Effects of T‑cadherin expression on B16F10 melanoma cells

  • Authors:
    • Xin‑Suo Duan
    • Jie Lu
    • Zhi‑Hua Ge
    • En‑Hong Xing
    • Hai‑Tao Lu
    • Li‑Xin Sun
  • View Affiliations

  • Published online on: January 30, 2013     https://doi.org/10.3892/ol.2013.1164
  • Pages: 1205-1210
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Melanoma is one of the most deadly skin cancers. T‑cadherin is an atypical member of the cadherin superfamily as it lacks the transmembrane and cytoplasmic domains and is anchored to cell membranes through glycosylphosphatidylinositol (GPI) anchors. T‑cadherin downregulation is associated with a poorer prognosis in various carcinomas, such as lung, ovarian, cervical and prostate cancer, while in the majority of cancer cell lines, T‑cadherin re‑expression inhibits cell proliferation and invasiveness, increases susceptibility in apoptosis and reduces tumor growth in in vivo models. The functional relevance of T‑cadherin gene expression in melanoma progression remains to be clarified. The present study was designed for this purpose. The T‑cadherin gene was transfected into B16F10 melanoma cells to express T‑cadherin in the cells which were originally deficient in T‑cadherin expression. The proliferation, invasiveness, apoptosis and cell cycle of the transfected B16F10 melanoma cells were analyzed. The present study showed that the expression of T‑cadherin in B16F10 melanoma cells markedly reduced cell proliferation and permeation through Matrigel‑coated membranes, representing invasiveness. The percentage of early apoptotic cells and cells in the G2/M phase of the cell cycle was markedly increased compared with either parental B16F10 (without transfection) or empty pEGFP‑N1 (without T‑cadherin gene)‑transfected B16F10 cells, suggesting G2/M arrest, with similarity between the parental and empty pEGFP‑N1‑transfected B16F10 cells. T‑cadherin is important in melanoma progression and may be a possible target for therapy in melanoma and certain other types of cancer.

Introduction

Melanoma is one of the most deadly skin cancers and the incidence of melanoma is continuing to increase world-wide (1). Cutaneous malignant melanoma (CMM) is the sixth most commonly diagnosed cancer in the USA in males and females (2). One person succumbs to from melanoma every hour in the USA. Advanced-stage cutaneous melanoma has a median survival time of <1 year. An important feature of melanoma is that the incidence rate is highest in lighter skinned patients and much lower in darker skinned individuals. Furthermore, melanoma strikes individuals before old age (median age, 52 years), almost a decade before the majority of solid tumors arise (e.g., breast, colon, lung or prostate). Metastasis is the main contributor to the high mortality. Once diagnosed with metastatic melanoma (AJCC stage IV), the majority of patients ultimately succumb to the disease within two years (3). The median survival following the onset of distant metastases is only six to nine months and the five-year survival rate is <5% (4).

One of the most significant factors associated with tumor invasiveness and metastasis is altered cell-cell adhesion, which determines cell polarity and is involved in cell differentiation, as well as the establishment and maintenance of tissue homeostasis. During oncogenesis, this organized adhesion is disturbed by genetic and epigenetic changes, resulting in changes in signaling, loss of contact inhibition and altered cell migration and stromal interactions. A major class of cell-cell adhesion molecules is the cadherin superfamily of transmembrane glycoproteins that typically mediate calcium-dependent homophilic intercellular adhesion (5). E-cadherin is a prototypic member of the cadherin superfamily and has been characterized as a potent suppressor of invasion and metastasis in studies dating back to the 1990s (6). Perturbations in cadherins have been associated with cancer, particularly invasion and metastasis (7). The decreased expression and abnormal cellular distribution of E-cadherin have been frequently observed to be associated with de-differentiation and invasiveness in a variety of human malignancies (8). T-cadherin (also known as CDH13 and H-cadherin) is an atypical cadherin superfamily member which is anchored in the membrane through a glycosylphosphatidylinositol (GPI) anchor instead of a transmembrane domain (9). T-cadherin has also been suggested to be involved in cancer progression (10,11). T-cadherin downregulation has been observed in various malignant tumors, including malignant melanoma (12), gallbladder carcinoma (13,14), ovarian carcinoma (15,16) and breast (17) and lung cancers (18,19). T-cadherin re-expression may suppress cell proliferation and invasiveness, increase sensitivity to apoptosis and decrease tumor growth (12,2022).

