Sox9 is a β-catenin-regulated transcription factor that enhances the colony-forming activity of squamous cell carcinoma cells

  • Authors:
    • Xue Mei Li
    • Yong Jun Piao
    • Kyung‑Cheol Sohn
    • Jeong-Min Ha
    • Myung Im
    • Young‑Joon Seo
    • Kyu Uang Whang
    • Jeung‑Hoon Lee
    • Young Lee
    • Chang Deok Kim
  • View Affiliations

  • Published online on: May 4, 2016     https://doi.org/10.3892/mmr.2016.5210
  • Pages: 337-342
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Squamous cell carcinoma (SCC) is a common skin cancer, of which the incidence is relatively high, ranking second among the non‑melanoma skin cancers. It is known that numerous intracellular signal regulators are involved in the pathogenesis of SCC. The Wnt/β-catenin signaling pathway serves an important role in cancer development. However, the downstream effectors of β‑catenin remain to be clearly elucidated yet. The present study investigated the functional importance of Wnt/β‑catenin signaling in cutaneous SCC. β‑catenin expression was reduced using recombinant adenovirus expressing specific microRNA (miR). Knockdown of β‑catenin resulted in a marked reduction of the colony-forming activity of the SCC cells, SCC12. In an attempt to identify the β‑catenin downstream genes, it was found that Sox9 was regulated by β‑catenin in SCC12 cells. Overexpression of a constitutively active form of β‑catenin led to the induction of Sox9, while knockdown of β‑catenin resulted in downregulation of Sox9. When the expression of Sox9 was reduced using specific miR, colony-forming activity of the SCC12 cells was significantly reduced. When Sox9 was overexpressed in cells where β‑catenin was knocked down, it partially restored the colony‑forming potential. Taken together, the present results suggested that Sox9 is a β-catenin downstream transcription factor and is positively involved in SCC development.

Introduction

Squamous cell carcinoma (SCC) is a common skin cancer originating from the upper layers of skin epidermis. The incidence of SCC is relatively high, ranking second among the non-melanoma skin cancers (1). Numerous factors are implicated in the pathogenesis of SCC, of which ultraviolet (UV) radiation is regarded as the most important environmental risk factor. UV radiation induces DNA damage and mutations of numerous susceptible genes, including the tumor suppressor, p53 (2). In addition, several intracellular signal regulators are involved in cancer development and progression. Examples include epidermal growth factor receptor, nuclear factor-κ-light-chain-enhancer of activated B cells and the Wnt/β-catenin signaling pathways (37).

In canonical Wnt/β-catenin signaling, binding of Wnt ligands to their cognate membrane receptors leads to the inactivation of the β-catenin degradation complex, resulting in the stabilization of cytoplasmic β-catenin. Once accumulated, β-catenin locates to the nucleus and interacts with the Lef-1/TCF family of DNA-binding proteins to generate a functional transcription factor complex (8). The pivotal role of Wnt/β-catenin signaling in cancer development has been previously described (911). The importance of β-catenin in skin cancer is supported by the fact that epidermis-specific ablation of the β-catenin gene results in the loss of cancer initiating cells and complete tumor regression (12). In addition, a previous genetic study showed that chromosome loci harboring Wnt signaling genes are frequently amplified in SCC (13), indicating a potential importance of Wnt/β-catenin signaling in cutaneous SCC.

Although the pivotal role of Wnt/β-catenin signaling in SCC is well recognized, the downstream effectors of β-catenin remain to be clearly elucidated. The present study identified Sox9 as a β-catenin-regulated gene and demonstrated that Sox9 has a potential role in the regulation of SCC cells.

Materials and methods

Cell culture

The SCC12 human squamous cell carcinoma cell line was provided by Professor Tae-Jin Yoon (Gyeongsang National University, Jinju, South Korea) and maintained in Dulbecco's modified Eagle's medium (DMEM), supplemented with 10% fetal bovine serum (FBS; Thermo Fisher Scientific, Inc., Waltham, MA, USA). Human keratinocytes were isolated from skin specimens then immortalized using a recombinant retrovirus expressing Simian virus 40 large T antigen (SV40T), as described in a previous report (14). SV40T-transformed human epidermal keratinocytes (SV-HEK) were maintained in keratinocyte-serum free medium (Thermo Fisher Scientific, Inc.), supplemented with bovine pituitary extract and recombinant human epidermal growth factor (Thermo Fisher Scientific, Inc.) (14). Human dermal fibroblasts were primary cultured and maintained in DMEM, supplemented with 10% FBS. Human melanocytes were cultured in Medium 254 (Cascade Biologics, Portland, OR, USA) and human melanocyte growth supplement (Cascade Biologics) (15,16). Cells were maintained at 37°C in an atmosphere 5% CO2 and 90% relative humidity.

