Skip to main content
Top
Published in: BMC Cancer 1/2016

Open Access 01-12-2016 | Research article

RASSF1 tumor suppressor gene in pancreatic ductal adenocarcinoma: correlation of expression, chromosomal status and epigenetic changes

Authors: Eliana Amato, Stefano Barbi, Matteo Fassan, Claudio Luchini, Caterina Vicentini, Matteo Brunelli, Giuseppe Malleo, Aldo Scarpa, Giorgio Malpeli

Published in: BMC Cancer | Issue 1/2016

Login to get access

Abstract

Background

The Ras Association Domain Family Member 1 (RASSF1) is one of the most frequently reported methylation-inactivated tumor suppressor genes in primary pancreatic ductal adenocarcinomas (PDAC). Limited information is still available about the impact of RASSF1 gene silencing on the expression of its different isoforms in neoplastic cells.

Methods

A series of 96 primary PDAC, with known clinico-pathological parameters, was tested for RASSF1 methylation status by methylation-specific PCR, RASSF1 locus copy number alterations by fluorescence in situ hybridization, and Rassf1a protein expression by immunohistochemistry. A further series of 14 xenografted primary PDAC and 8 PDAC-derived cell lines were tested to obtain a detailed methylation mapping of CpG islands A and C of the RASSF1 locus by pyrosequencing and to evaluate the expression of Rassf1 variants by qRT-PCR.

Results

Methylation of CpG island A of the RASSF1 gene was observed in 35 % of the tumors and allelic loss of RASSF1 locus was seen in 30 disomic and in 20 polysomic cases (52 %). Rassf1a immunohistochemical expression was downregulated in half of primary PDAC, and this downregulation was neither correlated with methylation of RASSF1 promoter nor with RASSF1 copy number alterations. RASSF1 status did not influence patients’ prognosis. The expression of the seven RASSF1 isoforms in xenografts and cell lines showed that RASSF1A, RASSF1B, and RASSF1C isoforms were present in all xenografts and cell lines, whereas RASSF1D, RASSF1E, and RASSF1F isoforms were variably expressed among samples. RASSF1G was never expressed in either xenografts or cell lines. The variable expression of RASSF1 isoforms in PDAC xenografts and cell lines was not dependent on RASSF1 methylation status of CpG islands A and C.

