Reduction of RUNX1 transcription factor activity by a CBFA2T3-mimicking peptide: application to B cell precursor acute lymphoblastic leukemia

Hélène Jakobczyk, Lydie Debaize, Benoit Soubise, Stéphane Avner, Jérémie Rouger-Gaudichon, Séverine Commet, Yan Jiang, Aurélien A Sérandour, Anne-Gaëlle Rio, Jason S Carroll, Christian Wichmann, Michael Lie-A-Ling, Georges Lacaud, Laurent Corcos, Gilles Salbert, Marie-Dominique Galibert, Virginie Gandemer, Marie-Bérengère Troadec, Hélène Jakobczyk, Lydie Debaize, Benoit Soubise, Stéphane Avner, Jérémie Rouger-Gaudichon, Séverine Commet, Yan Jiang, Aurélien A Sérandour, Anne-Gaëlle Rio, Jason S Carroll, Christian Wichmann, Michael Lie-A-Ling, Georges Lacaud, Laurent Corcos, Gilles Salbert, Marie-Dominique Galibert, Virginie Gandemer, Marie-Bérengère Troadec

Abstract

Background: B Cell Precursor Acute Lymphoblastic Leukemia (BCP-ALL) is the most common pediatric cancer. Identifying key players involved in proliferation of BCP-ALL cells is crucial to propose new therapeutic targets. Runt Related Transcription Factor 1 (RUNX1) and Core-Binding Factor Runt Domain Alpha Subunit 2 Translocated To 3 (CBFA2T3, ETO2, MTG16) are master regulators of hematopoiesis and are implicated in leukemia.

Methods: We worked with BCP-ALL mononuclear bone marrow patients' cells and BCP-ALL cell lines, and performed Chromatin Immunoprecipitations followed by Sequencing (ChIP-Seq), co-immunoprecipitations (co-IP), proximity ligation assays (PLA), luciferase reporter assays and mouse xenograft models.

Results: We demonstrated that CBFA2T3 transcript levels correlate with RUNX1 expression in the pediatric t(12;21) ETV6-RUNX1 BCP-ALL. By ChIP-Seq in BCP-ALL patients' cells and cell lines, we found that RUNX1 is recruited on its promoter and on an enhancer of CBFA2T3 located - 2 kb upstream CBFA2T3 promoter and that, subsequently, the transcription factor RUNX1 drives both RUNX1 and CBFA2T3 expression. We demonstrated that, mechanistically, RUNX1 and CBFA2T3 can be part of the same complex allowing CBFA2T3 to strongly potentiate the activity of the transcription factor RUNX1. Finally, we characterized a CBFA2T3-mimicking peptide that inhibits the interaction between RUNX1 and CBFA2T3, abrogating the activity of this transcription complex and reducing BCP-ALL lymphoblast proliferation.

Conclusions: Altogether, our findings reveal a novel and important activation loop between the transcription regulator CBFA2T3 and the transcription factor RUNX1 that promotes BCP-ALL proliferation, supporting the development of an innovative therapeutic approach based on the NHR2 subdomain of CBFA2T3 protein.

