Skip to main content
Top
Published in: BMC Cancer 1/2012

Open Access 01-12-2012 | Research article

Cancer specific promoter CpG Islands hypermethylation of HOP homeobox (HOPX) gene and its potential tumor suppressive role in pancreatic carcinogenesis

Authors: Mina Waraya, Keishi Yamashita, Hiroshi Katoh, Akira Ooki, Hiroshi Kawamata, Hiroshi Nishimiya, Kazunori Nakamura, Akira Ema, Masahiko Watanabe

Published in: BMC Cancer | Issue 1/2012

Login to get access

Abstract

Background

We have recently identified HOP hoemobox (HOPX) as a tumor suppressor gene candidate, characterized by tumor-specific promoter DNA hypermethylation in human cancers, and it can remarkably inhibit tumors’ aggressive phenotypes. In this current study, we for the first time examined methylation level of HOPX and tested the functional relevance in pancreatic cancer (PC).

Methods

Clinical features of HOPX promoter hypermethylation was investigated in 89 PC tissues, and immunohistochemistry was added. We also examined its functional relevance in phenotype assays such as soft agar, proliferation, invasion, and cell cycle analysis.

Results

PC tissues had HOPX gene hypermethylation as compared to the corresponding normal pancreas tissues, and its uniqueness was robust to discriminate tumor from normal tissues (AUC = 0.85, P < 0.0001). Unexpectedly, HOPX was increased in expression in tumor tissues, and immunohistochemistry revealed its predominant expression in the Langerhans islet cells, where HOPX was reduced in expression for PC cells with promoter hypermethylation. HOPX transfectants exhibited G1 arrest with subG1 accumulation, and inhibited tumor forming and invasive ability.

