DOI QR코드

DOI QR Code

Highly Expressed Integrin-α8 Induces Epithelial to Mesenchymal Transition-Like Features in Multiple Myeloma with Early Relapse

  • Ryu, Jiyeon (Cancer Research Institute, Seoul National University College of Medicine) ;
  • Koh, Youngil (Department of Internal Medicine, Seoul National University Hospital) ;
  • Park, Hyejoo (Cancer Research Institute, Seoul National University College of Medicine) ;
  • Kim, Dae Yoon (Cancer Research Institute, Seoul National University College of Medicine) ;
  • Kim, Dong Chan (Cancer Research Institute, Seoul National University College of Medicine) ;
  • Byun, Ja Min (Department of Internal Medicine, Seoul National University Hospital) ;
  • Lee, Hyun Jung (Department of Internal Medicine, Dongguk University Ilsan Hospital) ;
  • Yoon, Sung-Soo (Cancer Research Institute, Seoul National University College of Medicine)
  • Received : 2016.08.31
  • Accepted : 2016.12.05
  • Published : 2016.12.31

Abstract

Despite recent groundbreaking advances in multiple myeloma (MM) treatment, most MM patients ultimately experience relapse, and the relapse biology is not entirely understood. To define altered gene expression in MM relapse, gene expression profiles were examined and compared among 16 MM patients grouped by 12 months progression-free survival (PFS) after autologous stem cell transplantation. To maximize the difference between prognostic groups, patients at each end of the PFS spectrum (the four with the shortest PFS and four with the longest PFS) were chosen for additional analyses. We discovered that integrin-${\alpha}8$ (ITGA8) is highly expressed in MM patients with early relapse. The integrin family is well known to be involved in MM progression; however, the role of integrin-${\alpha}8$ is largely unknown. We functionally overexpressed integrin-${\alpha}8$ in MM cell lines, and surprisingly, stemness features including $HIF1{\alpha}$, VEGF, OCT4, and Nanog, as well as epithelial mesenchymal transition (EMT)-related phenotypes, including N-cadherin, Slug, Snail and CXCR4, were induced. These, consequently, enhanced migration and invasion abilities, which are crucial to MM pathogenesis. Moreover, the gain of integrin-${\alpha}8$ expression mediated drug resistance against melphalan and bortezomib, which are the main therapeutic agents in MM. The cBioPortal genomic database revealed that ITGA8 have significant tendency to co-occur with PDGFRA and PDGFRB and their mRNA expression were up-regulated in ITGA8 overexpressed MM cells. In summary, integrin-${\alpha}8$, which was up-regulated in MM of early relapse, mediates EMT-like phenotype, enhancing migration and invasion; therefore, it could serve as a potential marker of MM relapse and be a new therapeutic target.

