Skip to main content
Top
Published in: Molecular Cancer 1/2009

Open Access 01-12-2009 | Research

The novel RASSF6 and RASSF10 candidate tumour suppressor genes are frequently epigenetically inactivated in childhood leukaemias

Authors: Luke B Hesson, Thomas L Dunwell, Wendy N Cooper, Daniel Catchpoole, Anna T Brini, Raffaella Chiaramonte, Mike Griffiths, Andrew D Chalmers, Eamonn R Maher, Farida Latif

Published in: Molecular Cancer | Issue 1/2009

Login to get access

Abstract

Background

The Ras-assocation family (RASSF) of tumour suppressor genes (TSGs) contains 10 members that encode proteins containing Ras-assocation (RA) domains. Several members of the RASSF family are frequently epigenetically inactivated in cancer, however, their role in leukaemia has remained largely uninvestigated. Also, RASSF10 is a predicted gene yet to be experimentally verified. Here we cloned, characterised and demonstrated expression of RASSF10 in normal human bone marrow. We also determined the methylation status of CpG islands associated with RASSF1–10 in a series of childhood acute lymphocytic leukaemias (ALL) and normal blood and bone marrow samples.

Results

COBRA and bisulphite sequencing revealed RASSF6 and RASSF10 were the only RASSF members with a high frequency of leukaemia-specific methylation. RASSF6 was methylated in 94% (48/51) B-ALL and 41% (12/29) T-ALL, whilst RASSF10 was methylated in 16% (8/51) B-ALL and 88% (23/26) T-ALL. RASSF6 and RASSF10 expression inversely correlated with methylation which was restored by treatment with 5-aza-2'deoxycytidine (5azaDC).

