Open Access

EpCAM is essential for maintenance of the small intestinal epithelium architecture via regulation of the expression and localization of proteins that compose adherens junctions

  • Authors:
    • Guibin Chen
    • Yanhong Yang
    • Wanwan Liu
    • Li Huang
    • Lanxiang Yang
    • Yuting Lei
    • Huijuan Wu
    • Zili Lei
    • Jiao Guo
  • View Affiliations

  • Published online on: December 10, 2020     https://doi.org/10.3892/ijmm.2020.4815
  • Pages: 621-632
  • Copyright: © Chen 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

Epithelial cell adhesion molecule (EpCAM) is highly expressed in mammalian intestines, and is essential for maintaining the homeostasis of the intestinal epithelium. EpCAM protein is localized at tight junctions and the basolateral membrane of the intestinal epithelium, where it interacts with many cell adhesion molecules. To explore the molecular functions of EpCAM in regulating adherens junctions in the intestinal epithelium, EpCAM knockout embryos and newborn pups were analyzed. Hematoxylin and eosin staining was used to assess the histology of the duodenum, jejunum, ileum and colon from wild-type and EpCAM‑/‑ mice at E18.5, P0 and P3. The expression and localization of adherens junction‑associated genes and genes that encode the proteins that participate in the assembly of adherens junctions were measured at the mRNA and protein levels using qPCR, western blot analysis and immunofluorescence staining. The results showed that although there was no significant damage to the intestines of EpCAM‑/‑ mice at E18.5 and P0, they were significantly damaged at P3 in mutant mice. The expression of adherens junction‑associated genes in EpCAM mutant mice was normal at the mRNA level from E18.5 to P3, but their protein levels were gradually reduced and mislocalized from E18.5 to P3. The expression of nectin 1, which can regulate the assembly and adhesion activity of E‑cadherin, was also gradually reduced at both the mRNA and protein levels in the intestinal epithelium of EpCAM mutant mice from E18.5 to P3. In summary, the loss of EpCAM may cause the reduction and mislocalization of proteins that compose adherens junctions partly via the downregulation of nectin 1 in the intestines.

Introduction

EpCAM, the epithelial cell adhesion molecule, is a hemophilic Ca2+-independent cell-cell adhesion molecule, and it is expressed in different kinds of epithelial tissues (1-3). In addition to its function in cell adhesion, EpCAM has been reported to contribute to various biological processes, such as signaling, migration and proliferation (2,4). EpCAM has been considered to be a cell surface marker for many kinds of stem cells (5,6). Moreover, EpCAM is also highly expressed in epithelial tumor tissues (7). However, the molecular mechanisms of these processes remain to be elucidated.

EpCAM mutations in humans can cause congenital tufting enteropathy (CTE), a rare diarrheal disease that can cause neonatal death (8). Several EpCAM knockout mouse models have been reported to generate a CTE phenotype (9-11). From these reports, it can be concluded that EpCAM has important physiological functions in the intestines of human and mammalian animals. EpCAM was determined to be highly expressed in developing adult intestinal epithelium (2,10). In addition, EpCAM is also highly expressed in many kinds of stem cells, neoplastic tissues, and other normal epithelial tissues, such as embryonic stem cells, hepatocyte progenitor cells, and labyrinthine layer of placentas (1,12-14). EpCAM recruits proteins of the claudin family to maintain functional tight junctions in the intestinal epithelium (10,15,16). Loss of functional tight junctions in the intestinal epithelium may be one of the important causes of diarrheal disease in humans and mice with mutated EpCAM. EpCAM is not only localized to tight junctions but is also enriched at the basolateral membrane of the intestinal epithelium (2). This suggests that EpCAM may have some interactions with other types of cell junctions. However, the conclusions regarding the function of EpCAM in regulating adherens junctions are contradictory. Previous findings have shown that EpCAM may suppress E-cadherin-mediated cell aggregation by disrupting the association of E-cadherin with the cytoskeleton in cultured murine fibroblast L cells (17,18). It has also been reported that E-cadherin could exert opposing effects on EpCAM, especially in tumor tissues (2). In addition, E-cadherin protein was decreased (19) or mislocalized (20) in EpCAM-depleted human colorectal adenocarcinoma Caco-2 cell line. However, Wu et al found that the expression and localization of E-cadherin and β-catenin were still normal after EpCAM knockdown in human colorectal adenocarcinoma T84 cell line (16). Results from in vivo studies were also different. In biopsy specimens from children with CTE, the expression of E-cadherin was normal (21). Lei et al also found that the expression level and localization of E-cadherin and β-catenin were normal in the embryonic intestines of EpCAM knockout mice (10). However, it has been reported that E-cadherin and β-catenin proteins progressively lose cell membrane localization and accumulate in the cytoplasm in the intestinal epithelium of EpCAM knockout mice after birth (9). In EpCAM mutant zebrafish, the expression of E-cadherin, β-catenin and α-catenin was reduced in the enveloping layer, but the localization of these proteins was still normal (4). Moreover, global depletion of EpCAM in Xenopus embryos caused a significant decrease in C-cadherin at the protein level (19). The molecular roles of EpCAM in regulating adherens junctions, especially in mammalian models, are still unclear.