Keywords: AML1; CBFA2T3; Childhood leukemia; Driver loop; ETO2; Inhibitor; NHR2; RUNX1; Transcription factor.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
RUNX1 and CBFA2T3 mRNA level are positively correlated in ETV6-RUNX1 BCP-ALL. a and b The graphs represent the mRNA expression level of CBFA2T3 and RUNX1 in human pediatric ETV6-RUNX1 BCP-ALL bone marrow mononuclear cells and the other non-ETV6-RUNX1 BCP-ALL bone marrow mononuclear cells. Data of mRNA levels (expressed in Fragments Per Kilobase Million—FPKM) have been extracted from the St. Jude Children's Research Hospital RNA-Seq Pediatric Cancer Data Portal [31]. For a ****p < 0.0001 in t-test between both conditions, for b a Pearson correlation has been performed. ns, non significant. Mean data and numbers of samples (n) are indicated above each graph. c Correlation between RUNX1 and CBFA2T3 mRNA levels originating from REH cells and Nalm6 cells that overexpress or are depleted for RUNX1 or CBFA2T3. Statistical analysis has been performed using Pearson correlation. n, number of samples per condition
Fig. 2
Fig. 2
RUNX1 and CBFA2T3 interplay for their transcription. a and b ChIP-Seq profiles across the human RUNX1 (a) and CBFA2T3 (b) genes. Genomic tracks display ChIP-Seq profiles of RUNX1 from REH cells, Nalm6 cells and BCP-ALL patients, and of the histones H3K27ac, H3K4me3, and H3K4me1 from REH and Nalm6 cells. RUNX1 ChIP-Seq from bone marrow mononuclear cells isolated from three pre-B acute lymphoblastic leukemia patients (BCP-ALL) are also displayed. ChIP-Seq reads were aligned to the reference human genome version GRCh37 (hg19). Each genomic regions of RUNX1 and CBFA2T3 genes that are subsequently studied are indicated by boxes. ce Luciferase assays with a plasmid containing the RUNX1 promoter (chr21: 940,581–940,336(C), CBFA2T3 enhancer (chr16:89,045,181–89,045,538) (D) and a repetition of RUNX1-consensus motif (E) upstream a minimal promoter and a luciferase ORF, in presence of RUNX1 and CBFA2T3 expressing plasmids in HEK293 cells. Note that the HEK293 cells do not endogenously express RUNX1 and CBFA2T3. Luciferase levels (Firefly luciferase/Renilla luciferase) are represented using a scatter dot plot indicating the means and S.D. NS: non-significant, **p < 0.01, ***p < 0.001, ****p < 0.0001 in Mann–Whitney tests compared to the control condition
Fig. 3
Fig. 3
CBFA2T3 and RUNX1 colocalize. a, b, c, e and f Quantitation of protein co-localization per nucleus and visualized by Proximity Ligation Assay (PLA) dots in REH cells (a, e), Nalm6 cells (b, f) and BCP-ALL patient cells (c), presented with the mean values ± S.D. Antibodies used are indicated under each plot. Positive controls (total CBFA2T3, where primary antibodies against two different epitopes of CBFA2T3 were used) and negative controls (only one anti-CBFA2T3) were included. One representative experiment of at least two independent experiments is shown. The data are normalized against total CBFA2T3. The mean value is indicated above each plot. The positive threshold value is represented by the dotted line (set at two S.D over the background signal as described in [25]). NS: non-significant, * p < 0.05, **** p < 0.0001 in Fisher’s exact test compared to the negative control condition. d Co-immunoprecipitation (co-IP) using (left panel) IgG or RUNX1 antibody in REH cells, and (in right panel) anti-Flag antibody in HEK293 cells expressing RUNX1-Halotag and/or CBFA2T3-Flag plasmids. Western blots were performed with RUNX1 and CBFA2T3 antibodies. Molecular weights are indicated on the right. g Density plots of CBFA2T3 Chip-Seq signals into RUNX1-bound regions or random regions in REH and Nalm6 cells. h ChIP-Seq profiles across the human CBFA2T3 gene. Genomic tracks display ChIP-Seq profiles for RUNX1 and CBFA2T3 from REH cells. ChIP-Seq reads were aligned to the reference human genome version GRCh37 (hg19). Two enhancers at + 9 kb and − 2 kb have been focused on. i Logo corresponding to the RUNX1 enriched motif for CBFA2T3 regions in REH cells. REH and Nalm6 CBFA2T3 Chip-Seq have been analyzed with the Analysis of Motif Enrichment of the MEME suite [32]. The optimal enrichment p-value of the motif according to the Fisher’s exact test, adjusted for multiple tests using a Bonferroni correction is indicated
Fig. 4
Fig. 4