The functional relevance of T-cadherin gene expression in melanoma progression remains to be further clarified. Our previous studies using B16F10 melanoma cells focused on the immune mechanisms and used Ganoderma lucidum polysaccharides to demonstrate the enhancement of MHC class I and costimulatory molecules in B16F10 cells (23), promotion of lymphocyte activation by B16F10 cells (24), antagonism against lymphocyte suppression caused by the culture super-natants of B16F10 melanoma cells (25), induction of marked cytotoxicity against B16F10 cells in cytotoxic T cells (CTLs) with granzyme B and porforin and inhibition of tumori-genesis in wild-type B16F10 melanoma cells in vivo(26). Cell proliferation, apoptosis, cell cycle and invasiveness are important features associated with malignancy (27). In the present study, the T-cadherin gene was expressed in B16F10 melanoma cells, which are deficient in T-cadherin expression, by T-cadherin gene transfection to examine its effects on proliferation, apoptosis, cell cycle and invasiveness in B16F10 melanoma cells.

Materials and methods

Cell culture system

The mouse B16F10 melanoma cell line, which is derived from C57BL/6 mice, was used in this study as it is originally deficient in T-cadherin expression. The B16F10 melanoma cell line was purchased from KeyGen Biotech. (Nanjing, China) and was cultured in RPMI-1640 medium supplemented with fetal bovine serum (10%), penicillin (100 IU/ml) and streptomycin (100 μg/ml) at 37°C in humidified 5% CO2 atmosphere.

Immunocytochemistry

The cells were cultured on slides and fixed with cold acetone for 5 min, then rinsed with distilled water. The endogenous peroxidase activity was quenched with 3% hydrogen peroxide. After blocking with 10% normal serum, rabbit polyclonal primary antibody against T-cadherin (Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA) was added at a 1:100 dilution and incubated overnight at 4°C. The next day, the horseradish peroxidase labelled secondary antibody was applied for 1 h and staining was finalized with a diaminobenzidine solution to detect the target antigen. Slides were extensively washed with phosphate-buffered saline (PBS) between the stages and counterstained with hematoxylin prior to mounting. The slides were examined under a light microscope. The replacement of primary antibody with PBS was used as a negative control.

RNA extraction and RT-PCR

The total ribonucleic acids (RNAs) were extracted with TRIzol reagent (Invitrogen, Carlsbad, CA, USA) according to the manufacturer’s instructions. The RNA precipitates were dissolved in DEPC-treated double distilled water containing RNasin (R) ribonuclease inhibitor (Tiangen Biotech, Beijing, China). The concentration of RNA was determined spectrophotometrically (BioSpec-mini; Shimadzu, Kyoto, Japan). The messenger RNA (mRNA) of T-cadherin in the tissues for expression vector engineering was amplified by the reverse transcription polymerase chain reaction (RT-PCR) with an RT-PCR system (Tiangen Biotech) according to the manufacturer’s instructions. The levels of T-cadherin mRNA transcribed in the transfected cells were also detected by RT-PCR. To ensure that equal amounts of starting material were used in each RT-PCR reaction, RNA was reverse transcribed and amplified with β-actin-specific primers. The number of PCR amplification cycles was 34, to ensure that the amplification of all specific complementary deoxyribonucleic acid (cDNA) products was exponential. The specific primer sequences were as follows: T-cadherin sense primer, 5′-CCGGAATTCATGCAGCCGAG AACTCCGCT-3′ (for gene cloning) or 5′-TTCAGCAGAAA GTGTTCCATAT-3′ (for expression determination); T-cadherin antisense primer, 5′- CGCGGATCCTCACAGACA AGCTAAGCTGAAG-3′ (for gene cloning) or 5′-GTG CATGGACGAACAGAGT-3′ (for expression determination); β-actin sense primer, 5′-CCTCGCCTTTGCCGATCC-3′; and β-actin antisense primer, 5’-GACTGACTACCTCATGA AGATCC-3’. All of the products were electrophoresed on 2% agarose gel and stained with ethidium bromide. The expression intensities of the bands were quantified using ImageJ software (National Institutes of Health, Bethesda, MA, USA) and expressed as a ratio (T-cadherin vs. β-actin).

