Skip to main content

Epigenetic Disruption of the SLIT-ROBO Interactions in Human Cancer

  • Chapter
DNA Methylation, Epigenetics and Metastasis

Part of the book series: Cancer Metastasis — Biology and Treatment ((CMBT,volume 7))

Abstract

During the development of the nervous system, guidance cues are required to correctly direct the developing axons. These cues are highly conserved in evolution and may have diverse functions and receptors. The Slit proteins are members of these cues and along with their roundabout (robo) receptors, they act as repulsive cues for robo-expressing axons preventing them from crossing or re-crossing the midline. There is increasing evidence that the slit-robo interactions are not limited to axon guidance. The repulsive effect on axons due to slit-robo binding is mirrored in the immune system as well as in breast tumour cells; Slit proteins act as inhibitors of cell migration and invasion. Our group recently demonstrated that both SLIT and ROBO genes are inactivated in human cancers by promoter region CpG island hypermethylation with the subsequent silencing of gene expression. Restoring expression after treatment with a demethylating agent, provided further evidence that promoter hypermethylation was responsible for silencing SLIT-ROBO genes in several human cancers. Whilst Robo1 homozygous mutant mice die at birth due to incomplete lung development, the heterozygous mice show increased predisposition to tumour development concurrent with the inactivation of the remaining wild type Robo1 allele by promoter region CpG island hypermethylation. SEMA3B, another axon guidance molecule, was recently demonstrated to be epigenetically inactivated in human cancers and suppressed tumour growth. Hence, evidence is accumulating for the role of axon guidance molecules in human cancer development. Unlike mutational inactivation, epigenetic inactivation is a reversible event. This presents new and exciting opportunities for clinical management of cancer. In addition, promoter hypermethylation of genes is increasingly being developed as molecular biomarkers for non-invasive screens for early detection of cancer. Furthermore, the SLIT(s) gene products are secretary proteins, which may also lead to the development of novel therapeutic approaches. This chapter summarises the literature on SLIT-ROBO gene families in relation to human diseases.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 169.00
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 219.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 219.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Preview

Unable to display preview. Download preview PDF.

Unable to display preview. Download preview PDF.

References

  1. Seeger M., Tear G., Ferres-Marco D. and Goodman C. S. Mutations affecting growth cone guidance in Drosophila: genes necessary for guidance toward or away from the midline. Neuron, 1993; 10: 409–426.

    Article  PubMed  Google Scholar 

  2. Kidd T., Brose K., Mitchell K. J., Fetter R. D., Tessier-Lavigne M., Goodman C. S. and Tear, G. Roundabout controls axon crossing of the CNS midline and defines a novel subfamily of evolutionarily conserved guidance receptors. Cell, 1998; 92: 205–215.

    Article  PubMed  Google Scholar 

  3. Sundaresan V., Roberts I., Bateman A., Bankier A., Sheppard M., Hobbs C., Xiong J., Minna J., Latif F., Lerman M. and Rabbitts P. The DUTT1 gene, a novel NCAM family member is expressed in developing murine neural tissues and has an unusually broad pattern of expression. Mol. Cell Neurosci., 1998; 11: 29–35.

    Article  PubMed  Google Scholar 

  4. Bashaw G. J., Kidd T., Murray D., Pawson T. and Goodman, C. S. Repulsive axon guidance: Abelson and Enabled play opposing roles downstream of the roundabout receptor. Cell, 2000; 101: 703–715.

    Article  PubMed  Google Scholar 

  5. Stein E. and Tessier-Lavigne M. Hierarchical organization of guidance receptors: silencing of netrin attraction by slit through a Robo/DCC receptor complex. Science, 2001; 291: 1928–1938.

    Article  PubMed  Google Scholar 

  6. Wong K., Ren X. R., Huang Y. Z., Xie Y., Liu G., Saito H., Tang H., Wen L., Brady-Kalnay S. M., Mei L., Wu J. Y., Xiong W. C. and Rao Y. Signal transduction in neuronal migration: roles of GTPase activating proteins and the small GTPase Cdc42 in the Slit-Robo pathway. Cell, 2001, 107: 209–221.

