Skip to main content
Log in

Epigenetics of Multiple Myeloma Bone Disease

  • Molecular Biology of Bone Metastasis (H Taipaleenmäki and M Capulli, Section Editors)
  • Published:
Current Molecular Biology Reports Aims and scope Submit manuscript

Abstract

Purpose of Review

Multiple myeloma bone disease (MMDB) is a devastating clinical manifestation of multiple myeloma associated with excessive bone osteolysis, which results from enhanced osteoclastogenesis and suppression of bone marrow stromal cell (BMSC) differentiation into osteoblasts. Impaired osteogenesis and functional alterations of myeloma-exposed BMSCs (MM-BMSCs) during the course of disease evolution significantly contribute to myeloma growth, metastasis, and chemoresistance. This review highlights new studies demonstrating that epigenetic modalities including chromatin-mediated gene silencing and non-coding RNA contribute to pathogenesis of MM-BMSCs.

Recent Findings

Inhibitors targeting histone-modifying enzymes EZH2, JMJD3, HDACs, and BET proteins have been successfully used to revert osteogenic suppression of MM-BMSCs. Aberrant expression of non-coding RNA cause functional changes associated with senescence, osteogenic suppression, and tumor-promoting phenotype of MM-BMSCs.

Summary

Epigenetic events guiding transformations of the surrounding BMSC compartments are ultimately linked to disease onset and progression and open new therapeutic opportunity to target dissemination of MM tumors and reliably repair bone lesions.

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

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1

Similar content being viewed by others

References

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. Ronchetti D, Agnelli L, Pietrelli A, Todoerti K, Manzoni M, Taiana E, et al. A compendium of long non-coding RNAs transcriptional fingerprint in multiple myeloma. Sci Rep. 2018;8(1):6557. https://doi.org/10.1038/s41598-018-24701-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Ring ES, Lawson MA, Snowden JA, Jolley I, Chantry AD. New agents in the treatment of myeloma bone disease. Calcif Tissue Int. 2018;102(2):196–209. https://doi.org/10.1007/s00223-017-0351-7.

    Article  CAS  PubMed  Google Scholar 

  3. Terpos E, Ntanasis-Stathopoulos I, Gavriatopoulou M, Dimopoulos MA. Pathogenesis of bone disease in multiple myeloma: from bench to bedside. Blood Cancer J. 2018;8(1):7. https://doi.org/10.1038/s41408-017-0037-4.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Terpos E, Confavreux CB, Clezardin P. Bone antiresorptive agents in the treatment of bone metastases associated with solid tumours or multiple myeloma. Bonekey Rep. 2015;4:744. https://doi.org/10.1038/bonekey.2015.113.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Nishida H. Bone-targeted agents in multiple myeloma. Hematol Rep. 2018;10(1):7401. https://doi.org/10.4081/hr.2018.7401.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Gao F, Chiu SM, Motan DA, Zhang Z, Chen L, Ji HL, et al. Mesenchymal stem cells and immunomodulation: current status and future prospects. Cell Death Dis. 2016;7:e2062. https://doi.org/10.1038/cddis.2015.327.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Reagan MR, Rosen CJ. Navigating the bone marrow niche: translational insights and cancer-driven dysfunction. Nat Rev Rheumatol. 2016;12(3):154–68. https://doi.org/10.1038/nrrheum.2015.160.

    Article  CAS  PubMed  Google Scholar 

  8. • Adamik J, Galson DL, Roodman GD. Osteoblast suppression in multiple myeloma bone disease. J Bone Oncol. 2018;13:62–70 This paper highlights transcriptomic characterizations and functional changes of MM-BMSCs associated with osteogenic suppression, senescence, and tumor-supporting features.

    Article  Google Scholar 

  9. Mehdi SJ, Johnson SK, Epstein J, Zangari M, Qu P, Hoering A, et al. Mesenchymal stem cells gene signature in high-risk myeloma bone marrow linked to suppression of distinct IGFBP2-expressing small adipocytes. Br J Haematol. 2018. https://doi.org/10.1111/bjh.15669.