Immunohistochemistry

All human skin samples were obtained under the written informed consent of donors, in accordance with the Ethical Committee approval process of the Institutional Review Board of Chungnam National University School of Medicine. Paraffin sections of skin specimens were dewaxed, rehydrated and washed three times with phosphate-buffered saline (PBS). The tissue sections were subsequently incubated with proteinase K (Dako, Carpinteria, CA, USA) for 5 min at 37°C, and treated with H2O2 for 10 min at room temperature. The tissue sections were blocked in 0.1% Tween-20 and 1% bovine serum albumin (Sigma-Aldrich, St. Louis, MO, USA) in PBS for 30 min. Following blocking, the tissue sections were incubated at 4°C with the appropriate primary antibodies as follows: Rabbit polyclonal anti-β-catenin (1:100 dilution; cat. no. sc-7199; Santa Cruz Biotechnologies, Inc., Santa Cruz, CA, USA); mouse monoclonal anti-Sox9 (1:200 dilution; cat. no. ab76997; Abcam, Cambridge, MA, USA) overnight. Subsequently, the tissue sections were incubated with the following peroxidase-conjugated secondary antibodies (dilution, 1:1,000): Horseradish peroxidase (HRP)-conjugated polyclonal goat anti-rabbit immunoglobulin (Ig; cat. no. P0448) and HRP-conjugated polyclonal rabbit anti-mouse Ig (cat. no. P0161; both Dako), visualized using 3,3′-diaminobenzidine solution from the Chemmate envision detection kit (Dako) and photographed under a Diaphot inverted microscope (Nikon Corporation, Tokyo, Japan).

Western blot analysis

The cells were lysed in Proprep solution (Intron, Daejeon, Korea). The total protein was quantified using a bicinchoninic acid protein assay reagent (Pierce Biotechnology, Inc., Rockford, IL, USA). The samples were separated by 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (100 V for 1 h) and transferred onto nitrocellulose membranes (Thermo Fisher Scientific, Inc.). The membranes were then blocked using 5% skimmed milk in Tris-buffered saline and Tween-20 (Sigma-Aldrich) for 1 h at room temperature, and incubated (for 1 h at room temperature) with mouse monoclonal anti-Sox9 (1:200 dilution), rabbit polyclonal anti-β-catenin (1:100 dilution) and mouse monoclonal anti-β-actin [1:1,000 dilution (cat. no. A2228), Sigma-Aldrich] primary antibodies. Following primary antibody incubation, the membranes were incubated for 1 h at room temperature with the following secondary antibodies: Rabbit anti-mouse IgG (cat. no. ab97046) and goat anti-rabbit IgG (cat. no. .ab6721; both Abcam) and were visualized by enhanced chemiluminescence (Intron).

Production of recombinant adenovirus

The recombinant adenovirus expressing an 87-amino acid N-terminally truncated β-catenin (Ad/ΔN87-β-cat) or Sox9 (Ad/Sox9) were previously described (17,18). For knockdown experiments, the recombinant adenovirus expressing microRNA (miR) was generated. The target sequences for β-catenin and Sox9 were designed using BLOCK-iT™ RNAi Designer (Thermo Fisher Scientific, Inc.). The double-stranded DNA oligonucle-otides were synthesized and cloned into the parental vector, pcDNA6.2-GW (Thermo Fisher Scientific, Inc.). In this vector system, the miR sequence for the target gene is located downstream of emerald green fluorescent protein (EmGFP) coding sequence, allowing the identification of miR expressing cells by observing GFP under an IX71 fluorescent microscope (Olympus Corporation, Tokyo, Japan). This vector was termed pcDNA6.2-GW/EmGFP-miR. The expression cassette for miR was moved into the pENT/CMV vector and then adenovirus was generated. The miR sequences were as follows: β-catenin, top strand: 5′-TGCTGTCTGCATGCCCTCATCTAATGGTTTTGGCCACTGACTGACCATTAGATGGGCATGCAGA-3′ and bottom strand: 5′-CCTGTCTGCATGCCCATCTAATGGTCAGTCAGTGGCCAAAACCATTAGATGAGGGCATGCAGAC-3′; Sox9, top strand: 5′-TGCTGTGTTCTTGCTGGAGCCGTTGAGTTTTGGCCACTGACTGACTCAACGGCCAGCAAGAACA-3′ and bottom strand: 5′-CCTGTGTTCTTGCTGGCCGTTGAGTCAGTCAGTGGCCAAAACTCAACGGCTCCAGCAAGAACA-3′C. For adenovirus transduction, SCC12 cells were incubated with 10 multiplicity of infection of adenovirus for 6 h at 37°C in an atmosphere 5% CO2 and 90% relative humidity. The cells were replenished with fresh medium and incubated for a further 2 days.

