DOI QR코드

DOI QR Code

Aberrant Expression of the Autocrine Motility Factor Receptor Correlates with Poor Prognosis and Promotes Metastasis in Gastric Carcinoma

  • Huang, Zhen (Department of Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical University) ;
  • Zhang, Neng (Department of Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical University) ;
  • Zha, Lang (Department of Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical University) ;
  • Mao, Hong-Chao (Department of Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical University) ;
  • Chen, Xuan (Department of Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical University) ;
  • Xiang, Ji-Feng (Department of Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical University) ;
  • Zhang, Hua (Department of Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical University) ;
  • Wang, Zi-Wei (Department of Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical University)
  • Published : 2014.01.30

Abstract

AMFR, autocrine motility factor receptor, also called gp78, is a cell surface cytokine receptor which has a dual role as an E3 ubiquitin ligase in endoplasmic reticulum-associated degradation. AMFR expression is associated with tumor malignancy. We here investigated the clinical significance of AMFR and its role in metastasis and prognosis in gastric cancer. Expression of AMFR, E-cadherin and N-cadherin in cancer tissues and matched adjacent normal tissues from 122 gastric cancer (GC) patients undergoing surgical resection was assessed by immunohistochemistry. Levels of these molecules in 17 cases selected randomly were also analysed by Western blotting. AMFR expression was significantly increased in gastric cancer tissues, and associated with invasion depth and lymph node metastasis. Kaplan-Meier analysis showed AMFR expression correlated with poor overall survival and an increased risk of recurrence in the GC cases. Cox regression analysis suggested AMFR to be an independent predictor for overall and recurrence-free survival. E-cadherin expression was decreased in gastric cancer tissues; conversely, N-cadherin was increased. Expression of AMFR negatively correlated with E-cadherin expression, whereas N-cadherin expression showed a significant positive correlation with AMFR expression. AMFR might be involved in the regulation of epithelial-mesenchymal transition, with aberrant expression correlating with a poor prognosis and promoting invasion and metastasis in GCs.

