DOI QR코드

DOI QR Code

Lack of Prognostic Significance of SOCS-1 Expression in Colorectal Adenocarcinomas

  • Ayyildiz, Talat (Department of Gastroenterology, School of Medicine, Ondokuz Mayis University) ;
  • Dolar, Enver (Department of Gastroenterology, School of Medicine, Uludag University) ;
  • Adim, Saduman Balaban (Department of Pathology, School of Medicine, Uludag University) ;
  • Eminler, Ahmet Tarik (Department of Gastroenterology, School of Medicine, Uludag University) ;
  • Yerci, Omer (Department of Pathology, School of Medicine, Uludag University)
  • Published : 2014.10.23

Abstract

Introduction: Recent studies have indicated that down-regulation of the suppressor of cytokine signaling-1 (SOCS-1) gene results in tumor formation and that SOCS-1 acts as a tumor suppressor gene. SOCS-1 has been also suggested to function as a tumor suppressor with colorectal cancer. Objectives: In the present study, we aimed to determine the association of SOCS-1 expression in colorectal cancer tissues with clinicopathologic characteristics immunohistochemically and also to identify its prognostic significance. Materials and Methods: SOCS-1 expression was studied immunohistochemically in 67 patients diagnosed with resected colorectal carcinomas and 30 control subjects. Results: SOCS-1 expression was found in 46.3% of tumor tissues and 46.7% of the control group. Statistical analyses did not establish any significant association between SOCS-1 expression and clinicopathologic characteristics. Also, no significant association with SOCS-1 expression was found using progression-free survival and overall survival analyses (p=0.326 and p=0.360, respectively). Conclusions: Our results show that SOCS-1 has no prognostic significance in colorectal cancer.