Colony forming assay

The cells were trypsinized and counted using a hemocytometer. Following counting, ~1,000 cells were resuspended in DMEM, supplemented with 10% FBS, and were seeded into 100 mm culture dishes. The cells were incubated for 2–3 weeks and stained with crystal violet (Sigma-Alrdrich).

Statistical analysis

The data were statistically analyzed using one-way analysis of variance with SPSS software (v22.0; IBM SPSS, Chicago, IL, USA). P<0.01 was considered to indicate a statistically significant difference.

Results

In order to elucidate the importance of β-catenin in cutaneous SCC, the present study first determined the expression of β-catenin by immunohistochemistry. In normal skin, β-catenin immunoreactivity was observed in all epidermal layers, with a characteristic membrane staining pattern. In SCC, very intense immunostaining of β-catenin was observed, and notably, many of the SCC cells exhibited nuclear staining of β-catenin (Fig. 1A). These results suggested a fundamental role of β-catenin signaling in cutaneous SCC. The relative expression level of β-catenin in SCC12 cell line and skin-comprising cells, including SV-HEK, fibroblasts and melanocytes, was determined. Although it was not significant, the total β-catenin level was marginally increased in the SCC12 cells compared with the keratinocytes (Fig. 1B). To confirm the potential role of β-catenin, clonogenic assays were performed as an in vitro tumorigenic test (19). Following transduction of recombinant adenovirus expressing miR specific for β-catenin, gene knockdown was confirmed (Fig. 1C). Knockdown of β-catenin markedly reduced the colony-forming potential of SCC12 cells (Fig. 1D), supporting the pivotal role of β-catenin in the tumorigenesis of SCC.

The present study next attempted to identify the genes regulated by β-catenin. To this end, the present study transduced the recombinant adenovirus expressing N-terminal 87-amino acid truncated β-catenin (Ad/ΔN87-β-cat), a constitutively active form of β-catenin (20). It was found that Sox9 was induced by the overexpression of the constitutively active form of β-catenin. Conversely, knockdown of β-catenin resulted in downregulation of Sox9 (Fig. 2A). These results suggested that Sox9 is a β-catenin-regulated gene in SCC cells and has a potential role in tumorigenesis. Therefore, the expression of Sox9 was next determined in cutaneous SCC. Immunohistochemistry and western blotting showed that the expression of Sox9 was increased in cutaneous SCC tissue compared with normal skin tissue (Fig. 2B and C). Consistent with these data, the relative protein expression level of Sox9 was significantly increased in SCC12 cells compared with keratinocytes cultured in vitro (Fig. 2D).

The expression of Sox9 was reduced in SCC12 cells using the recombinant adenovirus expressing miR specific for Sox9 (Fig. 3A). The colony-forming potential of SCC12 cells was determined, and the data showed that knockdown of Sox9 significantly decreased the colony number (Fig. 3B). These results suggested that Sox9 is a potential transcription factor that can increase tumorigenicity of SCC cells.

To further confirm the association between β-catenin and Sox9, β-catenin levels were reduced and the expression of Sox9 was simultaneously increased using the recombinant adenovirus (Fig. 4A). Overexpression of Sox9 led to a slight increase of colony number in the control miR transduced group, however, it was not significant (lane 1 vs. 2; Fig. 4B). Consistent with previous data, knockdown of β-catenin resulted in a marked decrease of colony formation; however overexpression of Sox9 partially restored the colony-forming potential (lane 3 vs. 4; Fig. 4B). These results strengthened the notion that Sox9 is a β-catenin-downstream transcription factor and is involved in the development of SCC.

