MEF2 plays a significant role in the tumor inhibitory mechanism of encapsulated RENCA cells via EGF receptor signaling in target tumor cells

Prithy C Martis, Atira T Dudley, Melissa A Bemrose, Hunter L Gazda, Barry H Smith, Lawrence S Gazda, Prithy C Martis, Atira T Dudley, Melissa A Bemrose, Hunter L Gazda, Barry H Smith, Lawrence S Gazda

Abstract

Background: Agarose encapsulated murine renal adenocarcinoma cells (RENCA macrobeads) are currently being investigated in clinical trials as a treatment for therapy-resistant metastatic colorectal cancer. We have previously demonstrated the capacity of RENCA macrobeads to produce diffusible substances that markedly inhibit the proliferation of epithelial-derived tumor cells outside the macrobead environment. This study examined the molecular mechanisms underlying the observed inhibition in targeted tumor cells exposed to RENCA macrobeads.

Methods: We evaluated changes in transcription factor responses, participating intracellular signaling pathways and the involvement of specific cellular receptors in targeted tumor cells exposed to RENCA macrobeads.

Results: Factors secreted by RENCA macrobeads significantly up-regulated the activity of the MEF2 transcription factor as well as altered the transcription of MEF2b and MEF2d isoforms in targeted tumor cells. Suppression of individual or multiple MEF2 isoforms in target tumor cells markedly reduced the growth inhibitory effects of RENCA macrobeads. Furthermore, these effects were linked to the activation of the EGF receptor as attenuation of EGFR resulted in a substantial reduction of the cancer cell growth-inhibitory effect.

Conclusions: Since interruption of the EGFR signaling cascade did not eliminate RENCA macrobead-induced growth control, our data suggests that RENCA macrobeads exert their full growth inhibitory effects through the simultaneous activation of multiple signaling pathways. In contrast to a precision medicine approach targeting single molecular abnormalities, the RENCA macrobead functions as a biological-systems therapy to re-establish regulation in a highly dysfunctional and dysregulated cancer system.

Keywords: Cell therapy; EGFR; MEF2; Tumor inhibition.