    Article  PubMed  Google Scholar 

  7. Emerson M. M. and Van Vactor D. Robo is Abl to block N-Cadherin function. Nat. Cell Biol., 2002; 4: E227–E230.

    Article  PubMed  Google Scholar 

  8. Rhee J., Mahfooz N. S., Arregui C., Lilien J., Balsamo J. and VanBerkum M. F. Activation of the repulsive receptor Roundabout inhibits N-cadherin-mediated cell adhesion. Nat. Cell Biol., 2002; 4: 798–805.

    Article  PubMed  Google Scholar 

  9. Clark K., Hammond E. and Rabbitts P. Temporal and spatial expression of two isoforms of the Dutt1/Robo1 gene in mouse development. FEBS Lett., 2002, 523: 12–16.

    Article  PubMed  Google Scholar 

  10. Nagase T., Kikuno R., Nakayama M., Hirosawa M. and Ohara O. Prediction of the coding sequences of unidentified human genes. XVIII. The complete sequences of 100 new cDNA clones from brain which code for large proteins in vitro. DNA Res., 2000; 7: 273–281.

    PubMed  Google Scholar 

  11. Grieshammer U., Le M., Plump A. S., Wang F., Tessier-Lavigne M. and Martin G. R. SLIT2-mediated ROBO2 signaling restricts kidney induction to a single site. Dev. Cell, 2004; 6: 709–717.

    Article  PubMed  Google Scholar 

  12. Yuan S. S., Cox L. A., Dasika G. K. and Lee E. Y. Cloning and functional studies of a novel gene aberrantly expressed in RB-deficient embryos. Dev. Biol., 1999, 207: 62–75.

    Article  PubMed  Google Scholar 

  13. Yuan S. S., Yeh Y. T. and Lee E. Y. Pax-2 interacts with RB and reverses its repression on the promoter of Rig-1, a Robo member. Biochem. Biophys. Res. Commun., 2002, 296: 1019–1025.

    Article  PubMed  Google Scholar 

  14. Sabatier C., Plump A. S., Le M., Brose K., Tamada A., Murakami F., Lee E. Y. and Tessier-Lavigne M. The divergent Robo family protein rig-1/Robo3 is a negative regulator of slit responsiveness required for midline crossing by commissural axons. Cell, 2004; 117: 157–169.

    Article  PubMed  Google Scholar 

  15. Huminiecki L., Gorn M., Suchting S., Poulsom R. and Bicknell R. Magic roundabout is a new member of the roundabout receptor family that is endothelial specific and expressed at sites of active angiogenesis. Genomics, 2002; 79: 547–552.

    Article  PubMed  Google Scholar 

  16. Park K. W., Morrison C. M., Sorensen L. K., Jones C. A., Rao Y., Chien C. B., Wu J. Y., Urness L. D. and Li D. Y. Robo4 is a vascular-specific receptor that inhibits endothelial migration. Dev. Biol., 2003; 261: 251–267.

    Article  PubMed  Google Scholar 

  17. Suchting S., Heal P., Tahtis K., Stewart L. M. and Bicknell, R. Soluble Robo4 receptor inhibits in vivo angiogenesis and endothelial cell migration. FASEB J., 2004.

    Google Scholar 

  18. Brose K., Bland K. S., Wang K. H., Arnott D., Henzel W., Goodman C. S., Tessier-Lavigne M. and Kidd T. Slit proteins bind Robo receptors and have an evolutionarily conserved role in repulsive axon guidance. Cell, 1999; 96: 795–806.

    Article  PubMed  Google Scholar 

  19. Itoh A., Miyabayashi T., Ohno M. and Sakano S. Cloning and expressions of three mammalian homologues of Drosophila slit suggest possible roles for Slit in the formation and maintenance of the nervous system. Brain Res. Mol. Brain Res., 1998, 62: 175–186.

    Article  PubMed  Google Scholar 

  20. Li H. S., Chen J. H., Wu W., Fagaly T., Zhou L., Yuan W., Dupuis S., Jiang Z. H., Nash W., Gick C., Ornitz D. M., Wu J. Y. and Rao Y. Vertebrate slit, a secreted ligand for the transmembrane protein roundabout, is a repellent for olfactory bulb axons. Cell, 1999; 96: 807–818.