Keywords

References

  1. Berx G, van Roy F (2009). Involvement of members of the cadherin superfamily in cancer. Cold Spring Harb Perspect Biol, 1, a003129.
  2. Cai J, Zhao XL, Liu AW, Nian H, Zhang SH (2011). Apigenin inhibits hepatoma cell growth through alteration of gene expression patterns. Phytomedicine, 18, 366-73. https://doi.org/10.1016/j.phymed.2010.08.006
  3. Cavallaro U, Christofori G (2004). Cell adhesion and signalling by cadherins and Ig-CAMs in cancer. Nat Rev Cancer, 4, 118-32. https://doi.org/10.1038/nrc1276
  4. Chen B, Mariano J, Tsai YC, et al (2006). The activity of a human endoplasmic reticulum-associated degradation E3, gp78, requires its Cue domain, RING finger, and an E2-binding site. Proc Natl Acad Sci USA, 103, 341-6. https://doi.org/10.1073/pnas.0506618103
  5. Chiu CG, St-Pierre P, Nabi IR, Wiseman SM (2008). Autocrine motility factor receptor: a clinical review. Expert Rev Anticancer Ther, 8, 207-17. https://doi.org/10.1586/14737140.8.2.207
  6. Das R, Mariano J, Tsai YC, et al (2009). Allosteric activation of E2-RING finger-mediated ubiquitylation by a structurally defined specific E2-binding region of gp78. Mol Cell, 34, 674-85. https://doi.org/10.1016/j.molcel.2009.05.010
  7. Fairbank M, St-Pierre P, Nabi IR (2009). The complex biology of autocrine motility factor/ phosphoglucose isomerase (AMF/PGI) and its receptor, the gp78/AMFR E3 ubiquitin ligase. Mol Biosyst, 5, 793-801. https://doi.org/10.1039/b820820b
  8. Fang S, Ferrone M, Yang C, et al (2001). The tumor autocrine motility factor receptor, gp78, is a ubiquitin protein ligase implicated in degradation from the endoplasmic reticulumProc. Proc Natl Acad Sci USA, 98, 14422-7. https://doi.org/10.1073/pnas.251401598
  9. Funasaka T, Haga A, Raz A, Nagase H (2001). Tumor autocrine motility factor is an angiogenic factor that stimulates endothelial cell motility. Biochem Biophys Res Commun, 285, 118-28. https://doi.org/10.1006/bbrc.2001.5135
  10. Funasaka T, Haga A, Raz A, Nagase H (2002). Autocrine motility factor secreted by tumor cells upregulates vascular endothelial growth factor receptor (Flt-1) expression in endothelial cells. Int J Cancer, 101, 217-23. https://doi.org/10.1002/ijc.10617
  11. Funasaka T, Hu H, Yanagawa T, Hogan V, Raz A (2007). Downregulation of phosphoglucose isomerase/autocrine motility factor results in mesenchymal-to-epithelial transition of human lung fibrosarcoma cells. Cancer Res, 67, 4236-43. https://doi.org/10.1158/0008-5472.CAN-06-3935
  12. Graziosi L, Marino E, Cavazzoni E, Donini A (2013). Prognostic value of the seventh AJCC/UICC TNM classification of noncardia gastric cancer. World J Surg Oncol, 11, 103. https://doi.org/10.1186/1477-7819-11-103
  13. Haga A, Funasaka T, Niinaka Y, Raz A, Nagase H (2003). The autocrine motility factor signaling induces tumor apoptotic resistance which regulates apoptosome expression. Int J Cancer, 107, 707-14. https://doi.org/10.1002/ijc.11449
  14. Haga A, Niinaka Y, Raz A (2000). Phosphohexose isomerase/autocrine motility factor/neuroleukin/ maturation factor is a multifunctional phosphoprotein. Biochim Biophys Acta, 1480, 235-44. https://doi.org/10.1016/S0167-4838(00)00075-3
  15. Hanahan D, Weinberg RA (2011). Hallmarks of cancer: the next generation. Cell, 144, 646-74. https://doi.org/10.1016/j.cell.2011.02.013
  16. Hlubek F, Brabletz T, Budczies J, et al. Heterogeneous expression of Wnt/beta-catenin target genes within colorectal cancer. Int J Cancer, 121, 1941-8.
  17. Iiizumi M, Liu W, Pai SK, Furuta E, Watabe K (2008). Drug development against metastasis-related genes and their pathways: a rationale for cancer therapy. Biochim Biophys Acta, 1786, 87-104.