Engineering of human T-cadherin expression vector and cell transfection

The pEGFP-N1/T-cad, a plasmid vector encoding human T-cadherin, was generated by inserting T-cadherin cDNA into a pEGFP-N1 vector (Clontech Laboratories, Inc., Mountain View, CA, USA), containing a CMV promoter and SV40 early promoter, as well as the neomycin/kanamycin resistance gene of Tn5 which allows stably transfected eukaryotic cells to be selected using G418. The vector backbone also contains an SV40 origin of replication in mammalian cells. The multiple cloning site (MCS) is next to the immediate early promoter of CMV (PCMV IE). The human T-cadherin gene was cloned from the mRNA extracted with TRIzol reagent (Invitrogen) from human uterine smooth muscle tissue by performing RT-PCR using the primers described previously in which the EcoRI or BamHI restriction sites were included, digesting with EcoRI or BamHI and ligating into the pEGFP-N1. The pEGFP-N1/T-cad was transfected into Escherichia coli TOP10 for amplification and DNA sequencing was used to ensure the fidelity. Plasmid DNA was prepared using the TIANprep Mini Plasmid Kit (Tiangen Biotech) as per the manufacturer’s instructions. Plasmids were transfected into the mouse melanoma cell line B16F10 cells at 80% confluence and 5×105 cells per well in a six-well plate using the Lipofectamine™ 2000 (Invitrogen) according to the manufacturer’s instructions. The transfected cells which stably expressed T-cadherin were selected by incubating with G418 for >2 weeks. The stable expression of T-cadherin was confirmed by RT-PCR and immunohistochemistry.

Cell proliferation assay

Cell proliferation was measured by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay following a 48 h incubation in 96-well microculture plates with a volume of 200 μl/well. MTT solution (Sigma, St. Louis, MO, USA; 20 μl, 5 mg/ml) was added to each well 4 h before termination. After 4 h of incubation, the cells were lysed and the purple formazan crystals were solubilized with DMSO. The plate was analyzed on a microtitre plate reader at 490 nm and the absorbance was translated into a cell proliferation ratio for comparison: Cell proliferation ratio = (test absorbance / non-transfection control absorbance) × 100.

Detection of apoptosis via EGFP-annexin V and propidium iodide (PI) staining

Briefly, cells were cultured in six-well plates (Costar; Corning Inc., Corning, NY, USA) for 72 h at 37°C and 5% CO2. Cells (2×105) were collected from each well and incubated with 5 μl EGFP-annexin V and 5 μl PI (KeyGen Biotech.) at room temperature for 15 min in the dark. Subsequently, 500 μl annexin V binding buffer was added and flow cytometry was performed using a FACSCalibur flow cytometer (Becton Dickinson, Franklin Lakes, NJ, USA). Cells were considered to be apoptotic if they were annexin V+/PI (early apoptotic). Each analysis was performed using at least 10,000 events.

Analysis of cell cycle using PI staining

Cells were cultured in 6-well plates (Costar; Corning Inc.) for 72 h at 37°C and 5% CO2. The cells were then harvested, washed with PBS and resuspended in 75% ethanol at 4°C overnight to make the membranes porous. After washing with PBS, the cells were incubated with 0.5 ml PI solution (5 mg PI, 2 mg RNase, 1 ml Triton X-100, 65 ml NS and 100 mg citrate sodium; volume to 100 ml, pH 7.4) for 30 min at 4°C in the dark to stain the nuclear DNA. The percentages of cells in the G0/G1, S and G2/M phases of the cell cycle were identified by FACSCalibur flow cytometry (Becton Dickinson) excluding cell doublets. The analyses were performed using CellQuest software (Becton Dickinson). Each analysis was performed using at least 10,000 events.

