Skip to main content
Top
Published in: Tumor Biology 1/2016

01-01-2016 | Original Article

CDX2 inhibits pancreatic adenocarcinoma cell proliferation via promoting tumor suppressor miR-615-5p

Authors: Yuanyuan Jiang, Yan Zhang, Fuqing Li, Xiaolin Du, Jinping Zhang

Published in: Tumor Biology | Issue 1/2016

Login to get access

Abstract

CDX2 has recently been identified as a prognostic marker for pancreatic adenocarcinoma. However, the role and mechanism of CDX2 in progression of pancreatic adenocarcinoma are still elusive. In this study, we observed that CDX2 expression was much lower in mouse pancreatic adenocarcinoma tissues and pancreatic cancer cells. A network integrated by ChIPBase platform hinted that miR-615-5p, a most newly discovered tumor suppressor, was probably bound by CDX2 in the promoter region. Chromatin immunoprecipitation (ChIP)-qPCR assay showed that CDX2 exhibited a high capacity of binding to miR-615-5p promoter region compared to the negative control. Real-time PCR and western blotting analyses revealed that CDX2 overexpression caused inflation of miR-615-5p and depression of insulin-like growth factor 2 (IGF2), a direct target of miR-615-5p. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and EdU approaches showed that CDX2 overexpression markedly suppressed pancreatic adenocarcinoma cell proliferation. CDX2 small interfering RNA (siRNA) transfection showed an opposite effect on gene expression and cell proliferation to that of CDX2 overexpression. Collectively, CDX2 inhibited pancreatic adenocarcinoma cell proliferation via promoting tumor suppressor miR-615-5p. Our findings suggested a potential molecular target for pancreatic adenocarcinoma therapy.
Literature
1.
go back to reference Almeida R, Silva E, Santos-Silva F, Silberg DG, Wang J, De Bolós C, et al. Expression of intestine-specific transcription factors, CDX1 and CDX2, in intestinal metaplasia and gastric carcinomas. J Pathol. 2003;199:36–40.CrossRefPubMed Almeida R, Silva E, Santos-Silva F, Silberg DG, Wang J, De Bolós C, et al. Expression of intestine-specific transcription factors, CDX1 and CDX2, in intestinal metaplasia and gastric carcinomas. J Pathol. 2003;199:36–40.CrossRefPubMed
2.
go back to reference Guo R-J, Suh ER, Lynch JP. Review the role of Cdx proteins in intestinal development and cancer. Cancer Biol Ther. 2004;3:593–601.CrossRefPubMed Guo R-J, Suh ER, Lynch JP. Review the role of Cdx proteins in intestinal development and cancer. Cancer Biol Ther. 2004;3:593–601.CrossRefPubMed
3.
go back to reference Camilo V, Garrido M, Valente P, Ricardo S, Amaral AL, Barros R, et al. Differentiation reprogramming in gastric intestinal metaplasia and dysplasia: role of SOX2 and CDX2. Histopathology. 2015;66:343–50.CrossRefPubMed Camilo V, Garrido M, Valente P, Ricardo S, Amaral AL, Barros R, et al. Differentiation reprogramming in gastric intestinal metaplasia and dysplasia: role of SOX2 and CDX2. Histopathology. 2015;66:343–50.CrossRefPubMed
4.
go back to reference Chawengsaksophak K, James R, Hammond V, Köntgen F, Beck F. Homeosis and intestinal tumours in Cdx2 mutant mice. Nature. 1997;386:84–7.CrossRefPubMed Chawengsaksophak K, James R, Hammond V, Köntgen F, Beck F. Homeosis and intestinal tumours in Cdx2 mutant mice. Nature. 1997;386:84–7.CrossRefPubMed