Conclusion

This study shows the hypermethylation profile of RASSF genes in leukaemias is distinct from that of solid tumours and represents the first report of inactivation of RASSF6 or RASSF10 in cancer. These data show epigenetic inactivation of the candidate TSGs RASSF6 and RASSF10 is an extremely frequent event in the pathogenesis of childhood leukaemia. This study also warrants further investigation of the newly identified RASSF member RASSF10 and its potential role in leukaemia.
Appendix
Available only for authorised users
Literature
1.
go back to reference Downward J: Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer. 2003, 3: 11-22. 10.1038/nrc969.CrossRefPubMed Downward J: Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer. 2003, 3: 11-22. 10.1038/nrc969.CrossRefPubMed
2.
go back to reference Weyden van der L, Adams DJ: The Ras-association domain family (RASSF) members and their role in human tumourigenesis. Biochim Biophys Acta. 2007, 1776: 58-85.PubMedCentralPubMed Weyden van der L, Adams DJ: The Ras-association domain family (RASSF) members and their role in human tumourigenesis. Biochim Biophys Acta. 2007, 1776: 58-85.PubMedCentralPubMed
4.
go back to reference Khokhlatchev A, Rabizadeh S, Xavier R, Nedwidek M, Chen T, Zhang XF, Seed B, Avruch J: Identification of a novel Ras-regulated proapoptotic pathway. Curr Biol. 2002, 12: 253-265. 10.1016/S0960-9822(02)00683-8.CrossRefPubMed Khokhlatchev A, Rabizadeh S, Xavier R, Nedwidek M, Chen T, Zhang XF, Seed B, Avruch J: Identification of a novel Ras-regulated proapoptotic pathway. Curr Biol. 2002, 12: 253-265. 10.1016/S0960-9822(02)00683-8.CrossRefPubMed
5.
go back to reference Praskova M, Khoklatchev A, Ortiz-Vega S, Avruch J: Regulation of the MST1 kinase by autophosphorylation, by the growth inhibitory proteins, RASSF1 and NORE1, and by Ras. Biochem J. 2004, 381: 453-462. 10.1042/BJ20040025.PubMedCentralCrossRefPubMed Praskova M, Khoklatchev A, Ortiz-Vega S, Avruch J: Regulation of the MST1 kinase by autophosphorylation, by the growth inhibitory proteins, RASSF1 and NORE1, and by Ras. Biochem J. 2004, 381: 453-462. 10.1042/BJ20040025.PubMedCentralCrossRefPubMed
6.
go back to reference Vos MD, Dallol A, Eckfeld K, Allen NP, Donninger H, Hesson LB, Calvisi D, Latif F, Clark GJ: The RASSF1A tumor suppressor activates Bax via MOAP-1. J Biol Chem. 2006, 281: 4557-4563. 10.1074/jbc.M512128200.CrossRefPubMed Vos MD, Dallol A, Eckfeld K, Allen NP, Donninger H, Hesson LB, Calvisi D, Latif F, Clark GJ: The RASSF1A tumor suppressor activates Bax via MOAP-1. J Biol Chem. 2006, 281: 4557-4563. 10.1074/jbc.M512128200.CrossRefPubMed
7.
go back to reference Baksh S, Tommasi S, Fenton S, Yu VC, Martins LM, Pfeifer GP, Latif F, Downward J, Neel BG: The tumor suppressor RASSF1A and MAP-1 link death receptor signaling to Bax conformational change and cell death. Mol Cell. 2005, 18: 637-650. 10.1016/j.molcel.2005.05.010.CrossRefPubMed Baksh S, Tommasi S, Fenton S, Yu VC, Martins LM, Pfeifer GP, Latif F, Downward J, Neel BG: The tumor suppressor RASSF1A and MAP-1 link death receptor signaling to Bax conformational change and cell death. Mol Cell. 2005, 18: 637-650. 10.1016/j.molcel.2005.05.010.CrossRefPubMed
8.
go back to reference Matallanas D, Romano D, Yee K, Meissl K, Kucerova L, Piazzolla D, Baccarini M, Vass JK, Kolch W, O'Neill E: RASSF1A elicits apoptosis through an MST2 pathway directing proapoptotic transcription by the p73 tumor suppressor protein. Mol Cell. 2007, 27: 962-975. 10.1016/j.molcel.2007.08.008.PubMedCentralCrossRefPubMed Matallanas D, Romano D, Yee K, Meissl K, Kucerova L, Piazzolla D, Baccarini M, Vass JK, Kolch W, O'Neill E: RASSF1A elicits apoptosis through an MST2 pathway directing proapoptotic transcription by the p73 tumor suppressor protein. Mol Cell. 2007, 27: 962-975. 10.1016/j.molcel.2007.08.008.PubMedCentralCrossRefPubMed
9.
go back to reference O'Neill E, Rushworth L, Baccarini M, Kolch W: Role of the kinase MST2 in suppression of apoptosis by the proto-oncogene product Raf-1. Science. 2004, 306: 2267-2270. 10.1126/science.1103233.CrossRefPubMed O'Neill E, Rushworth L, Baccarini M, Kolch W: Role of the kinase MST2 in suppression of apoptosis by the proto-oncogene product Raf-1. Science. 2004, 306: 2267-2270. 10.1126/science.1103233.CrossRefPubMed