Conclusions

RASSF1 alterations occurring in PDAC mainly consist in variations of expression of the different isoforms. Different genetic mechanisms seem to contribute to RASSF1 deregulation in this setting, but RASSF1 methylation does not seem to substantially affect RASSF1 isoforms expression.
Appendix
Available only for authorised users
Literature
1.
go back to reference Nones K, Waddell N, Song S, Patch AM, Miller D, Johns A, et al. Genome-wide DNA methylation patterns in pancreatic ductal adenocarcinoma reveal epigenetic deregulation of SLIT-ROBO, ITGA2 and MET signaling. Int J Cancer. 2014;135:1110–8.PubMedCrossRef Nones K, Waddell N, Song S, Patch AM, Miller D, Johns A, et al. Genome-wide DNA methylation patterns in pancreatic ductal adenocarcinoma reveal epigenetic deregulation of SLIT-ROBO, ITGA2 and MET signaling. Int J Cancer. 2014;135:1110–8.PubMedCrossRef
2.
go back to reference Dammann R, Schagdarsurengin U, Liu L, Otto N, Gimm O, Dralle H, et al. Frequent RASSF1A promoter hypermethylation and K-ras mutations in pancreatic carcinoma. Oncogene. 2003;22:3806–12.PubMedCrossRef Dammann R, Schagdarsurengin U, Liu L, Otto N, Gimm O, Dralle H, et al. Frequent RASSF1A promoter hypermethylation and K-ras mutations in pancreatic carcinoma. Oncogene. 2003;22:3806–12.PubMedCrossRef
3.
go back to reference Dammann R, Li C, Yoon JH, Chin PL, Bates S, Pfeifer GP. Epigenetic inactivation of a RAS association domain family protein from the lung tumour suppressor locus 3p21.3. Nat Genet. 2000;25:315–9.PubMedCrossRef Dammann R, Li C, Yoon JH, Chin PL, Bates S, Pfeifer GP. Epigenetic inactivation of a RAS association domain family protein from the lung tumour suppressor locus 3p21.3. Nat Genet. 2000;25:315–9.PubMedCrossRef
4.
go back to reference Rodriguez-Viciana P, Sabatier C, McCormick F. Signaling specificity by Ras family GTPases is determined by the full spectrum of effectors they regulate. Mol Cell Biol. 2004;24:4943–54.PubMedPubMedCentralCrossRef Rodriguez-Viciana P, Sabatier C, McCormick F. Signaling specificity by Ras family GTPases is determined by the full spectrum of effectors they regulate. Mol Cell Biol. 2004;24:4943–54.PubMedPubMedCentralCrossRef
5.
go back to reference Shivakumar L, Minna J, Sakamaki T, Pestell R, White MA. The RASSF1A tumor suppressor blocks cell cycle progression and inhibits cyclin D1 accumulation. Mol Cell Biol. 2002;22:4309–18.PubMedPubMedCentralCrossRef Shivakumar L, Minna J, Sakamaki T, Pestell R, White MA. The RASSF1A tumor suppressor blocks cell cycle progression and inhibits cyclin D1 accumulation. Mol Cell Biol. 2002;22:4309–18.PubMedPubMedCentralCrossRef
6.
go back to reference Vos MD, Ellis CA, Bell A, Birrer MJ, Clark GJ. Ras uses the novel tumor suppressor RASSF1 as an effector to mediate apoptosis. J Biol Chem. 2000;275(46):35669–72.PubMedCrossRef Vos MD, Ellis CA, Bell A, Birrer MJ, Clark GJ. Ras uses the novel tumor suppressor RASSF1 as an effector to mediate apoptosis. J Biol Chem. 2000;275(46):35669–72.PubMedCrossRef
7.
go back to reference Arnette C, Efimova N, Zhu X, Clark GJ, Kaverina I. Microtubule segment stabilization by RASSF1A is required for proper microtubule dynamics and Golgi integrity. Mol Biol Cell. 2014;25:800–10.PubMedPubMedCentralCrossRef Arnette C, Efimova N, Zhu X, Clark GJ, Kaverina I. Microtubule segment stabilization by RASSF1A is required for proper microtubule dynamics and Golgi integrity. Mol Biol Cell. 2014;25:800–10.PubMedPubMedCentralCrossRef
8.
go back to reference Fu L, Zhang S. RASSF1A promotes apoptosis and suppresses the proliferation of ovarian cancer cells. Int J Mol Med. 2014;33:1153–60.PubMed Fu L, Zhang S. RASSF1A promotes apoptosis and suppresses the proliferation of ovarian cancer cells. Int J Mol Med. 2014;33:1153–60.PubMed