  10. • Berlier JL, Rethnam M, Banu Binte Abdul Majeed A, Suda T. Modification of the bone marrow MSC population in a xenograft model of early multiple myeloma. Biochem Biophys Res Commun. 2018. https://doi.org/10.1016/j.bbrc.2018.11.178 This paper illustrates that adipogenic changes in MSC population happen prior to significant engraftment of MM tumors.

  11. Trotter TN, Gibson JT, Sherpa TL, Gowda PS, Peker D, Yang Y. Adipocyte-lineage cells support growth and dissemination of multiple myeloma in bone. Am J Pathol. 2016;186(11):3054–63. https://doi.org/10.1016/j.ajpath.2016.07.012.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Silbermann R, Roodman GD. Current controversies in the management of myeloma bone disease. J Cell Physiol. 2016;231(11):2374–9. https://doi.org/10.1002/jcp.25351.

    Article  CAS  PubMed  Google Scholar 

  13. Lawrence M, Daujat S, Schneider R. Lateral thinking: how histone modifications regulate gene expression. Trends Genet. 2016;32(1):42–56. https://doi.org/10.1016/j.tig.2015.10.007.

    Article  CAS  PubMed  Google Scholar 

  14. Voigt P, Tee WW, Reinberg D. A double take on bivalent promoters. Genes Dev. 2013;27(12):1318–38. https://doi.org/10.1101/gad.219626.113.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Sadakierska-Chudy A, Filip M. A comprehensive view of the epigenetic landscape. Part II: histone post-translational modification, nucleosome level, and chromatin regulation by ncRNAs. Neurotox Res. 2015;27(2):172–97. https://doi.org/10.1007/s12640-014-9508-6.

    Article  CAS  PubMed  Google Scholar 

  16. Agirre X, Castellano G, Pascual M, Heath S, Kulis M, Segura V, et al. Whole-epigenome analysis in multiple myeloma reveals DNA hypermethylation of B cell-specific enhancers. Genome Res. 2015;25(4):478–87. https://doi.org/10.1101/gr.180240.114.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Agarwal P, Alzrigat M, Parraga AA, Enroth S, Singh U, Ungerstedt J, et al. Genome-wide profiling of histone H3 lysine 27 and lysine 4 trimethylation in multiple myeloma reveals the importance of polycomb gene targeting and highlights EZH2 as a potential therapeutic target. Oncotarget. 2016;7(6):6809–23. https://doi.org/10.18632/oncotarget.6843.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Zaidi SK, Frietze SE, Gordon JA, Heath JL, Messier T, Hong D, et al. Bivalent epigenetic control of oncofetal gene expression in cancer. Mol Cell Biol. 2017;37(23). https://doi.org/10.1128/MCB.00352-17.

  19. Park-Min KH. Epigenetic regulation of bone cells. Connect Tissue Res. 2017;58(1):76–89. https://doi.org/10.1080/03008207.2016.1177037.

    Article  CAS  PubMed  Google Scholar 

  20. •• Alzrigat M, Parraga AA, Jernberg-Wiklund H. Epigenetics in multiple myeloma: from mechanisms to therapy. Semin Cancer Biol. 2018;51:101–15. https://doi.org/10.1016/j.semcancer.2017.09.007 Well written reiview about epigenetic regulation and targeting in MM cells.

    Article  CAS  PubMed  Google Scholar 

  21. Hombach S, Kretz M. Non-coding RNAs: classification, biology and functioning. Adv Exp Med Biol. 2016;937:3–17. https://doi.org/10.1007/978-3-319-42059-2_1.