The NHR2 domain of CBFA2T3 is involved in the interaction between RUNX1 and CBFA2T3. a Representation of the full-length and truncated CBFA2T3 and RUNX1 proteins used to the functional domain analysis. NHR, Nervy Homology Region; TD, Transactivation domain; ID, inhibitory domain. b, c Co-immunoprecipitation (IP) using anti-Flag antibody in HEK293 cells expressing various full-length and truncated proteins for CBFA2T3 (b) and RUNX1 (c) described in (a). Western blots were performed with RUNX1, Myc and CBFA2T3 antibodies. Molecular weights are indicated on the right. d Quantitation of protein co-localization per nucleus and visualized by Proximity Ligation Assay (PLA) dots in HEK293 cells expressing CBFA2T3NHR2-Myc with either RUNX1-Halotag or CBFA2T3-Flag, presented with the mean values ± S.D. Antibodies used are anti-RUNX1, anti-Myc and anti-Flag. Positive control (total RUNX1) and negative control were included. The data are normalized against total RUNX1. The mean value is indicated above each plot. The positive threshold value is represented by the dotted line as described in [25]. ****p < 0.0001 in Fisher’s exact test compared to the negative control condition
Fig. 5
Fig. 5
Free NHR2 domain of CBFA2T3 disrupts the protein complex formed by RUNX1 and CBFA2T3. a and b Quantitation of RUNX1 and CBFA2T3 protein co-localization per nucleus and visualized by PLA dots in REH cells (a) and HEK293 cells (b) in presence or absence of CBFA2T3NHR2-Myc, presented with the mean values ± S.D. The mean value is indicated above each plot. The statistical analyses are run with parametric t-tests. *p < 0.1; ****p < 0.0001. The right panel shows a set of pictures of PLA between RUNX1 and CBFA2T3 for HEK293 cells in presence or absence of CBFA2T3NHR2-Myc. The PLA dots are in purple, nuclei are in blue (DAPI). Bar: 10 um. c Co-immunoprecipitation (IP) using anti-Flag antibody in HEK293 cells expressing RUNX1-Halotag, CBFA2T3-Flag or CBFA2T3NHR2-Myc. Western blots were performed with RUNX1, Myc and CBFA2T3 antibodies. Molecular weights are indicated on the right
Fig. 6
Fig. 6
Free NHR2 domain of CBFA2T3 inhibits the cooperative effect of CBFA2T3 on RUNX1 activity. ad Luciferase assays with the RUNX1-consensus motif repetition (a), the RUNX1 promoter (b), the CBFA2T3 enhancer (c) and the c-KIT enhancer (d) upstream a minimal promoter and a luciferase ORF, in presence of RUNX1 and CBFA2T3 or CBFA2T3NHR2 expressing plasmids in HEK293 cells. Luciferase levels (Firefly luciferase/Renilla luciferase) are represented using a scatter dot plot indicating the means and S.D. ns: non-significant, **p < 0.01, ***p < 0.001, ****p < 0.0001 in Mann–Whitney tests compared to the control condition. e Relative mRNA expression of RUNX1, CBFA2T3 and c-KIT measured by RT-qPCR in REH cells, and REH expressing CBFA2T3NHR2 cells (REH+NHR2). Results are presented in-terms of a fold change after normalizing with ABL mRNA. f Schematic representation of the activation loop between RUNX1 and CBFA2T3. Expression of the free NHR2 domain interrupts this loop
Fig. 7
Fig. 7
Expression of free NHR2 domain of CBFA2T3 delays BCP-ALL cell proliferation. a and b Proliferation curves from REHshcontrol, REHshRUNX1, REHshCBFA2T3−1, REHshCBFA2T3−2 (a), or REHshcontrol and REH+NHR2 cells (b). Three experiments are represented for each condition. For more readability, statistical analyses have been run only for the last day (day 7) and compared to the condition REHshcontrol. * p < 0.05. c, d Kaplan–Meier survival curves from immunodeficient NOD/scid IL2 Rg null mice xenografted with 100,000 cells of REH, REHshRUNX1, REHshCBFA2T3−1, REHshCBFA2T3−2 (C), or REH+NHR2 (D) (n = 7–8 per group). The general condition of mice was monitored daily until experiment ended. Mantel-Cox statistical tests compared to the condition REHshcontrol. **p < 0.01, ***p < 0.001. e We propose the model in which CBFA2T3 acts as an activator of RUNX1 transcription activity and sustains cell proliferation. Presence of free NHR2 domain disrupts this activation complex and slows down proliferation

References

    1. Levanon D, Groner Y. Structure and regulated expression of mammalian RUNX genes. Oncogene. 2004;23:4211–4219. doi: 10.1038/sj.onc.1207670.
    1. Okuda T, van Deursen J, Hiebert SW, Grosveld G, Downing JR. AML1, the target of multiple chromosomal translocations in human leukemia, is essential for normal fetal liver hematopoiesis. Cell. 1996;84:321–330. doi: 10.1016/S0092-8674(00)80986-1.