9.
go back to reference Liu L, Guo C, Dammann R, Tommasi S, Pfeifer GP. RASSF1A interacts with and activates the mitotic kinase Aurora-A. Oncogene. 2008;27:6175–86.PubMedCrossRef Liu L, Guo C, Dammann R, Tommasi S, Pfeifer GP. RASSF1A interacts with and activates the mitotic kinase Aurora-A. Oncogene. 2008;27:6175–86.PubMedCrossRef
10.
go back to reference Reeves ME, Baldwin SW, Baldwin ML, Chen ST, Moretz JM, Aragon RJ, et al. Ras-association domain family 1C protein promotes breast cancer cell migration and attenuates apoptosis. BMC Cancer. 2010;10:562.PubMedPubMedCentralCrossRef Reeves ME, Baldwin SW, Baldwin ML, Chen ST, Moretz JM, Aragon RJ, et al. Ras-association domain family 1C protein promotes breast cancer cell migration and attenuates apoptosis. BMC Cancer. 2010;10:562.PubMedPubMedCentralCrossRef
11.
go back to reference Reeves ME, Firek M, Chen ST, Amaar Y. The RASSF1 gene and the opposing effects of the RASSF1A and RASSF1C isoforms on cell proliferation and apoptosis. Mol Biol Int. 2013;2013:145096.PubMedPubMedCentralCrossRef Reeves ME, Firek M, Chen ST, Amaar Y. The RASSF1 gene and the opposing effects of the RASSF1A and RASSF1C isoforms on cell proliferation and apoptosis. Mol Biol Int. 2013;2013:145096.PubMedPubMedCentralCrossRef
12.
go back to reference van der Weyden L, Adams DJ. The Ras-association domain family (RASSF) members and their role in human tumourigenesis. Biochim Biophys Acta. 2007;1776:58–85.PubMedPubMedCentral van der Weyden L, Adams DJ. The Ras-association domain family (RASSF) members and their role in human tumourigenesis. Biochim Biophys Acta. 2007;1776:58–85.PubMedPubMedCentral
13.
go back to reference Guo W, Cui L, Wang C, Guo Y, Shen S, Kuang G, et al. Decreased expression of RASSF1A and up-regulation of RASSF1C is associated with esophageal squamous cell carcinoma. Clin Exp Metastasis. 2014;31:521–33.PubMedCrossRef Guo W, Cui L, Wang C, Guo Y, Shen S, Kuang G, et al. Decreased expression of RASSF1A and up-regulation of RASSF1C is associated with esophageal squamous cell carcinoma. Clin Exp Metastasis. 2014;31:521–33.PubMedCrossRef
14.
go back to reference Grawenda AM, O’Neill E. Clinical utility of RASSF1A methylation in human malignancies. Br J Cancer. 2015;113:372–81.PubMedCrossRef Grawenda AM, O’Neill E. Clinical utility of RASSF1A methylation in human malignancies. Br J Cancer. 2015;113:372–81.PubMedCrossRef
15.
go back to reference Donninger H, Clark JA, Monaghan MK, Schmidt ML, Vos M, Clark GJ. Cell cycle restriction is more important than apoptosis induction for RASSF1A protein tumor suppression. J Biol Chem. 2014;289:31287–95.PubMedPubMedCentralCrossRef Donninger H, Clark JA, Monaghan MK, Schmidt ML, Vos M, Clark GJ. Cell cycle restriction is more important than apoptosis induction for RASSF1A protein tumor suppression. J Biol Chem. 2014;289:31287–95.PubMedPubMedCentralCrossRef
16.
go back to reference Honda S, Haruta M, Sugawara W, Sasaki F, Ohira M, Matsunaga T, et al. The methylation status of RASSF1A promoter predicts responsiveness to chemotherapy and eventual cure in hepatoblastoma patients. Int J Cancer. 2008;123:1117–25.PubMedCrossRef Honda S, Haruta M, Sugawara W, Sasaki F, Ohira M, Matsunaga T, et al. The methylation status of RASSF1A promoter predicts responsiveness to chemotherapy and eventual cure in hepatoblastoma patients. Int J Cancer. 2008;123:1117–25.PubMedCrossRef
17.