    Article  CAS  PubMed  Google Scholar 

  22. Carthew RW, Sontheimer EJ. Origins and mechanisms of miRNAs and siRNAs. Cell. 2009;136(4):642–55. https://doi.org/10.1016/j.cell.2009.01.035.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Marchese FP, Raimondi I, Huarte M. The multidimensional mechanisms of long noncoding RNA function. Genome Biol. 2017;18(1):206. https://doi.org/10.1186/s13059-017-1348-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Ferreira HJ, Esteller M. Non-coding RNAs, epigenetics, and cancer: tying it all together. Cancer Metastasis Rev. 2018;37(1):55–73. https://doi.org/10.1007/s10555-017-9715-8.

    Article  CAS  PubMed  Google Scholar 

  25. Caracciolo D, Montesano M, Altomare E, Scionti F, Di Martino MT, Tagliaferri P, et al. The potential role of miRNAs in multiple myeloma therapy. Expert Rev Hematol. 2018;11(10):793–803. https://doi.org/10.1080/17474086.2018.1517041.

    Article  CAS  PubMed  Google Scholar 

  26. Nobili L, Ronchetti D, Agnelli L, Taiana E, Vinci C, Neri A. Long non-coding RNAs in multiple myeloma. Gene. 2018;9(2). https://doi.org/10.3390/genes9020069.

  27. •• Adamik J, Jin S, Sun Q, Zhang P, Weiss KR, Anderson JL, et al. EZH2 or HDAC1 Inhibition reverses multiple myeloma-induced epigenetic suppression of osteoblast differentiation. Mol Cancer Res. 2017;15(4):405–17. https://doi.org/10.1158/1541-7786.MCR-16-0242-T One of the first studies demonstrating that chromatin repression of the Runx2 gene in MM-BMSCs contributes to their osteogenic suppression.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Adamik J, Silbermann R, Marino S, Sun Q, Anderson JL, Zhou D, et al. XRK3F2 inhibition of p62-ZZ domain signaling rescues myeloma-induced GFI1-driven epigenetic repression of the Runx2 gene in pre-osteoblasts to overcome differentiation suppression. Front Endocrinol. 2018;9:344. https://doi.org/10.3389/fendo.2018.00344.

    Article  Google Scholar 

  29. Dudakovic A, Camilleri ET, Xu F, Riester SM, McGee-Lawrence ME, Bradley EW, et al. Epigenetic control of skeletal development by the histone methyltransferase Ezh2. J Biol Chem. 2015;290(46):27604–17. https://doi.org/10.1074/jbc.M115.672345.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Fang C, Qiao Y, Mun SH, Lee MJ, Murata K, Bae S, et al. Cutting edge: EZH2 promotes osteoclastogenesis by epigenetic silencing of the negative regulator IRF8. J Immunol. 2016;196(11):4452–6. https://doi.org/10.4049/jimmunol.1501466.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Adamik J, Silbermann R, Lontos K, Zhang P, Sun Q, Galson DL, et al. EZH2 inhibitor GSK126 exhibits osteo-anabolic properties in MM bone disease and synergizes with Bortezomib to inhibit MM cell viability. Blood. 2016;128(22):3247.

    Google Scholar 

  32. Zeng D, Liu M, Pan J. Blocking EZH2 methylation transferase activity by GSK126 decreases stem cell-like myeloma cells. Oncotarget. 2017;8(2):3396–411. https://doi.org/10.18632/oncotarget.13773.

    Article  PubMed  Google Scholar 

  33. Harding T, Swanson J, Van Ness B. EZH2 inhibitors sensitize myeloma cell lines to panobinostat resulting in unique combinatorial transcriptomic changes. Oncotarget. 2018;9(31):21930–42. https://doi.org/10.18632/oncotarget.25128.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Yang D, Okamura H, Nakashima Y, Haneji T. Histone demethylase Jmjd3 regulates osteoblast differentiation via transcription factors Runx2 and osterix. J Biol Chem. 2013;288(47):33530–41. https://doi.org/10.1074/jbc.M113.497040.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Richardson PG, Schlossman RL, Alsina M, Weber DM, Coutre SE, Gasparetto C, et al. PANORAMA 2: panobinostat in combination with bortezomib and dexamethasone in patients with relapsed and bortezomib-refractory myeloma. Blood. 2013;122(14):2331–7. https://doi.org/10.1182/blood-2013-01-481325.