Keywords

References

  1. Alsayed, Y., Ngo, H., Runnels, J., Leleu, X., Singha, U.K., Pitsillides, C.M., Spencer, J.A., Kimlinger, T., Ghobrial, J.M., Jia, X., et al. (2007). Mechanisms of regulation of CXCR4/SDF-1 (CXCL12)-dependent migration and homing in multiple myeloma. Blood 109, 2708-2717.
  2. Anguiano, A., Tuchman, S.A., Acharya, C., Salter, K., Gasparetto, C., Zhan, F., Dhodapkar, M., Nevins, J., Barlogie, B., Shaughnessy, J.D., Jr., et al. (2009). Gene expression profiles of tumor biology provide a novel approach to prognosis and may guide the selection of therapeutic targets in multiple myeloma. J. Clin. Oncol. 27, 4197-4203. https://doi.org/10.1200/JCO.2008.19.1916
  3. Arumugam, T., Ramachandran, V., Fournier, K.F., Wang, H., Marquis, L., Abbruzzese, J.L., Gallick, G.E., Logsdon, C.D., McConkey, D.J., and Choi, W. (2009). Epithelial to mesenchymal transition contributes to drug resistance in pancreatic cancer. Cancer Res. 69, 5820-5828. https://doi.org/10.1158/0008-5472.CAN-08-2819
  4. Avet-Loiseau, H. (2010). Ultra high-risk myeloma. Hematology / the Education Program of the American Society of Hematology. American Society of Hematology. Education Program 2010, 489-493.
  5. Azab, A.K., Runnels, J.M., Pitsillides, C., Moreau, A.S., Azab, F., Leleu, X., Jia, X., Wright, R., Ospina, B., Carlson, A.L., et al. (2009). CXCR4 inhibitor AMD3100 disrupts the interaction of multiple myeloma cells with the bone marrow microenvironment and enhances their sensitivity to therapy. Blood 113, 4341-4351. https://doi.org/10.1182/blood-2008-10-186668
  6. Azab, A.K., Hu, J., Quang, P., Azab, F., Pitsillides, C., Awwad, R., Thompson, B., Maiso, P., Sun, J.D., Hart, C.P., et al. (2012). Hypoxia promotes dissemination of multiple myeloma through acquisition of epithelial to mesenchymal transition-like features. Blood 119, 5782-5794. https://doi.org/10.1182/blood-2011-09-380410
  7. Batist, G., Wu, J.H., Spatz, A., Miller, W.H., Jr., Cocolakis, E., Rousseau, C., Diaz, Z., Ferrario, C., and Basik, M. (2011). Resistance to cancer treatment: the role of somatic genetic events and the challenges for targeted therapies. Front. Pharmacol. 2, 59.
  8. Bianchi, G., Richardson, P.G., and Anderson, K.C. (2015). Promising therapies in multiple myeloma. Blood 126, 300-310. https://doi.org/10.1182/blood-2015-03-575365
  9. Blagosklonny, M.V. (2006). Target for cancer therapy: proliferating cells or stem cells. Leukemia 20, 385-391. https://doi.org/10.1038/sj.leu.2404075
  10. Brandenberger, R., Schmidt, A., Linton, J., Wang, D., Backus, C., Denda, S., Muller, U., and Reichardt, L.F. (2001). Identification and characterization of a novel extracellular matrix protein nephronectin that is associated with integrin alpha8beta1 in the embryonic kidney. J. Cell Biol. 154, 447-458. https://doi.org/10.1083/jcb.200103069
  11. Broyl, A., Hose, D., Lokhorst, H., de Knegt, Y., Peeters, J., Jauch, A., Bertsch, U., Buijs, A., Stevens-Kroef, M., Beverloo, H.B., et al. (2010). Gene expression profiling for molecular classification of multiple myeloma in newly diagnosed patients. Blood 116, 2543-2553. https://doi.org/10.1182/blood-2009-12-261032
  12. Damiano, J.S., and Dalton, W.S. (2000). Integrin-mediated drug resistance in multiple myeloma. Leuk. Lymphoma 38, 71-81.
  13. Damiano, J.S., Cress, A.E., Hazlehurst, L.A., Shtil, A.A., and Dalton, W.S. (1999). Cell adhesion mediated drug resistance (CAM-DR): Role of integrins and resistance to apoptosis in human myeloma cell lines. Blood 93, 1658-1667.
  14. Denda, S., Muller, U., Crossin, K.L., Erickson, H.P., and Reichardt, L.F. (1998). Utilization of a soluble integrin-alkaline phosphatase chimera to characterize integrin alpha 8 beta 1 receptor interactions with tenascin: murine alpha 8 beta 1 binds to the RGD site in tenascin-C fragments, but not to native tenascin-C. Biochemistry 37, 5464-5474. https://doi.org/10.1021/bi9727489
  15. Desgrosellier, J.S., and Cheresh, D.A. (2010). Integrins in cancer: biological implications and therapeutic opportunities. Nat. Rev. Cancer 10, 9-22. https://doi.org/10.1038/nrc2748
  16. Draghici, S., Khatri, P., Tarca, A.L., Amin, K., Done, A., Voichita, C., Georgescu, C., and Romero, R. (2007). A systems biology approach for pathway level analysis. Genome Res. 17, 1537-1545. https://doi.org/10.1101/gr.6202607
  17. Eliceiri, B.P. (2001). Integrin and growth factor receptor crosstalk. Circulation Res. 89, 1104-1110. https://doi.org/10.1161/hh2401.101084