6.
go back to reference Bonhomme C, Duluc I, Martin E, Chawengsaksophak K, Chenard M, Kedinger M, et al. The Cdx2 homeobox gene has a tumour suppressor function in the distal colon in addition to a homeotic role during gut development. Gut. 2003;52:1465–71.CrossRefPubMedPubMedCentral Bonhomme C, Duluc I, Martin E, Chawengsaksophak K, Chenard M, Kedinger M, et al. The Cdx2 homeobox gene has a tumour suppressor function in the distal colon in addition to a homeotic role during gut development. Gut. 2003;52:1465–71.CrossRefPubMedPubMedCentral
7.
go back to reference Witek ME, Nielsen K, Walters R, Hyslop T, Palazzo J, Schulz S, et al. The putative tumor suppressor Cdx2 is overexpressed by human colorectal adenocarcinomas. Clin Cancer Res. 2005;11:8549–56.CrossRefPubMed Witek ME, Nielsen K, Walters R, Hyslop T, Palazzo J, Schulz S, et al. The putative tumor suppressor Cdx2 is overexpressed by human colorectal adenocarcinomas. Clin Cancer Res. 2005;11:8549–56.CrossRefPubMed
8.
go back to reference Kaimaktchiev V, Terracciano L, Tornillo L, Spichtin H, Stoios D, Bundi M, et al. The homeobox intestinal differentiation factor CDX2 is selectively expressed in gastrointestinal adenocarcinomas. Mod Pathol. 2004;17:1392–9.CrossRefPubMed Kaimaktchiev V, Terracciano L, Tornillo L, Spichtin H, Stoios D, Bundi M, et al. The homeobox intestinal differentiation factor CDX2 is selectively expressed in gastrointestinal adenocarcinomas. Mod Pathol. 2004;17:1392–9.CrossRefPubMed
9.
go back to reference De Lott LB, Morrison C, Suster S, Cohn DE, Frankel WL. CDX2 is a useful marker of intestinal-type differentiation: a tissue microarray-based study of 629 tumors from various sites. Arch Pathol Lab Med. 2005;129:1100–5.PubMed De Lott LB, Morrison C, Suster S, Cohn DE, Frankel WL. CDX2 is a useful marker of intestinal-type differentiation: a tissue microarray-based study of 629 tumors from various sites. Arch Pathol Lab Med. 2005;129:1100–5.PubMed
10.
go back to reference Verzi MP, Shin H, He HH, Sulahian R, Meyer CA, Montgomery RK, et al. Differentiation-specific histone modifications reveal dynamic chromatin interactions and partners for the intestinal transcription factor CDX2. Dev Cell. 2010;19:713–26.CrossRefPubMedPubMedCentral Verzi MP, Shin H, He HH, Sulahian R, Meyer CA, Montgomery RK, et al. Differentiation-specific histone modifications reveal dynamic chromatin interactions and partners for the intestinal transcription factor CDX2. Dev Cell. 2010;19:713–26.CrossRefPubMedPubMedCentral
11.
go back to reference VVang R, Gown AM, Lee-Shu-Fune Wu TS, Barry DTW, Yemelyanova A, Seidman JD, et al. Immunohistochemical expression of CDX2 in primary ovarian mucinous tumors and metastatic mucinous carcinomas involving the ovary: comparison with CK20 and correlation with coordinate expression of CK7. Mod Pathol. 2006;19:1421–8. VVang R, Gown AM, Lee-Shu-Fune Wu TS, Barry DTW, Yemelyanova A, Seidman JD, et al. Immunohistochemical expression of CDX2 in primary ovarian mucinous tumors and metastatic mucinous carcinomas involving the ovary: comparison with CK20 and correlation with coordinate expression of CK7. Mod Pathol. 2006;19:1421–8.