Transwell invasion assay

In vitro invasion assays were performed using 8-μm Transwell membranes (Corning Inc.) to measure tumor invasion. The Matrigel invasion chambers were prepared at 1:4 dilution and incubated for 1 h at 37°C and 5% CO2. Cells were washed with 1X PBS, resuspended in 0.1% fetal bovine serum (FBS)-RPMI-1640 and 2×105 cells (200 μl) were added to the Matrigel-coated upper chamber. RPMI-1640 culture medium containing 30% FBS was placed in the lower compartment of the chemotaxis chamber to function as a source of chemoattractants. The 24-well plastic culture plate was incubated at 37°C and 5% CO2 for 24 h. After incubation, the invasive cells which had passed through to the lower surface of the filter were fixed and stained with hematoxylin for microscope observation or quantified using an MTT assay in which the absorbance was translated into a cell ratio for comparison: Cell ratio = (test absorbance / non-transfection control absorbance) × 100.

Statistical analysis

The results, with the exception of the immunocytochemistry, were expressed as the mean ± SD of triplicate experiments (six experiments in the MTT cell proliferation assay) and statistical comparisons between the experimental groups and the control were performed using one-way analysis of variance (ANOVA) followed by Dunnett’s t-test. P<0.05 was considered to indicate statistically significant differences.

Results

Confirmation of cDNA fidelity and expression in transfected melanoma cells

The cDNA fidelity of the engineered human T-cadherin expression vector pEGFP-N1/T-cad was evaluated by DNA sequencing which showed that the cDNA was consistent with the genebank (NM_001257). The pEGFP-N1/T-cad was transfected into mouse B16F10 melanoma cells and the transfected cells which stably expressed human T-cadherin were selected by culturing with G418 for two weeks. The expression of T-cadherin was demonstrated by immunohisto-chemistry (Fig. 1A–C) and RT-PCR (Fig. 1D).

Cell proliferation in T-cadherin-transfected melanoma cells

It was shown by MTT assays that the proliferation in T-cadherin-transfected B16F10 melanoma cells was markedly lower than either the parental (without transfection) or empty pEGFP-N1 (without T-cadherin gene)-transfected B16F10 cells (P<0.05), while the proliferation in the parental and empty pEGFP-N1-transfected B16F10 cells was similar (P>0.05, Fig. 2E).

Apoptosis in T-cadherin-transfected melanoma cells

Flowcytometry FACS analysis with annexin V and PI staining was used to determine cell apoptosis (28). It was shown by EGFP-annexin V/PI staining and flow cytometry that the percentage of early apoptotic cells in T-cadherin-transfected B16F10 melanoma cells was significantly larger compared with either the parental or empty pEGFP-N1-transfected B16F10 cells (P<0.05), while the percentage of early apoptotic cells in the parental and empty pEGFP-N1-transfected B16F10 cells was similar (P>0.05, Fig. 2A–D).

Cell cycle in T-cadherin-transfected melanoma cells

The cell cycle may be analyzed by PI staining and flow cytometry analysis (29,30). It was shown by PI staining and flow cytometry analysis that the cell cycle in T-cadherin-transfected B16F10 melanoma cells was significantly different from either the parental or empty pEGFP-N1-transfected B16F10 cells. The cells in the G2/M phase were more abundant compared with either the parental or empty pEGFP-N1-transfected B16F10 cells (P<0.05), while the number of cells in the G2/M phase in the parental and empty pEGFP-N1-transfected B16F10 cells was similar (P>0.05, Fig. 3). This suggested that the T-cadherin-transfected B16F10 melanoma cells were arrested in the G2/M phase.

Invasiveness of T-cadherin-transfected melanoma cells

The Transwell assay is a useful method for determining cell migration (31) which represents invasiveness. It was shown by the Transwell invasion assay that the number of cells permeating the Matrigel-coated membranes in the T-cadherin-transfected B16F10 melanoma cells was significantly lower compared with either the parental or empty pEGFP-N1-transfected B16F10 cells (P<0.05). The number of cells permeating the Matrigel-coated membranes in the parental and empty pEGFP-N1-transfected B16F10 cells was similar (P>0.05, Fig. 4).

Discussion

Invasiveness and metastasis are the most important features of malignant tumors, including melanoma. Cell adhesion is closely associated with tumor invasiveness and metastasis and the cadherin superfamily is a superfamily of transmembrane glycoproteins that mediate calcium-dependent homophilic intercellular adhesion. E-cadherin is a prototypic member of the cadherin superfamily and is expressed by the majority normal epithelial tissues and a number of epithelium-derived cancer cells have lost E-cadherin expression (3234).