10.
go back to reference Sherwood V, Manbodh R, Sheppard C, Chalmers AD: RASSF7 is a member of a new family of RAS association domain-containing proteins and is required for completing mitosis. Mol Biol Cell. 2008, 19: 1772-1782. 10.1091/mbc.E07-07-0652.PubMedCentralCrossRefPubMed Sherwood V, Manbodh R, Sheppard C, Chalmers AD: RASSF7 is a member of a new family of RAS association domain-containing proteins and is required for completing mitosis. Mol Biol Cell. 2008, 19: 1772-1782. 10.1091/mbc.E07-07-0652.PubMedCentralCrossRefPubMed
11.
go back to reference Allen NP, Donninger H, Vos MD, Eckfeld K, Hesson L, Gordon L, Birrer MJ, Latif F, Clark GJ: RASSF6 is a novel member of the RASSF family of tumor suppressors. Oncogene. 2007, 26: 6203-6211. 10.1038/sj.onc.1210440.CrossRefPubMed Allen NP, Donninger H, Vos MD, Eckfeld K, Hesson L, Gordon L, Birrer MJ, Latif F, Clark GJ: RASSF6 is a novel member of the RASSF family of tumor suppressors. Oncogene. 2007, 26: 6203-6211. 10.1038/sj.onc.1210440.CrossRefPubMed
12.
go back to reference Hesson LB, Wilson R, Morton D, Adams C, Walker M, Maher ER, Latif F: CpG island promoter hypermethylation of a novel Ras-effector gene RASSF2A is an early event in colon carcinogenesis and correlates inversely with K-ras mutations. Oncogene. 2005, 24: 3987-3994. 10.1038/sj.onc.1208566.CrossRefPubMed Hesson LB, Wilson R, Morton D, Adams C, Walker M, Maher ER, Latif F: CpG island promoter hypermethylation of a novel Ras-effector gene RASSF2A is an early event in colon carcinogenesis and correlates inversely with K-ras mutations. Oncogene. 2005, 24: 3987-3994. 10.1038/sj.onc.1208566.CrossRefPubMed
13.
go back to reference Kozak M: Point mutations define a sequence flanking the AUG initiator codon that modulates translation by eukaryotic ribosomes. Cell. 1986, 44: 283-292. 10.1016/0092-8674(86)90762-2.CrossRefPubMed Kozak M: Point mutations define a sequence flanking the AUG initiator codon that modulates translation by eukaryotic ribosomes. Cell. 1986, 44: 283-292. 10.1016/0092-8674(86)90762-2.CrossRefPubMed
14.
go back to reference Kozak M: Interpreting cDNA sequences: some insights from studies on translation. Mamm Genome. 1996, 7: 563-574. 10.1007/s003359900171.CrossRefPubMed Kozak M: Interpreting cDNA sequences: some insights from studies on translation. Mamm Genome. 1996, 7: 563-574. 10.1007/s003359900171.CrossRefPubMed
15.
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: 3497-3508. 10.1158/0008-5472.CAN-04-4088.CrossRefPubMed Agathanggelou A, Cooper WN, Latif F: Role of the Ras-association domain family 1 tumor suppressor gene in human cancers. Cancer Res. 2005, 65: 3497-3508. 10.1158/0008-5472.CAN-04-4088.CrossRefPubMed
16.
go back to reference Ikeda M, Hirabayashi S, Fujiwara N, Mori H, Kawata A, Iida J, Bao Y, Sato Y, Iida T, Sugimura H, Hata Y: Ras-association domain family protein 6 induces apoptosis via both caspase-dependent and caspase-independent pathways. Exp Cell Res. 2007, 313: 1484-1495. 10.1016/j.yexcr.2007.02.013.CrossRefPubMed Ikeda M, Hirabayashi S, Fujiwara N, Mori H, Kawata A, Iida J, Bao Y, Sato Y, Iida T, Sugimura H, Hata Y: Ras-association domain family protein 6 induces apoptosis via both caspase-dependent and caspase-independent pathways. Exp Cell Res. 2007, 313: 1484-1495. 10.1016/j.yexcr.2007.02.013.CrossRefPubMed
17.
go back to reference Wiemels JL, Zhang Y, Chang J, Zheng S, Metayer C, Zhang L, Smith MT, Ma X, Selvin S, Buffler PA, Wiencke JK: RAS mutation is associated with hyperdiploidy and parental characteristics in pediatric acute lymphoblastic leukemia. Leukemia. 2005, 19: 415-419. 10.1038/sj.leu.2403641.CrossRefPubMed Wiemels JL, Zhang Y, Chang J, Zheng S, Metayer C, Zhang L, Smith MT, Ma X, Selvin S, Buffler PA, Wiencke JK: RAS mutation is associated with hyperdiploidy and parental characteristics in pediatric acute lymphoblastic leukemia. Leukemia. 2005, 19: 415-419. 10.1038/sj.leu.2403641.CrossRefPubMed