Discussion

In the present study, it was found that Sox9 was regulated by β-catenin in SCC12 cells. When β-catenin signaling was activated by the overexpression of stabilized β-catenin (ΔN87-β-Cat), Sox9 expression was increased. By contrast, knockdown of β-catenin using specific miR resulted in the downregulation of Sox9. Sox9 demonstrated tumorigenicity in terms of colony-forming activity. Therefore, the present data suggested that β-catenin-regulated Sox9 is one of the effector molecules that can positively affect the development of SCC.

Sox9 is a member of high mobility group box transcription factor family and serves a critical role in various biological events, including embryonic development, cell fate determination and lineage commitment (21,22). Germline mutation for Sox9 causes campomelic dysplasia, a disorder characterized by numerous skeletal abnormalities, defects in central nervous system and XY sex reversal (23,24). A potential role of Sox9 for cancer development has been previously recognized. For example, Sox9 supports breast tumor cell proliferation and directly contributes to the poor clinical outcomes associated with invasive breast cancer (25,26). In other examples, high levels of Sox9 were detected in colorectal cancer and Sox9 exhibits several properties, including the ability to promote proliferation, inhibit senescence and collaborate with other oncogenes in neoplastic transformation (27,28). Based on these findings, it is plausible that Sox9 can also affect cutaneous SCC development. The present study demonstrated that Sox9 was increased in cutaneous SCC, and that Sox9 enhanced the colony-forming activity of SCC12 cells. These data supported the notion that Sox9 is an important pro-oncogenic protein in SCC development.

The putative association between β-catenin and Sox9 has been demonstrated several times in other systems. For example, the Wnt signaling pathway promotes chondrocyte differentiation in a Sox9-dependent manner (29). Another example shows that Sox9 protein is expressed in the intestinal epithelium in a pattern characteristic of Wnt/β-catenin targets. Additionally, inhibition of β-catenin signaling by the over-expression of dominant negative TCF4 resulted in a marked decrease of Sox9 in human colon carcinoma cells (30). Finally, conditional inactivation of adenomatous polyposis coli, thereby activating Wnt/β-catenin signaling in mouse colon, is associated with the induction of Sox9 expression and initiation of crypt budding (31). In the present study, activation and inactivation of β-catenin signaling clearly affected Sox9 expression in a positive manner. Therefore, the present study suggested that Sox9 is an authentic β-catenin-downstream transcription factor and exerts its effect as a positive regulator for cutaneous SCC.

In conclusion, the present study demonstrated that Sox9 is a functional downstream effector of β-catenin in cutaneous SCC. These findings provided novel insights into the association between β-catenin and Sox9 in SCC, and may assist with developing novel therapeutic targets for skin cancer in the future.

Acknowledgments

The present study was supported by the Basic Science Research Program through the National Research Foundation of Korea funded by the Ministry of Education, Science and Technology (grant no. 2012R1A1A1041389).

References

1 

Alam M and Ratner D: Cutaneous squamous-cell carcinoma. N Engl J Med. 344:975–983. 2001. View Article : Google Scholar : PubMed/NCBI

2 

Brash DE, Rudolph JA, Simon JA, Lin A, McKenna GJ, Baden HP, Halperin AJ and Pontén J: A role for sunlight in skin cancer: UV-induced p53 mutations in squamous cell carcinoma. Proc Natl Acad Sci USA. 88:10124–10128. 1991. View Article : Google Scholar : PubMed/NCBI

3 

Kolev V, Mandinova A, Guinea-Viniegra J, Hu B, Lefort K, Lambertini C, Neel V, Dummer R, Wagner EF and Dotto GP: EGFR signalling as a negative regulator of Notch1 gene transcription and function in proliferating keratinocytes and cancer. Nat Cell Biol. 10:902–911. 2008. View Article : Google Scholar : PubMed/NCBI

4 

van Hogerlinden M, Rozell BL, Ahrlund-Richter L and Toftgård R: Squamous cell carcinomas and increased apoptosis in skin with inhibited Rel/nuclear factor-kappaB signaling. Cancer Res. 59:3299–3303. 1999.PubMed/NCBI

5 

Brasanac D, Boricic I, Todorovic V, Tomanovic N and Radojevic S: Cyclin A and beta-catenin expression in actinic keratosis, Bowen's disease and invasive squamous cell carcinoma of the skin. Br J Dermatol. 153:1166–1175. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Lyakhovitsky A, Barzilai A, Fogel M, Trau H and Huszar M: Expression of e-cadherin and beta-catenin in cutaneous squamous cell carcinoma and its precursors. Am J Dermatopathol. 26:372–378. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Doglioni C, Piccinin S, Demontis S, Cangi MG, Pecciarini L, Chiarelli C, Armellin M, Vukosavljevic T, Boiocchi M and Maestro R: Alterations of beta-catenin pathway in non-melanoma skin tumors: Loss of alpha-ABC nuclear reactivity correlates with the presence of beta-catenin gene mutation. Am J Pathol. 163:2277–2287. 2003. View Article : Google Scholar : PubMed/NCBI

8 

Hisken J and Behrens J: The Wnt signaling pathway. J Cell Sci. 113:3545–3546. 2000.