  18. Gialeli, C., Theocharis, A.D., and Karamanos, N.K. (2011). Roles of matrix metalloproteinases in cancer progression and their pharmacological targeting. FEBS J. 278, 16-27. https://doi.org/10.1111/j.1742-4658.2010.07919.x
  19. Guarino, M. (2007). Epithelial-mesenchymal transition and tumour invasion. Int. J. Biochem. Cell Biol. 39, 2153-2160. https://doi.org/10.1016/j.biocel.2007.07.011
  20. Guarino, M., Rubino, B., and Ballabio, G. (2007). The role of epithelial-mesenchymal transition in cancer pathology. Pathology 39, 305-318. https://doi.org/10.1080/00313020701329914
  21. Harousseau, J.L., Shaughnessy, J., Jr. and Richardson, P. (2004). Multiple myeloma. Hematology / the Education Program of the American Society of Hematology. American Society of Hematology. Education Program, 237-256.
  22. Hideshima, T., Chauhan, D., Hayashi, T., Podar, K., Akiyama, M., Gupta, D., Richardson, P., Munshi, N., and Anderson, K.C. (2002). The biological sequelae of stromal cell-derived factor-1alpha in multiple myeloma. Mol. Cancer Ther. 1, 539-544. https://doi.org/10.4161/cbt.1.5.174
  23. Kalluri, R., and Weinberg, R.A. (2009). The basics of epithelialmesenchymal transition. J. Clin. Invest. 119, 1420-1428. https://doi.org/10.1172/JCI39104
  24. Kuiper, R., Broyl, A., de Knegt, Y., van Vliet, M.H., van Beers, E.H., van der Holt, B., el Jarari, L., Mulligan, G., Gregory, W., Morgan, G., et al. (2012). A gene expression signature for high-risk multiple myeloma. Leukemia 26, 2406-2413. https://doi.org/10.1038/leu.2012.127
  25. Kyle, R.A., and Rajkumar, S.V. (2004). Multiple myeloma. The New Engl. J. Med. 351, 1860-1873. https://doi.org/10.1056/NEJMra041875
  26. Lobo, N.A., Shimono, Y., Qian, D., and Clarke, M.F. (2007). The biology of cancer stem cells. Ann. Rev. Cell Dev. Biol. 23, 675-699. https://doi.org/10.1146/annurev.cellbio.22.010305.104154
  27. Lonial, S. (2010). Relapsed multiple myeloma. Hematology / the Education Program of the American Society of Hematology. American Society of Hematology. Education Program 2010, 303-309.
  28. Matsui, W., Wang, Q., Barber, J.P., Brennan, S., Smith, B.D., Borrello, I., McNiece, I., Lin, L., Ambinder, R.F., Peacock, C., et al. (2008). Clonogenic multiple myeloma progenitors, stem cell properties, and drug resistance. Cancer Res. 68, 190-197. https://doi.org/10.1158/0008-5472.CAN-07-3096
  29. Menu, E., Asosingh, K., Indraccolo, S., De Raeve, H., Van Riet, I., Van Valckenborgh, E., Vande Broek, I., Fujii, N., Tamamura, H., Van Camp, B., et al. (2006). The involvement of stromal derived factor 1alpha in homing and progression of multiple myeloma in the 5TMM model. Haematologica 91, 605-612.
  30. Mitra, A., Mishra, L., and Li, S.L. (2015). EMT, CTCs and CSCs in tumor relapse and drug-resistance. Oncotarget 6, 10697-10711. https://doi.org/10.18632/oncotarget.4037
  31. Muller, U., Bossy, B., Venstrom, K., and Reichardt, L.F. (1995). Integrin alpha 8 beta 1 promotes attachment, cell spreading, and neurite outgrowth on fibronectin. Mol. Biol. Cell 6, 433-448. https://doi.org/10.1091/mbc.6.4.433
  32. Muz, B., de la Puente, P., Azab, F., Luderer, M. and Azab, A.K. (2014). Hypoxia promotes stem cell-like phenotype in multiple myeloma cells. Blood Cancer J. 4, e262. https://doi.org/10.1038/bcj.2014.82
  33. Neri, P., Ren, L., Azab, A.K., Brentnall, M., Gratton, K., Klimowicz, A.C., Lin, C., Duggan, P., Tassone, P., Mansoor, A., et al. (2011). Integrin beta7-mediated regulation of multiple myeloma cell adhesion, migration, and invasion. Blood 117, 6202-6213. https://doi.org/10.1182/blood-2010-06-292243
  34. Nooter, K., and Herweijer, H. (1991). Multidrug resistance (mdr) genes in human cancer. Brit. J. Cancer 63, 663-669. https://doi.org/10.1038/bjc.1991.152
  35. Nowakowski, G.S., Witzig, T.E., Dingli, D., Tracz, M.J., Gertz, M.A., Lacy, M.Q., Lust, J.A., Dispenzieri, A., Greipp, P.R., Kyle, R.A., et al. (2005). Circulating plasma cells detected by flow cytometry as a predictor of survival in 302 patients with newly diagnosed multiple myeloma. Blood 106, 2276-2279. https://doi.org/10.1182/blood-2005-05-1858
  36. Nurwidya, F., Takahashi, F., Murakami, A., and Takahashi, K. (2012). Epithelial mesenchymal transition in drug resistance and metastasis of lung cancer. Cancer Res. Treat. 44, 151-156. https://doi.org/10.4143/crt.2012.44.3.151
  37. Ooi, L.L., and Dunstan, C.R. (2009). CXCL12/CXCR4 axis in tissue targeting and bone destruction in cancer and multiple myeloma. J. Bone Miner. Res. 24, 1147-1149. https://doi.org/10.1359/jbmr.090503