go back to reference Liu L, Kron KJ, Pethe VV, Demetrashvili N, Nesbitt ME, Trachtenberg J, et al. Association of tissue promoter methylation levels of APC, TGFβ2, HOXD3 and RASSF1A with prostate cancer progression. Int J Cancer. 2011;129:2454–62.PubMedCrossRef Liu L, Kron KJ, Pethe VV, Demetrashvili N, Nesbitt ME, Trachtenberg J, et al. Association of tissue promoter methylation levels of APC, TGFβ2, HOXD3 and RASSF1A with prostate cancer progression. Int J Cancer. 2011;129:2454–62.PubMedCrossRef
18.
go back to reference Kim JS, Chae Y, Ha YS, Kim IY, Byun SS, Yun SJ, et al. Ras association domain family 1A: a promising prognostic marker in recurrent nonmuscle invasive bladder cancer. Clin Genitourin Cancer. 2012;10:114–20.PubMedCrossRef Kim JS, Chae Y, Ha YS, Kim IY, Byun SS, Yun SJ, et al. Ras association domain family 1A: a promising prognostic marker in recurrent nonmuscle invasive bladder cancer. Clin Genitourin Cancer. 2012;10:114–20.PubMedCrossRef
19.
go back to reference Kawai Y, Sakano S, Suehiro Y, Okada T, Korenaga Y, Hara T, et al. Methylation level of the RASSF1A promoter is an independent prognostic factor for clear-cell renal cell carcinoma. Ann Oncol. 2010;21:1612–7.PubMedCrossRef Kawai Y, Sakano S, Suehiro Y, Okada T, Korenaga Y, Hara T, et al. Methylation level of the RASSF1A promoter is an independent prognostic factor for clear-cell renal cell carcinoma. Ann Oncol. 2010;21:1612–7.PubMedCrossRef
20.
go back to reference Hesson L, Dallol A, Minna JD, Maher ER, Latif F. NORE1A, a homologue of RASSF1A tumour suppressor gene is inactivated in human cancers. Oncogene. 2003;22:947–54.PubMedCrossRef Hesson L, Dallol A, Minna JD, Maher ER, Latif F. NORE1A, a homologue of RASSF1A tumour suppressor gene is inactivated in human cancers. Oncogene. 2003;22:947–54.PubMedCrossRef
21.
go back to reference Qian ZR, Sano T, Yoshimoto K, Yamada S, Ishizuka A, Mizusawa N, et al. Inactivation of RASSF1A tumor suppressor gene by aberrant promoter hypermethylation in human pituitary adenomas. Lab Invest. 2005;85:464–73.PubMedCrossRef Qian ZR, Sano T, Yoshimoto K, Yamada S, Ishizuka A, Mizusawa N, et al. Inactivation of RASSF1A tumor suppressor gene by aberrant promoter hypermethylation in human pituitary adenomas. Lab Invest. 2005;85:464–73.PubMedCrossRef
22.
go back to reference Fischer JR, Ohnmacht U, Rieger N, Zemaitis M, Stoffregen C, Manegold C, et al. Prognostic significance of RASSF1A promoter methylation on survival of non-small cell lung cancer patients treated with gemcitabine. Lung Cancer. 2007;56:115–23.PubMedCrossRef Fischer JR, Ohnmacht U, Rieger N, Zemaitis M, Stoffregen C, Manegold C, et al. Prognostic significance of RASSF1A promoter methylation on survival of non-small cell lung cancer patients treated with gemcitabine. Lung Cancer. 2007;56:115–23.PubMedCrossRef
23.
go back to reference Agathanggelou A, Cooper WN, Latif F. Role of the Ras-association domain family 1 tumor suppressor gene in human cancers. Cancer Res. 2005;65:3497–508.PubMedCrossRef Agathanggelou A, Cooper WN, Latif F. Role of the Ras-association domain family 1 tumor suppressor gene in human cancers. Cancer Res. 2005;65:3497–508.PubMedCrossRef
24.
go back to reference Calhoun ES, Hucl T, Gallmeier E, West KM, Arking DE, Maitra A, et al. Identifying allelic loss and homozygous deletions in pancreatic cancer without matched normals using high-density single-nucleotide polymorphism arrays. Cancer Res. 2006;66:7920–8.PubMedCrossRef Calhoun ES, Hucl T, Gallmeier E, West KM, Arking DE, Maitra A, et al. Identifying allelic loss and homozygous deletions in pancreatic cancer without matched normals using high-density single-nucleotide polymorphism arrays. Cancer Res. 2006;66:7920–8.PubMedCrossRef
25.