Conclusion

Defective expression of HOPX which is consistent with promoter DNA hypermethylation may explain aggressive phenotype of pancreatic cancer, and intense expression of HOPX in the Langerhans cells may in turn uniquely contribute to pancreatic carcinogenesis.
Appendix
Available only for authorised users
Literature
2.
go back to reference Bird AP: CpG-rich islands and the function of DNA methylation. Nature. 1986, 321 (6067): 209-213. 10.1038/321209a0.CrossRefPubMed Bird AP: CpG-rich islands and the function of DNA methylation. Nature. 1986, 321 (6067): 209-213. 10.1038/321209a0.CrossRefPubMed
3.
go back to reference Goelz SE, Vogelstein B, Hamilton SR, Feinberg AP: Hypomethylation of DNA from benign and malignant human colon neoplasms. Science. 1985, 228 (4696): 187-190. 10.1126/science.2579435.CrossRefPubMed Goelz SE, Vogelstein B, Hamilton SR, Feinberg AP: Hypomethylation of DNA from benign and malignant human colon neoplasms. Science. 1985, 228 (4696): 187-190. 10.1126/science.2579435.CrossRefPubMed
4.
go back to reference Herman JG, Baylin SB: Gene silencing in cancer in association with promoter hypermethylation. N Engl J Med. 2003, 349 (21): 2042-2054. 10.1056/NEJMra023075.CrossRefPubMed Herman JG, Baylin SB: Gene silencing in cancer in association with promoter hypermethylation. N Engl J Med. 2003, 349 (21): 2042-2054. 10.1056/NEJMra023075.CrossRefPubMed
5.
go back to reference Yamashita K, Upadhyay S, Osada M, Hoque MO, Xiao Y, Mori M, Sato F, Meltzer SJ, Sidransky D: Pharmacologic unmasking of epigenetically silenced tumor suppressor genes in esophageal squamous cell carcinoma. Cancer Cell. 2002, 2 (6): 485-495. 10.1016/S1535-6108(02)00215-5.CrossRefPubMed Yamashita K, Upadhyay S, Osada M, Hoque MO, Xiao Y, Mori M, Sato F, Meltzer SJ, Sidransky D: Pharmacologic unmasking of epigenetically silenced tumor suppressor genes in esophageal squamous cell carcinoma. Cancer Cell. 2002, 2 (6): 485-495. 10.1016/S1535-6108(02)00215-5.CrossRefPubMed
6.
go back to reference Kim MS, Yamashita K, Baek JH, Park HL, Carvalho AL, Osada M, Hoque MO, Upadhyay S, Mori M, Moon C, et al: N-methyl-D-aspartate receptor type 2B is epigenetically inactivated and exhibits tumor-suppressive activity in human esophageal cancer. Cancer Res. 2006, 66 (7): 3409-3418. 10.1158/0008-5472.CAN-05-1608.CrossRefPubMed Kim MS, Yamashita K, Baek JH, Park HL, Carvalho AL, Osada M, Hoque MO, Upadhyay S, Mori M, Moon C, et al: N-methyl-D-aspartate receptor type 2B is epigenetically inactivated and exhibits tumor-suppressive activity in human esophageal cancer. Cancer Res. 2006, 66 (7): 3409-3418. 10.1158/0008-5472.CAN-05-1608.CrossRefPubMed
7.
go back to reference Yamashita K, Kim MS, Park HL, Tokumaru Y, Osada M, Inoue H, Mori M, Sidransky D: HOP/OB1/NECC1 promoter DNA is frequently hypermethylated and involved in tumorigenic ability in esophageal squamous cell carcinoma. Mol Cancer Res. 2008, 6 (1): 31-41. 10.1158/1541-7786.MCR-07-0213.CrossRefPubMed Yamashita K, Kim MS, Park HL, Tokumaru Y, Osada M, Inoue H, Mori M, Sidransky D: HOP/OB1/NECC1 promoter DNA is frequently hypermethylated and involved in tumorigenic ability in esophageal squamous cell carcinoma. Mol Cancer Res. 2008, 6 (1): 31-41. 10.1158/1541-7786.MCR-07-0213.CrossRefPubMed
8.
go back to reference Ooki A, Yamashita K, Kikuchi S, Sakuramoto S, Katada N, Kokubo K, Kobayashi H, Kim MS, Sidransky D, Watanabe M: Potential utility of HOP homeobox gene promoter methylation as a marker of tumor aggressiveness in gastric cancer. Oncogene. 2010, 29 (22): 3263-3275. 10.1038/onc.2010.76.CrossRefPubMed Ooki A, Yamashita K, Kikuchi S, Sakuramoto S, Katada N, Kokubo K, Kobayashi H, Kim MS, Sidransky D, Watanabe M: Potential utility of HOP homeobox gene promoter methylation as a marker of tumor aggressiveness in gastric cancer. Oncogene. 2010, 29 (22): 3263-3275. 10.1038/onc.2010.76.CrossRefPubMed
9.
