Cx32 reverses epithelial-mesenchymal transition in doxorubicin-resistant hepatocellular carcinoma

  • Authors:
    • Meiling Yu
    • Guangshu Han
    • Benquan Qi
    • Xiaoxiang Wu
  • View Affiliations

  • Published online on: February 17, 2017     https://doi.org/10.3892/or.2017.5462
  • Pages: 2121-2128
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Recently, epithelial-mesenchymal transition (EMT) has been reported to be an important mechanism of drug resistance in numerous types of cancer cells, including hepatocellular carcinoma (HCC). However, the underlying mechanisms remain to be fully elucidated. Connexin (Cx)32 plays a crucial role in hepatocarcinogenesis. The present study investigated the role of Cx32 in the regulation of chemotherapy-induced EMT in HCC. We found that the expression levels of Cx32 and E-cadherin were clearly decreased in HCC tissues compared with the corresponding paracancerous tissues, while the expression level of vimentin was significantly enhanced in HCC tissues. The expression of Cx32 had a strong correlation with the expression of E-cadherin and vimentin. In an in vitro study, a doxorubicin (DOX)-resistant liver cell line HepG2/DOX was established from parental HepG2 cells. The results showed that HepG2/DOX cells acquired EMT characteristics, with a decreased expression level of E-cadherin and an enhanced expression level of vimentin, and possessed high migratory abilities and invasiveness. Meanwhile, Cx32 was significantly decreased in the HepG2/DOX cells. Knockdown of Cx32 by shRNA in HepG2 cells induced EMT, while overexpression of Cx32 converted EMT to mesenchymal-epithelial transition (MET) in the HepG2/DOX cells. These results suggest that Cx32 is an important regulator of DOX-induced EMT in HCC. Cx32 could be considered as a novel target to reverse DOX resistance in HCC.

Introduction

Hepatocellular carcinoma (HCC) is the one of the most common human types of cancer with a high mortality rate worldwide (1). To date, surgical resection is still the best option for early-stage patients. However, most patients at the time of diagnosis are in the late stages. Thus, chemotherapy and radiation therapy are the main therapies for patients who are not suitable candidates for surgery (2). Doxorubicin (DOX) is a widely used chemotherapy agent for the clinical treatment of liver cancer. However, during clinical treatment, the development of drug resistance limits the application of DOX (3,4). Thus, it is necessary to investigate the underlying mechanism of drug resistance of HCC to DOX.

Studies suggest that the occurrence of epithelial-mesenchymal transition (EMT) has an intimate correlation with the process of tumor resistance (58). EMT is a special physiological process whereby epithelial cells transform into motile mesenchymal cells, with the downregulation of epithelial characteristics (E-cadherin), the enhancement of mesenchymal characteristics (vimentin, N-cadherin, Snail and Slug) and increased abilities of invasion and migration.

Connexins (Cxs) have been revealed to be proteins which form gap junction channels and mediate the communication between cells (911). The intercellular communication is necessary for managing various pathological processes of the human body, such as cell proliferation, differentiation, tissue homeostasis and wound healing (12). Cxs decrease significantly during the process of carcinogenesis and when cancer cells acquire drug resistance. Studies have indicated that Cx genes are tumor-suppressor genes (1315) and that recovering or increasing Cx expression is a new strategy that may be used for cancer therapy.

In the present study, we found that the DOX-resistant human liver cancer HepG2 cell line (HepG2/DOX) exhibited EMT characteristics and that the expression level of Cx32 was gradually decreased after HepG2 cells became resistant to DOX. We also demonstrated that Cx32 regulated EMT in HepG2 and HepG2/DOX cells. By detecting the expression levels of Cx32, E-cadherin and vimentin in HCC and the corresponding paracancerous tissues, the results revealed that the expression levels of Cx32 and E-cadherin were clearly decreased in HCC tissues when compared with the levels in the corresponding paracancerous tissues, while the expression of vimentin was significantly enhanced in the HCC tissues. Moreover, the expression of Cx32 had a strong correlation with the expression of E-cadherin and vimentin. These results suggest that Cx32 is an important regulator of DOX-induced EMT in HCC.