    1. Wang Q, Stacy T, Binder M, Marin-Padilla M, Sharpe AH, Speck NA. Disruption of the Cbfa2 gene causes necrosis and hemorrhaging in the central nervous system and blocks definitive hematopoiesis. Proc Natl Acad Sci USA. 1996;93:3444–3449. doi: 10.1073/pnas.93.8.3444.
    1. Ichikawa M, Asai T, Saito T, Seo S, Yamazaki I, Yamagata T, et al. AML-1 is required for megakaryocytic maturation and lymphocytic differentiation, but not for maintenance of hematopoietic stem cells in adult hematopoiesis. Nat Med. 2004;10:299–304. doi: 10.1038/nm997.
    1. Lie-A-Ling M, Mevel R, Patel R, Blyth K, Baena E, Kouskoff V, et al. RUNX1 Dosage in Development and Cancer. Mol Cells. 2020;43(2):126–138.
    1. Sood R, Kamikubo Y, Liu P. Role of RUNX1 in hematological malignancies. Blood. 2017;129(15):2070–2082. doi: 10.1182/blood-2016-10-687830.
    1. De Braekeleer E, Douet-Guilbert N, Morel F, Le Bris M-J, Férec C, De Braekeleer M. RUNX1 translocations and fusion genes in malignant hemopathies. Future Oncol Lond Engl. 2011;7(1):77–91. doi: 10.2217/fon.10.158.
    1. Sun C-C, Li S-J, Chen Z-L, Li G, Zhang Q, Li D-J. Expression and prognosis analyses of runt-related transcription factor family in human Leukemia. Mol Ther Oncolytics. 2019;12:103–111. doi: 10.1016/j.omto.2018.12.008.
    1. Yokota A, Huo L, Lan F, Wu J, Huang G. The clinical, molecular, and mechanistic basis of RUNX1 mutations identified in hematological malignancies. Mol Cells. 2020;43(2):145–152.
    1. Bartfeld D, Shimon L, Couture GC, Rabinovich D, Frolow F, Levanon D, et al. DNA recognition by the RUNX1 transcription factor is mediated by an allosteric transition in the RUNT domain and by DNA bending. Struct Lond Engl. 2002;10(10):1395–1407.
    1. Blumenthal E, Greenblatt S, Huang G, Ando K, Xu Y, Nimer SD. Covalent modifications of RUNX proteins: structure affects function. Adv Exp Med Biol. 2017;962:33–44. doi: 10.1007/978-981-10-3233-2_3.
    1. Davis JN, McGhee L, Meyers S. The ETO (MTG8) gene family. Gene. 2003;303:1–10. doi: 10.1016/S0378-1119(02)01172-1.
    1. Lutterbach B, Sun D, Schuetz J, Hiebert SW. The MYND motif is required for repression of basal transcription from the multidrug resistance 1 promoter by the t(8;21) fusion protein. Mol Cell Biol. 1998;18(6):3604–3611. doi: 10.1128/MCB.18.6.3604.
    1. Lenny N, Meyers S, Hiebert SW. Functional domains of the t(8;21) fusion protein, AML-1/ETO. Oncogene. 1995;11(9):1761–1769.
    1. Hunt A, Fischer M, Engel ME, Hiebert SW. Mtg16/Eto2 contributes to murine T-cell development. Mol Cell Biol. 2011;31(13):2544–2551. doi: 10.1128/MCB.01458-10.
    1. Goardon N, Lambert JA, Rodriguez P, Nissaire P, Herblot S, Thibault P, et al. ETO2 coordinates cellular proliferation and differentiation during erythropoiesis. EMBO J. 2006;25(2):357–366. doi: 10.1038/sj.emboj.7600934.
    1. Steinauer N, Guo C, Zhang J. Emerging roles of MTG16 in cell-fate control of hematopoietic stem cells and cancer. Stem Cells Int. 2017;2017:6301385. doi: 10.1155/2017/6301385.
    1. Wang J, Hoshino T, Redner RL, Kajigaya S, Liu JM. ETO, fusion partner in t(8;21) acute myeloid leukemia, represses transcription by interaction with the human N-CoR/mSin3/HDAC1 complex. Proc Natl Acad Sci USA. 1998;95(18):10860–10865. doi: 10.1073/pnas.95.18.10860.