  38. Pagnucco, G., Cardinale, G., and Gervasi, F. (2004). Targeting multiple myeloma cells and their bone marrow microenvironment. Ann. N Y Acad. Sci. 1028, 390-399. https://doi.org/10.1196/annals.1322.047
  39. Palumbo, A., and Anderson, K. (2011). Multiple Myeloma REPLY. New Engl. J. Med. 364, 2364-2364. https://doi.org/10.1056/NEJMc1104560
  40. Pandit, S., and Vesole, D.H. (2001). Relapsed multiple myeloma. Curr. Treat. Options Oncol. 2, 261-269. https://doi.org/10.1007/s11864-001-0040-6
  41. Potti, A., Dressman, H.K., Bild, A., Riedel, R.F., Chan, G., Sayer, R., Cragun, J., Cottrill, H., Kelley, M.J., Petersen, R., et al. (2006). Genomic signatures to guide the use of chemotherapeutics. Nat. Med. 12, 1294-1300. https://doi.org/10.1038/nm1491
  42. Rathinam, R., and Alahari, S.K. (2010). Important role of integrins in the cancer biology. Cancer Metastsis Rev. 29, 223-237. https://doi.org/10.1007/s10555-010-9211-x
  43. Reverter, F., Vegas, E., and Sanchez, P. (2010). Mining gene expression profiles: an integrated implementation of kernel principal component analysis and singular value decomposition. Genom. Proteom. Bioinform. 8, 200-210. https://doi.org/10.1016/S1672-0229(10)60022-8
  44. Richardson, P.G., Mitsiades, C.S., Hideshima, T. and Anderson, K.C. (2005) Novel biological therapies for the treatment of multiple myeloma. Best Pract. Res. Clin. Haematol. 18, 619-634. https://doi.org/10.1016/j.beha.2005.01.010
  45. Roccaro, A.M., Mishima, Y., Sacco, A., Moschetta, M., Tai, Y.T., Shi, J., Zhang, Y., Reagan, M.R., Huynh, D., Kawano, Y., et al. (2015). CXCR4 regulates extra-medullary myeloma through epithelial-mesenchymal-transition-like transcriptional activation. Cell. Rep. 12, 622-635. https://doi.org/10.1016/j.celrep.2015.06.059
  46. Saldanha, A.J. (2004). Java Treeview--extensible visualization of microarray data. Bioinformatics 20, 3246-3248. https://doi.org/10.1093/bioinformatics/bth349
  47. Sanz-Rodriguez, F., and Teixido, J. (2001). VLA-4-dependent myeloma cell adhesion. Leuk. Lymphoma 41, 239-245. https://doi.org/10.3109/10428190109057979
  48. Sanz-Rodriguez, F., Ruiz-Velasco, N., Pascual-Salcedo, D., and Teixido, J. (1999). Characterization of VLA-4-dependent myeloma cell adhesion to fibronectin and VCAM-1. Brit. J. Haematol. 107, 825-834. https://doi.org/10.1046/j.1365-2141.1999.01762.x
  49. Scherberich, A., Tucker, R.P., Degen, M., Brown-Luedi, M., Andres, A.C., and Chiquet-Ehrismann, R. (2005). Tenascin-W is found in malignant mammary tumors, promotes alpha8 integrindependent motility and requires p38MAPK activity for BMP-2 and TNF-alpha induced expression in vitro. Oncogene 24, 1525-1532. https://doi.org/10.1038/sj.onc.1208342
  50. Shang, Y., Cai, X., and Fan, D. (2013). Roles of epithelialmesenchymal transition in cancer drug resistance. Cur. Cancer Drug Targets 13, 915-929. https://doi.org/10.2174/15680096113136660097
  51. Singh, A., and Settleman, J. (2010). EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer. Oncogene 29, 4741-4751. https://doi.org/10.1038/onc.2010.215
  52. Thiery, J.P., Acloque, H., Huang, R.Y., and Nieto, M.A. (2009). Epithelial-mesenchymal transitions in development and disease. Cell 139, 871-890. https://doi.org/10.1016/j.cell.2009.11.007
  53. Thompson, E.W. and Haviv, I. (2011) The social aspects of EMTMET plasticity. Nat. Med. 17, 1048-1049. https://doi.org/10.1038/nm.2437
  54. Tsirakis, G., Pappa, C.A., Kanellou, P., Stratinaki, M.A., Xekalou, A., Psarakis, F.E., Sakellaris, G., Alegakis, A., Stathopoulos, E.N., and Alexandrakis, M.G. (2012) Role of platelet-derived growth factor-AB in tumour growth and angiogenesis in relation with other angiogenic cytokines in multiple myeloma. Hematol. Oncol. 30, 131-136. https://doi.org/10.1002/hon.1014
  55. Yagi, H., Tan, W., Dillenburg-Pilla, P., Armando, S., Amornphimoltham, P., Simaan, M., Weigert, R., Molinolo, A.A., Bouvier, M., and Gutkind, J.S. (2011) A synthetic biology approach reveals a CXCR4-G13-Rho signaling axis driving transendothelial migration of metastatic breast cancer cells. Sci. Signal. 4, ra60.
  56. Yu, J., Ustach, C., and Kim, H.R. (2003). Platelet-derived growth factor signaling and human cancer. J. Biochem. Mol. Biol. 36, 49-59.
  57. Zhan, F., Hardin, J., Kordsmeier, B., Bumm, K., Zheng, M., Tian, E., Sanderson, R., Yang, Y., Wilson, C., Zangari, M., et al. (2002). Global gene expression profiling of multiple myeloma, monoclonal gammopathy of undetermined significance, and normal bone marrow plasma cells. Blood 99, 1745-1757. https://doi.org/10.1182/blood.V99.5.1745