Unlike other members of the cadherin superfamily which contain transmembrane domains linking the extracellular portion of the molecules with the intracellular signaling pathways, T-cadherin is unique as it lacks the transmembrane and cytoplasmic domains and is anchored to cell membranes through a GPI anchor (9). Cell adhesion mediated by classical cadherins depends on the binding between cadherin extracellular domains presented on the surfaces of opposing cells and is regulated through intracellular associations with β- and α-catenins which affect the dynamics of the actin-based cytoskeleton (35,36). T-cadherin shares the extracellular five cadherin repeats with other cadherins but lacks the trans-membrane and cytosolic domains and instead attaches to the membrane via a GPI anchor (9). T-cadherin has been localized within lipid rafts of the plasma membrane, is targeted to the apical surface in polarized epithelial cells and redistributed to the leading edge of migrating cells (3739). T-cadherin mediates calcium-dependent cell adhesion and colocalizes with small trimeric G-proteins and Src family kinases in lipid rafts, where it may be involved in modulating signal transduction pathways (37,40,41). T-cadherin mediates calcium-dependent adhesion, although it is not concentrated at the cell-cell junctions of transfected cells in culture (9,42). Thus T-cadherin appears to be far less adhesive than classical cadherins and, consistent with its role in cell growth and migration, is likely to be involved in reversible and dynamic cell-cell adhesion/de-adhesion (43). T-cadherin has been shown to have diverse roles in physiology and pathophysiology, including as a negative guidance cue for motor axon projections, tumor suppressor factor in various types of cancer, atypical lipoprotein-binding protein and stimulator of angiogenesis (4446). Hence, in various studies T-cadherin has been shown to be involved in the regulation of proliferation, apoptosis and angiogenesis in normal tissues, as well as tumor growth (47).

T-cadherin is widely expressed in the brain and cardiovascular system, but is absent or strongly depleted in a number of cell cancer lines (18,20,21,48). Downregulation of T-cadherin gene expression associated with promoter hyper-methylation has been frequently reported in breast, lung and colon carcinomas (49). T-cadherin mediates the activation of PI3K/Akt/GSK3β signaling which protects endothelial cells from oxidative stress-induced apoptosis (45). T-cadherin downregulation is associated with poorer prognoses in various carcinomas, such as lung, ovarian, cervical and prostate cancer, while in the majority of cancer cell lines, T-cadherin re-expression inhibits cell proliferation and invasiveness, increases susceptibility to apoptosis and reduces tumor growth in in vivo models. In contrast to the majority cancer cell lines, T-cadherin overexpression in endothelial cells promotes proliferation and migration and has a pro-survival effect (10).

In the present study, it was shown that inducing the expression of T-cadherin by transfecting the T-cadherin gene into B16F10 melanoma cells markedly reduced cell proliferation and permeation through the Matrigel-coated membranes, representing invasiveness. The percentage of early apoptotic cells and the cells in the G2/M phase of the cell cycle was markedly increased compared with either the parental or empty pEGFP-N1-transfected B16F10 cells, with similarity between the parental and empty pEGFP-N1-transfected B16F10 cells. These properties suggest that T-cadherin may have an important role in melanoma progression and be a possible target for therapy in melanoma and certain other types of cancer.

References

1 

Erdei E and Torres SM: A new understanding in the epidemiology of melanoma. Expert Rev Anticancer Ther. 10:1811–1823. 2010. View Article : Google Scholar

2 

Jemal A, Siegel R, Ward E, Murray T, Xu J, Smigal C and Thun MJ: Cancer statistics, 2006. CA Cancer J Clin. 56:106–130. 2006. View Article : Google Scholar

3 

Balch CM, Buzaid AC, Atkins MB, Cascinelli N, Coit DG, Fleming ID, et al: A new American Joint Committee on Cancer staging system for cutaneous melanoma. Cancer. 88:1484–1491. 2000. View Article : Google Scholar : PubMed/NCBI

4 

Houghton AN and Polsky D: Focus on melanoma. Cancer Cell. 2:275–278. 2002. View Article : Google Scholar

5 

Wheelock MJ and Johnson KR: Cadherins as modulators of cellular phenotype. Annu Rev Cell Dev Biol. 19:207–235. 2003. View Article : Google Scholar : PubMed/NCBI

6 

van Roy F and Berx G: The cell-cell adhesion molecule E-cadherin. Cell Mol Life Sci. 65:3756–3788. 2008.