    1. Gandemer V, Rio A-G, de Tayrac M, Sibut V, Mottier S, Ly Sunnaram B, et al. Five distinct biological processes and 14 differentially expressed genes characterize TEL/AML1-positive leukemia. BMC Genomics. 2007;8:385. doi: 10.1186/1471-2164-8-385.
    1. Yeoh E-J, Ross ME, Shurtleff SA, Williams WK, Patel D, Mahfouz R, et al. Classification, subtype discovery, and prediction of outcome in pediatric acute lymphoblastic leukemia by gene expression profiling. Cancer Cell. 2002;1(2):133–143. doi: 10.1016/S1535-6108(02)00032-6.
    1. Ross ME, Zhou X, Song G, Shurtleff SA, Girtman K, Williams WK, et al. Classification of pediatric acute lymphoblastic leukemia by gene expression profiling. Blood. 2003;102(8):2951–2959. doi: 10.1182/blood-2003-01-0338.
    1. Hu Z, Gu X, Baraoidan K, Ibanez V, Sharma A, Kadkol S, et al. RUNX1 regulates corepressor interactions of PU.1. Blood. 2011;117(24):6498–6508. doi: 10.1182/blood-2010-10-312512.
    1. Meier N, Krpic S, Rodriguez P, Strouboulis J, Monti M, Krijgsveld J, et al. Novel binding partners of Ldb1 are required for haematopoietic. Development. 2006;133(24):4913–4923. doi: 10.1242/dev.02656.
    1. Stadhouders R, Cico A, Stephen T, Thongjuea S, Kolovos P, Baymaz HI, et al. Control of developmentally primed erythroid genes by combinatorial co-repressor actions. Nat Commun. 2015;6:8893. doi: 10.1038/ncomms9893.
    1. Debaize L, Jakobczyk H, Rio A-G, Gandemer V, Troadec M-B. Optimization of proximity ligation assay (PLA) for detection of protein interactions and fusion proteins in non-adherent cells: application to pre-B lymphocytes. Mol Cytogenet. 2017;10:27. doi: 10.1186/s13039-017-0328-2.
    1. Debaize L, Jakobczyk H, Avner S, Gaudichon J, Rio A-G, Sérandour AA, et al. Interplay between transcription regulators RUNX1 and FUBP1 activates an enhancer of the oncogene c-KIT and amplifies cell proliferation. Nucleic Acids Res. 2018;46(21):11214–11228. doi: 10.1093/nar/gky756.
    1. Kumar R, Cheney KM, McKirdy R, Neilsen PM, Schulz RB, Lee J, et al. CBFA2T3-ZNF652 corepressor complex regulates transcription of the E-box gene HEB. J Biol Chem. 2008;283(27):19026–19038. doi: 10.1074/jbc.M709136200.
    1. Fears S, Gavin M, Zhang DE, Hetherington C, Ben-David Y, Rowley JD, et al. Functional characterization of ETV6 and ETV6/CBFA2 in the regulation of the MCSFR proximal promoter. Proc Natl Acad Sci USA. 1997;94:1949–1954. doi: 10.1073/pnas.94.5.1949.
    1. Nordengrahn A, Gustafsdottir SM, Ebert K, Reid SM, King DP, Ferris NP, et al. Evaluation of a novel proximity ligation assay for the sensitive and rapid detection of foot-and-mouth disease virus. Vet Microbiol. 2008;127(3–4):227–236. doi: 10.1016/j.vetmic.2007.08.026.
    1. Arnaud M-P, Vallée A, Robert G, Bonneau J, Leroy C, Varin-Blank N, et al. CD9, a key actor in the dissemination of lymphoblastic leukemia, modulating CXCR4-mediated migration via RAC1 signaling. Blood. 2015;126(15):1802–1812. doi: 10.1182/blood-2015-02-628560.
    1. Zhou X, Edmonson MN, Wilkinson MR, Patel A, Wu G, Liu Y, et al. Exploring genomic alteration in pediatric cancer using ProteinPaint. Nat Genet. 2016;48(1):4–6. doi: 10.1038/ng.3466.
    1. McLeay RC, Bailey TL. Motif Enrichment Analysis: a unified framework and an evaluation on ChIP data. BMC Bioinform. 2010;11:165. doi: 10.1186/1471-2105-11-165.