In the present study, to investigate the functions of EpCAM in regulating adherens junctions in the mammalian intestinal epithelium, the expression of proteins that compose adherens junctions was assessed at both the mRNA and protein levels in different sections of the intestinal epithelium from EpCAM−/− and wild-type (WT) mice at E18.5 to P3, providing new evidence for the effects of EpCAM on adherens junctions in the intestinal epithelium.

Materials and methods

Animal treatment

All mouse experiments were approved by the Committee on Laboratory Animal Care and Use of Guangdong Pharmaceutical University [Guangzhou, China (gdpu2016073)].

EpCAM−/− mice were previously generated by CRISPR/Cas9 technology (22). Mice were housed in the specific pathogen-free (SPF) animal facility at 25°C with a 12-h light/dark cycle and 50-55% humidity. The animals had free access to water and food. Heterozygous males and females were mated and a total of 47 pairs of WT and EpCAM−/− embryos and pups were used for the current study. E18.5 embryos were harvested from pregnant females. E18.5 embryos and P0 and P3 pups were sacrificed, after which their intestines were collected for experiments. Pregnant mice were sacrificed by cervical dislocation. E18.5 embryos were obtained by dissecting pregnant females. E18.5 Embryos, P0 and P3 pups were sacrificed humanely by decapitation. The information of the number and genotype of mice used in the present work was summarized in Table SI.

Histological analysis

For histological analysis, duodenum, jejunum, ileum and colon tissues from WT and EpCAM−/− mice were fixed overnight with 4% paraformaldehyde in PBS at 4°C before being dehydrated and embedded in paraffin. Then, hematoxylin and eosin (H&E) staining was performed on 4-µm paraffin sections; the sections were incubated in hematoxylin for 2 min and eosin for 30 sec at room temperature. Images were captured using the PerkinElmer Automated Quantitative Pathology System.

Immunofluorescence staining

For immunofluorescence staining, duodenum, jejunum, ileum and colon tissues from WT and EpCAM−/− mice were fixed overnight with 4% para-formaldehyde in PBS at 4°C before being dehydrated and embedded in optimal cutting temperature compound (OCT) (Sakura Finetek). Then, 7-µm frozen sections were boiled in 10 mM citric acid (Merck) at pH 6.0 for 5 min. After exposure to goat serum blocking buffer (ZSGB-BIO, ZLI-9056) at room temperature for 1 h, the sections were incubated overnight at 4°C with primary antibodies and then with secondary antibodies at room temperature for 1 h. Primary antibodies were used as follows: Rabbit anti-E-cadherin (Cell Signaling Technology, Inc.; cat. no. 14472, 1:200), rabbit anti-p120-catenin (Santa Cruz Biotechnology, Inc.; cat. no. 15D2, 1:200), and rabbit anti-β-catenin (BD Biosciences; cat. no. 610154, 1:200). Immunofluorescence analysis was performed with Alexa Fluor 488-labeled donkey anti-rabbit IgG (H+L) secondary antibodies (Thermo Fisher Scientific, Inc.; cat. no. A21206, 1:1,000), and DAPI (Sigma-Aldrich; Merck KGaA; cat. no. D9564, 1:10,000) was used to stain the nuclei of tissues. Immunofluorescence images were observed using a PerkinElmer Automated Quantitative Pathology System.

Reverse transcription-quantitative PCR (RT-qPCR)

Total RNA from mouse small intestines was extracted using an RNAiso Plus kit (Takara; cat. no. 9109), which was subjected to reverse transcription using a PrimeScript™ RT Reagent Kit with gDNA Eraser (Takara; cat. no. RR047A) at 37°C for 15 min and 85°C for 5 sec. The sequences of primers for RT-qPCR are listed in Table SII and they were produced by Sangon Biotech Co., Ltd. RT-qPCR was conducted using a TB Green® Premix Ex Taq™ II Kit (Takara; cat. no. RR820A) and the PikoReal PCR system (Thermo Fisher Scientific, Inc.). The thermocycling program was 95°C for 30 sec, followed by 40 cycles of 95°C for 5 sec, 60°C for 20 sec and 65°C for 15 sec. Mouse GAPDH was used as an internal reference gene.