  18. Jemal A, Bray F, Center MM, et al (2011). Global Cancer Statistics. CA Cancer J Clin, 61, 69-90. https://doi.org/10.3322/caac.20107
  19. Joshi B, Li L, Nabi IR (2010). A role for KAI1 in promotion of cell proliferation and mammary gland hyperplasia by the gp78 ubiquitin ligase. J Biol Chem, 285, 8830-9. https://doi.org/10.1074/jbc.M109.074344
  20. Kostova Z, Tsai YC, Weissman AM (2007). Ubiquitin ligases, critical mediators of endoplasmic reticulum-associated degradation. Semin Cell Dev Biol, 18, 770-79. https://doi.org/10.1016/j.semcdb.2007.09.002
  21. Li FX, Zhang RP, Liang H, et al (2013). Validity and necessity of sub-classification of N3 in the 7th UICC TNM stage of gastric cancer. Asian Pac J Cancer Prev, 14, 2091-5. https://doi.org/10.7314/APJCP.2013.14.3.2091
  22. Liang JS, Kim T, Fang S, et al (2003). Overexpression of the tumor autocrine motility factor receptor Gp78, a ubiquitin protein ligase, results in increased ubiquitinylation and decreased secretion of apolipoprotein B100 in HepG2 cells. J Biol Chem, 278, 23984-8. https://doi.org/10.1074/jbc.M302683200
  23. Liotta LA, Mandler R, Murano G, et al (1986). Tumor cell autocrine motility factor. Proc Natl Acad Sci USA, 83, 3302-6. https://doi.org/10.1073/pnas.83.10.3302
  24. Miranti CK (2009). Controlling cell surface dynamics and signaling: how CD82/KAI1 suppresses metastasis. Cell Signal, 21, 196-211. https://doi.org/10.1016/j.cellsig.2008.08.023
  25. Morito D, Hirao K, Oda Y, et al (2008). Gp78 cooperates with RMA1 in endoplasmic reticulum-associated degradation of CFTRDeltaF508. Mol Biol Cell, 19, 1328-36. https://doi.org/10.1091/mbc.E07-06-0601
  26. Murray D, Horgan G, Macmathuna P, Doran P (2008). NET1-mediated RhoA activation facilitates lysophosphatidic acidinduced cell migration and invasion in gastric cancer. Br J Cancer, 99, 1322-9. https://doi.org/10.1038/sj.bjc.6604688
  27. Nakatsukasa K, Brodsky JL (2008). The recognition and retrotranslocation of misfolded proteins from the endoplasmic reticulum. Traffic, 9, 861-70. https://doi.org/10.1111/j.1600-0854.2008.00729.x
  28. Niinaka Y, Paku S, Haga A, Watanabe H, Raz A (1998). Expression and secretion of neuroleukin/phosphohexose isomerase/maturation factor as autocrine motility factor by tumor cells. Cancer Res, 58, 2667-74.
  29. Nussenbaum F, Herman IM (2010). Tumor angiogenesis: insights and innovations. J Oncol, 2010, 132641.
  30. Peng CW, Wang LW, Zeng WJ, Yang XJ, Li Y (2013). Evaluation of the staging systems for gastric cancer. J Surg Oncol, 108, 93-105. https://doi.org/10.1002/jso.23360
  31. Romagnolia A, Oliverio S, Evangelisti C, et al (2003). Neuroleukin inhibition sensitises neuronal cells to caspasedependent apoptosis. Biochem Biophys Res Commun, 302, 448-53. https://doi.org/10.1016/S0006-291X(03)00188-8
  32. Saito H, Fukumoto Y, Osaki T, et al (2008). Prognostic significance of the ratio between metastatic and dissected lymph nodes (n ratio) in patients with advanced gastric cancer. J Surg Oncol, 97, 132-5. https://doi.org/10.1002/jso.20929
  33. Shimizu K, Tani M, Watanabe H, et al (1999). The autocrine motility factor gene encodes a novel type of seven transmembrane protein. FEBS Lett, 456, 295-300. https://doi.org/10.1016/S0014-5793(99)00966-7
  34. Siegel R, Naishadham MA, Jemal A (2013). Cancer Statistics, 2013. CA Cancer J Clin, 63, 11-30. https://doi.org/10.3322/caac.21166
  35. Silletti S, Watanabe H, Hogan V, Nabi IR, Raz A (1991). Purification of B16-F1 melanoma autocrine motility factor and its receptor. Cancer Res, 51, 3507-11.
  36. Sjoblom T, Jones S, Wood LD, et al (2006). The consensus coding sequences of human breast and colorectal cancers. Science, 314, 268-74. https://doi.org/10.1126/science.1133427