    1. Gamou T, Kitamura E, Hosoda F, Shimizu K, Shinohara K, Hayashi Y, et al. The partner gene of AML1 in t(16;21) myeloid malignancies is a novel member of the MTG8(ETO) family. Blood. 1998;91(11):4028–4037. doi: 10.1182/blood.V91.11.4028.
    1. Chen-Wichmann L, Shvartsman M, Preiss C, Hockings C, Windisch R, Redondo Monte E, et al. Compatibility of RUNX1/ETO fusion protein modules driving CD34+ human progenitor cell expansion. Oncogene. 2019;38(2):261–272. doi: 10.1038/s41388-018-0441-7.
    1. Wichmann C, Chen L, Heinrich M, Baus D, Pfitzner E, Zornig M, et al. Targeting the Oligomerization Domain of ETO Interferes with RUNX1/ETO Oncogenic Activity in t(8;21)-Positive Leukemic Cells. Cancer Res. 2007;67(5):2280–2289. doi: 10.1158/0008-5472.CAN-06-3360.
    1. Chagraoui H, Kristiansen MS, Ruiz JP, Serra-Barros A, Richter J, Hall-Ponselé E, et al. SCL/TAL1 cooperates with Polycomb RYBP-PRC1 to suppress alternative lineages in blood-fated cells. Nat Commun. 2018;9(1):5375. doi: 10.1038/s41467-018-07787-6.
    1. Fujiwara T, Lee H-Y, Sanalkumar R, Bresnick EH. Building multifunctionality into a complex containing master regulators of hematopoiesis. Proc Natl Acad Sci USA. 2010;107(47):20429–20434. doi: 10.1073/pnas.1007804107.
    1. Kiefer CM, Lee J, Hou C, Dale RK, Lee YT, Meier ER, et al. Distinct Ldb1/NLI complexes orchestrate γ-globin repression and reactivation through ETO2 in human adult erythroid cells. Blood. 2011;118(23):6200–6208. doi: 10.1182/blood-2011-06-363101.
    1. Tracey LJ, Brooke-Bisschop T, Jansen PWTC, Campos EI, Vermeulen M, Justice MJ. The Pluripotency Regulator PRDM14 Requires Hematopoietic Regulator CBFA2T3 to Initiate Leukemia in Mice. Mol Cancer Res MCR. 2019;17(7):1468–1479. doi: 10.1158/1541-7786.MCR-18-1327.
    1. Cubedo E, Gentles AJ, Huang C, Natkunam Y, Bhatt S, Lu X, et al. Identification of LMO2 transcriptome and interactome in diffuse large B-cell lymphoma. Blood. 2012;119(23):5478–5491. doi: 10.1182/blood-2012-01-403154.
    1. Fujiwara T, Alqadi YW, Okitsu Y, Fukuhara N, Onishi Y, Ishizawa K, et al. Role of transcriptional corepressor ETO2 in erythroid cells. Exp Hematol. 2013;41(3):303–315.e1. doi: 10.1016/j.exphem.2012.10.015.
    1. Ptasinska A, Assi SA, Mannari D, James SR, Williamson D, Dunne J, et al. Depletion of RUNX1/ETO in t(8;21) AML cells leads to genome-wide changes in chromatin structure and transcription factor binding. Leukemia. 2012;26(8):1829–1841. doi: 10.1038/leu.2012.49.
    1. Granja JM, Klemm S, McGinnis LM, Kathiria AS, Mezger A, Corces MR, et al. Single-cell multiomic analysis identifies regulatory programs in mixed-phenotype acute leukemia. Nat Biotechnol. 2019;37(12):1458–1465. doi: 10.1038/s41587-019-0332-7.
    1. Soler E, Andrieu-Soler C, de Boer E, Bryne JC, Thongjuea S, Stadhouders R, et al. The genome-wide dynamics of the binding of Ldb1 complexes during erythroid differentiation. Genes Dev. 2010;24(3):277–289. doi: 10.1101/gad.551810.
    1. Sanda T, Lawton LN, Barrasa MI, Fan ZP, Kohlhammer H, Gutierrez A, et al. Core transcriptional regulatory circuit controlled by the TAL1 complex in human T cell acute lymphoblastic leukemia. Cancer Cell. 2012;22(2):209–221. doi: 10.1016/j.ccr.2012.06.007.