    Article  PubMed  Google Scholar 

  21. Wu J. Y., Feng L., Park H. T., Havlioglu N., Wen L., Tang H., Bacon K. B., Jiang Z., Zhang X. and Rao Y. The neuronal repellent Slit inhibits leukocyte chemotaxis induced by chemotactic factors. Nature, 2001; 410: 948–952.

    Article  PubMed  Google Scholar 

  22. Wang B., Xiao Y., Ding B. B., Zhang N., Yuan X., Gui L., Qian K. X., Duan S., Chen Z., Rao Y. and Geng J. G. Induction of tumor angiogenesis by Slit-Robo signaling and inhibition of cancer growth by blocking Robo activity. Cancer Cell, 2003; 4: 19–29.

    Article  PubMed  Google Scholar 

  23. Plump A. S., Erskine L., Sabatier C., Brose K., Epstein C. J., Goodman C. S., Mason C. A. and Tessier-Lavigne M. Slit1 and Slit2 cooperate to prevent premature midline crossing of retinal axons in the mouse visual system. Neuron, 2002; 33: 219–232.

    Article  PubMed  Google Scholar 

  24. Liu J., Zhang L., Wang D., Shen H., Jiang M., Mei P., Hayden P. S., Sedor J. R. and Hu H. Congenital diaphragmatic hernia, kidney agenesis and cardiac defects associated with Slit3-deficiency in mice. Mech. Dev., 2003; 120: 1059–1070.

    Article  PubMed  Google Scholar 

  25. Yuan W., Rao Y., Babiuk R. P., Greer J. J., Wu J. Y. and Ornitz D. M. A genetic model for a central (septum transversum) congenital diaphragmatic hernia in mice lacking Slit3. Proc. Natl. Acad. Sci. U.S.A, 2003; 100: 5217–5222.

    Article  PubMed  Google Scholar 

  26. Liu Z., Patel K., Schmidt H., Andrews W., Pini A. and Sundaresan V. Extracellular Ig domains 1 and 2 of Robo are important for ligand (Slit) binding. Mol. Cell Neurosci., 2004; 26: 232–240.

    Article  PubMed  Google Scholar 

  27. Battye R., Stevens A., Perry R. L. and Jacobs J. R. Repellent signaling by Slit requires the leucine-rich repeats. J. Neurosci., 2001; 21: 4290–4298.

    PubMed  Google Scholar 

  28. Liang Y., Annan R. S., Carr S. A., Popp S., Mevissen M., Margolis R. K. and Margolis R. U. Mammalian homologues of the Drosophila slit protein are ligands of the heparan sulfate proteoglycan glypican-1 in brain. J. Biol. Chem., 1999; 274: 17885–17892.

    Article  PubMed  Google Scholar 

  29. Anselmo M. A., Dalvin S., Prodhan P., Komatsuzaki K., Aidlen J. T., Schnitzer J. J., Wu J. Y. and Kinane, T. B. Slit and robo: expression patterns in lung development. Gene Expr. Patterns., 2003; 3: 13–19.

    Article  PubMed  Google Scholar 

  30. Xian J., Clark K. J., Fordham R., Pannell R., Rabbitts T. H. and Rabbitts P. H. Inadequate lung development and bronchial hyperplasia in mice with a targeted deletion in the Dutt1/Robo1 gene. Proc. Natl. Acad. Sci. U.S.A, 2001; 98: 15062–15066.

    Article  PubMed  Google Scholar 

  31. Todd S., Franklin W. A., Varella-Garcia M., Kennedy T., Hilliker C. E., Jr., Hahner L., Anderson M., Wiest J. S., Drabkin H. A. and Gemmill R. M. Homozygous deletions of human chromosome 3p in lung tumors. Cancer Res., 1997; 57: 1344–1352.

    PubMed  Google Scholar 

  32. Kok K., Naylor S. L. and Buys C. H. Deletions of the short arm of chromosome 3 in solid tumors and the search for suppressor genes. Adv. Cancer Res., 1997; 71: 27–92.