7 

Takeichi M: Cadherins in cancer: implications for invasion and metastasis. Curr Opin Cell Biol. 5:806–811. 1993. View Article : Google Scholar : PubMed/NCBI

8 

Christofori G and Semb H: The role of the cell-adhesion molecule E-cadherin as a tumour-suppressor gene. Trends Biochem Sci. 24:73–76. 1999. View Article : Google Scholar : PubMed/NCBI

9 

Ranscht B and Dours-Zimmermann MT: T-cadherin, a novel cadherin cell adhesion molecule in the nervous system lacks the conserved cytoplasmic region. Neuron. 7:391–402. 1991. View Article : Google Scholar : PubMed/NCBI

10 

Andreeva AV and Kutuzov MA: Cadherin 13 in cancer. Genes Chromosomes Cancer. 49:775–790. 2010.PubMed/NCBI

11 

Philippova M, Joshi MB, Kyriakakis E, Pfaff D, Erne P and Resink TJ: A guide and guard: the many faces of T-cadherin. Cell Signal. 21:1035–1044. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Kuphal S, Martyn AC, Pedley J, Crowther LM, Bonazzi VF, Parsons PG, et al: H-cadherin expression reduces invasion of malignant melanoma. Pigment Cell Melanoma Res. 22:296–306. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Adachi Y, Takeuchi T, Nagayama T, Ohtsuki Y and Furihata M: Zeb1-mediated T-cadherin repression increases the invasive potential of gallbladder cancer. FEBS Lett. 583:430–436. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Maruyama R, Toyooka S, Toyooka KO, Harada K, Virmani AK, Zöchbauer-Müller S, et al: Aberrant promoter methylation profile of bladder cancer and its relationship to clinicopathological features. Cancer Res. 61:8659–8663. 2001.PubMed/NCBI

15 

Kawakami M, Staub J, Cliby W, Hartmann L, Smith DI and Shridhar V: Involvement of H-cadherin (CDH13) on 16q in the region of frequent deletion in ovarian cancer. Int J Oncol. 15:715–720. 1999.PubMed/NCBI

16 

Ehrlich M, Woods CB, Yu MC, Dubeau L, Yang F, Campan M, et al: Quantitative analysis of associations between DNA hypermethylation, hypomethylation, and DNMT RNA levels in ovarian tumors. Oncogene. 25:2636–2645. 2006. View Article : Google Scholar : PubMed/NCBI

17 

Toyooka KO, Toyooka S, Virmani AK, Sathyanarayana UG, Euhus DM, Gilcrease M, et al: Loss of expression and aberrant methylation of the CDH13 (H-cadherin) gene in breast and lung carcinomas. Cancer Res. 61:4556–4560. 2001.PubMed/NCBI

18 

Sato M, Mori Y, Sakurada A, Fujimura S and Horii A: The H-cadherin (CDH13) gene is inactivated in human lung cancer. Hum Genet. 103:96–101. 1998. View Article : Google Scholar : PubMed/NCBI

19 

Brock MV, Hooker CM, Ota-Machida E, Han Y, Guo M, Ames S, et al: DNA methylation markers and early recurrence in stage I lung cancer. N Engl J Med. 358:1118–1128. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Zhong Y, Delgado Y, Gomez J, Lee SW and Perez-Soler R: Loss of H-cadherin protein expression in human non-small cell lung cancer is associated with tumorigenicity. Clin Cancer Res. 7:1683–1687. 2001.PubMed/NCBI

21 

Lee SW, Reimer CL, Campbell DB, Cheresh P, Duda RB and Kocher O: H-cadherin expression inhibits in vitro invasiveness and tumor formation in vivo. Carcinogenesis. 19:1157–1159. 1998. View Article : Google Scholar : PubMed/NCBI

22 

Chan DW, Lee JM, Chan PC and Ng IO: Genetic and epigenetic inactivation of T-cadherin in human hepatocellular carcinoma cells. Int J Cancer. 123:1043–1052. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Sun LX, Lin ZB, Duan XS, Lu J, Ge ZH, Li XF, et al: Enhanced MHC class I and costimulatory molecules on B16F10 cells by Ganoderma lucidum polysaccharides. J Drug Target. 20:582–592. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Sun LX, Lin ZB, Li XJ, Li M, Lu J, Duan XS, et al: Promoting effects of Ganoderma lucidum polysaccharides on B16F10 cells to activate lymphocytes. Basic Clin Pharmacol Toxicol. 108:149–154. 2011.