    1. Huret J-L, Ahmad M, Arsaban M, Bernheim A, Cigna J, Desangles F, et al. Atlas of genetics and cytogenetics in oncology and haematology in 2013. Nucleic Acids Res. 2013;41(D1):D920–D924. doi: 10.1093/nar/gks1082.
    1. Hayashi K, Abe N, Watanabe T, Obinata M, Ito M, Sato T, et al. Overexpression of AML1 transcription factor drives thymocytes into the CD8 single-positive lineage. J Immunol. 2001;1(167):4957–4965. doi: 10.4049/jimmunol.167.9.4957.
    1. Blyth K, Slater N, Hanlon L, Bell M, Mackay N, Stewart M, et al. Runx1 promotes B-cell survival and lymphoma development. Blood Cells Mol Dis. 2009;43:12–19. doi: 10.1016/j.bcmd.2009.01.013.
    1. Ben-Ami O, Friedman D, Leshkowitz D, Goldenberg D, Orlovsky K, Pencovich N, et al. Addiction of t(8;21) and inv(16) acute myeloid leukemia to native RUNX1. Cell Rep. 2013;4(6):1131–1143. doi: 10.1016/j.celrep.2013.08.020.
    1. Steinauer N, Guo C, Huang C, Wong M, Tu Y, Freter CE, et al. Myeloid translocation gene CBFA2T3 directs a relapse gene program and determines patient-specific outcomes in AML. Blood Adv. 2019;3(9):1379–1393. doi: 10.1182/bloodadvances.2018028514.
    1. Wichmann C, Chen L, Heinrich M, Baus D, Pfitzner E, Zörnig M, et al. Targeting the oligomerization domain of ETO interferes with RUNX1/ETO oncogenic activity in t(8;21)-positive leukemic cells. Cancer Res. 2007;67(5):2280–2289. doi: 10.1158/0008-5472.CAN-06-3360.
    1. Wichmann C, Becker Y, Chen-Wichmann L, Vogel V, Vojtkova A, Herglotz J, et al. Dimer-tetramer transition controls RUNX1/ETO leukemogenic activity. Blood. 2010;116(4):603–613. doi: 10.1182/blood-2009-10-248047.
    1. Bartel Y, Grez M, Wichmann C. Interference with RUNX1/ETO leukemogenic function by cell-penetrating peptides targeting the NHR2 oligomerization domain. BioMed Res Int. 2013;2013:297692. doi: 10.1155/2013/297692.
    1. Schanda J, Lee C-W, Wohlan K, Müller-Kuller U, Kunkel H, Coco IQ-L, et al. Suppression of RUNX1/ETO oncogenic activity by a small molecule inhibitor of tetramerization. Haematologica. 2017;102(5):e170–e174. doi: 10.3324/haematol.2016.161570.
    1. Thirant C, Lopez C, Malinge S, Mercher T. Molecular pathways driven by ETO2-GLIS2 in aggressive pediatric leukemia. Mol Cell Oncol. 2017;4(6):e1345351. doi: 10.1080/23723556.2017.1345351.
    1. Chyla BJ, Moreno-Miralles I, Steapleton MA, Thompson MA, Bhaskara S, Engel M, et al. Deletion of Mtg16, a target of t(16;21), alters hematopoietic progenitor cell proliferation and lineage allocation. Mol Cell Biol. 2008;28(20):6234–6247. doi: 10.1128/MCB.00404-08.
    1. Zhang J, Kalkum M, Yamamura S, Chait BT, Roeder RG. E protein silencing by the leukemogenic AML1-ETO fusion protein. Science. 2004;305(5688):1286–1289. doi: 10.1126/science.1097937.
    1. Gow C-H, Guo C, Wang D, Hu Q, Zhang J. Differential involvement of E2A-corepressor interactions in distinct leukemogenic pathways. Nucleic Acids Res. 2014;42(1):137–152. doi: 10.1093/nar/gkt855.
    1. Hamlett I, Draper J, Strouboulis J, Iborra F, Porcher C, Vyas P. Characterization of megakaryocyte GATA1-interacting proteins: the corepressor ETO2 and GATA1 interact to regulate terminal megakaryocyte maturation. Blood. 2008;112(7):2738–2749. doi: 10.1182/blood-2008-03-146605.

Source: PubMed

3
Abonnieren