    PubMed  Google Scholar 

  33. Drabkin H. A., Mendez M. J., Rabbitts P. H., Varkony T., Bergh J., Schlessinger J., Erickson P. and Gemmill R. M. Characterization of the submicroscopic deletion in the small-cell lung carcinoma (SCLC) cell line U2020. Genes Chromosomes. Cancer, 1992; 5: 67–74.

    Google Scholar 

  34. Latif F., Tory K., Modi W. S., Graziano S. L., Gamble G., Douglas J., Heppell-Parton, A. C., Rabbitts, P. H., Zbar, B., and Lerman, M. I. Molecular characterization of a large homozygous deletion in the small cell lung cancer cell line U2020: a strategy for cloning the putative tumor suppressor gene. Genes Chromosomes. Cancer, 1992; 5: 119–127.

    Google Scholar 

  35. Rabbitts P., Bergh J., Douglas J., Collins F. and Waters J. A submicroscopic homozygous deletion at the D3S3 locus in a cell line isolated from a small cell lung carcinoma. Genes Chromosomes. Cancer, 1990; 2: 231–238.

    Google Scholar 

  36. Todd S., Franklin W. A., Varella-Garcia M., Kennedy T., Hilliker C. E., Jr., Hahner L., Anderson M., Wiest J. S., Drabkin H. A. and Gemmill R. M. Homozygous deletions of human chromosome 3p in lung tumors. Cancer Res., 1997; 57: 1344–1352.

    PubMed  Google Scholar 

  37. Sundaresan V., Chung G., Heppell-Parton A., Xiong J., Grundy C., Roberts I., James L., Cahn A., Bench A., Douglas J., Minna J., Sekido Y., Lerman M., Latif F., Bergh J., Li H., Lowe N., Ogilvie D. and Rabbitts P. Homozygous deletions at 3p12 in breast and lung cancer. Oncogene, 1998; 17: 1723–1729.

    Article  PubMed  Google Scholar 

  38. Kang Y. S., Cheong H. M., Moon Y., Lee I. B., Kim S. M., Kim H. S., Jun S. Y., Jung S. K., Kim J. S., Choi J. H., Cho H. E., Son J. S., Min N. Y. and Lee K. H. Molecular genetic characterization of a Korean split hand/split foot malformation (SHFM). Mol. Cells, 2004; 17: 397–403.

    PubMed  Google Scholar 

  39. Meijer I. A., Cossette P., Roussel J., Benard M., Toupin S. and Rouleau G. A. A novel locus for pure recessive hereditary spastic paraplegia maps to 10q22.1–10q24.1. Ann. Neurol., 2004; 56: 579–582.

    Article  PubMed  Google Scholar 

  40. Lo N. C., Cusano R., Gigli G. L., Forabosco P., Valente M., Ravazzolo R., Diomedi M. and Seri M. Genetic heterogeneity in inherited spastic paraplegia associated with epilepsy. Am. J. Med. Genet., 2003; 117A: 116–121.

    Google Scholar 

  41. Shivapurkar N., Virmani A. K., Wistuba I. I., Milchgrub S., Mackay B., Minna J. D. and Gazdar A. F. Deletions of chromosome 4 at multiple sites are frequent in malignant mesothelioma and small cell lung carcinoma. Clin. Cancer Res., 1999; 5: 17–23.

    PubMed  Google Scholar 

  42. Shivapurkar N., Sood S., Wistuba I. I., Virmani A. K., Maitra A., Milchgrub S., Minna J. D. and Gazdar A. F. Multiple regions of chromosome 4 demonstrating allelic losses in breast carcinomas. Cancer Res., 1999; 59: 3576–3580.

    PubMed  Google Scholar 

  43. Shivapurkar N., Maitra A., Milchgrub S. and Gazdar A. F. Deletions of chromosome 4 occur early during the pathogenesis of colorectal carcinoma. Hum. Pathol., 2001; 32: 169–177.

    Article  PubMed  Google Scholar 

  44. Sherwood J. B., Shivapurkar N., Lin W. M., Ashfaq R., Miller D. S., Gazdar A. F. and Muller C. Y. Chromosome 4 deletions are frequent in invasive cervical cancer and differ between histologic variants. Gynecol. Oncol., 2000; 79: 90–96.