25 

Sun LX, Lin ZB, Duan XS, Lu J, Ge ZH, Li XJ, et al: Ganoderma lucidum polysaccharides antagonize the suppression on lymphocytes induced by culture supernatants of B16F10 melanoma cells. J Pharm Pharmacol. 63:725–735. 2011. View Article : Google Scholar

26 

Sun LX, Lin ZB, Duan XS, Lu J, Ge ZH, Song YX, et al: Stronger cytotoxicity in CTLs with granzyme B and porforin was induced by Ganoderma lucidum polysaccharides acting on B16F10 cells. Biomed Prev Nutr. 2:113–118. 2012. View Article : Google Scholar

27 

Hufbauer M, Lazić D, Reinartz M, Akgül B, Pfister H and Weissenborn SJ: Skin tumor formation in human papillomavirus 8 transgenic mice is associated with a deregulation of oncogenic miRNAs and their tumor suppressive targets. J Dermatol Sci. 64:7–15. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Liang Y and Sundberg JP: SHARPIN regulates mitochondria-dependent apoptosis in keratinocytes. J Dermatol Sci. 63:148–153. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Lai WW, Hsiao YP, Chung JG, Wei YH, Cheng YW and Yang JH: Synergistic phototoxic effects of glycolic acid in a human keratinocyte cell line (HaCaT). J Dermatol Sci. 64:191–198. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Chang SP, Shen SC, Lee WR, Yang LL and Chen YC: Imatinib mesylate induction of ROS-dependent apoptosis in melanoma B16F0 cells. J Dermatol Sci. 62:183–191. 2011. View Article : Google Scholar : PubMed/NCBI

31 

Dong D, Jiang M, Xu X, Guan M, Wu J, Chen Q and Xiang L: The effects of NB-UVB on the hair follicle-derived neural crest stem cells differentiating into melanocyte lineage in vitro. J Dermatol Sci. 66:20–28. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Batlle E, Sancho E, Francí C, Domínguez D, Monfar M, Baulida J, et al: The transcription factor snail is a repressor of E-cadherin gene expression in epithelial tumour cells. Nat Cell Biol. 2:84–89. 2000. View Article : Google Scholar : PubMed/NCBI

33 

Comijn J, Berx G, Vermassen P, Verschueren K, van Grunsven L, Bruyneel E, et al: The two-handed E box binding zinc finger protein SIP1 downregulates E-cadherin and induces invasion. Mol Cell. 7:1267–1278. 2001. View Article : Google Scholar : PubMed/NCBI

34 

Wheelock MJ, Soler AP and Knudsen KA: Cadherin junctions in mammary tumors. J Mammary Gland Biol Neoplasia. 6:275–285. 2001. View Article : Google Scholar : PubMed/NCBI

35 

Goodwin M and Yap AS: Classical cadherin adhesion molecules: coordinating cell adhesion, signaling and the cytoskeleton. J Mol Histol. 35:839–844. 2004. View Article : Google Scholar : PubMed/NCBI

36 

Drees F, Pokutta S, Yamada S, Nelson WJ and Weis WI: Alpha-catenin is a molecular switch that binds E-cadherin-beta-catenin and regulates actin-filament assembly. Cell. 123:903–915. 2005. View Article : Google Scholar : PubMed/NCBI

37 

Doyle DD, Goings GE, Upshaw-Earley J, Page E, Ranscht B and Palfrey HC: T-cadherin is a major glycophosphoinositol-anchored protein associated with noncaveolar detergent-insoluble domains of the cardiac sarcolemma. J Biol Chem. 273:6937–6943. 1998. View Article : Google Scholar