    Article  CAS  PubMed  Google Scholar 

  36. Xu S, De Veirman K, Evans H, Santini GC, Vande Broek I, Leleu X, et al. Effect of the HDAC inhibitor vorinostat on the osteogenic differentiation of mesenchymal stem cells in vitro and bone formation in vivo. Acta Pharmacol Sin. 2013;34(5):699–709. https://doi.org/10.1038/aps.2012.182.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Mithraprabhu S, Kalff A, Chow A, Khong T, Spencer A. Dysregulated class I histone deacetylases are indicators of poor prognosis in multiple myeloma. Epigenetics. 2014;9(11):1511–20. https://doi.org/10.4161/15592294.2014.983367.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Harada T, Oda A, Grondin Y, Teramachi J, Bat-Erdene A, Iwasa M, et al. The critical role of HDAC1-IRF4-Pim-2 axis in myeloma cell growth and survival: therapeutic impacts of targeting the HDAC1-IRF4-Pim-2 axis. Blood. 2018;132(Suppl 1):1939. https://doi.org/10.1182/blood-2018-99-114086.

    Article  Google Scholar 

  39. Harada T, Ohguchi H, Grondin Y, Kikuchi S, Sagawa M, Tai YT, et al. HDAC3 regulates DNMT1 expression in multiple myeloma: therapeutic implications. Leukemia. 2017;31(12):2670–7. https://doi.org/10.1038/leu.2017.144.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Ho M, Liu J, Kalbasi A, Harada T, Hideshima T, Dowling P, et al. Blocking HDAC3 in bone marrow stromal cells has direct anti-multiple myeloma effect and modulates T cell function. Blood. 2018;130(Suppl 1):4429.

    Google Scholar 

  41. Schroeder TM, Westendorf JJ. Histone deacetylase inhibitors promote osteoblast maturation. J Bone Miner Res. 2005;20(12):2254–63. https://doi.org/10.1359/JBMR.050813.

    Article  CAS  PubMed  Google Scholar 

  42. Stathis A, Bertoni F. BET proteins as targets for anticancer treatment. Cancer Discov. 2018;8(1):24–36. https://doi.org/10.1158/2159-8290.CD-17-0605.

    Article  CAS  PubMed  Google Scholar 

  43. Chaidos A, Caputo V, Gouvedenou K, Liu B, Marigo I, Chaudhry MS, et al. Potent antimyeloma activity of the novel bromodomain inhibitors I-BET151 and I-BET762. Blood. 2014;123(5):697–705. https://doi.org/10.1182/blood-2013-01-478420.

    Article  CAS  PubMed  Google Scholar 

  44. Stubbs MC, Burn TC, Sparks R, Maduskuie T, Diamond S, Rupar M, et al. The novel bromodomain and extraterminal domain inhibitor INCB054329 induces vulnerabilities in myeloma cells that inform rational combination strategies. Clin Cancer Res. 2018;25:300–11. https://doi.org/10.1158/1078-0432.CCR-18-0098.

    Article  PubMed  Google Scholar 

  45. Siegel MB, Liu SQ, Davare MA, Spurgeon SE, Loriaux MM, Druker BJ, et al. Small molecule inhibitor screen identifies synergistic activity of the bromodomain inhibitor CPI203 and bortezomib in drug resistant myeloma. Oncotarget. 2015;6(22):18921–32. https://doi.org/10.18632/oncotarget.4214.