Materials and methods

Immunohistochemical samples

HCC and paracancerous tissue specimens were collected from 40 HCC patients who underwent surgical resection at the Hepatobiliary Surgical Department of The First Affiliated Hospital of Bengbu Medical College (Bengbu, China) from February 2014 to June 2015. Paracancerous tissue was the non-cancerous liver tissue located 3–5 cm from the primary tumor. No patients received radiotherapy, chemotherapy, immunotherapy or other related treatment before surgery. For all patients, complete clinical and pathological data were obtained. All specimens were fixed with neutral formalin solution, paraffin-embedded and continuously sectioned to a 5-µm thickness. The specimen collection protocol was approved by the Medical Ethics Committee of the Bengbu Medical College.

Immunohistochemistry

Specimens were embedded in 10% paraffin and cut by a microtome into 5-µm sections. All paraffin sections were routinely deparaffinized and rehydrated in xylene, and graded ethanol solutions. Endogenous peroxidase activity was blocked by 0.3% H2O2 in methanol for 20 min. Then, the sections were incubated with the primary antibodies [Cx32 (1:100), E-cadherin (1:400) and vimentin (1:100); Cell Signaling Technology, Danvers, MA, USA] at 4̊C overnight. Subsequently, the sections were exposed to the biotin-labeled secondary antibody for 20 min. Immunostaining was visualized with 3,3-diaminobenzidine (DAB) and hematoxylin counterstaining. Phosphate-buffered saline (PBS) instead of a primary antibody was used as the negative control. Positive staining was observed and images were captured with an optical microscope. The stained cells were scored as follows: negative (−), stained cells <5%; positive (+), stained cells 5–10%; positive (++), stained cells >10–50%; positive (+++), stained cells >50–75%; positive (++++), stained cells >75%.

Materials

The HepG2 cell line was obtained from Shanghai Bai Li Biological Technology Co., Ltd. (Shanghai, China). Cell culture reagents were obtained from Invitrogen (Carlsbad, CA, USA). DOX and dimethyl sulfoxide (DMSO) were obtained from Sigma-Aldrich (St. Louis, MO, USA). Antibodies for E-cadherin, vimentin, β-actin and the secondary antibodies were obtained from Cell Signaling Technology. shRNA-Cx32, pcDNA-Cx32 and Lipofectamine™ 2000 were obtained from Shanghai GenePharma Co., Ltd. (Shanghai, China).

MTT assay

Cell viability was evaluated by an MTT assay as previously described (16). The cell inhibition rate was calculated to determine the standard pharmacological activity at OD490 nm. Briefly, cells were harvested and seeded in 96-well plates. Various concentrations of DOX were added into the wells followed by 24 h of incubation (at 37̊C with 5% CO2). Then, 20 µl of 5 mg/ml MTT was added into each well after the removal of the culture medium. Next, the cells were incubated for an additional 4 h. Subsequently, 150 µl of DMSO was used after discarding the culture medium. Plates were placed on a plate shaker for 10 min and the absorbance was assessed by a microplate reader at 490 nm.

Western blotting

Cell proteins were isolated using protein lysis buffer at 4̊C. Equal amounts of cell protein samples were separated by SDS-polyacrylamide (10%) gel electrophoresis, and then transferred to polyvinylidene fluoride (PVDF) membranes. Then, the membranes were blocked using 5% bovine serum albumin in Tris-buffered saline Tween-20 (TBST) for 2 h. Subsequently, the membranes were incubated with the specific antibodies overnight, followed by incubation with a secondary antibody for 2 h. Finally, the protein bands were exposed using ECL-detecting reagents.