9 

Zhang Y, Liu B, Zhao Q, Hou T and Huang X: Nuclear local-izaiton of β-catenin is associated with poor survival and chemo-/radioresistance in human cervical squamous cell cancer. Int J Clin Exp Pathol. 7:3908–3917. 2014.

10 

Li P, Cao Y, Li Y, Zhou L, Liu X and Geng M: Expression of Wnt-5a and β-catenin in primary hepatocellular carcinoma. Int J Clin Exp Pathol. 7:3190–3195. 2014.

11 

Cui J, Xi H, Cai A, Bian S, Wei B and Chen L: Decreased expression of Sox7 correlates with the upregulation of the Wnt/β-catenin signaling pathway and the poor survival of gastric cancer patients. Int J Mol Med. 34:197–204. 2014.PubMed/NCBI

12 

Malanchi I, Peinado H, Kassen D, Hussenet T, Metzger D, Chambon P, Huber M, Hohl D, Cano A, Birchmeier W and Huelsken J: Cutaneous cancer stem cell maintenance is dependent on beta-catenin signalling. Nature. 452:650–653. 2008. View Article : Google Scholar : PubMed/NCBI

13 

Popp S, Waltering S, Herbst C, Moll I and Boukamp P: UV-B-type mutations and chromosomal imbalances indicate common pathways for the development of Merkel and skin squamous cell carcinomas. Int J Cancer. 99:352–360. 2002. View Article : Google Scholar : PubMed/NCBI

14 

Choi HI, Sohn KC, Hong DK, Lee Y, Kim CD, Yoon TJ, Park JW, Jung S, Lee JH and Lee YH: Melanosome uptake is associated with the proliferation and differentiation of keratinocytes. Arch Dermatol Res. 306:59–66. 2014. View Article : Google Scholar

15 

Je YJ, Choi DK, Sohn KC, Kim HR, Im M, Lee Y, Lee JH, Kim CD and Seo YJ: Inhibitory role of Id1 on TGF-β-induced collagen expression in human dermal fibroblasts. Biochem Biophys Res Commun. 444:81–85. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Lee JS, Kim DH, Choi DK, Kim CD, Ahn GB, Yoon TY, Lee JH and Lee JY: Comparison of gene expression profiles between keratinocytes, melanocytes and fibroblasts. Ann Dermatol. 25:36–45. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Sohn KC, Shi G, Jang S, Choi DK, Lee Y, Yoon TJ, Park H, Hwang C, Kim HJ, Seo YJ, et al: Pitx2, a beta-catenin-regulated transcription factor, regulates the differentiation of outer root sheath cells cultured in vitro. J Dermatol Sci. 54:6–11. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Shi G, Sohn KC, Li Z, Choi DK, Park YM, Kim JH, Fan YM, Nam YH, Kim S, Im M, et al: Expression and functional role of Sox9 in human epidermal keratinocytes. PLoS One. 8:e543552013. View Article : Google Scholar : PubMed/NCBI

19 

Franken NA, Rodermond HM, Stap J, Haveman J and van Bree C: Clonogenic assay of cells in vitro. Nat Protoc. 1:2315–2319. 2006. View Article : Google Scholar

20 

Gat U, DasGupta R, Degenstein L and Fuchs E: De Novo hair follicle morphogenesis and hair tumors in mice expressing a truncated beta-catenin in skin. Cell. 95:605–614. 1998. View Article : Google Scholar : PubMed/NCBI

21 

Lefebvre V, Dumitriu B, Penzo-Méndez A, Han Y and Pallavi B: Control of cell fate and differentiation by Sry-related high-mobility-group box (Sox) transcription factors. Int J Biochem Cell Biol. 39:2195–2214. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Sarkar A and Hochedlinger K: The sox family of transcription factors: Versatile regulators of stem and progenitor cell fate. Cell Stem Cell. 12:15–30. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Foster JW, Dominguez-Steglich MA, Guioli S, Kwok C, Weller PA, Stevanović M, Weissenbach J, Mansour S, Young ID, Goodfellow PN, et al: Campomelic dysplasia and autosomal sex reversal caused by mutations in an SRY-related gene. Nature. 372:525–530. 1994. View Article : Google Scholar : PubMed/NCBI