go back to reference Iacobuzio-Donahue CA, van der Heijden MS, Baumgartner MR, Troup WJ, Romm JM, Doheny K, et al. Large-scale allelotype of pancreaticobiliary carcinoma provides quantitative estimates of genome-wide allelic loss. Cancer Res. 2004;64:871–5.PubMedCrossRef Iacobuzio-Donahue CA, van der Heijden MS, Baumgartner MR, Troup WJ, Romm JM, Doheny K, et al. Large-scale allelotype of pancreaticobiliary carcinoma provides quantitative estimates of genome-wide allelic loss. Cancer Res. 2004;64:871–5.PubMedCrossRef
26.
go back to reference Harada T, Chelala C, Bhakta V, Chaplin T, Caulee K, Baril P, et al. Genome-wide DNA copy number analysis in pancreatic cancer using high-density single nucleotide polymorphism arrays. Oncogene. 2008;27:1951–60.PubMedPubMedCentralCrossRef Harada T, Chelala C, Bhakta V, Chaplin T, Caulee K, Baril P, et al. Genome-wide DNA copy number analysis in pancreatic cancer using high-density single nucleotide polymorphism arrays. Oncogene. 2008;27:1951–60.PubMedPubMedCentralCrossRef
27.
go back to reference Sorio C, Bonora A, Orlandini S, Moore PS, Capelli P, Cristofori P, et al. Successful xenografting of cryopreserved primary pancreatic cancers. Virchows Arch. 2001;438:154–8.PubMedCrossRef Sorio C, Bonora A, Orlandini S, Moore PS, Capelli P, Cristofori P, et al. Successful xenografting of cryopreserved primary pancreatic cancers. Virchows Arch. 2001;438:154–8.PubMedCrossRef
28.
go back to reference Moore PS, Sipos B, Orlandini S, Sorio C, Real FX, Lemoine NR, et al. Genetic profile of 22 pancreatic carcinoma cell lines. Analysis of K-ras, p53, p16 and DPC4/Smad4. Virchows Arch. 2001;439:798–802.PubMedCrossRef Moore PS, Sipos B, Orlandini S, Sorio C, Real FX, Lemoine NR, et al. Genetic profile of 22 pancreatic carcinoma cell lines. Analysis of K-ras, p53, p16 and DPC4/Smad4. Virchows Arch. 2001;439:798–802.PubMedCrossRef
29.
go back to reference Luchini C, Parcesepe P, Nottegar A, Parolini C, Mafficini A, Remo A, et al. CD71 in gestational pathology: a versatile immunohistochemical marker with new possible applications. Appl Immunohistochem Mol Morphol. 2015 Apr 21. [Epub ahead of print]. Luchini C, Parcesepe P, Nottegar A, Parolini C, Mafficini A, Remo A, et al. CD71 in gestational pathology: a versatile immunohistochemical marker with new possible applications. Appl Immunohistochem Mol Morphol. 2015 Apr 21. [Epub ahead of print].
31.
go back to reference Hu L, Chen G, Yu H, Qiu X. Clinicopathological significance of RASSF1A reduced expression and hypermethylation in hepatocellular carcinoma. Hepatol Int. 2010;4:423–32.PubMedPubMedCentralCrossRef Hu L, Chen G, Yu H, Qiu X. Clinicopathological significance of RASSF1A reduced expression and hypermethylation in hepatocellular carcinoma. Hepatol Int. 2010;4:423–32.PubMedPubMedCentralCrossRef
32.
go back to reference Lichter P, Ledbetter SA, Ledbetter DH, Ward DC. Fluorescence in situ hybridization with Alu and L1 polymerase chain reaction probes for rapid characterization of human chromosomes in hybrid cell lines. Proc Natl Acad Sci U S A. 1990;87:6634–8.PubMedPubMedCentralCrossRef Lichter P, Ledbetter SA, Ledbetter DH, Ward DC. Fluorescence in situ hybridization with Alu and L1 polymerase chain reaction probes for rapid characterization of human chromosomes in hybrid cell lines. Proc Natl Acad Sci U S A. 1990;87:6634–8.PubMedPubMedCentralCrossRef
33.
go back to reference Pelosi G, Del Curto B, Trubia M, Nicholson AG, Manzotti M, Veronesi G, et al. 3q26 Amplification and polysomy of chromosome 3 in squamous cell lesions of the lung: a fluorescence in situ hybridization study. Clin Cancer Res. 2007;13:1995–2004.PubMedCrossRef Pelosi G, Del Curto B, Trubia M, Nicholson AG, Manzotti M, Veronesi G, et al. 3q26 Amplification and polysomy of chromosome 3 in squamous cell lesions of the lung: a fluorescence in situ hybridization study. Clin Cancer Res. 2007;13:1995–2004.PubMedCrossRef