12.
go back to reference Zhou ZC, Wang J, Cai ZH, Zhang QH, Cai ZX, Wu JH. Association between vitamin D receptor gene Cdx2 polymorphism and breast cancer susceptibility. Tumor Biol. 2013;34:3437–41.CrossRef Zhou ZC, Wang J, Cai ZH, Zhang QH, Cai ZX, Wu JH. Association between vitamin D receptor gene Cdx2 polymorphism and breast cancer susceptibility. Tumor Biol. 2013;34:3437–41.CrossRef
13.
go back to reference Bari MF, Brown H, Nicholson AG, Kerr KM, Gosney JR, Wallace WA, et al. BAI3, CDX2 and VIL1: a panel of three antibodies to distinguish small cell from large cell neuroendocrine lung carcinomas. Histopathology. 2014;64:547–56.CrossRefPubMed Bari MF, Brown H, Nicholson AG, Kerr KM, Gosney JR, Wallace WA, et al. BAI3, CDX2 and VIL1: a panel of three antibodies to distinguish small cell from large cell neuroendocrine lung carcinomas. Histopathology. 2014;64:547–56.CrossRefPubMed
15.
go back to reference Sun Y, Zhang T, Wang C, Jin X, Jia C, Yu S, et al. MiRNA-615-5p functions as a tumor suppressor in pancreatic ductal adenocarcinoma by targeting AKT2. PLoS One. 2015;10, e0128257.CrossRefPubMedPubMedCentral Sun Y, Zhang T, Wang C, Jin X, Jia C, Yu S, et al. MiRNA-615-5p functions as a tumor suppressor in pancreatic ductal adenocarcinoma by targeting AKT2. PLoS One. 2015;10, e0128257.CrossRefPubMedPubMedCentral
16.
go back to reference Mineno J, Okamoto S, Ando T, Sato M, Chono H, Izu H, et al. The expression profile of microRNAs in mouse embryos. Nucleic Acids Res. 2006;34:1765–71.CrossRefPubMedPubMedCentral Mineno J, Okamoto S, Ando T, Sato M, Chono H, Izu H, et al. The expression profile of microRNAs in mouse embryos. Nucleic Acids Res. 2006;34:1765–71.CrossRefPubMedPubMedCentral
17.
go back to reference Chiang HR, Schoenfeld LW, Ruby JG, Auyeung VC, Spies N, Baek D, et al. Mammalian microRNAs: experimental evaluation of novel and previously annotated genes. Genes Dev. 2010;24:992–1009.CrossRefPubMedPubMedCentral Chiang HR, Schoenfeld LW, Ruby JG, Auyeung VC, Spies N, Baek D, et al. Mammalian microRNAs: experimental evaluation of novel and previously annotated genes. Genes Dev. 2010;24:992–1009.CrossRefPubMedPubMedCentral
18.
go back to reference El Tayebi H, Hosny K, Esmat G, Breuhahn K, Abdelaziz AI. miR-615-5p is restrictedly expressed in cirrhotic and cancerous liver tissues and its overexpression alleviates the tumorigenic effects in hepatocellular carcinoma. FEBS Lett. 2012;586:3309–16.CrossRefPubMed El Tayebi H, Hosny K, Esmat G, Breuhahn K, Abdelaziz AI. miR-615-5p is restrictedly expressed in cirrhotic and cancerous liver tissues and its overexpression alleviates the tumorigenic effects in hepatocellular carcinoma. FEBS Lett. 2012;586:3309–16.CrossRefPubMed
19.
go back to reference Gao W, Gu Y, Li Z, Cai H, Peng Q, Tu M, et al. miR-615-5p is epigenetically inactivated and functions as a tumor suppressor in pancreatic ductal adenocarcinoma. Oncogene. 2014;34:1629–40.CrossRefPubMed Gao W, Gu Y, Li Z, Cai H, Peng Q, Tu M, et al. miR-615-5p is epigenetically inactivated and functions as a tumor suppressor in pancreatic ductal adenocarcinoma. Oncogene. 2014;34:1629–40.CrossRefPubMed