Conflict of interest statement

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

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

Figures

Fig. 1
Fig. 1
Factors secreted by RENCA macrobeads alter the transcription factor activity and expression of MEF2. (a) RENCA cells, transiently transfected with pathway-focused transcription factor-responsive luciferase reporter constructs were exposed to naïve media or 5-day conditioned media from > 18 wk. RENCA macrobeads. Fold-change, calculated based on normalized luciferase activity of the conditioned media (CM) response relative to the naïve media response, is graphed in descending order. Columns, mean (n = 3); bar, SD. Dotted lines at 2 and 0.5 on the y-axis indicate the threshold for two-fold up- and down-regulation respectively. (b) MEF2 reporter activity in RENCA cells exposed to 5-day RENCA macrobead-conditioned media. Reporter activity in response to naïve media was used as a control. Fold-change was calculated for each sample relative to the naïve media sample. Each column represents the mean (n = 6–8) ± SD (primary axis). Mean inhibitory response of RENCA macrobeads on freely growing RENCA cells; red circles (n = 3) ± SD (secondary axis). (c) RENCA cells were co-cultured with > 18 wk. RENCA macrobeads for 5 days. RENCA cells cultured in naïve media served as a control. Total RNA was isolated and subjected to real-time PCR analysis for the expression of MEF2a, MEF2b, MEF2c and MEF2d. MEF2c was not detected in RENCA cells. Each column represents the mean (n = 3) ± SD. *p < 0.001, compared with naïve media
Fig. 2
Fig. 2
MEF2 is integral in mediating the anti-proliferative effect of RENCA macrobeads. RENCA cells were transiently transfected with two-rounds of 1 μM non-targeting siRNA, MEF2a, MEF2b, MEF2d or combined MEF2a, b and d siRNAs (MEF2 pool) for 72 h followed by culture in naïve media or together with > 18 wk. RENCA macrobeads in a cell culture insert system for 5 days. Each column represents the mean absorbance value (n = 3) ± SD following neutral red staining, with growth inhibition calculated as the percent difference in absorbance between the indicated conditions. *p < 0.005 compared with naïve media
Fig. 3
Fig. 3
RENCA macrobeads regulate the epidermal growth factor receptor in both murine and human cell lines. In-cell Western analysis of total EGFR, phosphorylated EGFR (pY1068) and overlay (normalized pY1068/EGFR) in (a) RENCA cells, and (b) DU145 cells following exposure to 5-day conditioned media (CM) from RENCA macrobeads at 1–2 weeks or > 18 weeks of age as indicated. Cells cultured in naïve media were used as a control. Each column represents the mean normalized intensity in arbitrary units (AU) (n = 6–8) ± SD. *p < 0.001, compared with naïve media
Fig. 4
Fig. 4
Gefitinib limits basal and RENCA macrobead-mediated EGFR activity and contributes to an additive inhibitory effect in DU145 cells. In-cell Western analysis of total EGFR, phosphorylated EGFR (pY1068) and overlay (normalized pY1068/EGFR) in DU145 cells pre-treated with gefitinib (GF) or 0.01% DMSO for 2 h at the indicated concentrations followed by culture in (a) naïve media, or (b) 5-day conditioned media from > 18 wk. RENCA macrobeads. Each column represents the mean normalized intensity in arbitrary units (AU) (n = 6–8) ± SD. *p < 0.001, compared with vehicle control. (c) DU145 cells pre-treated with gefitinib or 0.01% DMSO were cultured with naïve media or together with > 18 wk. RENCA macrobeads in a cell culture insert system for 5 days. Cells cultured in naïve media were used as a control. Histogram represents percent survival calculated as the percent absorbance of co-cultured samples relative to naïve media samples. Each column represents the mean survival (n = 3) ± SD
Fig. 5
Fig. 5
EGFR blockade results in partial attenuation of RENCA macrobead-mediated growth inhibition. (a) In-cell Western analysis of total EGFR, phosphorylated EGFR (pY1068) and overlay (normalized pY1068/EGFR) in DU145/GR cells pre-treated with 1 μM gefitinib (GF) or 0.01% DMSO followed by culture in naïve or 5-day conditioned media from > 18 wk. RENCA macrobeads. Each column represents the mean normalized intensity in arbitrary units (AU) (n = 6–8) ± SD. *p < 0.001, compared with vehicle control or conditioned media. (b) DU145/GR cells pre-treated with 1 μM gefitinib or 0.01% DMSO were cultured with naïve media or together with > 18 wk. RENCA macrobeads in a cell culture insert system for 5 days. Histogram represents percent survival calculated as the percent absorbance of co-cultured samples relative to naïve media samples. Each column represents the mean survival (n = 3) ± SD
Fig. 6
Fig. 6
RENCA macrobeads modulate the expression of MEF2 isoforms through EGF receptor signaling. MEF2A, MEF2B, MEF2C and MEF2D expression was assessed by qRT-PCR on (a) DU145 or (b) DU145/GR cells cultured in naïve media or together with > 18 wk. RENCA macrobeads alone or pre-treated with 1 μM gefitinib followed by culture in media as described for 5 days. MEF2B expression was not detected in DU145 or DU145/GR cells. Each column represents the mean (n = 3) ± SD. *p < 0.005, compared with naïve media or gefitinib in naïve media