피인용 문헌

  1. Established Models and New Paradigms for Hypoxia-Driven Cancer-Associated Bone Disease vol.102, pp.2, 2018, https://doi.org/10.1007/s00223-017-0352-6
  2. KAT3B-p300 and H3AcK18/H3AcK14 levels are prognostic markers for kidney ccRCC tumor aggressiveness and target of KAT inhibitor CPTH2 vol.10, pp.1, 2018, https://doi.org/10.1186/s13148-018-0473-4
  3. The role of integrins in acute leukemias and potential as targets for therapy vol.1, pp.3, 2018, https://doi.org/10.4103/tme.tme_4_19
  4. Are Integrins Still Practicable Targets for Anti-Cancer Therapy? vol.11, pp.7, 2019, https://doi.org/10.3390/cancers11070978
  5. Expression and Prognostic Analysis of Integrins in Gastric Cancer vol.2020, pp.None, 2016, https://doi.org/10.1155/2020/8862228
  6. Exosomes-Mediated Transfer of Itga2 Promotes Migration and Invasion of Prostate Cancer Cells by Inducing Epithelial-Mesenchymal Transition vol.12, pp.8, 2016, https://doi.org/10.3390/cancers12082300
  7. The Analysis of Gene Expression Data Incorporating Tumor Purity Information vol.12, pp.None, 2016, https://doi.org/10.3389/fgene.2021.642759
  8. Treatment of Multiple Myeloma and the Role of Melphalan in the Era of Modern Therapies-Current Research and Clinical Approaches vol.10, pp.9, 2021, https://doi.org/10.3390/jcm10091841
  9. Anti-androgen therapy induces transcriptomic reprogramming in metastatic castration-resistant prostate cancer in a murine model vol.1867, pp.7, 2016, https://doi.org/10.1016/j.bbadis.2021.166151