Keywords

References

  1. Abdel-Rahman WM, Nieminen TT, Shoman S, Eissa S, Peltomaki P (2014). Loss of p15INK4b expression in colorectal cancer is linked to ethnic origin. Asian Pac J Cancer Prev, 15, 2083-7. https://doi.org/10.7314/APJCP.2014.15.5.2083
  2. Ahuja N, Issa JP (2000). Aging, methylation and cancer. Histol Histopathol, 15, 835-42.
  3. Andre T, Boni C, Navarro M, et al (2009). Improved overall survival with oxaliplatin, fluorouracil, and leucovorin as adjuvant treatment in stage II or III colon cancer in the MOSAIC trial. J Clin Oncol, 27, 3109-16. https://doi.org/10.1200/JCO.2008.20.6771
  4. Boursi B, Arber N (2007). Current and future clinical strategies in colon cancer prevention and the emerging role of chemoprevention. Curr Pharm Des, 13, 2274-82. https://doi.org/10.2174/138161207781368783
  5. Chai SK, Nichols GL, Rothman P (1997). Constitutive activation of JAKs and STATs in BCR-Abl-expressing cell lines and peripheral blood cells derived from leukemic patients. J Immunol, 159, 4720-28.
  6. Chan AT, Baba Y, Shima K, et al (2010). Cathepsin B expression and survival in colon cancer: Implications for molecular detection of neoplasia. Cancer Epidemiol Biomark Prev, 19, 2777-85. https://doi.org/10.1158/1055-9965.EPI-10-0529
  7. Colussi D, Brandi G, Bazzoli F, Ricciardiello L (2013). Molecular pathways involved in colorectal cancer: implications for disease behavior and prevention. Int J Mol Sci, 14, 16365-85. https://doi.org/10.3390/ijms140816365
  8. Denlinger CS, Cohen SJ (2007). Progress in the development of prognostic and predictive markers for gastrointestinal malignancies. Curr Treat Options Oncol, 8, 339-51. https://doi.org/10.1007/s11864-007-0045-x
  9. Dong H, Tang J, Li LH, et al (2013). Serum carbohydrate antigen 19-9 as an indicator of liver metastasis in colorectal carcinoma cases. Asian Pac J Cancer Prev, 14, 909-13. https://doi.org/10.7314/APJCP.2013.14.2.909
  10. Fang WJ, Zheng Y, Wu LM, et al (2012). Genome-wide analysis of aberrant DNA methylation for identification of potential biomarkers in colorectal cancer patients. Asian Pac J Cancer Prev, 13, 1917-21. https://doi.org/10.7314/APJCP.2012.13.5.1917
  11. Farabegoli F, Ceccarelli C, Santini D, Taffurelli M (2005). Suppressor of cytokine signalling 2(SOCS-2) expression in breast carcinoma. J Clin Pathol, 58, 1046-50. https://doi.org/10.1136/jcp.2004.024919
  12. Firestein R, Shima K, Nosho K, et al (2010). CDK8 expression in 470 colorectal cancers in relation to beta-catenin activation, other molecular alterations and patient survival. Int J Cancer, 126, 2863-73.
  13. Fujitake S, Hibi K, Okochi O, et al (2004). Aberrant methylation of SOCS-1 was observed in younger colorectal cancer patients. J Gastroenterol, 39, 120-4. https://doi.org/10.1007/s00535-003-1262-0
  14. Fukushima N, Sato N, Sahin F, et al (2003). Aberrant methylation of suppressor of cytokine signalling-1 (SOCS-1) gene in pancreatic ductal neoplasms. Br J Cancer, 89, 338-43. https://doi.org/10.1038/sj.bjc.6601039
  15. Grady WM, Carethers JM (2008). Genomic and epigenetic instability in colorectal cancer pathogenesis. Gastroenterol, 135, 1079-99. https://doi.org/10.1053/j.gastro.2008.07.076
  16. Hanada T, Kobayashi T, Chinen T, et al (2006). IFN gamma dependent, spontaneous development of colorectal carcinomas in SOCS-1-deficient mice. J Exp Med, 203, 1391-7. https://doi.org/10.1084/jem.20060436
  17. He B, You L, Xu Z, et al (2003). SOCS-3 is frequently silenced by hypermethylation and suppresses cell growth inhuman lung cancer. Proc Natl Acad Sci USA, 100, 14133-8. https://doi.org/10.1073/pnas.2232790100
  18. Imamura Y, Morikawa T, Liao X, et al (2012). Specific mutations in KRAS codons 12 and 13, and patient prognosis in 1075 BRAF wild-type colorectal cancers. Clin Cancer Res, 18, 4753-63. https://doi.org/10.1158/1078-0432.CCR-11-3210
  19. Issa JP (2008). Colon cancer: it's CIN or CIMP. Clin Cancer Res, 14, 5939-40. https://doi.org/10.1158/1078-0432.CCR-08-1596
  20. Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D (2011). Global cancer statistics. CA Cancer J Clin, 61, 69-90. https://doi.org/10.3322/caac.20107
  21. Kawazoe Y, Naka T, Fujimoto M, et al (2001). Signal transducer and activator of transcription (STAT)-induced STAT inhibitor 1 (SSI-1)/suppressor of cytokine signaling 1 (SOCS-1) inhibits insulin signal transduction pathway through modulating insulin receptor substrate 1 (IRS-1) phosphorylation. J Exp Med, 193, 263-9. https://doi.org/10.1084/jem.193.2.263
  22. Kishimoto T, Kikutani H (2001). Knocking the SOCS off a tumor suppressor. Nature Genetics, 28, 4-5 .
  23. Komazaki T, Nagai H, Emi M, et al (2004). Hypermethylationassociated inactivation of the SOCS-1gene, a JAK/STAT inhibitor, in human pancreatic cancers. Jpn J Clin Oncol, 34, 191-4. https://doi.org/10.1093/jjco/hyh035
  24. Lee DW, Han SW, Lee HJ, et al (2013). Prognostic implication of mucinous histology in colorectal cancer patients treated with adjuvant FOLFOX chemotherapy. Br J Cancer, 108, 1978-84. https://doi.org/10.1038/bjc.2013.232
  25. Li HY, Zhang Y, Cai JH, Bian HL (2013). MicroRNA-451 inhibits growth of human colorectal carcinoma cells via downregulation of Pi3k/Akt pathway. Asian Pac J Cancer Prev, 14, 3631-4. https://doi.org/10.7314/APJCP.2013.14.6.3631