References

    1. Fialkow PJ. Clonal origin of human tumors. Annu Rev Med. 1979;30:135–143. doi: 10.1146/annurev.me.30.020179.001031.
    1. Burrell RA, McGranahan N, Bartek J, Swanton C. The causes and consequences of genetic heterogeneity in cancer evolution. Nature. 2013;501(7467):338–345. doi: 10.1038/nature12625.
    1. Haber DA, Bell DW, Sordella R, Kwak EL, Godin-Heymann N, Sharma SV, et al. Molecular targeted therapy of lung cancer: EGFR mutations and response to EGFR inhibitors. Cold Spring Harb Symp Quant Biol. 2005;70:419–426. doi: 10.1101/sqb.2005.70.043.
    1. Grigsby PW, Herr HW. Urethral tumors. In: Vogelsang NJ, Scardino, PT, Shipley WU et al., editors. Comprehensive textbook of genitourinary oncology. Baltimore: Williams & Wilkins; 1996. p. 1117–1123.
    1. Pui CH, Pei D, Campana D, Cheng C, Sandlund JT, Bowman WP, et al. A revised definition for cure of childhood acute lymphoblastic leukemia. Leukemia. 2014;28(12):2336–2343. doi: 10.1038/leu.2014.142.
    1. Sawyers C. Targeted cancer therapy. Nature. 2004;432(7015):294–297. doi: 10.1038/nature03095.
    1. Spano D, Heck C, De Antonellis P, Christofori G, Zollo M. Molecular networks that regulate cancer metastasis. Semin Cancer Biol. 2012;22(3):234–249. doi: 10.1016/j.semcancer.2012.03.006.
    1. Smith BH, Gazda LS, Conn BL, Jain K, Asina S, Levine DM, et al. Hydrophilic agarose macrobead cultures select for outgrowth of carcinoma cell populations that can restrict tumor growth. Cancer Res. 2011;71(3):725–735. doi: 10.1158/0008-5472.CAN-10-2258.
    1. Kastenhuber ER, Lowe SW. Putting p53 in context. Cell. 2017;170(6):1062–1078. doi: 10.1016/j.cell.2017.08.028.
    1. Mills KD. Tumor suppression: putting p53 in context. Cell Cycle. 2013;12(22):3461–3462. doi: 10.4161/cc.26806.
    1. Jain K, Yang H, Cai BR, Haque B, Hurvitz AI, Diehl C, et al. Retrievable, replaceable, macroencapsulated pancreatic islet xenografts. Long-term engraftment without immunosuppression. Transplantation. 1995;59(3):319–324. doi: 10.1097/00007890-199502000-00002.
    1. Dumpala PR, Holdcraft RW, Martis PC, Laramore MA, Parker TS, Levine DM, et al. Retention of gene expression in porcine islets after agarose encapsulation and long-term culture. Biochem Biophys Res Commun. 2016;476(4):580–585. doi: 10.1016/j.bbrc.2016.05.165.
    1. Gazda LS, Martis PC, Laramore MA, Bautista MA, Dudley A, Vinerean HV, et al. Treatment of agarose-agarose RENCA macrobeads with docetaxel selects for OCT4(+) cells with tumor-initiating capability. Cancer Biol Ther. 2013;14(12):1147–1157. doi: 10.4161/cbt.26455.
    1. Wakeling AE, Guy SP, Woodburn JR, Ashton SE, Curry BJ, Barker AJ, et al. ZD1839 (Iressa): an orally active inhibitor of epidermal growth factor signaling with potential for cancer therapy. Cancer Res. 2002;62(20):5749–5754.
    1. Jones HE, Goddard L, Gee JM, Hiscox S, Rubini M, Barrow D, et al. Insulin-like growth factor-I receptor signalling and acquired resistance to gefitinib (ZD1839; Iressa) in human breast and prostate cancer cells. Endocr Relat Cancer. 2004;11(4):793–814. doi: 10.1677/erc.1.00799.
    1. Di Giorgio E, Clocchiatti A, Piccinin S, Sgorbissa A, Viviani G, Peruzzo P, et al. MEF2 is a converging hub for histone deacetylase 4 and phosphatidylinositol 3-kinase/Akt-induced transformation. Mol Cell Biol. 2013;33(22):4473–4491. doi: 10.1128/MCB.01050-13.
    1. Potthoff MJ, Olson EN. MEF2: a central regulator of diverse developmental programs. Development. 2007;134(23):4131–4140. doi: 10.1242/dev.008367.
    1. Wilker PR, Kohyama M, Sandau MM, Albring JC, Nakagawa O, Schwarz JJ, et al. Transcription factor Mef2c is required for B cell proliferation and survival after antigen receptor stimulation. Nat Immunol. 2008;9(6):603–612. doi: 10.1038/ni.1609.
    1. Ying CY, Dominguez-Sola D, Fabi M, Lorenz IC, Hussein S, Bansal M, et al. MEF2B mutations lead to deregulated expression of the oncogene BCL6 in diffuse large B cell lymphoma. Nat Immunol. 2013;14(10):1084–1092. doi: 10.1038/ni.2688.