    Article  PubMed  PubMed Central  Google Scholar 

  46. Diaz T, Rodriguez V, Lozano E, Mena MP, Calderon M, Rosinol L, et al. The BET bromodomain inhibitor CPI203 improves lenalidomide and dexamethasone activity in in vitro and in vivo models of multiple myeloma by blockade of Ikaros and MYC signaling. Haematologica. 2017;102(10):1776–84. https://doi.org/10.3324/haematol.2017.164632.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Shi J, Song S, Han H, Xu H, Huang M, Qian C, et al. Potent activity of the bromodomain inhibitor OTX015 in multiple myeloma. Mol Pharm. 2018;15(9):4139–47. https://doi.org/10.1021/acs.molpharmaceut.8b00554.

    Article  CAS  PubMed  Google Scholar 

  48. Raimondi L, De Luca A, Morelli E, Giavaresi G, Tagliaferri P, Tassone P, et al. MicroRNAs: novel crossroads between myeloma cells and the bone marrow microenvironment. Biomed Res Int. 2016;2016:6504593–12. https://doi.org/10.1155/2016/6504593.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Reagan MR, Mishima Y, Glavey SV, Zhang Y, Manier S, Lu ZN, et al. Investigating osteogenic differentiation in multiple myeloma using a novel 3D bone marrow niche model. Blood. 2014;124(22):3250–9. https://doi.org/10.1182/blood-2014-02-558007.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. De Veirman K, Wang J, Xu S, Leleu X, Himpe E, Maes K, et al. Induction of miR-146a by multiple myeloma cells in mesenchymal stromal cells stimulates their pro-tumoral activity. Cancer Lett. 2016;377(1):17–24. https://doi.org/10.1016/j.canlet.2016.04.024.

    Article  CAS  PubMed  Google Scholar 

  51. Cheng Q, Li X, Liu J, Ye Q, Chen Y, Tan S, et al. Multiple myeloma-derived exosomes regulate the functions of mesenchymal stem cells partially via modulating miR-21 and miR-146a. Stem Cells Int. 2017;2017:9012152–9. https://doi.org/10.1155/2017/9012152.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Pitari MR, Rossi M, Amodio N, Botta C, Morelli E, Federico C, et al. Inhibition of miR-21 restores RANKL/OPG ratio in multiple myeloma-derived bone marrow stromal cells and impairs the resorbing activity of mature osteoclasts. Oncotarget. 2015;6(29):27343–58. https://doi.org/10.18632/oncotarget.4398.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Pan J, Sun Y, Zhang N, Li J, Ta F, Wei W, et al. Characteristics of BAFF and APRIL factor expression in multiple myeloma and clinical significance. Oncol Lett. 2017;14(3):2657–62. https://doi.org/10.3892/ol.2017.6528.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Umezu T, Imanishi S, Azuma K, Kobayashi C, Yoshizawa S, Ohyashiki K, et al. Replenishing exosomes from older bone marrow stromal cells with miR-340 inhibits myeloma-related angiogenesis. Blood Adv. 2017;1(13):812–23. https://doi.org/10.1182/bloodadvances.2016003251.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Xu S, Cecilia Santini G, De Veirman K, Vande Broek I, Leleu X, De Becker A, et al. Upregulation of miR-135b is involved in the impaired osteogenic differentiation of mesenchymal stem cells derived from multiple myeloma patients. PLoS One. 2013;8(11):e79752. https://doi.org/10.1371/journal.pone.0079752.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Schaap-Oziemlak AM, Raymakers RA, Bergevoet SM, Gilissen C, Jansen BJ, Adema GJ, et al. MicroRNA hsa-miR-135b regulates mineralization in osteogenic differentiation of human unrestricted somatic stem cells. Stem Cells Dev. 2010;19(6):877–85. https://doi.org/10.1089/scd.2009.0112.