18.
go back to reference Paulsson K, Horvat A, Strombeck B, Nilsson F, Heldrup J, Behrendtz M, Forestier E, Andersson A, Fioretos T, Johansson B: Mutations of FLT3, NRAS, KRAS, and PTPN11 are frequent and possibly mutually exclusive in high hyperdiploid childhood acute lymphoblastic leukemia. Genes Chromosomes Cancer. 2008, 47: 26-33. 10.1002/gcc.20502.CrossRefPubMed Paulsson K, Horvat A, Strombeck B, Nilsson F, Heldrup J, Behrendtz M, Forestier E, Andersson A, Fioretos T, Johansson B: Mutations of FLT3, NRAS, KRAS, and PTPN11 are frequent and possibly mutually exclusive in high hyperdiploid childhood acute lymphoblastic leukemia. Genes Chromosomes Cancer. 2008, 47: 26-33. 10.1002/gcc.20502.CrossRefPubMed
19.
go back to reference Liang DC, Shih LY, Fu JF, Li HY, Wang HI, Hung IJ, Yang CP, Jaing TH, Chen SH, Liu HC: K-Ras mutations and N-Ras mutations in childhood acute leukemias with or without mixed-lineage leukemia gene rearrangements. Cancer. 2006, 106: 950-956. 10.1002/cncr.21687.CrossRefPubMed Liang DC, Shih LY, Fu JF, Li HY, Wang HI, Hung IJ, Yang CP, Jaing TH, Chen SH, Liu HC: K-Ras mutations and N-Ras mutations in childhood acute leukemias with or without mixed-lineage leukemia gene rearrangements. Cancer. 2006, 106: 950-956. 10.1002/cncr.21687.CrossRefPubMed
20.
go back to reference Richter AM, Pfeifer GP, Dammann RH: The RASSF proteins in cancer; from epigenetic silencing to functional characterization. Biochim Biophys Acta. 2009, Richter AM, Pfeifer GP, Dammann RH: The RASSF proteins in cancer; from epigenetic silencing to functional characterization. Biochim Biophys Acta. 2009,
21.
go back to reference Reeves N, Posakony JW: Genetic programs activated by proneural proteins in the developing Drosophila PNS. Dev Cell. 2005, 8: 413-425. 10.1016/j.devcel.2005.01.020.CrossRefPubMed Reeves N, Posakony JW: Genetic programs activated by proneural proteins in the developing Drosophila PNS. Dev Cell. 2005, 8: 413-425. 10.1016/j.devcel.2005.01.020.CrossRefPubMed
22.
go back to reference Nybakken K, Vokes SA, Lin TY, McMahon AP, Perrimon N: A genome-wide RNA interference screen in Drosophila melanogaster cells for new components of the Hh signaling pathway. Nat Genet. 2005, 37: 1323-1332. 10.1038/ng1682.CrossRefPubMed Nybakken K, Vokes SA, Lin TY, McMahon AP, Perrimon N: A genome-wide RNA interference screen in Drosophila melanogaster cells for new components of the Hh signaling pathway. Nat Genet. 2005, 37: 1323-1332. 10.1038/ng1682.CrossRefPubMed
23.
go back to reference Hesson L, Bieche I, Krex D, Criniere E, Hoang-Xuan K, Maher ER, Latif F: Frequent epigenetic inactivation of RASSF1A and BLU genes located within the critical 3p21.3 region in gliomas. Oncogene. 2004, 23: 2408-2419. 10.1038/sj.onc.1207407.CrossRefPubMed Hesson L, Bieche I, Krex D, Criniere E, Hoang-Xuan K, Maher ER, Latif F: Frequent epigenetic inactivation of RASSF1A and BLU genes located within the critical 3p21.3 region in gliomas. Oncogene. 2004, 23: 2408-2419. 10.1038/sj.onc.1207407.CrossRefPubMed
24.
go back to reference Eckfeld K, Hesson L, Vos MD, Bieche I, Latif F, Clark GJ: RASSF4/AD037 is a potential ras effector/tumor suppressor of the RASSF family. Cancer Res. 2004, 64: 8688-8693. 10.1158/0008-5472.CAN-04-2065.CrossRefPubMed Eckfeld K, Hesson L, Vos MD, Bieche I, Latif F, Clark GJ: RASSF4/AD037 is a potential ras effector/tumor suppressor of the RASSF family. Cancer Res. 2004, 64: 8688-8693. 10.1158/0008-5472.CAN-04-2065.CrossRefPubMed
Metadata
Title
The novel RASSF6 and RASSF10 candidate tumour suppressor genes are frequently epigenetically inactivated in childhood leukaemias
Authors
Luke B Hesson
Thomas L Dunwell
Wendy N Cooper
Daniel Catchpoole
Anna T Brini
Raffaella Chiaramonte
Mike Griffiths
Andrew D Chalmers
Eamonn R Maher
Farida Latif
Publication date
01-12-2009
Publisher
BioMed Central
Published in
Molecular Cancer / Issue 1/2009
Electronic ISSN: 1476-4598
DOI
https://doi.org/10.1186/1476-4598-8-42

Other articles of this Issue 1/2009

Molecular Cancer 1/2009 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