34.
go back to reference Pizzi S, Azzoni C, Bottarelli L, Campanini N, D’Adda T, Pasquali C, et al. RASSF1A promoter methylation and 3p21.3 loss of heterozygosity are features of foregut, but not midgut and hindgut, malignant endocrine tumours. J Pathol. 2005;206:409–16.PubMedCrossRef Pizzi S, Azzoni C, Bottarelli L, Campanini N, D’Adda T, Pasquali C, et al. RASSF1A promoter methylation and 3p21.3 loss of heterozygosity are features of foregut, but not midgut and hindgut, malignant endocrine tumours. J Pathol. 2005;206:409–16.PubMedCrossRef
35.
go back to reference Colella S, Shen L, Baggerly KA, Issa JP, Krahe R. Sensitive and quantitative universal Pyrosequencing methylation analysis of CpG sites. Biotechniques. 2003;35:146–50.PubMed Colella S, Shen L, Baggerly KA, Issa JP, Krahe R. Sensitive and quantitative universal Pyrosequencing methylation analysis of CpG sites. Biotechniques. 2003;35:146–50.PubMed
36.
go back to reference Malpeli G, Amato E, Dandrea M, Fumagalli C, Debattisti V, Boninsegna L, et al. Methylation-associated down-regulation of RASSF1A and up-regulation of RASSF1C in pancreatic endocrine tumors. BMC Cancer. 2011;11:351.PubMedPubMedCentralCrossRef Malpeli G, Amato E, Dandrea M, Fumagalli C, Debattisti V, Boninsegna L, et al. Methylation-associated down-regulation of RASSF1A and up-regulation of RASSF1C in pancreatic endocrine tumors. BMC Cancer. 2011;11:351.PubMedPubMedCentralCrossRef
37.
go back to reference Ishimura N, Yamasawa K, Karim Rumi MA, Kadowaki Y, Ishihara S, Amano Y, et al. BRAF and K-ras gene mutations in human pancreatic cancers. Cancer Lett. 2003;199:169–73.PubMedCrossRef Ishimura N, Yamasawa K, Karim Rumi MA, Kadowaki Y, Ishihara S, Amano Y, et al. BRAF and K-ras gene mutations in human pancreatic cancers. Cancer Lett. 2003;199:169–73.PubMedCrossRef
38.
go back to reference Calhoun ES, Jones JB, Ashfaq R, Adsay V, Baker SJ, Valentine V, et al. BRAF and FBXW7 (CDC4, FBW7, AGO, SEL10) mutations in distinct subsets of pancreatic cancer: potential therapeutic targets. Am J Pathol. 2003;163:1255–60.PubMedPubMedCentralCrossRef Calhoun ES, Jones JB, Ashfaq R, Adsay V, Baker SJ, Valentine V, et al. BRAF and FBXW7 (CDC4, FBW7, AGO, SEL10) mutations in distinct subsets of pancreatic cancer: potential therapeutic targets. Am J Pathol. 2003;163:1255–60.PubMedPubMedCentralCrossRef
39.
go back to reference Bosman FT, Carneiro F, Hruban RH, Theise ND. WHO classification of tumours of the digestive system. Lyon: IARC Press; 2010. Bosman FT, Carneiro F, Hruban RH, Theise ND. WHO classification of tumours of the digestive system. Lyon: IARC Press; 2010.
40.
go back to reference Haruta M, Kamijo T, Nakagawara A, Kaneko Y. RASSF1A methylation may have two biological roles in neuroblastoma tumorigenesis depending on the ploidy status and age of patients. Cancer Lett. 2014;348:167–76.PubMedCrossRef Haruta M, Kamijo T, Nakagawara A, Kaneko Y. RASSF1A methylation may have two biological roles in neuroblastoma tumorigenesis depending on the ploidy status and age of patients. Cancer Lett. 2014;348:167–76.PubMedCrossRef
Metadata
Title
RASSF1 tumor suppressor gene in pancreatic ductal adenocarcinoma: correlation of expression, chromosomal status and epigenetic changes
Authors
Eliana Amato
Stefano Barbi
Matteo Fassan
Claudio Luchini
Caterina Vicentini
Matteo Brunelli
Giuseppe Malleo
Aldo Scarpa
Giorgio Malpeli
Publication date
01-12-2016
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2016
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-016-2048-0

Other articles of this Issue 1/2016

BMC Cancer 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