    Article  PubMed  Google Scholar 

  45. Pershouse M. A., El Naggar A. K., Hurr K., Lin H., Yung W. K. and Steck P. A. Deletion mapping of chromosome 4 in head and neck squamous cell carcinoma. Oncogene, 1997; 14: 369–373.

    PubMed  Google Scholar 

  46. Polascik T. J., Cairns P., Chang W. Y., Schoenberg M. P. and Sidransky, D. Distinct regions of allelic loss on chromosome 4 in human primary bladder carcinoma. Cancer Res., 1995; 55: 5396–5399.

    PubMed  Google Scholar 

  47. Faulkner S. W. and Friedlander M. L. Molecular genetic analysis of malignant ovarian germ cell tumors. Gynecol. Oncol., 2000; 77: 283–288.

    Article  PubMed  Google Scholar 

  48. von Knobloch R., Bugert P., Jauch A., Kalble T, and Kovacs G. Allelic changes at multiple regions of chromosome 5 are associated with progression of urinary bladder cancer. J. Pathol., 2000; 190: 163–168.

    Article  PubMed  Google Scholar 

  49. Shi Y., Zhao X., Yu L., Tao R., Tang J., La Y., Duan Y., Gao B., Gu N., Xu Y., Feng G., Zhu S., Liu H., Salter H. and He L. Genetic structure adds power to detect schizophrenia susceptibility at SLIT3 in the Chinese Han population. Genome Res., 2004; 14: 1345–1349.

    Article  PubMed  Google Scholar 

  50. Brose K. and Tessier-Lavigne M. Slit proteins: key regulators of axon guidance, axonal branching, and cell migration. Curr. Opin. Neurobiol., 2000; 10: 95–102.

    Article  PubMed  Google Scholar 

  51. Kanellis J., Garcia G. E., Li P., Parra G., Wilson C. B., Rao Y., Han, S., Smith C. W., Johnson R. J., Wu J. Y. and Feng L. Modulation of inflammation by slit protein in vivo in experimental crescentic glomerulonephritis. Am. J. Pathol., 2004; 165: 341–352.

    PubMed  Google Scholar 

  52. Xian J., Aitchison A., Bobrow L., Corbett G., Pannell R., Rabbitts T. and Rabbitts P. Targeted disruption of the 3p12 gene, Dutt1/Robo1, predisposes mice to lung adenocarcinomas and lymphomas with methylation of the gene promoter. Cancer Res., 2004; 64: 6432–6437.

    Article  PubMed  Google Scholar 

  53. Dallol A., Da Silva N. F., Viacava P., Minna J. D., Bieche I., Maher E. R. and Latif F. SLIT2, a human homologue of the Drosophila Slit2 gene, has tumor suppressor activity and is frequently inactivated in lung and breast cancers. Cancer Res., 2002; 62: 5874–5880.

    PubMed  Google Scholar 

  54. Dallol A., Morton D., Maher E. R. and Latif F. SLIT2 axon guidance molecule is frequently inactivated in colorectal cancer and suppresses growth of colorectal carcinoma cells. Cancer Res., 2003; 63: 1054–1058.

    PubMed  Google Scholar 

  55. Dallol A., Krex D., Hesson L., Eng C., Maher E. R. and Latif, F. Frequent epigenetic inactivation of the SLIT2 gene in gliomas. Oncogene, 2003; 22: 4611–4616.

    Article  PubMed  Google Scholar 

  56. Prasad A., Fernandis A. Z., Rao Y. and Ganju R. K. Slit protein-mediated inhibition of CXCR4-induced chemotactic and chemoinvasive signaling pathways in breast cancer cells. J. Biol. Chem., 2004; 279: 9115–9124.

    Article  PubMed  Google Scholar 

  57. Balkwill F. The significance of cancer cell expression of the chemokine receptor CXCR4. Semin.Cancer Biol., 2004; 14: 171–179.

    Article  PubMed  Google Scholar 

  58. Wang R. Y., Gehrke C. W. and Ehrlich M. Comparison of bisulfite modification of 5-methyldeoxycytidine and deoxycytidine residues. Nucleic Acids Res., 1980; 8: 4777–4790.