20.
go back to reference Ma Y, Yu S, Zhao W, Lu Z, Chen J. miR-27a regulates the growth, colony formation and migration of pancreatic cancer cells by targeting Sprouty2. Cancer Lett. 2010;298:150–8.CrossRefPubMed Ma Y, Yu S, Zhao W, Lu Z, Chen J. miR-27a regulates the growth, colony formation and migration of pancreatic cancer cells by targeting Sprouty2. Cancer Lett. 2010;298:150–8.CrossRefPubMed
21.
go back to reference Hafeez BB, Mustafa A, Fischer JW, Singh A, Zhong W, Shekhani MO, et al. α-Mangostin: a dietary antioxidant derived from the pericarp of Garcinia mangostana L. inhibits pancreatic tumor growth in xenograft mouse model. Antioxid Redox Signal. 2014;21:682–99.CrossRefPubMedPubMedCentral Hafeez BB, Mustafa A, Fischer JW, Singh A, Zhong W, Shekhani MO, et al. α-Mangostin: a dietary antioxidant derived from the pericarp of Garcinia mangostana L. inhibits pancreatic tumor growth in xenograft mouse model. Antioxid Redox Signal. 2014;21:682–99.CrossRefPubMedPubMedCentral
22.
go back to reference Bansal D, Scholl C, Fröhling S, McDowell E, Lee BH, Döhner K, et al. Cdx4 dysregulates Hox gene expression and generates acute myeloid leukemia alone and in cooperation with Meis1a in a murine model. Proc Natl Acad Sci U S A. 2006;103:16924–9.CrossRefPubMedPubMedCentral Bansal D, Scholl C, Fröhling S, McDowell E, Lee BH, Döhner K, et al. Cdx4 dysregulates Hox gene expression and generates acute myeloid leukemia alone and in cooperation with Meis1a in a murine model. Proc Natl Acad Sci U S A. 2006;103:16924–9.CrossRefPubMedPubMedCentral
23.
go back to reference Rawat VP, Thoene S, Naidu VM, Arseni N, Heilmeier B, Metzeler K, et al. Overexpression of CDX2 perturbs HOX gene expression in murine progenitors depending on its N-terminal domain and is closely correlated with deregulated HOX gene expression in human acute myeloid leukemia. Blood. 2008;111:309–19.CrossRefPubMed Rawat VP, Thoene S, Naidu VM, Arseni N, Heilmeier B, Metzeler K, et al. Overexpression of CDX2 perturbs HOX gene expression in murine progenitors depending on its N-terminal domain and is closely correlated with deregulated HOX gene expression in human acute myeloid leukemia. Blood. 2008;111:309–19.CrossRefPubMed
24.
go back to reference Liu H, Shi J, Anandan V, Wang HL, Diehl D, Blansfield J, et al. Reevaluation and identification of the best immunohistochemical panel (pVHL, Maspin, S100P, IMP-3) for ductal adenocarcinoma of the pancreas. Arch Pathol Lab Med. 2012;136:601–9.CrossRefPubMed Liu H, Shi J, Anandan V, Wang HL, Diehl D, Blansfield J, et al. Reevaluation and identification of the best immunohistochemical panel (pVHL, Maspin, S100P, IMP-3) for ductal adenocarcinoma of the pancreas. Arch Pathol Lab Med. 2012;136:601–9.CrossRefPubMed
25.
go back to reference Werling RW, Yaziji H, Bacchi CE, Gown AM. CDX2, a highly sensitive and specific marker of adenocarcinomas of intestinal origin: an immunohistochemical survey of 476 primary and metastatic carcinomas. Am J Surg Pathol. 2003;27:303–10.CrossRefPubMed Werling RW, Yaziji H, Bacchi CE, Gown AM. CDX2, a highly sensitive and specific marker of adenocarcinomas of intestinal origin: an immunohistochemical survey of 476 primary and metastatic carcinomas. Am J Surg Pathol. 2003;27:303–10.CrossRefPubMed