    Article  CAS  PubMed  Google Scholar 

  57. Hao M, Zang M, Zhao L, Deng S, Xu Y, Qi F, et al. Serum high expression of miR-214 and miR-135b as novel predictor for myeloma bone disease development and prognosis. Oncotarget. 2016;7(15):19589–600. https://doi.org/10.18632/oncotarget.7319.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Tsukamoto S, Lovendorf MB, Park J, Salem KZ, Reagan MR, Manier S, et al. Inhibition of microRNA-138 enhances bone formation in multiple myeloma bone marrow niche. Leukemia. 2018;32(8):1739–50. https://doi.org/10.1038/s41375-018-0161-6.

    Article  CAS  PubMed  Google Scholar 

  59. Rastgoo N, Pourabdollah M, Abdi J, Reece D, Chang H. Dysregulation of EZH2/miR-138 axis contributes to drug resistance in multiple myeloma by downregulating RBPMS. Leukemia. 2018;32:2471–82. https://doi.org/10.1038/s41375-018-0140-y.

    Article  CAS  PubMed  Google Scholar 

  60. Eskildsen T, Taipaleenmaki H, Stenvang J, Abdallah BM, Ditzel N, Nossent AY, et al. MicroRNA-138 regulates osteogenic differentiation of human stromal (mesenchymal) stem cells in vivo. Proc Natl Acad Sci U S A. 2011;108(15):6139–44. https://doi.org/10.1073/pnas.1016758108.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Liang J, Zhang Y, Jiang G, Liu Z, Xiang W, Chen X, et al. MiR-138 induces renal carcinoma cell senescence by targeting EZH2 and is downregulated in human clear cell renal cell carcinoma. Oncol Res. 2013;21(2):83–91. https://doi.org/10.3727/096504013X13775486749218.

    Article  CAS  PubMed  Google Scholar 

  62. Zhu Z, Tang J, Wang J, Duan G, Zhou L, Zhou X. MiR-138 acts as a tumor suppressor by targeting EZH2 and enhances cisplatin-induced apoptosis in osteosarcoma cells. PLoS One. 2016;11(3):e0150026. https://doi.org/10.1371/journal.pone.0150026.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. •• Berenstein R, Blau O, Nogai A, Waechter M, Slonova E, Schmidt-Hieber M, et al. Multiple myeloma cells alter the senescence phenotype of bone marrow mesenchymal stromal cells under participation of the DLK1-DIO3 genomic region. BMC Cancer. 2015;15:68. https://doi.org/10.1186/s12885-015-1078-3 One of the earlier studies demonstrating involvment of DNA hypomethylation and senescence in pathogenesis of BM-BMSCs.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Blau O, Berenstein R, Wächter M, Nogai A, Künel A, Bayanova M, et al. Molecular aberrations in bone marrow stromal cells in multiple myeloma. 2018. https://doi.org/10.5772/intechopen.77179.

  65. Guo J, Zhao Y, Fei C, Zhao S, Zheng Q, Su J, et al. Dicer1 downregulation by multiple myeloma cells promotes the senescence and tumor-supporting capacity and decreases the differentiation potential of mesenchymal stem cells. Cell Death Dis. 2018;9(5):512. https://doi.org/10.1038/s41419-018-0545-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Navarro A, Diaz T, Tovar N, Pedrosa F, Tejero R, Cibeira MT, et al. A serum microRNA signature associated with complete remission and progression after autologous stem-cell transplantation in patients with multiple myeloma. Oncotarget. 2015;6(3):1874–83. https://doi.org/10.18632/oncotarget.2761.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Shen X, Guo Y, Yu J, Qi J, Shi W, Wu X, et al. miRNA-202 in bone marrow stromal cells affects the growth and adhesion of multiple myeloma cells by regulating B cell-activating factor. Clin Exp Med. 2016;16(3):307–16. https://doi.org/10.1007/s10238-015-0355-4.