go back to reference Chen F, Kook H, Milewski R, Gitler AD, Lu MM, Li J, Nazarian R, Schnepp R, Jen K, Biben C, et al: Hop is an unusual homeobox gene that modulates cardiac development. Cell. 2002, 110 (6): 713-723. 10.1016/S0092-8674(02)00932-7.CrossRefPubMed Chen F, Kook H, Milewski R, Gitler AD, Lu MM, Li J, Nazarian R, Schnepp R, Jen K, Biben C, et al: Hop is an unusual homeobox gene that modulates cardiac development. Cell. 2002, 110 (6): 713-723. 10.1016/S0092-8674(02)00932-7.CrossRefPubMed
10.
go back to reference Kook H, Yung WW, Simpson RJ, Kee HJ, Shin S, Lowry JA, Loughlin FE, Yin Z, Epstein JA, Mackay JP: Analysis of the structure and function of the transcriptional coregulator HOP. Biochemistry. 2006, 45 (35): 10584-10590. 10.1021/bi060641s.CrossRefPubMed Kook H, Yung WW, Simpson RJ, Kee HJ, Shin S, Lowry JA, Loughlin FE, Yin Z, Epstein JA, Mackay JP: Analysis of the structure and function of the transcriptional coregulator HOP. Biochemistry. 2006, 45 (35): 10584-10590. 10.1021/bi060641s.CrossRefPubMed
11.
go back to reference Asanoma K, Matsuda T, Kondo H, Kato K, Kishino T, Niikawa N, Wake N, Kato H: NECC1, a candidate choriocarcinoma suppressor gene that encodes a homeodomain consensus motif. Genomics. 2003, 81 (1): 15-25. 10.1016/S0888-7543(02)00011-3.CrossRefPubMed Asanoma K, Matsuda T, Kondo H, Kato K, Kishino T, Niikawa N, Wake N, Kato H: NECC1, a candidate choriocarcinoma suppressor gene that encodes a homeodomain consensus motif. Genomics. 2003, 81 (1): 15-25. 10.1016/S0888-7543(02)00011-3.CrossRefPubMed
12.
go back to reference Asanoma K, Kato H, Inoue T, Matsuda T, Wake N: Analysis of a candidate gene associated with growth suppression of choriocarcinoma and differentiation of trophoblasts. J Reprod Med. 2004, 49 (8): 617-626.PubMed Asanoma K, Kato H, Inoue T, Matsuda T, Wake N: Analysis of a candidate gene associated with growth suppression of choriocarcinoma and differentiation of trophoblasts. J Reprod Med. 2004, 49 (8): 617-626.PubMed
13.
go back to reference Chen Y, Petersen S, Pacyna-Gengelbach M, Pietas A, Petersen I: Identification of a novel homeobox-containing gene, LAGY, which is downregulated in lung cancer. Oncology. 2003, 64 (4): 450-458. 10.1159/000070306.CrossRefPubMed Chen Y, Petersen S, Pacyna-Gengelbach M, Pietas A, Petersen I: Identification of a novel homeobox-containing gene, LAGY, which is downregulated in lung cancer. Oncology. 2003, 64 (4): 450-458. 10.1159/000070306.CrossRefPubMed
14.
go back to reference Chen Y, Pacyna-Gengelbach M, Deutschmann N, Niesporek S, Petersen I: Homeobox gene HOP has a potential tumor suppressive activity in human lung cancer. Int J Cancer. 2007, 121 (5): 1021-1027. 10.1002/ijc.22753.CrossRefPubMed Chen Y, Pacyna-Gengelbach M, Deutschmann N, Niesporek S, Petersen I: Homeobox gene HOP has a potential tumor suppressive activity in human lung cancer. Int J Cancer. 2007, 121 (5): 1021-1027. 10.1002/ijc.22753.CrossRefPubMed
15.
go back to reference Yamaguchi S, Asanoma K, Takao T, Kato K, Wake N: Homeobox gene HOPX is epigenetically silenced in human uterine endometrial cancer and suppresses estrogen-stimulated proliferation of cancer cells by inhibiting serum response factor. Int J Cancer. 2009, 124 (11): 2577-2588. 10.1002/ijc.24217.CrossRefPubMed Yamaguchi S, Asanoma K, Takao T, Kato K, Wake N: Homeobox gene HOPX is epigenetically silenced in human uterine endometrial cancer and suppresses estrogen-stimulated proliferation of cancer cells by inhibiting serum response factor. Int J Cancer. 2009, 124 (11): 2577-2588. 10.1002/ijc.24217.CrossRefPubMed
16.
go back to reference Nishihira T, Hashimoto Y, Katayama M, Mori S, Kuroki T: Molecular and cellular features of esophageal cancer cells. J Cancer Res Clin Oncol. 1993, 119 (8): 441-449. 10.1007/BF01215923.CrossRefPubMed Nishihira T, Hashimoto Y, Katayama M, Mori S, Kuroki T: Molecular and cellular features of esophageal cancer cells. J Cancer Res Clin Oncol. 1993, 119 (8): 441-449. 10.1007/BF01215923.CrossRefPubMed
17.
go back to reference Livak KJ, Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) Method. Methods. 2001, 25 (4): 402-408. 10.1006/meth.2001.1262.CrossRefPubMed Livak KJ, Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) Method. Methods. 2001, 25 (4): 402-408. 10.1006/meth.2001.1262.CrossRefPubMed