38 

Koller E and Ranscht B: Differential targeting of T- and N-cadherin in polarized epithelial cells. J Biol Chem. 271:30061–30067. 1996. View Article : Google Scholar : PubMed/NCBI

39 

Philippova M, Ivanov D, Tkachuk V, Erne P and Resink TJ: Polarisation of T-cadherin to the leading edge of migrating vascular cells in vitro: a function in vascular cell motility? Histochem Cell Biol. 120:353–360. 2003. View Article : Google Scholar : PubMed/NCBI

40 

Kinch MS, Petch L, Zhong C and Burridge K: E-cadherin engagement stimulates tyrosine phosphorylation. Cell Adhes Commun. 4:425–437. 1997. View Article : Google Scholar : PubMed/NCBI

41 

Philippova MP, Bochkov VN, Stambolsky DV, Tkachuk VA and Resink TJ: T-cadherin and signal-transducing molecules co-localize in caveolin-rich membrane domains of vascular smooth muscle cells. FEBS Lett. 429:207–210. 1998. View Article : Google Scholar : PubMed/NCBI

42 

Vestal DJ and Ranscht B: Glycosyl phosphatidylinositol-anchored T-cadherin mediates calcium-dependent, homophilic cell adhesion. J Cell Biol. 119:451–461. 1992. View Article : Google Scholar : PubMed/NCBI

43 

Dames SA, Bang E, Haüssinger D, Ahrens T, Engel J and Grzesiek S: Insights into the low adhesive capacity of human T-cadherin from the NMR structure of Its N-terminal extra-cellular domain. J Biol Chem. 283:23485–23495. 2008. View Article : Google Scholar : PubMed/NCBI

44 

Takeuchi T and Ohtsuki Y: Recent progress in T-cadherin (CDH13, H-cadherin) research. Histol Histopathol. 16:1287–1293. 2001.PubMed/NCBI

45 

Joshi MB, Philippova M, Ivanov D, Allenspach R, Erne P and Resink TJ: T-cadherin protects endothelial cells from oxidative stress-induced apoptosis. FASEB J. 19:1737–1739. 2005.PubMed/NCBI

46 

Fredette BJ, Miller J and Ranscht B: Inhibition of motor axon growth by T-cadherin substrata. Development. 122:3163–3171. 1996.PubMed/NCBI

47 

Rubina K, Sysoeva V, Semina E, Kalinina N, Yurlova E, Khlebnikova A and Molochkov V: Malignant transformation in skin is associated with the loss of T-cadherin expression in human keratinocytes and heterogeneity in T-cadherin expression in tumor vasculature. Tumor Angiogenesis. Ran S: InTech; New York, NY: pp. 135–166. 2012

48 

Lee SW: H-cadherin, a novel cadherin with growth inhibitory functions and diminished expression in human breast cancer. Nat Med. 2:776–782. 1996. View Article : Google Scholar : PubMed/NCBI

49 

Toyooka S, Toyooka KO, Harada K, Miyajima K, Makarla P, Sathyanarayana UG, et al: Aberrant methylation of the CDH13 (H-cadherin) promoter region in colorectal cancers and adenomas. Cancer Res. 62:3382–3386. 2002.PubMed/NCBI

Related Articles

Journal Cover

April 2013
Volume 5 Issue 4

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
Duan XS, Lu J, Ge ZH, Xing EH, Lu HT and Sun LX: Effects of T‑cadherin expression on B16F10 melanoma cells. Oncol Lett 5: 1205-1210, 2013
APA
Duan, X., Lu, J., Ge, Z., Xing, E., Lu, H., & Sun, L. (2013). Effects of T‑cadherin expression on B16F10 melanoma cells. Oncology Letters, 5, 1205-1210. https://doi.org/10.3892/ol.2013.1164
MLA
Duan, X., Lu, J., Ge, Z., Xing, E., Lu, H., Sun, L."Effects of T‑cadherin expression on B16F10 melanoma cells". Oncology Letters 5.4 (2013): 1205-1210.
Chicago
Duan, X., Lu, J., Ge, Z., Xing, E., Lu, H., Sun, L."Effects of T‑cadherin expression on B16F10 melanoma cells". Oncology Letters 5, no. 4 (2013): 1205-1210. https://doi.org/10.3892/ol.2013.1164