    Article  CAS  PubMed  Google Scholar 

  68. • Faict S, Muller J, De Veirman K, De Bruyne E, Maes K, Vrancken L, et al. Exosomes play a role in multiple myeloma bone disease and tumor development by targeting osteoclasts and osteoblasts. Blood Cancer J. 2018;8(11):105. https://doi.org/10.1038/s41408-018-0139-7 Study which nicely illustrates role for exosomes in MM-tumor microenvironment.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Huszar JM, Payne CJ. MIR146A inhibits JMJD3 expression and osteogenic differentiation in human mesenchymal stem cells. FEBS Lett. 2014;588(9):1850–6. https://doi.org/10.1016/j.febslet.2014.03.057.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Roodman GD. Pathogenesis of myeloma bone disease. J Cell Biochem. 2010;109(2):283–91. https://doi.org/10.1002/jcb.22403.

    Article  CAS  PubMed  Google Scholar 

  71. Pawlyn C, Bright MD, Buros AF, Stein CK, Walters Z, Aronson LI, et al. Overexpression of EZH2 in multiple myeloma is associated with poor prognosis and dysregulation of cell cycle control. Blood Cancer J. 2017;7(3):e549. https://doi.org/10.1038/bcj.2017.27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. •• McHugh D, Gil J. Senescence and aging: causes, consequences, and therapeutic avenues. J Cell Biol. 2018;217(1):65–77. https://doi.org/10.1083/jcb.201708092 Well written review about the topic of senescence.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Andre T, Meuleman N, Stamatopoulos B, De Bruyn C, Pieters K, Bron D, et al. Evidences of early senescence in multiple myeloma bone marrow mesenchymal stromal cells. PLoS One. 2013;8(3):e59756. https://doi.org/10.1371/journal.pone.0059756.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Ozcan S, Alessio N, Acar MB, Toprak G, Gonen ZB, Peluso G, et al. Myeloma cells can corrupt senescent mesenchymal stromal cells and impair their anti-tumor activity. Oncotarget. 2015;6(37):39482–92. https://doi.org/10.18632/oncotarget.5430.

    Article  PubMed  PubMed Central  Google Scholar 

  75. Li S, Jiang Y, Li A, Liu X, Xing X, Guo Y, et al. Telomere length is positively associated with the expression of IL6 and MIP1alpha in bone marrow mesenchymal stem cells of multiple myeloma. Mol Med Rep. 2017;16(3):2497–504. https://doi.org/10.3892/mmr.2017.6885.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Kanehira M, Fujiwara T, Nakajima S, Okitsu Y, Onishi Y, Fukuhara N, et al. An lysophosphatidic acid receptors 1 and 3 axis governs cellular senescence of mesenchymal stromal cells and promotes growth and vascularization of multiple myeloma. Stem Cells. 2017;35(3):739–53. https://doi.org/10.1002/stem.2499.

    Article  CAS  PubMed  Google Scholar 

  77. De Cauwer A, Mariotte A, Sibilia J, Bahram S, Georgel P. DICER1: a key player in rheumatoid arthritis, at the crossroads of cellular stress, innate immunity, and chronic inflammation in aging. Front Immunol. 2018;9:1647. https://doi.org/10.3389/fimmu.2018.01647.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Guo J, Li X, Zhao Y, Fei C, Chang C, Zhao S, et al. Reduction of Dicer1 by multiple myeloma cells in mesenchymal stem cells promotes cellular senescence and tumor-supporting effect and decreases the differentiation. Blood. 2017;130(Suppl 1):4399.

    Google Scholar 

  79. Malek E, Kim BG, Driscoll JJ. Identification of long non-coding RNAs deregulated in multiple myeloma cells resistant to proteasome inhibitors. Genes. 2016;7(10). https://doi.org/10.3390/genes7100084.

