinoma cell proliferation and invasion. Oncol Lett 23: 145, 2022
APA
Yang, X., Li, Y., Liu, G., Zha, W., & Liu, Y. (2022). Cadherin‑16 inhibits thyroid carcinoma cell proliferation and invasion. Oncology Letters, 23, 145. https://doi.org/10.3892/ol.2022.13265
MLA
Yang, X., Li, Y., Liu, G., Zha, W., Liu, Y."Cadherin‑16 inhibits thyroid carcinoma cell proliferation and invasion". Oncology Letters 23.5 (2022): 145.
Chicago
Yang, X., Li, Y., Liu, G., Zha, W., Liu, Y."Cadherin‑16 inhibits thyroid carcinoma cell proliferation and invasion". Oncology Letters 23, no. 5 (2022): 145. https://doi.org/10.3892/ol.2022.13265