24 

Schafer AJ, Foster JW, Kwok C, Weller PA, Guioli S and Goodfellow PN: Campomelic dysplasia with XY sex reversal: Diverse phenotypes resulting from mutations in a single gene. Ann NY Acad Sci. 785:137–149. 1996. View Article : Google Scholar : PubMed/NCBI

25 

Chakravarty G, Moroz K, Makridakis NM, Lloyd SA, Galvez SE, Canavello PR, Lacey MR, Agrawal K and Mondal D: Prognostic significance of cytoplasmic SOX9 in invasive ductal carcinoma and metastatic breast cancer. Exp Biol Med (Maywood). 236:145–155. 2011. View Article : Google Scholar

26 

Wang H, He L, Ma F, Regan MM, Balk SP, Richardson AL and Yuan X: SOX9 regulates low density lipoprotein receptor-related protein 6 (LRP6) and T-cell factor 4 (TCF4) expression and Wnt/β-catenin activation in breast cancer. J Biol Chem. 288:6478–6487. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Matheu A, Collado M, Wise C, Manterola L, Cekaite L, Tye AJ, Canamero M, Bujanda L, Schedl A, Cheah KS, et al: Oncogenicity of the developmental transcription factor Sox9. Cancer Res. 72:1301–1315. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Panza A, Pazienza V, Ripoli M, Benegiamo G, Gentile A, Valvano MR, Augello B, Merla G, Prattichizzo C, Tavano F, et al: Interplay between SOX9, β-catenin and PPARγ activation in colorectal cancer. Biochim Biophys Acta. 1833:1853–1865. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Yano F, Kugimiya F, Ohba S, Ikeda T, Chikuda H, Ogasawara T, Ogata N, Takato T, Nakamura K, Kawaguchi H and Chung UI: The canonical Wnt signaling pathway promotes chondrocyte differentiation in a Sox9-dependent manner. Biochem Biophys Res Commun. 333:1300–1308. 2005. View Article : Google Scholar : PubMed/NCBI

30 

Blache P, van de Wetering M, Duluc I, Domon C, Berta P, Freund JN, Clevers H and Jay P: SOX9 is an intestine crypt transcription factor, is regulated by the Wnt pathway, and represses the CDX2 and MUC2 genes. J Cell Biol. 166:37–47. 2004. View Article : Google Scholar : PubMed/NCBI

31 

Feng Y, Sentani K, Wiese A, Sands E, Green M, Bommer GT, Cho KR and Fearon ER: Sox9 induction, ectopic Paneth cells, and mitotic spindle axis defects in mouse colon adenomatous epithelium arising from conditional biallelic Apc inactivation. Am J Pathol. 183:493–503. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2016
Volume 14 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li XM, Piao YJ, Sohn KC, Ha J, Im M, Seo YJ, Whang KU, Lee JH, Lee Y, Kim CD, Kim CD, et al: Sox9 is a β-catenin-regulated transcription factor that enhances the colony-forming activity of squamous cell carcinoma cells. Mol Med Rep 14: 337-342, 2016
APA
Li, X.M., Piao, Y.J., Sohn, K., Ha, J., Im, M., Seo, Y. ... Kim, C.D. (2016). Sox9 is a β-catenin-regulated transcription factor that enhances the colony-forming activity of squamous cell carcinoma cells. Molecular Medicine Reports, 14, 337-342. https://doi.org/10.3892/mmr.2016.5210
MLA
Li, X. M., Piao, Y. J., Sohn, K., Ha, J., Im, M., Seo, Y., Whang, K. U., Lee, J., Lee, Y., Kim, C. D."Sox9 is a β-catenin-regulated transcription factor that enhances the colony-forming activity of squamous cell carcinoma cells". Molecular Medicine Reports 14.1 (2016): 337-342.
Chicago
Li, X. M., Piao, Y. J., Sohn, K., Ha, J., Im, M., Seo, Y., Whang, K. U., Lee, J., Lee, Y., Kim, C. D."Sox9 is a β-catenin-regulated transcription factor that enhances the colony-forming activity of squamous cell carcinoma cells". Molecular Medicine Reports 14, no. 1 (2016): 337-342. https://doi.org/10.3892/mmr.2016.5210