  80. Peng S, Cao L, He S, Zhong Y, Ma H, Zhang Y, et al. An overview of long noncoding RNAs involved in bone regeneration from mesenchymal stem cells. Stem Cells Int. 2018;2018:8273648–11. https://doi.org/10.1155/2018/8273648.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Zhang W, Dong R, Diao S, Du J, Fan Z, Wang F. Differential long noncoding RNA/mRNA expression profiling and functional network analysis during osteogenic differentiation of human bone marrow mesenchymal stem cells. Stem Cell Res Ther. 2017;8(1):30. https://doi.org/10.1186/s13287-017-0485-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Zhuang W, Ge X, Yang S, Huang M, Zhuang W, Chen P, et al. Upregulation of lncRNA MEG3 promotes osteogenic differentiation of mesenchymal stem cells from multiple myeloma patients by targeting BMP4 transcription. Stem Cells. 2015;33(6):1985–97. https://doi.org/10.1002/stem.1989.

    Article  CAS  PubMed  Google Scholar 

  83. Li Z, Jin C, Chen S, Zheng Y, Huang Y, Jia L, et al. Long non-coding RNA MEG3 inhibits adipogenesis and promotes osteogenesis of human adipose-derived mesenchymal stem cells via miR-140-5p. Mol Cell Biochem. 2017;433(1–2):51–60. https://doi.org/10.1007/s11010-017-3015-z.

    Article  CAS  PubMed  Google Scholar 

  84. Li B, Chen P, Qu J, Shi L, Zhuang W, Fu J, et al. Activation of LTBP3 gene by a long noncoding RNA (lncRNA) MALAT1 transcript in mesenchymal stem cells from multiple myeloma. J Biol Chem. 2014;289(42):29365–75. https://doi.org/10.1074/jbc.M114.572693.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Li B, Han H, Song S, Fan G, Xu H, Zhou W, et al. HOXC10 regulates osteogenesis of mesenchymal stromal cells through interaction with its natural antisense transcript lncHOXC-AS3. Stem Cells. 2018. https://doi.org/10.1002/stem.2925.

  86. Xie X, Xiao Y, Huang X. Homeobox C10 knockdown suppresses cell proliferation and promotes cell apoptosis in osteosarcoma cells through regulating caspase 3. Blood Adv. 2018;11:473–82. https://doi.org/10.2147/OTT.S143440.

    Article  Google Scholar 

  87. Li B, Xu H, Han H, Song S, Zhang X, Ouyang L, et al. Exosome-mediated transfer of lncRUNX2-AS1 from multiple myeloma cells to MSCs contributes to osteogenesis. Oncogene. 2018;37(41):5508–19. https://doi.org/10.1038/s41388-018-0359-0.

    Article  CAS  PubMed  Google Scholar 

  88. Terpos E, Christoulas D, Gavriatopoulou M, Dimopoulos MA. Mechanisms of bone destruction in multiple myeloma. Eur J Cancer Care. 2017;26(6). https://doi.org/10.1111/ecc.12761.

  89. •• Schinke C, Qu P, Mehdi SJ, Hoering A, Epstein J, Johnson SK, et al. The pattern of mesenchymal stem cell expression is an independent marker of outcome in multiple myeloma. Clin Cancer Res. 2018;24(12):2913–9. https://doi.org/10.1158/1078-0432.CCR-17-2627 This paper defines MM-BMSC-specific gene signatures used for scoring MM stages and predictions of progression free survival outcome.

    Article  CAS  PubMed  Google Scholar 

  90. Xu S, De Veirman K, De Becker A, Vanderkerken K, Van Riet I. Mesenchymal stem cells in multiple myeloma: a therapeutical tool or target? Leukemia. 2018;32(7):1500–14. https://doi.org/10.1038/s41375-018-0061-9.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Juraj Adamik.

Ethics declarations

Conflict of Interest

The authors do not have potential conflict of interest that impacts this manuscript.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

This article is part of the Topical Collection on Molecular Biology of Bone Metastasis

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pulugulla, S.H., Adamik, J. Epigenetics of Multiple Myeloma Bone Disease. Curr Mol Bio Rep 5, 86–96 (2019). https://doi.org/10.1007/s40610-019-0117-2

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40610-019-0117-2

Keywords

Navigation