18.
go back to reference Waraya M, Yamashita K, Katagiri H, Ishii K, Takahashi Y, Furuta K, Watanabe M: Preoperative serum CA19-9 and dissected peripancreatic tissue margin as determiners of long-term survival in pancreatic cancer. Ann Surg Oncol. 2009, 16 (5): 1231-1240. 10.1245/s10434-009-0415-7.CrossRefPubMed Waraya M, Yamashita K, Katagiri H, Ishii K, Takahashi Y, Furuta K, Watanabe M: Preoperative serum CA19-9 and dissected peripancreatic tissue margin as determiners of long-term survival in pancreatic cancer. Ann Surg Oncol. 2009, 16 (5): 1231-1240. 10.1245/s10434-009-0415-7.CrossRefPubMed
19.
go back to reference Dammann R, Schagdarsurengin U, Liu L, Otto N, Gimm O, Dralle H, Boehm BO, Pfeifer GP, Hoang-Vu C: Frequent RASSF1A promoter hypermethylation and K-ras mutations in pancreatic carcinoma. Oncogene. 2003, 22 (24): 3806-3812. 10.1038/sj.onc.1206582.CrossRefPubMed Dammann R, Schagdarsurengin U, Liu L, Otto N, Gimm O, Dralle H, Boehm BO, Pfeifer GP, Hoang-Vu C: Frequent RASSF1A promoter hypermethylation and K-ras mutations in pancreatic carcinoma. Oncogene. 2003, 22 (24): 3806-3812. 10.1038/sj.onc.1206582.CrossRefPubMed
21.
go back to reference Hennig R, Ding XZ, Adrian TE: On the role of the islets of Langerhans in pancreatic cancer. Histol Histopathol. 2004, 19 (3): 999-1011.PubMed Hennig R, Ding XZ, Adrian TE: On the role of the islets of Langerhans in pancreatic cancer. Histol Histopathol. 2004, 19 (3): 999-1011.PubMed
22.
go back to reference Pour PM, Weide L, Liu G, Kazakoff K, Scheetz M, Toshkov I, Ikematsu Y, Fienhold MA, Sanger W: Experimental evidence for the origin of ductal-type adenocarcinoma from the islets of Langerhans. Am J Pathol. 1997, 150 (3): 2167-2180.PubMedPubMedCentral Pour PM, Weide L, Liu G, Kazakoff K, Scheetz M, Toshkov I, Ikematsu Y, Fienhold MA, Sanger W: Experimental evidence for the origin of ductal-type adenocarcinoma from the islets of Langerhans. Am J Pathol. 1997, 150 (3): 2167-2180.PubMedPubMedCentral
23.
go back to reference Schmied BM, Ulrich AB, Matsuzaki H, Li C, Friess H, Bochler MW, Andron-Sandberg A, Adrian TE, Pour PM: Alteration of the Langerhans islets in pancreatic cancer patients. Int J Pancreatol. 2000, 28 (3): 187-197. 10.1385/IJGC:28:3:187.CrossRefPubMed Schmied BM, Ulrich AB, Matsuzaki H, Li C, Friess H, Bochler MW, Andron-Sandberg A, Adrian TE, Pour PM: Alteration of the Langerhans islets in pancreatic cancer patients. Int J Pancreatol. 2000, 28 (3): 187-197. 10.1385/IJGC:28:3:187.CrossRefPubMed
24.
go back to reference Schmied BM, Ulrich AB, Matsuzaki H, Li C, Pour PM: In vitro pancreatic carcinogenesis. Ann Oncol. 1999, 10 (4): 41-45. 10.1023/A:1008328517563.CrossRefPubMed Schmied BM, Ulrich AB, Matsuzaki H, Li C, Pour PM: In vitro pancreatic carcinogenesis. Ann Oncol. 1999, 10 (4): 41-45. 10.1023/A:1008328517563.CrossRefPubMed
25.
go back to reference El-Ghamari M, Bergmann F, Schmied BM, Weitz J, Ulrich A: Islet cells contribute to pancreatic carcinogenesis in an animal model. Pancreas. 2011, 40 (2): 242-246. 10.1097/MPA.0b013e3182016a08.CrossRefPubMed El-Ghamari M, Bergmann F, Schmied BM, Weitz J, Ulrich A: Islet cells contribute to pancreatic carcinogenesis in an animal model. Pancreas. 2011, 40 (2): 242-246. 10.1097/MPA.0b013e3182016a08.CrossRefPubMed
26.
go back to reference Takahashi M, Furihata M, Akimitsu N, Watanabe M, Kaul S, Yumoto N, Okada T: A highly bone marrow metastatic murine breast cancer model established through in vivo selection exhibits enhanced anchorage-independent growth and cell migration mediated by ICAM-1. Clin Exp Metastasis. 2008, 25 (5): 517-529. 10.1007/s10585-008-9163-5.CrossRefPubMed Takahashi M, Furihata M, Akimitsu N, Watanabe M, Kaul S, Yumoto N, Okada T: A highly bone marrow metastatic murine breast cancer model established through in vivo selection exhibits enhanced anchorage-independent growth and cell migration mediated by ICAM-1. Clin Exp Metastasis. 2008, 25 (5): 517-529. 10.1007/s10585-008-9163-5.CrossRefPubMed
27.