Transwell assay

After Transwell chambers in 24-well plates were coated with 1 mg/ml of Matrigel matrix or not, cells were harvested and 5×105 cells/ml were seeded into the Transwell chambers. The culture medium was removed after 24 h of incubation (at 37̊C with 5% CO2). Then, the cells were fixed with 4% paraformaldehyde for 15 min and stained with 1% crystal violet for another 15 min. Subsequently, the inner-cells were wiped off with a swab and the cells were counted in random fields using a light microscope.

Cell transfection

Cells were randomly divided into 3 groups: the control, the negative control (transfected with an empty plasmid) and the shRNA-Cx32 or pcDNA-Cx32 group (transfected with Cx32-gene plasmids). According to the manufacturers instructions, following incubation of cells in 6-well plates for 24 h, shRNA-Cx32 or pcDNA-Cx32 genes were transfected into HepG2 or HepG2/DOX cells using Lipofectamine 2000, and then the cells were co-cultured with serum-free opti-MEM medium for 6 h. Subsequently, the old culture was discarded and was replaced with normal culture medium.

Statistical analysis

An unpaired Students t-test with SigmaPlot 10.0 software (Jandel Scientific, San Rafael, CA, USA) was used to assess the statistical data. Data are presented as the means ± SEM. Differences with P<0.05 are considered to have statistical significance. SPSS 17.0 was used to analyze the relationship between Cx32, E-cadherin and vimentin.

Results

Expression levels of Cx32, E-cadherin and vimentin in HCC specimens, and the corresponding paracancerous tissues

Forty patients (31 males and 9 females) were enrolled in the present study. The expression levels of Cx32, E-cadherin and vimentin in HCC tissues and adjacent non-tumorous liver samples were examined using immunohistochemistry (Fig. 1A-C). The results revealed that the expression levels of Cx32 and E-cadherin (an epithelial marker) were significantly decreased in the HCC tissues when compared to levels in the corresponding paracancerous tissues, while the expression of vimentin (a mesenchymal marker) was enhanced in the HCC specimens (Fig. 1D-F; Table I). HCC samples (55 and 40%) showed decreased Cx32 and E-cadherin expression respectively, while 62.5% of the HCC samples showed positive expression of vimentin.

Table I.

Positive rates of Cx32, E-cadherin and vimentin in the HCC and paracancerous tissues.

Table I.

Positive rates of Cx32, E-cadherin and vimentin in the HCC and paracancerous tissues.

HCC tissues (n=40)Paracancerous tissues (n=40)


Variant++++++++++%++++++++++%FisherP-value
Cx322211  16045.0  82  124580.032.207 <0.001a
E-cadherin1611  85060.0  3517  9692.546.567 <0.001a
Vimentin15  314  5362.5334  3  0017.511.9080.046a

a P<0.05 vs. paracancerous tissues. Cx32, connexin 32; HCC, hepatocellular carcinoma.

Spearman correlation analysis was used to observe the relationship between Cx32, E-cadherin and vimentin in the HCC samples. Table II shows that there is a strong correlation between Cx32, E-cadherin and vimentin in HCC tissues. The expression level of E-cadherin was positively correlated with the expression of Cx32, while the expression of vimentin was negatively correlated with the expression of Cx32.

Table II.

Correlation analysis of Cx32, E-cadherin and vimentin in the HCC samples.

Table II.

Correlation analysis of Cx32, E-cadherin and vimentin in the HCC samples.

Cx32

VariablesSpearman indicesP-value
E-cadherin   0.812 <0.001a
Vimentin−0.502   0.001a

a P<0.05 vs. Cx32. Cx32, Connexin 32; HCC, hepatocellularcarcinoma.

HepG2/DOX cell line is established

The HepG2/DOX cell line was induced via serially increasing DOX concentrations. An MTT assay was used to investigate the cytotoxicity of DOX in HepG2 cells (sensitive to DOX) and HepG2/DOX (resistant to DOX) cells. The results in Fig. 2 showed that the IC50 value of DOX in the HepG2 cells was 0.182 µg/ml, while the IC50 value of DOX in the HepG2/DOX cells was 2.058 µg/ml. Thus, the HepG2/DOX cells demonstrated an 11.31-fold higher resistance to DOX than the HepG2 cells.