    PubMed  Google Scholar 

  59. Clark S. J., Harrison J., Paul C. L. and Frommer M. High sensitivity mapping of methylated cytosines. Nucleic Acids Res., 1994; 22: 2990–2997.

    PubMed  Google Scholar 

  60. Herman J. G. and Baylin S. B. Gene silencing in cancer in association with promoter hypermethylation. N. Engl. J. Med., 2003; 349: 2042–2054.

    Article  PubMed  Google Scholar 

  61. Esteller M. CpG island hypermethylation and tumor suppressor genes: a booming present, a brighter future. Oncogene, 2002; 21: 5427–5440.

    Article  PubMed  Google Scholar 

  62. Agathanggelou A., Honorio S., Macartney D. P., Martinez A., Dallol A., Rader J., Fullwood P., Chauhan A., Walker R., Shaw J. A., Hosoe S., Lerman M. I., Minna J. D., Maher E. R. and Latif F. Methylation associated inactivation of RASSF1A from region 3p21.3 in lung, breast and ovarian tumours. Oncogene, 2001; 20: 1509–1518.

    Article  PubMed  Google Scholar 

  63. Dammann R., Schagdarsurengin U., Strunnikova M., Rastetter M., Seidel C., Liu L., Tommasi S., and Pfeifer, G. P. Epigenetic inactivation of the Ras-association domain family 1 (RASSF1A) gene and its function in human carcinogenesis. Histol. Histopathol., 2003; 18: 665–677.

    PubMed  Google Scholar 

  64. Dammann R., Li C., Yoon J. H., Chin P. L., Bates S. and Pfeifer G. P. Epigenetic inactivation of a RAS association domain family protein from the lung tumour suppressor locus 3p21.3. Nat. Genet., 2000; 25: 315–319.

    Article  PubMed  Google Scholar 

  65. Ruter B., Wijermans P. W. and Lubbert M. DNA methylation as a therapeutic target in hematologic disorders: recent results in older patients with myelodysplasia and acute myeloid leukemia. Int. J. Hematol., 2004; 80: 128–135.

    Article  PubMed  Google Scholar 

  66. Villar-Garea A. and Esteller M. Histone deacetylase inhibitors: understanding a new wave of anticancer agents. Int. J. Cancer, 2004; 112: 171–178.

    Article  PubMed  Google Scholar 

  67. Dallol A., Forgacs E., Martinez A., Sekido Y., Walker R., Kishida T., Rabbitts P., Maher E. R., Minna J. D. and Latif F. Tumour specific promoter region methylation of the human homologue of the Drosophila Roundabout gene DUTT1 (ROBO1) in human cancers. Oncogene, 2002; 21: 3020–3028.

    Article  PubMed  Google Scholar 

  68. Chen W., Cooper T. K., Zahnow C. A., Overholtzer M., Zhao Z., Ladanyi M., Karp J. E., Gokgoz N., Wunder J. S., Andrulis I. L., Levine A. J., Mankowski J. L. and Baylin S. B. Epigenetic and genetic loss of Hic1 function accentuates the role of p53 in tumorigenesis. Cancer Cell, 2004; 6: 387–398.

    Article  PubMed  Google Scholar 

  69. Chen W. Y., Zeng X., Carter M. G., Morrell C. N., Chiu Yen R. W., Esteller M., Watkins D. N., Herman J. G., Mankowski J. L. and Baylin S. B. Heterozygous disruption of Hic1 predisposes mice to a gender-dependent spectrum of malignant tumors. Nat. Genet., 2003; 33: 197–202.

    Article  PubMed  Google Scholar 

  70. Knudson A. G., Jr., Hethcote H. W. and Brown B. W. Mutation and childhood cancer: a probabilistic model for the incidence of retinoblastoma. Proc. Natl. Acad. Sci. U.S.A, 1975; 72: 5116–5120.