  26. Liao X, Lochhead P, Nishihara R, et al (2012). Aspirin use, tumor PIK3CA mutation, and colorectal-cancer survival. N Engl J Med, 367, 1596-06. https://doi.org/10.1056/NEJMoa1207756
  27. Lin SY, Yeh KT, Chen WT, et al (2004). Promoter CpG methylation of tumor suppressor genes in colorectal cancer and its relationship to clinical features. Oncol Rep, 11, 341-8.
  28. Melzner I, Bucur AJ, Bruderlein S, et al (2005). Biallelic mutation of SOCS-1 impairs JAK2 degradation and sustains phospho-JAK2 action in the MedB-1 mediastinal lymphoma line. Blood, 105, 2535-42. https://doi.org/10.1182/blood-2004-09-3701
  29. Morikawa T, Kuchiba A, Liao X, et al (2012). Tumor TP53 expression status, body mass index and prognosis in colorectal cancer. Int J Cancer, 131, 1169-78. https://doi.org/10.1002/ijc.26495
  30. Nagai H, Naka T, Terada Y, et al (2003) Hypermethylation associated with inactivation of the SOCS-1 gene, a JAK/STAT inhibitor, in human hepatoblastomas. J Hum Genet. 48, 65-9. https://doi.org/10.1007/s100380300008
  31. Nagai H, Naka T, Terada Y, et al (2003). Hypermethylation associated with inactivation of the SOCS-1gene, a JAK/STAT inhibitor, in human hepatoblastomas. J Hum Genet, 48, 65-9. https://doi.org/10.1007/s100380300008
  32. Ogino S, Nosho K, Irahara N, et al (2009). A cohort study of cyclin D1 expression and prognosis in 602 colon cancer cases. Clin Cancer Res, 15, 4431-38. https://doi.org/10.1158/1078-0432.CCR-08-3330
  33. Ogino S, Nosho K, Kirkner GJ, et al (2009). CpG island methylator phenotype, microsatellite instability, BRAF mutation and clinical outcome in colon cancer. Gut, 58, 90-96. https://doi.org/10.1136/gut.2008.155473
  34. Oshino Y, Kuraoka K, Nakayama H, et al (2004). Epigenetic inactiviation of SOCS-1 by CpG island hypermethylation in human gastric carsinoma. Int J Cancer, 112, 1003-9. https://doi.org/10.1002/ijc.20521
  35. Rakesh K, Agrawal DK (2005). Controlling cytokine signaling by constitutive inhibitors. Biochem Pharmacol, 70, 649-57. https://doi.org/10.1016/j.bcp.2005.04.042
  36. Rottapel R, Ilangumaran S, Neale C, et al (2002). The tumor suppressor activity of SOCS-1. Oncogene, 21, 4351-62. https://doi.org/10.1038/sj.onc.1205537
  37. Shima K, Morikawa T, Yamauchi M, et al (2011). TGFBR2 and BAX mononucleotide tract mutations, microsatellite instability, and prognosis in 1072 colorectal cancers. PLoS One, 6, 25062. https://doi.org/10.1371/journal.pone.0025062
  38. Siegel R, DeSantis C, Virgo K, et al (2012). Cancer treatment and survivorship statistics, 2012. CA Cancer J Clin, 62, 220-41. https://doi.org/10.3322/caac.21149
  39. Siegel R,Ward E, Brawley O, Jemal A (2011). Cancer statistics, 2011: the impact of eliminating socioeconomic and racial disparities on premature cancer deaths. CA Cancer J Clin, 61, 212-36. https://doi.org/10.3322/caac.20121
  40. Slattery ML, Lundgreen A, Kadlubar SA, Bondurant KL, Wolff RK (2011). JAK/STAT/SOCS-signaling pathway and colon and rectal cancer. Mol Carcinog, 52, 155-66.
  41. Sutherland KD, Lindeman GJ, Choong DY, et al (2004). Differential hypermethylation of SOCS genes in ovarian and breast carsinomas. Oncogene, 23, 7726-33. https://doi.org/10.1038/sj.onc.1207787
  42. To KF, Chan MW, Leung WK, et al (2004). Constitutional activation of IL-6-mediated JAK/STAT pathway through hypermethylation of SOCS-1 in human gastric cancer cell line. Br J Cancer, 91, 1335-41. https://doi.org/10.1038/sj.bjc.6602133
  43. Toyota M, Issa JP (1999). CpG island methylator phenotypes in aging and cancer. Semin Cancer Biol, 9, 349-57.
  44. Walsh JM, Terdiman JP (2003). Colorectal cancer screening: scientific review. JAMA, 289, 1288-96. https://doi.org/10.1001/jama.289.10.1288
  45. Watanabe D, Ezoe S, Fujimoto M, et al (2004). Suppressor of cytokine signalling-1 gene silencing in acute myeloid leukaemia and human haematopoietic cell lines. Br J Haematol, 126, 726-35. https://doi.org/10.1111/j.1365-2141.2004.05107.x
  46. Wilkinson N, Scott-Conner CE (2008). Surgical therapy for colorectal adenocarcinoma. Gastroenterol Clin North Am, 37, 253-67. https://doi.org/10.1016/j.gtc.2007.12.012
  47. Wolpin BM, Mayer RJ (2008). Systemic treatment of colorectal cancer. Gastroenterology, 134, 1296-310. https://doi.org/10.1053/j.gastro.2008.02.098
  48. Yang YF, Zhang XY, Yang M, et al (2014). Prognostic role of nucleophosmin in colorectal carcinomas. Asian Pac J Cancer Prev, 15, 2021-6. https://doi.org/10.7314/APJCP.2014.15.5.2021
  49. Yoshikawa H, Matsubara K, Qian GS, et al (2001). SOCS-1, a negative regulator of the JAK/STAT pathway, is silenced by methylation in human hepatocellular carcinoma and shows growth-suppression activity. Nat Genet, 28, 29-35.
  50. Yoshimura A, Nishinakamura H, Matsumura Y, Hanada T (2005). Negative regulation of cytokine signaling and immune responses by SOCS proteins. Arthritis Res Ther, 7, 100-10. https://doi.org/10.1186/ar1741
  51. Zlobec I, Lugli A (2008). Prognostic and predictive factors in colorectal cancer. J Clin Pathol, 61, 561-69.

Cited by

  1. Tumour-promoting role of SOCS1 in colorectal cancer cells vol.5, pp.1, 2015, https://doi.org/10.1038/srep14301
  2. SOCS1 is associated with clinical progression and acts as an oncogenic role in triple-negative breast cancer vol.70, pp.4, 2018, https://doi.org/10.1002/iub.1728