    1. Di Giorgio E, Franforte E, Cefalu S, Rossi S, Dei Tos AP, Brenca M, et al. The co-existence of transcriptional activator and transcriptional repressor MEF2 complexes influences tumor aggressiveness. PLoS Genet. 2017;13(4):e1006752. doi: 10.1371/journal.pgen.1006752.
    1. Pon JR, Marra MA. MEF2 transcription factors: developmental regulators and emerging cancer genes. Oncotarget. 2016;7(3):2297–2312. doi: 10.18632/oncotarget.6223.
    1. Zhang M, Truscott J, Davie J. Loss of MEF2D expression inhibits differentiation and contributes to oncogenesis in rhabdomyosarcoma cells. Mol Cancer. 2013;12(1):150. doi: 10.1186/1476-4598-12-150.
    1. Ma L, Liu J, Liu L, Duan G, Wang Q, Xu Y, et al. Overexpression of the transcription factor MEF2D in hepatocellular carcinoma sustains malignant character by suppressing G2-M transition genes. Cancer Res. 2014;74(5):1452–1462. doi: 10.1158/0008-5472.CAN-13-2171.
    1. Di Giorgio E, Gagliostro E, Clocchiatti A, Brancolini C. The control operated by the cell cycle machinery on MEF2 stability contributes to the downregulation of CDKN1A and entry into S phase. Mol Cell Biol. 2015;35(9):1633–1647. doi: 10.1128/MCB.01461-14.
    1. Black BL, Olson EN. Transcriptional control of muscle development by myocyte enhancer factor-2 (MEF2) proteins. Annu Rev Cell Dev Biol. 1998;14:167–196. doi: 10.1146/annurev.cellbio.14.1.167.
    1. Aude-Garcia C, Collin-Faure V, Bausinger H, Hanau D, Rabilloud T, Lemercier C. Dual roles for MEF2A and MEF2D during human macrophage terminal differentiation and c-Jun expression. Biochem J. 2010;430(2):237–244. doi: 10.1042/BJ20100131.
    1. Blaeser F, Ho N, Prywes R, Chatila TA. Ca(2+)-dependent gene expression mediated by MEF2 transcription factors. J Biol Chem. 2000;275(1):197–209. doi: 10.1074/jbc.275.1.197.
    1. She QB, Solit DB, Ye Q, O'Reilly KE, Lobo J, Rosen N. The BAD protein integrates survival signaling by EGFR/MAPK and PI3K/Akt kinase pathways in PTEN-deficient tumor cells. Cancer Cell. 2005;8(4):287–297. doi: 10.1016/j.ccr.2005.09.006.
    1. Chen GJ, Karajannis MA, Newcomb EW, Zagzag D. Overexpression and activation of epidermal growth factor receptor in hemangioblastomas. J Neuro-Oncol. 2010;99(2):195–200. doi: 10.1007/s11060-010-0125-9.
    1. Hyatt DC, Ceresa BP. Cellular localization of the activated EGFR determines its effect on cell growth in MDA-MB-468 cells. Exp Cell Res. 2008;314(18):3415–3425. doi: 10.1016/j.yexcr.2008.08.020.
    1. Chan DLH, Segelov E, Wong RS, Smith A, Herbertson RA, Li BT, et al. Epidermal growth factor receptor (EGFR) inhibitors for metastatic colorectal cancer. Cochrane Database Syst Rev. 2017;6:CD007047.
    1. Rothenberg ML, LaFleur B, Levy DE, Washington MK, Morgan-Meadows SL, Ramanathan RK, et al. Randomized phase II trial of the clinical and biological effects of two dose levels of gefitinib in patients with recurrent colorectal adenocarcinoma. J Clin Oncol. 2005;23(36):9265–9274. doi: 10.1200/JCO.2005.03.0536.
    1. Smith BH, Parikh T, Andrada ZP, Fahey TJ, Berman N, Wiles M, et al. First-in-human phase 1 trial of agarose beads containing murine RENCA cells in advanced solid tumors. Cancer Growth Metastasis. 2016;9:9–20. doi: 10.4137/CGM.S39442.
    1. Nazarian A, Sureshbabu S, Andrada ZP, Thomas J, Arreglado A, Berman N, et al. 18F-FDG PET/CT evaluation of tumor response to the implantation of RENCA macrobeads (RMB) in phase I and II clinical trials [INDBB 10091] in advanced, treatment-resistant metastatic colorectal cancer (mCRC) J Clin Oncol. 2017;35(Suppl 15):e15046–e1504e. doi: 10.1200/JCO.2017.35.15_suppl.e15046.
    1. Ocean AJ, Parikh T, Berman N, Escalon J, Shah MA, Andrada Z, et al. Phase I/II trial of intraperitoneal implantation of agarose-agarose macrobeads (MB) containing mouse renal adenocarcinoma cells (RENCA) in patients (pts) with advanced colorectal cancer (CRC) J Clin Oncol. 2013;31(Suppl 15):e14517–e1451e.
    1. Smith BH, Gazda LS, Conn BL, Jain K, Asina S, Levine DM, et al. Three-dimensional culture of mouse renal carcinoma cells in agarose macrobeads selects for a subpopulation of cells with cancer stem cell or cancer progenitor properties. Cancer Res. 2011;71(3):716–724. doi: 10.1158/0008-5472.CAN-10-2254.

Source: PubMed

3
Se inscrever