    PubMed  Google Scholar 

  71. Astuti D., Da Silva N. F., Dallol A., Gentle D., Martinsson T., Kogner P., Grundy R., Kishida T., Yao M., Latif F. and Maher E. R. SLIT2 promoter methylation analysis in neuroblastoma, Wilms’ tumour and renal cell carcinoma. Br.J.Cancer, 2004; 90: 515–521.

    Article  PubMed  Google Scholar 

  72. Dickinson R. E., Dallol A, Bieche I., Krex D., Morton D., Maher E. R. and Latif F. Epigenetic inactivation of SLIT3 and SLIT1 genes in human cancers. Br. J. Cancer 2004; (In Press).

    Google Scholar 

  73. Yu T. W. and Bargmann C. I. Dynamic regulation of axon guidance. Nat. Neurosci., 2001; 4Suppl: 1169–1176.

    Article  Google Scholar 

  74. Lerman M. I. and Minna J. D. The 630-kb lung cancer homozygous deletion region on human chromosome 3p21.3: identification and evaluation of the resident candidate tumor suppressor genes. The International Lung Cancer Chromosome 3p21.3 Tumor Suppressor Gene Consortium. Cancer Res., 60: 6116–6133, 2000.

    PubMed  Google Scholar 

  75. Kuroki T., Trapasso F., Yendamuri S., Matsuyama A., Alder H., Williams N. N., Kaiser L. R. and Croce C. M. Allelic loss on chromosome 3p21.3 and promoter hypermethylation of semaphorin 3B in non-small cell lung cancer. Cancer Res., 2003; 63: 3352–3355.

    PubMed  Google Scholar 

  76. de Lange R., Dimoudis N. and Weidle U. H. Identification of genes associated with enhanced metastasis of a large cell lung carcinoma cell line. Anticancer Res., 2003; 23: 187–194.

    PubMed  Google Scholar 

  77. Tomizawa Y., Sekido Y., Kondo M., Gao B., Yokota J., Roche J., Drabkin H., Lerman M. I., Gazdar A. F. and Minna J. D. Inhibition of lung cancer cell growth and induction of apoptosis after reexpression of 3p21.3 candidate tumor suppressor gene SEMA3B. Proc. Natl. Acad. Sci. U.S.A, 2001; 98: 13954–13959.

    Article  PubMed  Google Scholar 

  78. Tse C., Xiang R. H., Bracht T. and Naylor S. L. Human Semaphorin 3B (SEMA3B) located at chromosome 3p21.3 suppresses tumor formation in an adenocarcinoma cell line. Cancer Res., 2002; 62: 542–546.

    PubMed  Google Scholar 

  79. Castro-Rivera E., Ran S., Thorpe P. and Minna J. D. Semaphorin 3B (SEMA3B) induces apoptosis in lung and breast cancer, whereas VEGF165 antagonizes this effect. Proc. Natl. Acad. Sci. U.S.A, 2004; 101: 11432–11437.

    Article  PubMed  Google Scholar 

  80. Xiang R. H., Hensel C. H., Garcia D. K., Carlson H. C., Kok K., Daly M. C., Kerbacher K., van den B. A., Veldhuis P., Buy, C. H. and Naylor S. L. Isolation of the human semaphorin III/F gene (SEMA3F) at chromosome 3p21, a region deleted in lung cancer. Genomics, 1996; 32: 39–48.

    Article  PubMed  Google Scholar 

  81. Brambilla E., Constantin B., Drabkin H. and Roche J. Semaphorin SEMA3F localization in malignant human lung and cell lines: A suggested role in cell adhesion and cell migration. Am. J. Pathol., 2000; 156: 939–950.

    PubMed  Google Scholar 

  82. Nasarre P., Constantin B., Rouhaud L., Harnois T., Raymond G., Drabkin H. A., Bourmeyster N. and Roche J. Semaphorin SEMA3F and VEGF have opposing effects on cell attachment and spreading. Neoplasia., 2003, 5: 83–92.

    PubMed  Google Scholar 

  83. Xiang R., Davalos A. R., Hensel C. H., Zhou X. J., Tse C. and Naylor S. L. Semaphorin 3F gene from human 3p21.3 suppresses tumor formation in nude mice. Cancer Res., 2002; 62: 2637–2643,.