Western blot analysis

The small and large intestines of mice were collected and washed in cold PBS (HyClone). Then, they were lysed in radioimmunoprecipitation assay lysis buffer containing 1% PMSF as well as 1% protease inhibitor cocktail (Dalian Meilun Biotechnology Co., Ltd.), and centrifuged at 13,680 × g at 4°C for 30 min, and the supernatant was frozen at -80°C. Protein concentration was determined by BCA assay (Beyotime), and equal amounts of protein (20 µg) were subjected to SDS-PAGE on a 10 or 12% gel. The separated proteins were transferred electrophoretically to a PVDF membrane, after which the membrane was blocked with 5% non-fat milk at room temperature for 1 h and then incubated with primary antibodies including E-cadherin (Cell Signaling Technology, Inc.; cat. no. 14472, 1:1,000), p120-catenin (Santa Cruz Biotechnology, Inc.; cat. no. 15D2, 1:1,000), β-catenin (BD Biosciences; cat. no. 610154, 1:1,000), nectin 1 (Abcam; cat. no. ab66985), and α-catenin (Sigma-Aldrich; Merck KGaA; cat. no. C2081) at 4°C overnight. Subsequently, the membrane was incubated with horseradish peroxidase-labeled antibodies [goat anti-rabbit IgG H&L (HRP) (Abcam; cat. no. ab6721, 1:2,000) and goat anti-mouse IgG H&L (HRP) (Abcam; cat. no. ab6789, 1:5,000)] at room temperature for 1 h. The resultant signals were detected using enhanced chemiluminescence reagent (Bio-Rad Laboratories, Inc.; cat. no. 170-5060). Western blotting bands were quantitatively analyzed using Lane 1d software (version 5.1.0.0; SageCreation).

Statistical analysis

Statistical differences were determined using SPSS software (23.0). Unpaired two sample t-test was used to determine the difference among groups. The data are presented as the mean ± standard deviation. P<0.05 was considered to indicate a statistically significant difference.

Results

The architecture of intestines from postnatal EpCAM mutant mice was gradually broken down during development

To study the functions of EpCAM in vivo, EpCAM−/− mice that we previously generated through CRISPR-Cas9 technology were used software used for densitometry (22). The phenotype of EpCAM−/− mice is similar to that of EpCAM mutant mice, which were generated using traditional gene targeting methods (9,10,15). The newborn EpCAM−/− mice were indistinguishable from their WT and heterozygous littermates at P0. However, there was no increase in the body size of EpCAM−/− pups during development, and the body weight of P3 EpCAM−/− pups was significantly lower than that of their littermates (Fig. 1A).

Since a loss-of-function mutation in EpCAM can cause CTE in patients (8) and the intestines were seriously affected in previously reported EpCAM mutant mouse models (9), the morphological and histological changes of intestines of EpCAM−/− mice from E18.5 to P3 were investigated in this study. It was first confirmed that the EpCAM protein was completely lost in the intestines of EpCAM−/− mice (Fig. 1B). The morphology of the intestines from EpCAM−/− mice looked normal for both E18.5 (Fig. 1C) and P0 (data not shown). Then, it became abnormal at P3. The length of the intestines of mutant mice at P3 was significantly shorter than that for WT pups at P3. The diameter of the ileum from the WT pups was thinner than that of the duodenum and jejunum, but in EpCAM−/− pups, the diameter of the ileum became larger than that of the duodenum and jejunum. Blood could be observed in the intestinal lumen of P3 EpCAM−/− mice (Fig. 1C). Tufts of villi could be detected in the small intestines of EpCAM−/− mice from E18.5 to P3, as shown by H&E staining (Figs. 1D and S1). There was no significant damage to villi from EpCAM−/− mice at E18.5 and P0 (Fig. S1). However, the intestinal epithelium was gradually broken down during development of the mutant mice after birth, and the size of intestinal epithelial cells in mutant mice at P3 became very large (Fig. 1D).

Proteins that compose adherens junctions gradually mislocalize in the epithelial cells of the small intestine in postnatal EpCAM mutant mice during development

To investigate whether the composition and functions of adherens junctions (AJs) in the intestinal epithelium were affected by the loss of EpCAM, the proteins that compose the AJs were analyzed via immunofluorescence (IF) staining. E-cadherin, β-catenin and p120 were only found to be localized to the membrane between epithelial cells in the intestines of WT mice from E18.5 to P3 (Figs. 2Figure 3-4). The localization of these proteins was normal in the intestines of EpCAM−/− mice at E18.5, and the IF signal of these proteins was even higher in the duodenum and jejunum of EpCAM−/− mice than that in the WT mice at this stage (Figs. 2A, 3A, and 4A, and S2A, S3A, and S4A). Since the intestinal tight junctions of EpCAM−/− mice were impaired at the E18.5 stage (10), the intestinal epithelium was affected, although it was still not clear at the morphology level. The higher IF signal was induced by the affected tissues of the EpCAM−/− mice. However, some of these proteins began to be detected in the cytoplasm of epithelial cells in the duodenum and jejunum of EpCAM−/− pups at P0 (Figs. 2B, 3B, and 4B, and S2B, S3B, and S4B). At P3, the signals of these proteins clearly localized to the cytoplasm of epithelial cells in the duodenum and jejunum of EpCAM−/− pups. The localization of E-cadherin, β-catenin and p120 in the epithelium of the ileum and colon in EpCAM−/− mice was still normal from E18.5 to P3. The signal of these proteins was not detected in the nucleus of the epithelial cells of the intestines of either WT or EpCAM−/− mice from E18.5 to P3 (Figs. 2C, 3C, and 4C, and S2C, S3C, and S4C).

Proteins that compose adherens junctions are gradually reduced in the intestinal epithelial cells of postnatal EpCAM mutant mice during development

The expression levels of proteins that compose adherens junctions in the small intestines were further analyzed by western blot analysis. The protein levels of E-cadherin, β-catenin and p120 in the small intestines of EpCAM−/− mice at E18.5 was slightly lower than that of WT mice, but their reduction was significant (Fig. 5A). At P0, the reduction in the protein levels of E-cadherin, β-catenin and p120 in the small intestines of EpCAM−/− mice was more apparent (Fig. 5B). At P3, only approximately half as much p120 protein was observed in the small intestines of EpCAM−/− pups, and the protein levels of E-cadherin and β-catenin in the small intestines of EpCAM−/− pups were reduced by more than half compared with the levels in WT pups (Fig. 5C).

mRNA expression of proteins that compose adherens junctions was normal in the intestines of postnatal EpCAM mutant mice during development

To examine how EpCAM regulated the localization and expression of proteins that compose adherens junctions, their gene expression was assessed at the mRNA level by qPCR. At E18.5 and P3, the expression of E-cadherin, β-catenin and p120 mRNA in the small intestines was similar between WT and EpCAM−/− mice (Fig. 6). The expression of E-cadherin mRNA in the small intestines of EpCAM−/− pups was increased compared with that of WT pups at the same stage, and the expression of β-catenin mRNA in the small intestines of EpCAM−/− mice was reduced, but these changes were not significant (Fig. 6).

Expression of nectin 1 became gradually reduced in the intestines of postnatal EpCAM mutant mice during development

The expression of genes that encoded proteins that compose adherens junctions was further assessed. Nectin 1, also known as PVRL1, which can regulate the assembly and adhesion activity of E-cadherin (23), was found to be significantly changed in the intestines of EpCAM−/− mice (Fig. 7A and B). The qPCR results showed that the expression of nectin 1 mRNA in the small intestines of EpCAM−/− mice was significantly reduced at P0 and P3, although it was still normal at E18.5 (Fig. 7A). The expression of nectin 1 protein in the small intestines of EpCAM−/− mice was normal at E18.5 and P0, but compared to control mice, there was only half as much nectin 1 protein in the intestines of P3 EpCAM−/− mice (Fig. 7B).

Discussion

In the present study, we confirmed that the adherens junctions in the upper part of the small intestinal epithelium of postnatal EpCAM−/− mice were gradually affected, but they were still normal in the lower part of the intestinal epithelium in EpCAM−/− mice. The results of H&E staining showed that the architecture of the duodenum and jejunum of EpCAM−/− mice at P3 was broken down, but the architecture of the ileum and colon was not affected even at P3. Moreover, the expression level and localization of E-cadherin, β-catenin and p120 were also gradually affected in the duodenum and jejunum of EpCAM−/− mice from E18.5 to P3 stages, but they were not affected in the ileum and colon of EpCAM−/− mice even at P3 stage. Finally, the expression of nectin 1 was reduced at both the mRNA and protein levels, which may be one of the important reasons for the breakdown of adherens junctions in the intestinal epithelium of postnatal EpCAM−/− mice. These results first showed the detailed process and mechanism of the adherens junctions breakdown in the intestinal epithelium of EpCAM−/− mice.

The intestine was the most severely damaged organ in the EpCAM mutant mice, and the mutation of EpCAM could also cause CTE in human. Therefore, we only assessed the AJs in the intestines at the current study. The assumption was that the AJs of some other organs in the EpCAM mutant mice may also be affected slightly, and we aim to test AJs in other tissues of EpCAM−/− mice in future studies. The function of EpCAM in maintaining the architecture of the intestinal epithelium has been confirmed by several studies, since it was found that the mutation of EpCAM could cause CTE (8,9-11,24). Total or partial villus atrophy and crypt hyperplasia could be found in the epithelium of small intestines from CTE patients (8,25). Lei et al found that the intestines of EpCAM knockout mice were relatively normal at the neonatal stage, as revealed by both macroscopic and histological data, but the disruption of mucosal architecture and sloughing of epithelial cells could be detected in EpCAM knockout mice at P5, especially in the duodenum and jejunum (10). Guerra et al also reported the increasing severity of villous atrophy and an increase in epithelial tufts in EpCAM mutant intestines (9). In the present study, significant changes in the intestinal epithelium of EpCAM−/− mice also occurred after birth. The upper part of the intestinal epithelium was broken down, but the lower part was normal even at P3. These results demonstrated that the intestinal architecture breakdown occurred earlier in the upper parts of the intestines than it did in the lower parts. Since the EpCAM mutation could impair tight junction formation in the intestinal epithelium, Lei et al suggested that the abundant proteases in the upper parts of the small intestines might be the cause of this phenotype (10). In the present study, we hypothesize that these proteases could easily penetrate the upper parts of the intestinal epithelium in the EpCAM mutant and cause damage. Therefore, many swollen cells appeared in the upper parts of the small intestines of EpCAM−/− mice, which could exacerbate the architectural breakdown of the upper parts of the small intestines in the EpCAM mutant. Therefore, the compositions of adherens junctions in the duodenum and jejunum of EpCAM mutant mice may be affected by proteases.