HepG2/DOX cells undergo EMT

The expression of major markers of EMT in the HepG2 cells and HepG2/DOX cells were observed by western blotting. Results revealed that epithelial marker E-cadherin was significantly decreased in the HepG2/DOX cells compared with that in the HepG2 cells. In contrast, the expression level of mesenchymal marker vimentin was clearly increased in the HepG2/DOX cells (Fig. 3A-C). In order to further observe the changes of EMT in HepG2/DOX cells, the invasive and migratory abilities of the HepG2/DOX and HepG2 cells were detected by Transwell assays. Fig. 3D-F revealed that compared to the HepG2 cells, the HepG2/DOX cells demonstrated higher invasion and migration potential. This result suggests that acquired resistance could enhance the invasion and migration abilities of HepG2 cells. All the aforementioned results demonstrated that the HepG2/DOX cells acquired an EMT phenotype.

Acquired resistance of DOX decreases the expression of Cx32

We investigated the expression of Cx32 by western blotting in HepG2 and HepG2/DOX cells. As shown in Fig. 4, the HepG2 cells showed a significantly increased expression level of Cx32 compared with that in the HepG2/DOX cells. This result demonstrated that Cx32 may play an important role in the mechanism of acquired resistance of HepG2 cells to DOX.

Cx32 regulates EMT in HCC
Inhibition of Cx32 induces EMT in HepG2 cells

The aforementioned results showed that the HepG2/DOX cells initiated EMT with decreased expression of Cx32. Then, we observed the effect of Cx32 on DOX-induced EMT in two ways: inhibition of Cx32 expression with shRNA-Cx32 in HepG2 cells, and overexpression of Cx32 with pcDNA-Cx32 in HepG2/DOX cells.

First, we decreased the expression of Cx32 in HepG2 cells by shRNA-Cx32. As shown in Fig. 5A and B, after transfecting the HepG2 cells with shRNA-Cx32, the expression of Cx32 was significantly decreased in the shRNA-Cx32 group when compared with the control or negative control group. Then, to explore the effect of Cx32 on EMT in HepG2 cells, we also detected the expression of EMT markers (E-cadherin and vimentin) by western blotting as well as the invasion and migration abilities of the cells by Transwell assays. The results in Fig. 5A and C demonstrated that the expression of E-cadherin in the shRNA-Cx32 group was clearly decreased when compared with the control or negative control group. Inversely, vimentin was significantly enhanced (Fig. 5A and D). The results of Transwell assays also revealed that the HepG2 cells acquired enhanced invasive and migratory abilities after the inhibition of the expression of Cx32 (Fig. 6A and D). These results suggest that downregulation of Cx32 by shRNA-Cx32 induced EMT in the HepG2 cells, which indicated that Cx32 is partly responsible for the initiation of EMT in HepG2 cells.

Overexpression of Cx32 converts EMT to mesenchymal-epithelial transition (MET) in HepG2/DOX cells

To further observe the role of Cx32 in the mechanism of DOX-induced EMT, we used a Cx32 gene fragment to overexpress Cx32 in HepG2/DOX cells which were resistant to DOX. After HepG2/DOX cells were transfected with pcDNA-Cx32 for 24 h, the expression level of Cx32 was detected by western blotting. The result revealed that the pcDNA-Cx32 group showed significantly increased expression of Cx32 when compared to that of the control or the negative control group (Fig. 5E and F). To explore the effect of the upregulation of Cx32 on EMT in HepG2/DOX cells, EMT markers E-cadherin and vimentin as well as the invasion and migration abilities of the cells were also examined. As shown in Fig. 5E and G, E-cadherin was significantly enhanced in the pcDNA-Cx32 group, while vimentin was obviously decreased (Fig. 5E and H). The invasion and migration abilities also decreased after transfecting HepG2/DOX cells with Cx32 (Fig. 6E-H). These findings indicate that overexpression of Cx32 reverses EMT to MET in HepG2/DOX cells.