    PubMed  Google Scholar 

  84. Catalano A., Caprari P., Rodilossi S., Betta P., Castellucci M., Casazza A., Tamagnone L. and Procopio A. Cross-talk between vascular endothelial growth factor and semaphorin-3A pathway in the regulation of normal and malignant mesothelial cell proliferation. FASEB J., 2004; 18: 358–360.

    PubMed  Google Scholar 

  85. Fearon E. R., Cho K. R., Nigro J. M., Kern S. E., Simons J. W., Ruppert J. M., Hamilton S. R., Preisinger A. C., Thomas G. and Kinzler K. W. Identification of a chromosome 18q gene that is altered in colorectal cancers. Science, 1990; 247: 49–56.

    PubMed  Google Scholar 

  86. Sato K., Tamura G., Tsuchiya T., Endoh Y., Usuba O., Kimura W. and Motoyama, T. Frequent loss of expression without sequence mutations of the DCC gene in primary gastric cancer. Br. J. Cancer, 2001; 85: 199–203.

    Article  PubMed  Google Scholar 

  87. Fazeli A., Dickinson S. L., Hermiston M. L., Tighe R. V., Steen R. G., Small C. G., Stoeckli E. T., Keino-Masu K., Masu M., Rayburn H., Simons J., Bronson R. T., Gordon J. I., Tessier-Lavigne M. and Weinberg R. A. Phenotype of mice lacking functional Deleted in colorectal cancer (Dcc) gene. Nature, 1997; 386: 796–804.

    Article  PubMed  Google Scholar 

  88. Llambi F., Causeret F., Bloch-Gallego E. and Mehlen P. Netrin-1 acts as a survival factor via its receptors UNC5H and DCC. EMBO J., 2001; 20: 2715–2722.

    Article  PubMed  Google Scholar 

  89. Mazelin L., Bernet A., Bonod-Bidaud C., Pays L., Arnaud S., Gespach C., Bredesen D. E., Scoazec J. Y. and Mehlen P. Netrin-1 controls colorectal tumorigenesis by regulating apoptosis. Nature, 2004; 431: 80–84.

    Article  PubMed  Google Scholar 

  90. Fearon E. R. and Vogelstein B. A genetic model for colorectal tumorigenesis. Cell, 1990; 61: 759–767.

    Article  PubMed  Google Scholar 

  91. Bilder D. Epithelial polarity and proliferation control: links from the Drosophila neoplastic tumor suppressors. Genes Dev., 2004; 18: 1909–1925.

    Article  PubMed  Google Scholar 

  92. St John M. A. and Xu T. Understanding human cancer in a fly? Am. J. Hum. Genet., 1997; 61: 1006–1010.

    Article  PubMed  Google Scholar 

  93. Giot L., Bader J. S., Brouwer C., Chaudhuri A., Kuang B., Li, Y., Hao Y. L., Ooi C. E., Godwin B., Vitols E., Vijayadamodar G., Pochart P., Machineni H., Welsh M., Kong Y., Zerhusen B., Malcolm R., Varrone Z., Collis A., Minto M., Burgess S., McDaniel L., Stimpson E., Spriggs F., Williams J., Neurath K., Ioime N., Agee M., Voss E., Furtak K., Renzulli R., Aanensen N., Carrolla S., Bickelhaupt E., Lazovatsky Y., DaSilva A., Zhong J., Stanyon C. A., Finley R. L., Jr., White K. P., Braverman M., Jarvie T., Gold S., Leach M., Knight J., Shimkets R. A., McKenna M. P., Chant J. and Rothberg J. M. A protein interaction map of Drosophila melanogaster. Science, 2003; 302: 1727–1736.

    Article  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2005 Springer

About this chapter

Cite this chapter

Dallol, A., Dickinson, R.E., Latif, F. (2005). Epigenetic Disruption of the SLIT-ROBO Interactions in Human Cancer. In: Esteller, M. (eds) DNA Methylation, Epigenetics and Metastasis. Cancer Metastasis — Biology and Treatment, vol 7. Springer, Dordrecht. https://doi.org/10.1007/1-4020-3642-6_8

Download citation

Publish with us

Policies and ethics