  37. Song BL, Sever N, DeBose-Boyd RA (2005). Gp78, a membrane-anchored ubiquitin ligase, associates with Insig-1 and couples sterol-regulated ubiquitination to degradation of HMG CoA reductase. Mol Cell, 19, 829-40. https://doi.org/10.1016/j.molcel.2005.08.009
  38. Sud R, Wells D, Talbot IC, Delhanty JD (2001). Genetic alterations in gastric cancers from British patients. Cancer Genet Cytogenet, 126, 111-9. https://doi.org/10.1016/S0165-4608(00)00397-6
  39. Tahara E (2000). Molecular aspects of invasion and metastasis of stomach cancer. Verh Dtsch Ges Pathol, 84, 43-9.
  40. Thiery JP, Acloque H, Huang RY, Nieto MA (2009). Epithelialmesenchymal transitions in development and disease. Cell, 139, 871-90. https://doi.org/10.1016/j.cell.2009.11.007
  41. Tsai YC, Mendoza A, Mariano JM, et al (2007). The ubiquitin ligase gp78 promotes sarcoma metastasis by targeting KAI1 for degradation. Nat Med, 13, 1504-9. https://doi.org/10.1038/nm1686
  42. Tsutsumi S, Gupta SK, Hogan V, Collard JG, Raz A (2002). Activation of small GTPase Rho is required for autocrine motility factor signaling. Cancer Res, 62, 4484-90.
  43. Tsutsumi S, Yanagawa T, Shimura T, et al (2003). Regulation of cell proliferation by autocrine motility factor/phosphoglucose isomerase signaling. J Biol Chem, 278, 32165-72. https://doi.org/10.1074/jbc.M304537200
  44. Wallace TA, Prueitt RL, Yi M, et al (2008). Tumor immunobiological differences in prostate cancer between African-American and European-American men. Cancer Res, 68, 927-36. https://doi.org/10.1158/0008-5472.CAN-07-2608
  45. Watanabe H, Carmi P, Hogan V, et al (1991). Purification of human tumor cell autocrine motility factor and molecular cloning of its receptor. J Biol Chem, 266, 13442-8.
  46. Watanabe H, Takehama K, Date M, Shinozaki T, Raz A (1996). Tumor cell motility factor is the neuroleukin/ phosphohexose isomerase polypeptide. Cancer Res, 56, 2960-3.
  47. Wei D, Gong W, Kanai M, et al (2005). Drastic down-regulation of kruppel-like factor 4 expression is critical in human gastric cancer development and progression. Cancer Res, 65, 2746-54. https://doi.org/10.1158/0008-5472.CAN-04-3619
  48. World Health Organization (2008). The global burden of disease: 2004 update. Geneva: World Health Organization.
  49. Yilmaz M, Christofori G (2009). EMT, the cytoskeleton, and cancer cell invasion. Cancer Metastasis Rev, 28, 15-33. https://doi.org/10.1007/s10555-008-9169-0
  50. Zha L, Zhang J, Tang W, et al (2013). HMGA2 elicits EMT by activating the Wnt/$\beta$-catenin pathway in gastric cancer. Dig Dis Sci, 58, 724-33. https://doi.org/10.1007/s10620-012-2399-6
  51. Zhang N, Zhang J, Wang ZW, Zha L, Huang Z (2012). Altered expression of Kruppel-like factor 4 and $\beta$-catenin in human gastric cancer. Oncol Lett, 3, 1017-22.
  52. Zhong X, Shen Y, Ballar P, et al (2004). AAA ATPase p97/ valosin-containing protein interacts with gp78, a ubiquitin ligase for endoplasmic reticulum-associated degradation. J Biol Chem, 279, 45676-84. https://doi.org/10.1074/jbc.M409034200

Cited by

  1. N-cadherin, a novel prognostic biomarker, drives malignant progression of colorectal cancer vol.12, pp.2, 2015, https://doi.org/10.3892/mmr.2015.3687
  2. Inhibitory Effects of Dunning Rat Prostate Tumor Fluid on Proliferation of the Metastatic MAT-LyLu Cell Line vol.16, pp.2, 2015, https://doi.org/10.7314/APJCP.2015.16.2.831
  3. Autocrine motility factor receptor promotes the proliferation of human acute monocytic leukemia THP-1 cells vol.36, pp.3, 2015, https://doi.org/10.3892/ijmm.2015.2267
  4. Overexpression of E3 ubiquitin ligase tripartite motif 32 correlates with a poor prognosis in patients with gastric cancer vol.13, pp.5, 2017, https://doi.org/10.3892/ol.2017.5806