Discussion

In recent years, studies have focused on the role of EMT in the acquired resistance of chemotherapeutics drugs (16,1720), and acquisition of EMT has recently been proposed as an important mechanism for drug resistance in cancer cells. For example, cisplatin-resistance in lung cancer cells, cisplatin-resistance in cervical cancer cells, crizotinib-resistant anaplastic lymphoma kinase (ALK)-positive lung cancer cells, gemcitabine-resistant hepatoma cells, tamoxifen-resistant MCF7 breast cancer cells all possess EMT characteristics. Recently, targeting EMT has been considered as a new strategy to combat cancer drug resistance (21). In the present study, we also found that during the process of acquisition of DOX resistance, HepG2/DOX cells exhibited an EMT phenotype.

Cxs have been reported to be the components of gap-junctional communication channels and important tumor suppressors (9,10,22). Recently, 21 connexins and related genes have been found in the human body (23), among them, Cx43, Cx26 and Cx32 have the most extensive distribution. Normal hepatocytes express Cx32 and to a lesser extent Cx26, which represent ~90 and 5%, respectively, of the total Cx amount in rat and human livers. Cx32 is uniformly distributed throughout the liver, while Cx26 is mainly expressed in the periportal acinar area (24). During the process of hepatocarcinogenesis, both Cx32 and Cx26 are decreased (24). In the present study, Cx26 was not detected in the HepG2 and HepG2/DOX cells (data not shown), while the aforementioned 2 cell lines expressed different levels of Cx32. Thus, the research focused on the effect of Cx32, but not Cx26, on DOX-induced EMT. Reports showed that Cx32 demonstrated a suppressive effect on liver inflammation, fibrosis, hepatocarcinogenesis and hepatoma cell metastasis (2528). Nakashima et al also found that the expression of Cx32 in hepatocellular carcinoma (HCC) patients was significantly decreased compared with the non-HCC patients (27). Cx32 overexpression decreased the malignant phenotype of liver tumors (29, 30). The present study reveals a new role of Cx32 in the regulation of cancer metastasis and drug resistance in HCC.

In our previous study (16), we found that cisplatin (CDDP)-resistant cell line A549/CDDP acquired an EMT phenotype. Overexpression of Cx43 converted EMT to MET and enhanced the sensitivity of CDDP to A549/CDDP cells. The results revealed that Cx43 was correlated with EMT in A549/CDDP cells. The present study further investigated the relationship between Cx and EMT in chemotherapy drug-resistant cells. We used HepG2 cells to establish the DOX-resisistant cell line. The present study demonstrated that when the HepG2 cells became resistant to DOX, the HepG2 cells acquired an EMT phenotype, meanwhile, Cx32 was significantly decreased in the HepG2/DOX cells. We also found that downregulation of Cx32 by shRNA-Cx32 induced EMT in the HepG2 cells, while overexpression of Cx32 reversed EMT in the HepG2/DOX cells. Thus, the results in HCC were consistent with the results in human lung adenocarcinoma, which suggest that Cx-induced EMT may be responsible for drug resistance in cancer cells and Cx is an important target for overcoming drug resistance when cells initiate EMT.

Cx hemichannels can supply a pathway for cellular signaling between cells and their extracellular environment (3133). The messengers that diffuse through Cx hemichannels usually include adenosine triphosphate (ATP), glutamate and glutathione. Studies have shown that both ATP and glutathione participate in the regulation of EMT in different cells (3439). Thus, an inference is that ATP and/or glutathione may be responsible for Cx32-mediated EMT in HCC. Studies may be performed in the future to observe the role of ATP and glutathione in Cx32-mediated EMT in HCC.

