Page 268 - Haematologica March 2020
P. 268

F. Chatonnet et al.
cer and favor tumorigenesis induced by the Epstein-Barr virus-derived latent membrane protein 1.48,49 FAM72 genes are highly expressed in TP53 mutated cancer cells, whose growth has been shown to be dependent on FAM72D.50,51 Interestingly, cancer cell growth dependency on FAM72D has also been demonstrated for cells with mutated or copy number variants of p300, p19Arf and CDKI2A,51 suggest- ing that FAM72D is required for the growth of different cancer types. Corroborating the tumorigenic potential of FAM72 proteins, two insertional mutagenesis screens in mouse identified the mouse SRGAP2/FAM72A locus as a driver of chronic myeloid leukemia progression and growth factor-independent leukemogenesis.52,53 Hence, TET-mediated demethylation of the FAM72D upstream region is likely coupled to the proliferation potential of MM cells.
We have defined here a MM-specific hydroxymethy- lome that favors a survival/proliferation program relying, at least in part, on the cooperative actions of enhancers and super-enhancers controlling FAM72D, GAS2, DEP-
TOR and MYC expression (Figure 6). In accordance with the observation that FAM72 expression is predictive to the sensitivity of MM cells to different treatments, its evalua- tion in patients could help to tailor therapy.
Funding
GS was supported by a grant from La Ligue Contre le Cancer (Grand Ouest committee). The work was partially funded by the hematology laboratory of Rennes University Hospital. AP was supported by PhD training grants from Région Bretagne and Ligue Nationale Contre le Cancer. JM was supported by grants from French INCA (Institut National du Cancer) Institute (PLBIO15-256), PLBIO2018-PIT-MM, LR-FEDER Hemodiag, Fondation de France (201400047510), ITMO Cancer (MM&TT), SIRIC Montpellier (INCa-DGOS-Inserm 6045) and Labex EpiGenMed.
Acknowledgments
The authors thank the BioGenouest GEH sequencing plate- form (https://geh.univ-rennes1.fr/) from the UMS Biosit.
References
1. Zhan F, Huang Y, Colla S, et al. The molecu- lar classification of multiple myeloma. Blood. 2006;108(6):2020-2028.
2. Bolli N, Avet-Loiseau H, Wedge DC, et al. Heterogeneity of genomic evolution and mutational profiles in multiple myeloma. Nat Commun. 2014;5:2997.
3. Lohr JG, Stojanov P, Carter SL, et al. Widespread genetic heterogeneity in multi- ple myeloma: implications for targeted ther- apy. Cancer Cell. 2014;25(1):91-101.
4. Delmore JE, Issa GC, Lemieux ME, et al. BET bromodomain inhibition as a therapeu- tic strategy to target c-Myc. Cell. 2011;146(6):904-917.
5. Moreaux J, Reme T, Leonard W, et al. Gene expression-based prediction of myeloma cell sensitivity to histone deacetylase inhibitors. Br J Cancer. 2013;109(3):676-685.
6. Herviou L, Kassambara A, Boireau S, et al. PRC2 targeting is a therapeutic strategy for EZ score defined high risk multiple myelo- ma patients and overcome resistance to IMiDs. Clin Epigenetics. 2018;10(1):121.
7. Sawyer JR, Tricot G, Mattox S, Jagannath S, Barlogie B. Jumping translocations of chro- mosome 1q in multiple myeloma: evidence for a mechanism involving decondensation of pericentromeric heterochromatin. Blood. 1998;91(5):1732-1741.
8. Hanamura I, Stewart JP, Huang Y, et al. Frequent gain of chromosome 1q21 in plas- ma-cell dyscrasias detected by fluorescence in situ hybridization: incidence increases from MGUS to relapsed myeloma and is related to prognosis and disease progression following tandem stem-cell transplantation. Blood. 2006;108(5):1724-1732.
9. Walker BA, Leone PE, Chiecchio L, et al. A compendium of myeloma-associated chro- mosomal copy number abnormalities and their prognostic value. Blood. 2010;116(15):e56-65.
10. Marchesini M, Ogoti Y, Fiorini E, et al. ILF2 is a regulator of RNA splicing and DNA damage response in 1q21 amplified multiple myeloma. Cancer Cell. 2017;32(1):88-100.
11. Paiva B, Puig N, Cedena MT, et al. Differentiation stage of myeloma plasma cells: biological and clinical significance. Leukemia. 2017;31(2):382-392.
12. Wu X, Zhang Y. TET-mediated active DNA demethylation: mechanism, function and beyond. Nat Rev Genet. 2017;18(9):517-534.