26.
go back to reference Chu PG, Schwarz RE, Lau SK, Yen Y, Weiss LM. Immunohistochemical staining in the diagnosis of pancreatobiliary and ampulla of Vater adenocarcinoma: application of CDX2, CK17, MUC1, and MUC2. Am J Surg Pathol. 2005;29:359–67.CrossRefPubMed Chu PG, Schwarz RE, Lau SK, Yen Y, Weiss LM. Immunohistochemical staining in the diagnosis of pancreatobiliary and ampulla of Vater adenocarcinoma: application of CDX2, CK17, MUC1, and MUC2. Am J Surg Pathol. 2005;29:359–67.CrossRefPubMed
27.
go back to reference Frohling S, Scholl C, Bansal D, Huntly BJ. HOX gene regulation in acute myeloid leukemia: CDX marks the spot? Cell Cycle. 2007;6:2241–5.CrossRefPubMed Frohling S, Scholl C, Bansal D, Huntly BJ. HOX gene regulation in acute myeloid leukemia: CDX marks the spot? Cell Cycle. 2007;6:2241–5.CrossRefPubMed
28.
go back to reference Fraggetta F, Pelosi G, Cafici A, Scollo P, Nuciforo P, Viale G. CDX2 immunoreactivity in primary and metastatic ovarian mucinous tumours. Virchows Arch. 2003;443:782–6.CrossRefPubMed Fraggetta F, Pelosi G, Cafici A, Scollo P, Nuciforo P, Viale G. CDX2 immunoreactivity in primary and metastatic ovarian mucinous tumours. Virchows Arch. 2003;443:782–6.CrossRefPubMed
29.
go back to reference Xiao W, Hong H, Awadallah A, Zhou L, Xin W. Utilization of CDX2 expression in diagnosing pancreatic ductal adenocarcinoma and predicting prognosis. PLoS One. 2014;9, e86853.CrossRefPubMedPubMedCentral Xiao W, Hong H, Awadallah A, Zhou L, Xin W. Utilization of CDX2 expression in diagnosing pancreatic ductal adenocarcinoma and predicting prognosis. PLoS One. 2014;9, e86853.CrossRefPubMedPubMedCentral
30.
go back to reference Helu XM, De Leon DD. IGF2 role in trastuzumab-resistance in JIMT-1 cells. Cancer Res. 2013;73:538.CrossRef Helu XM, De Leon DD. IGF2 role in trastuzumab-resistance in JIMT-1 cells. Cancer Res. 2013;73:538.CrossRef
31.
go back to reference Dong X, Li Y, Tang H, Chang P, Hess KR, Abbruzzese JL, et al. Insulin-like growth factor axis gene polymorphisms modify risk of pancreatic cancer. Cancer Epidemiol. 2012;36:206–11.CrossRefPubMed Dong X, Li Y, Tang H, Chang P, Hess KR, Abbruzzese JL, et al. Insulin-like growth factor axis gene polymorphisms modify risk of pancreatic cancer. Cancer Epidemiol. 2012;36:206–11.CrossRefPubMed
32.
go back to reference Helu XM, De Leon D. Inhibition of HER2 and IGF2 triggers cell death in trastuzumab-resistant HER2 positive JIMT1 cells. Cancer Res. 2014;74:4244.CrossRef Helu XM, De Leon D. Inhibition of HER2 and IGF2 triggers cell death in trastuzumab-resistant HER2 positive JIMT1 cells. Cancer Res. 2014;74:4244.CrossRef
Metadata
Title
CDX2 inhibits pancreatic adenocarcinoma cell proliferation via promoting tumor suppressor miR-615-5p
Authors
Yuanyuan Jiang
Yan Zhang
Fuqing Li
Xiaolin Du
Jinping Zhang
Publication date
01-01-2016
Publisher
Springer Netherlands
Published in
Tumor Biology / Issue 1/2016
Print ISSN: 1010-4283
Electronic ISSN: 1423-0380
DOI
https://doi.org/10.1007/s13277-015-3900-6

Other articles of this Issue 1/2016

Tumor Biology 1/2016 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine