DEPTOR maintains plasma cell differentiation and favorably affects prognosis in multiple myeloma

Dalia Quwaider, Luis A Corchete, Irena Misiewicz-Krzeminska, María E Sarasquete, José J Pérez, Patryk Krzeminski, Noemí Puig, María Victoria Mateos, Ramón García-Sanz, Ana B Herrero, Norma C Gutiérrez, Dalia Quwaider, Luis A Corchete, Irena Misiewicz-Krzeminska, María E Sarasquete, José J Pérez, Patryk Krzeminski, Noemí Puig, María Victoria Mateos, Ramón García-Sanz, Ana B Herrero, Norma C Gutiérrez

Abstract

Background: The B cell maturation process involves multiple steps, which are controlled by relevant pathways and transcription factors. The understanding of the final stages of plasma cell (PC) differentiation could provide new insights for therapeutic strategies in multiple myeloma (MM). Here, we explore the role of DEPTOR, an mTOR inhibitor, in the terminal differentiation of myeloma cells, and its potential impact on patient survival.

Methods: The expression level of DEPTOR in MM cell lines and B cell populations was measured by real-time RT-PCR, and/or Western blot analysis. DEPTOR protein level in MM patients was quantified by capillary electrophoresis immunoassay. RNA interference was used to downregulate DEPTOR in MM cell lines.

Results: DEPTOR knockdown in H929 and MM1S cell lines induced dedifferentiation of myeloma cells, as demonstrated by the upregulation of PAX5 and BCL6, the downregulation of IRF4, and a clear reduction in cell size and endoplasmic reticulum mass. This effect seemed to be independent of mTOR signaling, since mTOR substrates were not affected by DEPTOR knockdown. Additionally, the potential for DEPTOR to be deregulated in MM by particular miRNAs was investigated. The ectopic expression of miR-135b and miR-642a in myeloma cell lines substantially diminished DEPTOR protein levels, and caused dedifferentiation of myeloma cells. Interestingly, the level of expression of DEPTOR protein in myeloma patients was highly variable, the highest levels being associated with longer progression-free survival.

Conclusions: Our results demonstrate for the first time that DEPTOR expression is required to maintain myeloma cell differentiation and that high level of its expression are associated with better outcome. Primary samples used in this study correspond to patients entered into GEM2010 trial (registered at www.clinicaltrials.gov as #NCT01237249, 4 November 2010).

Keywords: B lymphocyte differentiation; DEPTOR; Multiple myeloma; Plasma cell development; miRNAs.

Figures

Fig. 1
Fig. 1
DEPTOR expression. aDEPTOR expression in normal B lymphocytes (NBL), normal PC (NPC), and myeloma cells obtained from gene expression arrays (GSE6691 at GEO repository). bDEPTOR expression detected by qRT-PCR in immature B cells, naive B cells, memory B cells, and NPC. (*p ˂ 0.05, **p ˂ 0.01, ***p ˂ 0.001)
Fig. 2
Fig. 2
DEPTOR knockdown modulates transcription factors and markers related to B cell maturation. a mRNA levels of the indicated genes in H929 and MM1S determined by qRT-PCR 48 h after DEPTOR knockdown. b Western blot of DEPTOR, IRF4, and kappa and lambda light chain in H929 and MM1S. c Flow cytometry analysis of CD38, CD138, and cytoplasmic kappa light chain in H929 after DEPTOR knockdown (siDEPTOR) compared with non-targeting control (siNT); left panel: representative histograms; right panel: mean fluorescence intensity values (MFI). All results are presented as the means ± SD of three different experiments. (*p ˂ 0.05, **p ˂ 0.01, ***p ˂ 0.001)
Fig. 3
Fig. 3
DEPTOR knockdown induces dedifferentiation of myeloma cells. a Giemsa stain of H929 and MM1S after DEPTOR knockdown. b Immunofluorescence of H929 and MM1S cells stained with ER-Tracker™ Red dye. c Average maximum diameter of H929 and MM1S transfected with non-targeting (siNT) and DEPTOR siRNA (siDEPTOR) measured from two independent experiments. At least 100 cells per experiment were counted. d Cell cycle analysis 48 h after DEPTOR knockdown. The percentages of cells in different phases of the cell cycle are indicated. e Cell size of H929 48 h after DEPTOR knockdown. Representative dot plots showing forward-scatter (FSC) versus side-scatter (SSC). All results are presented as the means ± SD of three different experiments. (*p ˂ 0.05, **p ˂ 0.01, ***p ˂ 0.001)
Fig. 4
Fig. 4
Dedifferentiation of myeloma cells after DEPTOR silencing is independent of mTOR signaling. a Levels of mTORC1 and mTORC2 substrates after DEPTOR knockdown in H929 and MM1S. All experiments were carried out 48 h post-transfection. b Western blot of H929 transfected with siNT or siDEPTOR and cultured with control medium and rapamycin at 10 nM
Fig. 5
Fig. 5
DEPTOR expression is controlled by miR135b and miR642a in MM. aDEPTOR mRNA levels measured by qRT-PCR (upper panel). Levels of DEPTOR in MM cell lines detected by Western blot (lower panel). b miR135b and miR642a expression in NPC and MM patients measured by qRT-PCR. microRNA expression was normalized to RNU43 (2−Δct). Two-tailed Welch’s t test p values for miR135b and miR642a were calculated (0.0081 and 0.0001, respectively). c Schematic diagram of the miR642a and miR135b predicted site on the DEPTOR 3′UTR, dark squares on DEPTOR 3′UTR WT, and DEPTOR 3′UTR MUT represents fragments cloned in pmiRGlo plasmid. d Luciferase activity in HEK293 cells cotransfected with pre-miR-NC or pre-miR135b/ miR642a and plasmid pmiR-Glo with the putative miR135b/miR642a binding site of DEPTOR cloned downstream of the luciferase reporter gene. Luciferase activity was normalized using Renilla. All results are presented as the means ± SD of three different experiments. (*p ˂ 0.05, **p ˂ 0.01, ***p ˂ 0.001)
Fig. 6
Fig. 6
miR642a and miR135b regulate DEPTOR expression in MM. a miR-135b and miR-642a expression in MM cell lines measured by qRT-PCR. microRNA expression was normalized with respect to RNU43 (2−Δct). b Western blot of DEPTOR in H929 cells 72 h post-transfection with NC or miR-135b/miR-642a. c Western blot of DEPTOR in U266 and JJN3 transfected with NC or miR135b/miR642a inhibitors. (*p ˂ 0.05, **p ˂ 0.01, ***p ˂ 0.001)
Fig. 7
Fig. 7
Upregulation of miR135b and miR642a results in myeloma cell dedifferentiation through negative regulation of DEPTOR. a Western blot of DEPTOR, IRF4, and kappa light chain in H929 cells 72 h post-transfection with NC or miR-135b/miR-642a. b Giemsa stain of H929 cells transfected with NC or miR-135b/miR-642a. c Immunofluorescence of H929 cells stained with ER tracker
Fig. 8
Fig. 8
DEPTOR is differentially expressed in MM and its overexpression is associated with longer survival. a Levels of DEPTOR in MM patients assessed using capillary electrophoresis immunoassay. b Kaplan–Meier curves for progression-free survival (PFS) in 24 MM patients treated according to GEM2010

References

    1. Kyle RA, Rajkumar SV. Multiple myeloma. N Engl J Med. 2004;351:1860–73. doi: 10.1056/NEJMra041875.
    1. Somasundaram R, Prasad MA, Ungerback J, Sigvardsson M. Transcription factor networks in B-cell differentiation link development to acute lymphoid leukemia. Blood. 2015;126:144–52. doi: 10.1182/blood-2014-12-575688.
    1. Familiades J, Bousquet M, Lafage-Pochitaloff M, Béné M-C, Beldjord K, De Vos J, et al. PAX5 mutations occur frequently in adult B-cell progenitor acute lymphoblastic leukemia and PAX5 haploinsufficiency is associated with BCR-ABL1 and TCF3-PBX1 fusion genes: a GRAALL study. Leukemia. 2009;23:1989–98. doi: 10.1038/leu.2009.135.
    1. Traverse-Glehen A, Verney A, Baseggio L, Felman P, Callet-Bauchu E, Thieblemont C, et al. Analysis of BCL-6, CD95, PIM1, RHO/TTF and PAX5 mutations in splenic and nodal marginal zone B-cell lymphomas suggests a particular B-cell origin. Leukemia. 2007;21:1821–4. doi: 10.1038/sj.leu.2404706.
    1. Kirk SJ, Cliff JM, Thomas JA, Ward TH. Biogenesis of secretory organelles during B cell differentiation. J Leukoc Biol. 2010;87:245–55. doi: 10.1189/jlb.1208774.
    1. King LB, Corley RB. Characterization of a presecretory phase in B-cell differentiation. Proc Natl Acad Sci U S A. 1989;86:2814–8. doi: 10.1073/pnas.86.8.2814.
    1. Jourdan M, Caraux A, Caron G, Robert N, Fiol G, Reme T, et al. Characterization of a transitional preplasmablast population in the process of human B cell to plasma cell differentiation. J Immunol. 2011;187:3931–41. doi: 10.4049/jimmunol.1101230.
    1. Gass JN, Gunn KE, Sriburi R, Brewer JW. Stressed-out B cells? Plasma-cell differentiation and the unfolded protein response. Trends Immunol. 2004;25:17–24. doi: 10.1016/j.it.2003.11.004.
    1. Nutt SL, Taubenheim N, Hasbold J, Corcoran LM, Hodgkin PD. The genetic network controlling plasma cell differentiation. Semin Immunol. 2011;23:341–9. doi: 10.1016/j.smim.2011.08.010.
    1. Calame KL, Lin KI, Tunyaplin C. Regulatory mechanisms that determine the development and function of plasma cells. Annu Rev Immunol. 2003;21:205–30. doi: 10.1146/annurev.immunol.21.120601.141138.
    1. Shapiro-Shelef M, Calame K. Regulation of plasma-cell development. Nat Rev. 2005;5:230–42.
    1. Nutt SL, Hodgkin PD, Tarlinton DM, Corcoran LM. The generation of antibody-secreting plasma cells. Nat Rev. 2015;15:160–71.
    1. Peterson TR, Laplante M, Thoreen CC, Sancak Y, Kang SA, Kuehl WM, et al. DEPTOR is an mTOR inhibitor frequently overexpressed in multiple myeloma cells and required for their survival. Cell. 2009;137:873–86. doi: 10.1016/j.cell.2009.03.046.
    1. Gutierrez NC, Ocio EM, de Las Rivas J, Maiso P, Delgado M, Ferminan E, et al. Gene expression profiling of B lymphocytes and plasma cells from Waldenstrom’s macroglobulinemia: comparison with expression patterns of the same cell counterparts from chronic lymphocytic leukemia, multiple myeloma and normal individuals. Leukemia. 2007;21:541–9. doi: 10.1038/sj.leu.2404520.
    1. Catena V, Fanciulli M. Deptor: not only a mTOR inhibitor. J Exp Clin Cancer Res CR. 2017;36:12. doi: 10.1186/s13046-016-0484-y.
    1. Wang Z, Zhong J, Inuzuka H, Gao D, Shaik S, Sarkar FH, et al. An evolving role for DEPTOR in tumor development and progression. Neoplasia N Y N. 2012;14:368–75. doi: 10.1593/neo.12542.
    1. Carrasco DR, Tonon G, Huang Y, Zhang Y, Sinha R, Feng B, et al. High-resolution genomic profiles define distinct clinico-pathogenetic subgroups of multiple myeloma patients. Cancer Cell. 2006;9:313–25. doi: 10.1016/j.ccr.2006.03.019.
    1. Misiewicz-Krzeminska I, Sarasquete ME, Quwaider D, Krzeminski P, Ticona FV, Paino T, et al. Restoration of microRNA-214 expression reduces growth of myeloma cells through positive regulation of P53 and inhibition of DNA replication. Haematologica. 2013;98:640–8. doi: 10.3324/haematol.2012.070011.
    1. Budczies J, Klauschen F, Sinn BV, Gyorffy B, Schmitt WD, Darb-Esfahani S, et al. Cutoff Finder: a comprehensive and straightforward Web application enabling rapid biomarker cutoff optimization. PLoS One. 2012;7:e51862. doi: 10.1371/journal.pone.0051862.
    1. Klein U, Casola S, Cattoretti G, Shen Q, Lia M, Mo T, et al. Transcription factor IRF4 controls plasma cell differentiation and class-switch recombination. Nat Immunol. 2006;7:773–82. doi: 10.1038/ni1357.
    1. Hurt EM, Wiestner A, Rosenwald A, Shaffer AL, Campo E, Grogan T, et al. Overexpression of c-maf is a frequent oncogenic event in multiple myeloma that promotes proliferation and pathological interactions with bone marrow stroma. Cancer Cell. 2004;5:191–9. doi: 10.1016/S1535-6108(04)00019-4.
    1. Eychene A, Rocques N, Pouponnot C. A new MAFia in cancer. Nat Rev. 2008;8:683–93. doi: 10.1038/nrc2460.
    1. Gabrea A, Bergsagel PL, Chesi M, Shou Y, Kuehl WM. Insertion of excised IgH switch sequences causes overexpression of cyclin D1 in a myeloma tumor cell. Mol Cell. 1999;3:119–23. doi: 10.1016/S1097-2765(00)80180-X.
    1. Herath NI, Rocques N, Garancher A, Eychene A, Pouponnot C. GSK3-mediated MAF phosphorylation in multiple myeloma as a potential therapeutic target. Blood Cancer J. 2014;4:e175. doi: 10.1038/bcj.2013.67.
    1. Lombardi L, Poretti G, Mattioli M, Fabris S, Agnelli L, Bicciato S, et al. Molecular characterization of human multiple myeloma cell lines by integrative genomics: insights into the biology of the disease. Genes Chromosomes Cancer. 2007;46:226–38. doi: 10.1002/gcc.20404.
    1. Chang H, Qi Q, Xu W, Patterson B. c-Maf nuclear oncoprotein is frequently expressed in multiple myeloma. Leukemia. 2007;21:1572–4. doi: 10.1038/sj.leu.2404669.
    1. Gutierrez NC, Sarasquete ME, Misiewicz-Krzeminska I, Delgado M, Rivas JDL, Ticona FV, et al. Deregulation of microRNA expression in the different genetic subtypes of multiple myeloma and correlation with gene expression profiling. Leukemia. 2010;24:629–37. doi: 10.1038/leu.2009.274.
    1. Boyd KD, Walker BA, Wardell CP, Ross FM, Gregory WM, Davies FE, et al. High expression levels of the mammalian target of rapamycin inhibitor DEPTOR are predictive of response to thalidomide in myeloma. Leuk Lymphoma. 2010;51:2126–9. doi: 10.3109/10428194.2010.509893.
    1. Todoerti K, Agnelli L, Fabris S, Lionetti M, Tuana G, Mosca L, et al. Transcriptional characterization of a prospective series of primary plasma cell leukemia revealed signatures associated with tumor progression and poorer outcome. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2013;19:3247–58.
    1. Goasguen JE, Zandecki M, Mathiot C, Scheiff JM, Bizet M, Ly-Sunnaram B, et al. Mature plasma cells as indicator of better prognosis in multiple myeloma. New methodology for the assessment of plasma cell morphology. Leuk Res. 1999;23:1133–40. doi: 10.1016/S0145-2126(99)00132-0.
    1. Shaffer AL, Lin KI, Kuo TC, Yu X, Hurt EM, Rosenwald A, et al. Blimp-1 orchestrates plasma cell differentiation by extinguishing the mature B cell gene expression program. Immunity. 2002;17:51–62. doi: 10.1016/S1074-7613(02)00335-7.
    1. Jr JEF. Self-perpetuating states in signal transduction: positive feedback, double-negative feedback and bistability. Curr Opin Cell Biol. 2002;14:140–8. doi: 10.1016/S0955-0674(02)00314-9.
    1. Monod J, Jacob F. Teleonomic mechanisms in cellular metabolism, growth, and differentiation. Cold Spring Harb Symp Quant Biol. 1961;26:389–401. doi: 10.1101/SQB.1961.026.01.048.
    1. Leung-Hagesteijn C, Erdmann N, Cheung G, Keats JJ, Stewart AK, Reece DE, et al. Xbp1s-negative tumor B cells and pre-plasmablasts mediate therapeutic proteasome inhibitor resistance in multiple myeloma. Cancer Cell. 2013;24:289–304. doi: 10.1016/j.ccr.2013.08.009.
    1. van Keimpema M, Gruneberg LJ, Mokry M, van Boxtel R, van Zelm MC, Coffer P, et al. The forkhead transcription factor FOXP1 represses human plasma cell differentiation. Blood. 2015;126:2098–109. doi: 10.1182/blood-2015-02-626176.
    1. Fujita N, Jaye DL, Geigerman C, Akyildiz A, Mooney MR, Boss JM, et al. MTA3 and the Mi-2/NuRD complex regulate cell fate during B lymphocyte differentiation. Cell. 2004;119:75–86. doi: 10.1016/j.cell.2004.09.014.
    1. Mikkola I, Heavey B, Horcher M, Busslinger M. Reversion of B cell commitment upon loss of Pax5 expression. Science. 2002;297:110–3. doi: 10.1126/science.1067518.
    1. Bettigole SE, Glimcher LH. Endoplasmic reticulum stress in immunity. Annu Rev Immunol. 2015;33:107–38. doi: 10.1146/annurev-immunol-032414-112116.
    1. Agrawal P, Reynolds J, Chew S, Lamba DA, Hughes RE. DEPTOR is a stemness factor that regulates pluripotency of embryonic stem cells. J Biol Chem. 2014;289:31818–26. doi: 10.1074/jbc.M114.565838.
    1. Powell JD, Delgoffe GM. The mammalian target of rapamycin: linking T cell differentiation, function, and metabolism. Immunity. 2010;33:301–11. doi: 10.1016/j.immuni.2010.09.002.
    1. Chapman NM, Chi H. mTOR links environmental signals to T cell fate decisions. Front Immunol. 2015;5:686. doi: 10.3389/fimmu.2014.00686.
    1. Benhamron S, Tirosh B. Direct activation of mTOR in B lymphocytes confers impairment in B-cell maturation andloss of marginal zone B cells. Eur J Immunol. 2011;41:2390–6. doi: 10.1002/eji.201041336.
    1. Pichiorri F, Suh SS, Ladetto M, Kuehl M, Palumbo T, Drandi D, et al. MicroRNAs regulate critical genes associated with multiple myeloma pathogenesis. Proc Natl Acad Sci U S A. 2008;105:12885–90. doi: 10.1073/pnas.0806202105.
    1. Lionetti M, Biasiolo M, Agnelli L, Todoerti K, Mosca L, Fabris S, et al. Identification of microRNA expression patterns and definition of a microRNA/mRNA regulatory network in distinct molecular groups of multiple myeloma. Blood. 2009;114:e20–6. doi: 10.1182/blood-2009-08-237495.
    1. Kawano Y, Fujiwara S, Wada N, Izaki M, Yuki H, Okuno Y, et al. Multiple myeloma cells expressing low levels of CD138 have an immature phenotype and reduced sensitivity to lenalidomide. Int J Oncol. 2012;41:876–84.
    1. Kuroda Y, Sakai A, Okikawa Y, Munemasa S, Katayama Y, Hyodo H, et al. The maturation of myeloma cells correlates with sensitivity to chemotherapeutic agents. Int J Hematol. 2005;81:335–41. doi: 10.1532/IJH97.04189.
    1. Iriyama N, Miura K, Hatta Y, Uchino Y, Kurita D, Takahashi H, et al. Plasma cell maturity as a predictor of prognosis in multiple myeloma. Med Oncol. 2016;33:87. doi: 10.1007/s12032-016-0803-3.

Source: PubMed

3
Abonnieren