13. Sérandour AA, Avner S, Oger F, et al. Dynamic hydroxymethylation of deoxyri- bonucleic acid marks differentiation-associ- ated enhancers. Nucleic Acids Res. 2012;40(17):8255-8265.
14. Caron G, Hussein M, Kullis M, et al. Cell- cycle-dependent reconfiguration of the DNA methylome during terminal differenti- ation of human B cells into plasma cells. Cell Rep. 2015;13(5):1059-1071.
15. Lio C-W, Shukla V, Samaniego-Castruita D, et al. TET enzymes augment activation- induced deaminase (AID) expression via 5- hydroxymethylcytosine modifications at the Aicda superenhancer. Sci Immunol. 2019;4(34).
16. Jin Y, Chen K, De Paepe A, et al. Active enhancer and chromatin accessibility land- scapes chart the regulatory network of pri- mary multiple myeloma. Blood. 2018;131(19):2138-2150.
17. Agirre X, Castellano G, Pascual M, et al. Whole-epigenome analysis in multiple myeloma reveals DNA hypermethylation of B cell-specific enhancers. Genome Res. 2015;25(4):478-487.
18. Viziteu E, Klein B, Basbous J, et al. RECQ1 helicase is involved in replication stress sur- vival and drug resistance in multiple myelo- ma. Leukemia. 2017;31(10):2104-2113.
19. Requirand G, Robert N, Boireau S, et al. BrdU incorporation in multiparameter flow cytometry: A new cell cycle assessment approach in multiple myeloma. Cytometry B Clin Cytom. 2019;96(3):209-214.
20. Barlogie B, Tricot G, Rasmussen E, et al. Total therapy 2 without thalidomide in comparison with total therapy 1: role of intensified induction and posttransplanta- tion consolidation therapies. Blood. 2006;107(7):2633-2638.
21. Pineda-Roman M, Zangari M, Haessler J, et al. Sustained complete remissions in multi-
ple myeloma linked to bortezomib in total therapy 3: comparison with total therapy 2. Br J Haematol. 2008;140(6):625-634.
22. Mulligan G, Mitsiades C, Bryant B, et al. Gene expression profiling and correlation with outcome in clinical trials of the protea- some inhibitor bortezomib. Blood. 2007;109(8):3177-3188.
23. Song CX, Szulwach KE, Fu Y, et al. Selective chemical labeling reveals the genome-wide distribution of 5-hydroxymethylcytosine. Nat Biotechnol. 2011;29(1):68–72.
24. Sérandour AA, Avner S, Mahé EA, et al. Single-CpG resolution mapping of 5- hydroxymethylcytosine by chemical label- ing and exonuclease digestion identifies evo- lutionarily unconserved CpGs as TET tar- gets. Genome Biol. 2016;17:56.
25. Sérandour AA, Avner S, Salbert G. Coupling exonuclease digestion with selective chemi- cal labeling for base-resolution mapping of 5-hydroxymethylcytosine in genomic DNA. Bio Protoc. 2018;8(5):2747.
26. Liu T, Ortiz JA, Taing L, et al. Cistrome: an integrative platform for transcriptional regu- lation studies. Genome Biol. 2011;12(8):R83.
27. Zhang Z, Chang CW, Goh WL, Sung WK, Cheung E. Centdist: discovery of co-associ- ated factors by motif distribution. Nucleic Acids Res. 2011;39(Web Server issue):W391- 399.
28. McLean CY, Bristor D, Hiller M, et al. GREAT improves functional interpretation of cis-regulatory regions. Nat Biotechnol. 2010;28(5):495-501.
29. Stewart SK, Morris TJ, Guilhamon P, et al. oxBS-450K: a method for analysing hydrox- ymethylation using 450K beadchips. Methods. 2015;72:9-15.
30. Shaffer AL, Emre NC, Lamy L, et al. 2008. IRF4 addiction in multiple myeloma. Nature. 2008;454(7201):226-231.
31. LiuZ,XuJ,HeJ,etal.Acriticalroleof autocrine sonic hedgehog signaling in human CD138+ myeloma cell survival and drug resistance. Blood. 2014;124(13):2061- 2071.
32. Jundt F, Probsting KS, Anagnostopoulos I, et al. Jagged1-induced Notch signaling drives proliferation of multiple myeloma cells.
782
haematologica | 2020; 105(3)


































































































   266   267   268   269   270