Several proteins that compose adherens junctions have been demonstrated to be essential for maintaining the homeostasis and morphogenesis of intestines. Specific knocking out E-cadherin in the intestinal epithelium of adult mice can cause hemorrhagic diarrhea because of the abnormal intestinal epithelial architecture (26), and deletion of E-cadherin from the developing mouse intestinal epithelium causes death shortly after birth because of the impairment of intestinal morphogenesis (27,28). Transgenic expression of a dominant-negative N-cadherin (DN-cadherin) in the mouse small intestine can cause loss of endogenous E-cadherin protein and induce Crohn-like inflammatory bowel disease at the age of 3 months, with most of these mice developing adenomas within 6 months (29). Hermiston et al also found that the forced expression of E-cadherin in the mouse intestinal epithelium resulted in slower cellular migration from crypts to villi (30). Knocking out p120 in the mouse intestinal epithelium resulted in mucosal damage and inflammation, leading to bleeding and death within 3 weeks of birth (31). Smalley-Freed et al found that limited ablation of p120-catenin in the adult mouse intestinal epithelium could promote adenoma formation by an indirect non-cell autonomous mechanism (32), and Short et al confirmed that p120 was an obligate haploinsufficient tumor suppressor in intestinal neoplasia, as shown by a conditional p120 knockout in Apc-sensitized mouse models (33). β-catenin is the key molecule in the Wnt signaling pathway (34), and it is essential for many biological processes in the intestines, such as maintenance of intestinal stem cells, inflammation and carcinogenesis (15,35-38). Therefore, any factor that could affect the expression or localization of proteins that compose adherens junctions in the intestines would affect the homeostasis of the intestinal epithelium. Since EpCAM is essential for maintaining the homeostasis of the intestinal epithelium, previous studies have tried to elucidate the changes in proteins that compose adherens junctions in the EpCAM mutant intestines (9,10,21), but their conclusions were based on different developmental stages and different parts of the intestines. Furthermore, intestinal tissues from CTE patients are very difficult to obtain. In the present study, we compared the whole intestines of WT and EpCAM−/− mice at E18.5, P0 and P3, and we found that the E-cadherin, p120 and β-catenin proteins in the duodenum and jejunum of P0 EpCAM−/− mice started to decrease and mislocalize at P0, and they were seriously affected at P3, but these proteins in the ileum and colon were not affected even at P3. These results demonstrated that EpCAM could protect the architecture of the duodenum and jejunum by maintaining the expression and localization of proteins that compose adherens junctions in these areas. The mRNA levels of these adherens junction-associated genes were not decreased, so we concluded that EpCAM may regulate the expression of these genes at the post-transcriptional level.

E-cadherin, β-catenin, and p120 are three important proteins which composed adherens junctions, and the inter-actions between them have been clearly studied. There is a multiple complex at adherens junctions, and the binding sides of β-catenin and p120 on E-cadherin are very clear now (39). If EpCAM could directly interact with one of these proteins in the multiple complex of the adherens junctions, it could be detected by Co-IP experiments. However, Wu et al have found that EpCAM and E-cadherin were not tightly associated in T84 cells through Co-IP experiments (16). It means that EpCAM is not directly associated with one of the proteins in the multiple complex of the adherens junctions.

Nectins are a type of immunogroblin (Ig)-like cell adhesion molecule, and they can regulate the formation of adherens junctions and tight junctions (40,41). We found that the mRNA level of nectin 1 was significantly reduced at the P0 and P3 stages, while its protein level was also significantly reduced at the P3 stage in EpCAM−/− mice. Sato et al reported that nectin 1 could regulate the assembly and adhesion activity of E-cadherin in MDCK cells (23). Martinez-Rico et al found that nectin 1 had important roles in the regulation of E-cadherin-based adhesion (42). We hypothesize that the reduction of nectin 1 may be one of the important reasons for the decrease and mislocalization of proteins that compose adherens junctions. It has been reported that EpCAM could control at least four independent signaling pathways (1-3), and the expression of nectin 1 may control one or more of these signaling pathways. Furthermore, the impaired tight junctions of the intestinal epithelium from EpCAM−/− mice may disrupt intracellular signaling pathways which are essential to regulate the nectin 1 expression. Our previous study had already shown that claudin proteins were downregulated in the intestines of EpCAM mutant mice (10).

In summary, we examined the expression of claudin-7 at both mRNA and protein levels after obtaining the current EpCAM−/− model through CRISPR/Cas9 technology. The results yielded were similar to those of our previous studies. Since the current study focused on adherens junctions of the intestinal epithelium, the claudin-7 results were not included. Nevertheless, further investigations are needed to examine the manner in which EpCAM regulates the expression of nectin 1.

Supplementary Data

Funding

This study was supported by the National Natural Science Foundation of China (grant nos. 81830113, 81803912, 31671520, and 81530102), the Major Basic and Applied Basic Research Projects of Guangdong Province of China (grant no. 2019B030302005), the National Key R&D plan 'Research on modernization of traditional Chinese medicine' (grant no. 2018YFC1704200), the Opening Foundation of the Key Laboratory of Regenerative Biology, the Guangzhou Institutes of Biomedicine and Health, the Chinese Academy of Sciences (grant no. KLRB201807), the Science and Technology Planning Project of Guangzhou City (grant no. 201803010069), the Scientific Research Project of the Administration of Traditional Chinese Medicine of Guangdong Province (grant no. 20182079), the Characteristic Innovation Project (Natural Science) of the Education Department of Guangdong Province and the 'Innovation Strong School Project' of Guangdong Pharmaceutical University (grant no. 2017KTSCX102), and the Science and Technology Project of Yue-Xiu District of Guangzhou (grant no. 2018-WS-011).

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

JG and ZL designed the study and conceived its execution. ZL and YY analyzed and interpreted the results, and wrote and revised the manuscript. GC, WL and LH maintained the mouse model and performed all qPCR and immunostaining experiments. YY, LY, YL, and HW performed the H&E staining and western blots. All authors read the final manuscript and approved it.

Ethics approval and consent to participate

All mouse experiments were approved by the Committee on Laboratory Animal Care and Use of Guangdong Pharmaceutical University [Guangzhou, China (gdpu2016073)].

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

References

1 

Balzar M, Winter MJ, de Boer CJ and Litvinov SV: The biology of the 17-1A antigen (Ep-CAM). J Mol Med (Berl). 77:699–712. 1999. View Article : Google Scholar

2 

Huang L, Yang Y, Yang F, Liu S, Zhu Z, Lei Z and Guo J: Functions of EpCAM in physiological processes and diseases (Review). Int J Mol Med. 42:1771–1785. 2018.PubMed/NCBI

3 

Schnell U, Cirulli V and Giepmans BN: EpCAM: Structure and function in health and disease. Biochim Biophys Acta. 1828:1989–2001. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Slanchev K, Carney TJ, Stemmler MP, Koschorz B, Amsterdam A, Schwarz H and Hammerschmidt M: The epithelial cell adhesion molecule EpCAM is required for epithelial morphogenesis and integrity during zebrafish epiboly and skin development. PLoS Genet. 5:e10005632009. View Article : Google Scholar : PubMed/NCBI

5 

Kamimoto K, Kaneko K, Kok CY, Okada H, Miyajima A and Itoh T: Heterogeneity and stochastic growth regulation of biliary epithelial cells dictate dynamic epithelial tissue remodeling. Elife. 5:e150342016. View Article : Google Scholar : PubMed/NCBI

6 

Schmelzer E, Zhang L, Bruce A, Wauthier E, Ludlow J, Yao HL, Moss N, Melhem A, McClelland R, Turner W, et al: Human hepatic stem cells from fetal and postnatal donors. J Exp Med. 204:1973–1987. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Carneiro FP, Muniz-Junqueira MI, De Vasconcelos Carneiro M, De Araújo Oliveira Í, Soares AC, De Vargas Haar N, Takano GHS, De Sousa Vianna LM, De Carvalho Caldas G, Vieira DLM, et al: Anti-EpCAM antibodies for detection of metastatic carcinoma in effusions and peritoneal wash. Oncol Lett. 18:2019–2024. 2019.PubMed/NCBI

8 

Sivagnanam M, Mueller JL, Lee H, Chen Z, Nelson SF, Turner D, Zlotkin SH, Pencharz PB, Ngan BY, Libiger O, et al: Identification of EpCAM as the gene for congenital tufting enteropathy. Gastroenterology. 135:429–437. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Guerra E, Lattanzio R, La Sorda R, Dini F, Tiboni GM, Piantelli M and Alberti S: mTrop1/Epcam knockout mice develop congenital tufting enteropathy through dysregulation of intestinal E-cadherin/β-catenin. PLoS One. 7:e493022012. View Article : Google Scholar

10 

Lei Z, Maeda T, Tamura A, Nakamura T, Yamazaki Y, Shiratori H, Yashiro K, Tsukita S and Hamada H: EpCAM contributes to formation of functional tight junction in the intestinal epithelium by recruiting claudin proteins. Dev Biol. 371:136–145. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Mueller JL, McGeough MD, Peña CA and Sivagnanam M: Functional consequences of EpCam mutation in mice and men. Am J Physiol Gastrointest Liver Physiol. 306:G278–G288. 2014. View Article : Google Scholar :

12 

Ng V, Ang S, Chan J and Choo A: Characterization of epithelial cell adhesion molecule as a surface marker on undifferentiated human embryonic stem cells. Stem Cells. 28:29–35. 2010. View Article : Google Scholar

13 

Gerlach J, Foka H, Thompson R, Gridelli B and Schmelzer E: Epithelial cell adhesion molecule fragments and signaling in primary human liver cells. J Cell Physiol. 233:4841–4851. 2018. View Article : Google Scholar

14 

Nagao K, Zhu J, Heneghan M, Hanson J, Morasso M, Tessarollo L, Mackem S and Udey MC: Abnormal placental development and early embryonic lethality in EpCAM-null mice. PLoS One. 4:e85432009. View Article : Google Scholar :

15 

Das S, Yu S, Sakamori R, Vedula P, Feng Q, Flores J, Hoffman A, Fu J, Stypulkowski E, Rodriguez A, et al: Rab8a vesicles regulate Wnt ligand delivery and Paneth cell maturation at the intestinal stem cell niche. Development. 142:2147–2162. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Wu CJ, Mannan P, Lu M and Udey MC: Epithelial cell adhesion molecule (EpCAM) regulates claudin dynamics and tight junctions. J Biol Chem. 288:12253–12268. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Litvinov SV, Balzar M, Winter MJ, Bakker HA, Briaire-de Bruijn IH, Prins F, Fleuren GJ and Warnaar SO: Epithelial cell adhesion molecule (Ep-CAM) modulates cell-cell interactions mediated by classic cadherins. J Cell Biol. 139:1337–1348. 1997. View Article : Google Scholar

18 

Winter MJ, Nagelkerken B, Mertens AE, Rees-Bakker HA, Briaire-de Bruijn IH and Litvinov SV: Expression of Ep-CAM shifts the state of cadherin-mediated adhesions from strong to weak. Exp Cell Res. 285:50–58. 2003. View Article : Google Scholar : PubMed/NCBI

19 

Maghzal N, Kayali HA, Rohani N, Kajava AV and Fagotto F: EpCAM controls actomyosin contractility and cell adhesion by direct inhibition of PKC. Dev Cell. 27:263–277. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Salomon J, Gaston C, Magescas J, Duvauchelle B, Canioni D, Sengmanivong L, Mayeux A, Michaux G, Campeotto F, Lemale J, et al: Contractile forces at tricellular contacts modulate epithelial organization and monolayer integrity. Nat Commun. 8:139982017. View Article : Google Scholar : PubMed/NCBI

21 

Patey N, Scoazec JY, Cuenod-Jabri B, Canioni D, Kedinger M, Goulet O and Brousse N: Distribution of cell adhesion molecules in infants with intestinal epithelial dysplasia (tufting enter-opathy). Gastroenterology. 113:833–843. 1997. View Article : Google Scholar : PubMed/NCBI

22 

Yang Y, Liu S, Lei Z, Chen G, Huang L, Yang F, Lei Y, Liu Y, Yang L, Liu W, et al: Circular RNA profile in liver tissue of EpCAM knockout mice. Int J Mol Med. 44:1063–1077. 2019.PubMed/NCBI

23 

Sato T, Fujita N, Yamada A, Ooshio T, Okamoto R, Irie K and Takai Y: Regulation of the assembly and adhesion activity of E-cadherin by nectin and afadin for the formation of adherens junctions in Madin-Darby canine kidney cells. J Biol Chem. 281:5288–5299. 2006. View Article : Google Scholar

24 

Pathak SJ, Mueller JL, Okamoto K, Das B, Hertecant J, Greenhalgh L, Cole T, Pinsk V, Yerushalmi B, Gurkan OE, et al: EPCAM mutation update: Variants associated with congenital tufting enteropathy and Lynch syndrome. Hum Mutat. 40:142–161. 2019. View Article : Google Scholar :

25 

Sherman PM, Mitchell DJ and Cutz E: Neonatal enteropathies: Defining the causes of protracted diarrhea of infancy. J Pediatr Gastroenterol Nutr. 38:16–26. 2004. View Article : Google Scholar

26 

Schneider MR, Dahlhoff M, Horst D, Hirschi B, Trülzsch K, Müller-Höcker J, Vogelmann R, Allgäuer M, Gerhard M, Steininger S, et al: A key role for E-cadherin in intestinal homeo-stasis and Paneth cell maturation. PLoS One. 5:e143252010. View Article : Google Scholar

27 

Bondow BJ, Faber ML, Wojta KJ, Walker EM and Battle MA: E-cadherin is required for intestinal morphogenesis in the mouse. Dev Biol. 371:1–12. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Gloushankova NA, Rubtsova SN and Zhitnyak IY: Cadherin-mediated cell-cell interactions in normal and cancer cells. Tissue Barriers. 5:e13569002017. View Article : Google Scholar : PubMed/NCBI

29 

Hermiston ML and Gordon JI: Inflammatory bowel disease and adenomas in mice expressing a dominant negative N-cadherin. Science. 270:1203–1207. 1995. View Article : Google Scholar : PubMed/NCBI

30 

Hermiston ML, Wong MH and Gordon JI: Forced expression of E-cadherin in the mouse intestinal epithelium slows cell migration and provides evidence for nonautonomous regulation of cell fate in a self-renewing system. Genes Dev. 10:985–996. 1996. View Article : Google Scholar : PubMed/NCBI

31 

Smalley-Freed WG, Efimov A, Short SP, Jia P, Zhao Z, Washington MK, Robine S, Coffey RJ and Reynolds AB: Adenoma formation following limited ablation of p120-catenin in the mouse intestine. PLoS One. 6:e198802011. View Article : Google Scholar : PubMed/NCBI

32 

Smalley-Freed WG, Efimov A, Burnett PE, Short SP, Davis MA, Gumucio DL, Washington MK, Coffey RJ and Reynolds AB: p120-catenin is essential for maintenance of barrier function and intestinal homeostasis in mice. J Clin Invest. 120:1824–1835. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Short SP, Kondo J, Smalley-Freed WG, Takeda H, Dohn MR, Powell AE, Carnahan RH, Washington MK, Tripathi M, Payne DM, et al: p120-Catenin is an obligate haploinsufficient tumor suppressor in intestinal neoplasia. J Clin Invest. 127:4462–4476. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Nusse R and Clevers H: Wnt/β-catenin signaling, disease, and emerging therapeutic modalities. Cell. 169:985–999. 2017. View Article : Google Scholar : PubMed/NCBI

35 

Chiacchiera F, Rossi A, Jammula S, Piunti A, Scelfo A, Ordóñez-Morán P, Huelsken J, Koseki H and Pasini D: Polycomb complex PRC1 preserves intestinal stem cell identity by sustaining Wnt/β-catenin transcriptional activity. Cell Stem Cell. 18:91–103. 2016. View Article : Google Scholar

36 

Ahmad R, Kumar B, Chen Z, Chen X, Müller D, Lele SM, Washington MK, Batra SK, Dhawan P and Singh AB: Loss of claudin-3 expression induces IL6/gp130/Stat3 signaling to promote colon cancer malignancy by hyperactivating Wnt/β-catenin signaling. Oncogene. 36:6592–6604. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Janeckova L, Fafilek B, Krausova M, Horazna M, Vojtechova M, Alberich-Jorda M, Sloncova E, Galuskova K, Sedlacek R, Anderova M and Korinek V: Wnt signaling inhibition deprives small intestinal stem cells of clonogenic capacity. Genesis. 54:101–114. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Sun X, Yao L, Liang H, Wang D, He Y, Wei Y, Ye L, Wang K, Li L, Chen J, et al: Intestinal epithelial PKM2 serves as a safeguard against experimental colitis via activating β-catenin signaling. Mucosal Immunol. 12:1280–1290. 2019. View Article : Google Scholar : PubMed/NCBI

39 

Kobielak A and Fuchs E: Alpha-catenin: At the junction of inter-cellular adhesion and actin dynamics. Nat Rev Mol Cell Biol. 5:614–625. 2004. View Article : Google Scholar : PubMed/NCBI

40 

Shimono Y, Rikitake Y, Mandai K, Mori M and Takai Y: Immunoglobulin superfamily receptors and adherens junctions. Subcell Biochem. 60:137–170. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Takai Y, Ikeda W, Ogita H and Rikitake Y: The immunoglobulin-like cell adhesion molecule nectin and its associated protein afadin. Annu Rev Cell Dev Biol. 24:309–342. 2008. View Article : Google Scholar : PubMed/NCBI

42 

Martinez-Rico C, Pincet F, Perez E, Thiery JP, Shimizu K, Takai Y and Dufour S: Separation force measurements reveal different types of modulation of E-cadherin-based adhesion by nectin-1 and -3. J Biol Chem. 280:4753–4760. 2005. View Article : Google Scholar

Related Articles

Journal Cover

February-2021
Volume 47 Issue 2

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen G, Yang Y, Liu W, Huang L, Yang L, Lei Y, Wu H, Lei Z and Guo J: EpCAM is essential for maintenance of the small intestinal epithelium architecture via regulation of the expression and localization of proteins that compose adherens junctions. Int J Mol Med 47: 621-632, 2021
APA
Chen, G., Yang, Y., Liu, W., Huang, L., Yang, L., Lei, Y. ... Guo, J. (2021). EpCAM is essential for maintenance of the small intestinal epithelium architecture via regulation of the expression and localization of proteins that compose adherens junctions. International Journal of Molecular Medicine, 47, 621-632. https://doi.org/10.3892/ijmm.2020.4815
MLA
Chen, G., Yang, Y., Liu, W., Huang, L., Yang, L., Lei, Y., Wu, H., Lei, Z., Guo, J."EpCAM is essential for maintenance of the small intestinal epithelium architecture via regulation of the expression and localization of proteins that compose adherens junctions". International Journal of Molecular Medicine 47.2 (2021): 621-632.
Chicago
Chen, G., Yang, Y., Liu, W., Huang, L., Yang, L., Lei, Y., Wu, H., Lei, Z., Guo, J."EpCAM is essential for maintenance of the small intestinal epithelium architecture via regulation of the expression and localization of proteins that compose adherens junctions". International Journal of Molecular Medicine 47, no. 2 (2021): 621-632. https://doi.org/10.3892/ijmm.2020.4815