go back to reference Katoh H, Yamashita K, Waraya M, Margalit O, Ooki A, Tamaki H, Sakagami H, Kokubo K, Sidransky D, Watanabe M: Epigenetic silencing of HOPX promotes cancer progression in colorectal cancer. Neoplasia. 2012, 14 (7): 559-571.CrossRefPubMedPubMedCentral Katoh H, Yamashita K, Waraya M, Margalit O, Ooki A, Tamaki H, Sakagami H, Kokubo K, Sidransky D, Watanabe M: Epigenetic silencing of HOPX promotes cancer progression in colorectal cancer. Neoplasia. 2012, 14 (7): 559-571.CrossRefPubMedPubMedCentral
28.
go back to reference Kook H, Lepore JJ, Gitler AD, Lu MM, Wing-Man Yung W, Mackay J, Zhou R, Ferrari V, Gruber P, Epstein JA: Cardiac hypertrophy and histone deacetylase-dependent transcriptional repression mediated by the atypical homeodomain protein Hop. J Clin Invest. 2003, 112 (6): 863-871.CrossRefPubMedPubMedCentral Kook H, Lepore JJ, Gitler AD, Lu MM, Wing-Man Yung W, Mackay J, Zhou R, Ferrari V, Gruber P, Epstein JA: Cardiac hypertrophy and histone deacetylase-dependent transcriptional repression mediated by the atypical homeodomain protein Hop. J Clin Invest. 2003, 112 (6): 863-871.CrossRefPubMedPubMedCentral
29.
go back to reference Yin Z, Gonzales L, Kolla V, Rath N, Zhang Y, Lu MM, Kimura S, Ballard PL, Beers MF, Epstein JA, et al: Hop functions downstream of Nkx2.1 and GATA6 to mediate HDAC-dependent negative regulation of pulmonary gene expression. Am J Physiol Lung Cell Mol Physiol. 2006, 291 (2): L191-L199. 10.1152/ajplung.00385.2005.CrossRefPubMed Yin Z, Gonzales L, Kolla V, Rath N, Zhang Y, Lu MM, Kimura S, Ballard PL, Beers MF, Epstein JA, et al: Hop functions downstream of Nkx2.1 and GATA6 to mediate HDAC-dependent negative regulation of pulmonary gene expression. Am J Physiol Lung Cell Mol Physiol. 2006, 291 (2): L191-L199. 10.1152/ajplung.00385.2005.CrossRefPubMed
30.
go back to reference Kee HJ, Kim JR, Nam KI, Park HY, Shin S, Kim JC, Shimono Y, Takahashi M, Jeong MH, Kim N, et al: Enhancer of polycomb1, a novel homeodomain only protein-binding partner, induces skeletal muscle differentiation. J Biol Chem. 2007, 282 (10): 7700-7709.CrossRefPubMed Kee HJ, Kim JR, Nam KI, Park HY, Shin S, Kim JC, Shimono Y, Takahashi M, Jeong MH, Kim N, et al: Enhancer of polycomb1, a novel homeodomain only protein-binding partner, induces skeletal muscle differentiation. J Biol Chem. 2007, 282 (10): 7700-7709.CrossRefPubMed
31.
go back to reference Attwooll C, Oddi S, Cartwright P, Prosperini E, Agger K, Steensgaard P, Wagener C, Sardet C, Moroni MC, Helin K: A novel repressive E2F6 complex containing the polycomb group protein, EPC1, that interacts with EZH2 in a proliferation-specific manner. J Biol Chem. 2005, 280 (2): 1199-1208.CrossRefPubMed Attwooll C, Oddi S, Cartwright P, Prosperini E, Agger K, Steensgaard P, Wagener C, Sardet C, Moroni MC, Helin K: A novel repressive E2F6 complex containing the polycomb group protein, EPC1, that interacts with EZH2 in a proliferation-specific manner. J Biol Chem. 2005, 280 (2): 1199-1208.CrossRefPubMed
32.
go back to reference Aoto T, Saitoh N, Sakamoto Y, Watanabe S, Nakao M: Polycomb group protein-associated chromatin is reproduced in post-mitotic G1 phase and is required for S phase progression. J Biol Chem. 2008, 283 (27): 18905-18915. 10.1074/jbc.M709322200.CrossRefPubMed Aoto T, Saitoh N, Sakamoto Y, Watanabe S, Nakao M: Polycomb group protein-associated chromatin is reproduced in post-mitotic G1 phase and is required for S phase progression. J Biol Chem. 2008, 283 (27): 18905-18915. 10.1074/jbc.M709322200.CrossRefPubMed
Metadata
Title
Cancer specific promoter CpG Islands hypermethylation of HOP homeobox (HOPX) gene and its potential tumor suppressive role in pancreatic carcinogenesis
Authors
Mina Waraya
Keishi Yamashita
Hiroshi Katoh
Akira Ooki
Hiroshi Kawamata
Hiroshi Nishimiya
Kazunori Nakamura
Akira Ema
Masahiko Watanabe
Publication date
01-12-2012
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2012
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/1471-2407-12-397

Other articles of this Issue 1/2012

BMC Cancer 1/2012 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