In summary, the present study is the first to establish the role of Cx32 in the occurrence of EMT in liver carcinoma cells. We demonstrated that the upregulation of the expression of Cx32 in HepG2/DOX cells inhibited EMT and enhanced DOX-induced cytotoxicity. Therefore, Cx32 may be a new target to potentiate DOX cytotoxicity in hepatic carcinoma in the future.

Acknowledgements

The present study was supported by the National Natural Science Foundation of Anhui (grant no. 1508085QH151) and the Natural Science Foundation of the Provincial Education Department of Anhui (no. KJ2015A147), the visiting project of the Provincial Education Department of Anhui (gxfxZD2016142), the National Natural Science Foundation of China (no. 81001457), and the Foundation of Bengbu Medical College (nos. Byycxz1422 and Byky1407ZD).

References

1 

Siegel R, Naishadham D and Jemal A: Cancer statistics, 2012. CA Cancer J Clin. 62:10–29. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Zhu AX: Systemic therapy of advanced hepatocellular carcinoma: How hopeful should we be? Oncologist. 11:790–800. 2006. View Article : Google Scholar : PubMed/NCBI

3 

Asghar U and Meyer T: Are there opportunities for chemotherapy in the treatment of hepatocellular cancer? J Hepatol. 56:686–695. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Xu F, Wang F, Yang T, Sheng Y, Zhong T and Chen Y: Differential drug resistance acquisition to doxorubicin and paclitaxel in breast cancer cells. Cancer Cell Int. 14:5382014. View Article : Google Scholar : PubMed/NCBI

5 

Harada K, Ferdous T and Ueyama Y: Establishment of 5-fluorouracil-resistant oral squamous cell carcinoma cell lines with epithelial to mesenchymal transition changes. Int J Oncol. 44:1302–1308. 2014.PubMed/NCBI

6 

Haider M, Zhang X, Coleman I, Ericson N, True LD, Lam HM, Brown LG, Ketchanji M, Nghiem B, Lakely B, et al: Epithelial mesenchymal-like transition occurs in a subset of cells in castration resistant prostate cancer bone metastases. Clin Exp Metastasis. 33:239–248. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Xu H, Pirisi L and Creek KE: Six1 overexpression at early stages of HPV16-mediated transformation of human keratinocytes promotes differentiation resistance and EMT. Virology. 474:144–153. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Dinicola S, Pasqualato A, Proietti S, Masiello MG, Palombo A, Coluccia P, Canipari R, Catizone A, Ricci G, Harrath AH, et al: Paradoxical E-cadherin increase in 5FU-resistant colon cancer is unaffected during mesenchymal-epithelial reversion induced by γ-secretase inhibition. Life Sci. 145:174–183. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Zhou JZ and Jiang JX: Gap junction and hemichannel-independent actions of connexins on cell and tissue functions - an update. FEBS Lett. 588:1186–1192. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Aasen T: Connexins: Junctional and non-junctional modulators of proliferation. Cell Tissue Res. 360:685–699. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Jiang JX and Gu S: Gap junction- and hemichannel-independent actions of connexins. Biochim Biophys Acta. 1711:208–214. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Diezmos EF, Bertrand PP and Liu L: Purinergic signaling in gut inflammation: The role of connexins and pannexins. Front Neurosci. 10:3112016. View Article : Google Scholar : PubMed/NCBI

13 

Cronier L, Crespin S, Strale PO, Defamie N and Mesnil M: Gap junctions and cancer: New functions for an old story. Antioxid Redox Signal. 11:323–338. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Kandouz M and Batist G: Gap junctions and connexins as therapeutic targets in cancer. Expert Opin Ther Targets. 14:681–692. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Trosko JE and Ruch RJ: Gap junctions as targets for cancer chemoprevention and chemotherapy. Curr Drug Targets. 0.3:465–482. 2002. View Article : Google Scholar

16 

Yu M, Zhang C, Li L, Dong S, Zhang N and Tong X: Cx43 reverses the resistance of A549 lung adenocarcinoma cells to cisplatin by inhibiting EMT. Oncol Rep. 31:2751–2758. 2014.PubMed/NCBI

17 

Song J and Li Y: miR-25-3p reverses epithelial-mesenchymal transition via targeting Sema4C in cisplatin-resistance cervical cancer cells. Cancer Sci. Oct 15–2016.(Epub ahead of print). doi: 10.1111/cas.13104.

18 

Gao HX, Yan L, Li C, Zhao LM and Liu W: miR-200c regulates crizotinib-resistant ALK-positive lung cancer cells by reversing epithelial-mesenchymal transition via targeting ZEB1. Mol Med Rep. 14:4135–4143. 2016.PubMed/NCBI

19 

Wu Q, Wang R, Yang Q, Hou X, Chen S, Hou Y, Chen C, Yang Y, Miele L, Sarkar FH, et al: Chemoresistance to gemcitabine in hepatoma cells induces epithelial-mesenchymal transition and involves activation of PDGF-D pathway. Oncotarget. 4:1999–2009. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Hiscox S, Jiang WG, Obermeier K, Taylor K, Morgan L, Burmi R, Barrow D and Nicholson RI: Tamoxifen resistance in MCF7 cells promotes EMT-like behaviour and involves modulation of β-catenin phosphorylation. Int J Cancer. 118:290–301. 2006. View Article : Google Scholar : PubMed/NCBI

21 

Du B and Shim JS: Targeting epithelial-mesenchymal transition (EMT) to overcome drug resistance in cancer. Molecules. 21:212016. View Article : Google Scholar

22 

Kay CW, Ursu D, Sher E and King AE: The role of Cx36 and Cx43 in 4-aminopyridine-induced rhythmic activity in the spinal nociceptive dorsal horn: An electrophysiological study in vitro. Physiol Rep. 4:42016. View Article : Google Scholar

23 

Unger VM, Kumar NM, Gilula NB and Yeager M: Three-dimensional structure of a recombinant gap junction membrane channel. Science. 283:1176–1180. 1999. View Article : Google Scholar : PubMed/NCBI

24 

Maes M, Decrock E, Cogliati B, Oliveira AG, Marques PE, Dagli ML, Menezes GB, Mennecier G, Leybaert L, Vanhaecke T, et al: Connexin and pannexin (hemi)channels in the liver. Front Physiol. 4:4052014. View Article : Google Scholar : PubMed/NCBI

25 

De Maio A, Gingalewski C, Theodorakis NG and Clemens MG: Interruption of hepatic gap junctional communication in the rat during inflammation induced by bacterial lipopolysaccharide. Shock. 14:53–59. 2000. View Article : Google Scholar : PubMed/NCBI

26 

Sagawa H, Naiki-Ito A, Kato H, Naiki T, Yamashita Y, Suzuki S, Sato S, Shiomi K, Kato A, Kuno T, et al: Connexin 32 and luteolin play protective roles in non-alcoholic steatohepatitis development and its related hepatocarcinogenesis in rats. Carcinogenesis. 36:1539–1549. 2015.PubMed/NCBI

27 

Nakashima Y, Ono T, Yamanoi A, El-Assal ON, Kohno H and Nagasue N: Expression of gap junction protein connexin32 in chronic hepatitis, liver cirrhosis, and hepatocellular carcinoma. J Gastroenterol. 39:763–768. 2004. View Article : Google Scholar : PubMed/NCBI

28 

Zhao B, Zhao W, Wang Y, Xu Y, Xu J, Tang K, Zhang S, Yin Z, Wu Q and Wang X: Connexin32 regulates hepatoma cell metastasis and proliferation via the p53 and Akt pathways. Oncotarget. 6:10116–10133. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Yang J, Ichikawa A and Tsuchiya T: A novel function of connexin 32: Marked enhancement of liver function in a hepatoma cell line. Biochem Biophys Res Commun. 307:80–85. 2003. View Article : Google Scholar : PubMed/NCBI

30 

Muramatsu A, Iwai M, Morikawa T, Tanaka S, Mori T, Harada Y and Okanoue T: Influence of transfection with connexin 26 gene on malignant potential of human hepatoma cells. Carcinogenesis. 23:351–358. 2002. View Article : Google Scholar : PubMed/NCBI

31 

Decrock E, Vinken M, De Vuyst E, Krysko DV, Herde DK, Vanhaecke T, Vandenabeele P, Rogiers V and Leybaert L: Connexin-related signaling in cell death: To live or let die? Cell Death Differ. 16:524–536. 2009. View Article : Google Scholar : PubMed/NCBI

32 

Chandrasekhar A and Bera AK: Hemichannels: Permeants and their effect on development, physiology and death. Cell Biochem Funct. 30:89–100. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Kar R, Batra N, Riquelme MA and Jiang JX: Biological role of connexin intercellular channels and hemichannels. Arch Biochem Biophys. 524:2–15. 2012. View Article : Google Scholar : PubMed/NCBI

34 

Unahabhokha T, Chanvorachote P, Sritularak B, Kitsongsermthon J and Pongrakhananon V: Gigantol Inhibits Epithelial to Mesenchymal Process in Human Lung Cancer Cells. Evid Based Complement Alternat Med. 2016:45616742016. View Article : Google Scholar : PubMed/NCBI

35 

Park SJ, Choi YS, Lee S, Lee YJ, Hong S, Han S and Kim BC: BIX02189 inhibits TGF-β1-induced lung cancer cell metastasis by directly targeting TGF-β type I receptor. Cancer Lett. 381:314–322. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Zhou Q, Abraham AD, Li L, Babalmorad A, Bagby S, Arcaroli JJ, Hansen RJ, Valeriote FA, Gustafson DL, Schaack J, et al: Topoisomerase IIα mediates TCF-dependent epithelial-mesenchymal transition in colon cancer. Oncogene. 35:4990–4999. 2016. View Article : Google Scholar : PubMed/NCBI

37 

Wang HJ, Zhu J and Zheng GY: Role of glutathione and other antioxidants in the inhibition of apoptosis and mesenchymal transition in rabbit lens epithelial cells. Genet Mol Res. 13:7149–7156. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Zhang J, Qiu M, Ma Y, Bu Y, Yang L and Tang X: Advanced oxidation protein products induce epithelial-to-mesenchymal transition in cultured human proximal tubular epithelial cells via oxidative stress. Nan Fang Yi Ke Da Xue Xue Bao. 34:659–663. 2014.(In Chinese). PubMed/NCBI

39 

Ryoo IG, Shin DH, Kang KS and Kwak MK: Involvement of Nrf2-GSH signaling in TGFβ1-stimulated epithelial-to-mesenchymal transition changes in rat renal tubular cells. Arch Pharm Res. 38:272–281. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April-2017
Volume 37 Issue 4

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yu M, Han G, Qi B and Wu X: Cx32 reverses epithelial-mesenchymal transition in doxorubicin-resistant hepatocellular carcinoma. Oncol Rep 37: 2121-2128, 2017
APA
Yu, M., Han, G., Qi, B., & Wu, X. (2017). Cx32 reverses epithelial-mesenchymal transition in doxorubicin-resistant hepatocellular carcinoma. Oncology Reports, 37, 2121-2128. https://doi.org/10.3892/or.2017.5462
MLA
Yu, M., Han, G., Qi, B., Wu, X."Cx32 reverses epithelial-mesenchymal transition in doxorubicin-resistant hepatocellular carcinoma". Oncology Reports 37.4 (2017): 2121-2128.
Chicago
Yu, M., Han, G., Qi, B., Wu, X."Cx32 reverses epithelial-mesenchymal transition in doxorubicin-resistant hepatocellular carcinoma". Oncology Reports 37, no. 4 (2017): 2121-2128. https://doi.org/10.3892/or.2017.5462