Page 123 - 2019_08-Haematologica-web
P. 123

Oncogenic mechanisms of ZEB1 and LM02 in T-ALL
tions found in patients and the observed spontaneous T- cell lymphoma development in a mouse model expressing a carboxyterminal truncated form of ZEB1. This is in con- trast with the oncogenic role we have described for Zeb2 in T-ALL.21 In this study, we demonstrated that pre-leukemic T-cell differentiation is also affected by Zeb2 overexpres- sion. Maintenance of Zeb2 expression results in a partial differentiation block or delay at an early DN3C stage, coin- ciding with the T-cell developmental stage at which Zeb2 expression is normally downregulated in mice, as well as in humans. This delay could be partially rescued by decreas- ing IL7R pathway activation, suggesting that the delay is caused by an inability of the Zeb2-overexpressing thymo- cytes to downregulate IL7R expression. Interestingly, other mouse models have shown a similar block in T-cell differ- entiation at this key transition point, including mice that overexpress the IL7r.35,42 CD2-LMO2tg animals accumulate a similar aberrant DN3 population at a young age. Notably, our detailed flow cytometric analysis revealed that LMO2- overexpressing cells are delayed slightly earlier in T-cell dif- ferentiation compared to the Zeb2-overexpressing thymo- cytes, at the DN3A stage, and before T-cell rearrangement (data not shown).
As ZEB have been previously associated with the acqui- sition of cancer stem cell properties, we hypothesized that the cause of spontaneous T-ALL formation in R26-Zeb2tg could be due to acquired self-renewal of pre-leukemic thy- mocytes, as previously demonstrated for LMO2. This was supported by the fact that both proteins bind similar reg- ulatory elements, and that both factors can drive murine T-ALL development with a similar immature expression profile and increased stem cell properties. Nevertheless, using thymocyte transplantation experiments, we demon- strated that R26-Zeb2tg thymocytes are not able to recon- stitute an irradiated thymus and therefore have no pre-
leukemic self-renewal capacity. These data indicate differ- ent oncogenic mechanism for ZEB2 and LMO2. Upon transplantation, we observed that the survival of R26- Zeb2tg thymocytes was longer than that of wildtype thy- mocytes, with this difference most probably being associ- ated with increased IL7R signaling. Whether the increase in Il7r expression is involved in tumor initiation in R26- Zeb2tg animals remains to be determined.
Finally, we used a late-acting CD4-Cre line to show that Zeb2 could also transform thymic precursor cells at later stages during their development, suggesting that the delay at the DN3C stage is not essential for leukemic transfor- mation. Interestingly, a forward genetic screening used the same CD4-Cre line to activate the “sleeping beauty trans- posase” and induce oncogenic transformation specifically at later stages of T-cell development, which resulted in T- ALL with an immature expression profile.43 These data indicate that cases of immature murine T-ALL can also originate in cells beyond the DN stage. Furthermore, our data suggest that a Zeb2-transformed T-ALL with an immature/stem cell expression profile can originate from a more differentiated cell without self-renewal capacity.
In summary, we conclude that multiple oncogenes such as ZEB2 and LMO2 are able to induce subtypes of imma- ture murine T-ALL via distinct oncogenic mechanisms of action. In addition, Zeb2 can also drive T-cell transforma- tion in more differentiated T-cell precursor cells with sim- ilar kinetics.
Acknowledgments
This work was supported by the Australian NHMRC (grant 1047995 to JJH), the Worldwide Cancer Research Fund, the Swiss Bridge Foundation, Stand Up Against Cancer Fund, the Ghent University Special Research Fund and the Fund for Scientific Research-Flanders.
References
1. Rothenberg EV. T cell lineage commitment: identity and renunciation. J Immunol. 2011;186(12):6649-6655.
2. Morris GP, Allen PM. How the TCR bal- ances sensitivity and specificity for the recognition of self and pathogens. Nat Immunol. 2012;13(2):121-128.
3. Meijerink JP. Genetic rearrangements in rela- tion to immunophenotype and outcome in T-cell acute lymphoblastic leukaemia. Best Pract Res Clin Haematol. 2010;23(3):307- 318.
4. Van Vlierberghe P, Ferrando A. The molecu- lar basis of T cell acute lymphoblastic leukemia. J Clin Invest. 2012;122(10):3398- 3406.
5. Engel I, Murre C. The function of E- and Id proteins in lymphocyte development. Nat Rev Immunol. 2001;1(3):193-199.
6. Wang LH, Baker NE. E Proteins and ID pro- teins: helix-loop-helix partners in develop- ment and disease. Dev Cell. 2015;35(3):269- 280.
7. Wadman I, Li J, Bash RO, et al. Specific in vivo association between the bHLH and LIM proteins implicated in human T cell leukemia. EMBO J. 1994;13(20):4831-4839.
8. Wadman IA, Osada H, Grutz GG, et al. The LIM-only protein Lmo2 is a bridging mole- cule assembling an erythroid, DNA-binding complex which includes the TAL1, E47, GATA-1 and Ldb1/NLI proteins. EMBO J. 1997;16(11):3145-3157.
9. Boehm T, Foroni L, Kaneko Y, Perutz MF, Rabbitts TH. The rhombotin family of cys- teine-rich LIM-domain oncogenes: distinct members are involved in T-cell transloca- tions to human chromosomes 11p15 and 11p13. Proc Natl Acad Sci U S A. 1991;88(10):4367-4371.
10. Van Vlierberghe P, van Grotel M, Beverloo HB, et al. The cryptic chromosomal deletion del(11)(p12p13) as a new activation mecha- nism of LMO2 in pediatric T-cell acute lym- phoblastic leukemia. Blood. 2006;108(10): 3520-3529.
11. Goossens S, Van Vlierberghe P. Novel onco- genic noncoding mutations in T-ALL. Blood. 2017;129(24):3140-3142.
12. Fisch P, Boehm T, Lavenir I, et al. T-cell acute lymphoblastic lymphoma induced in trans- genic mice by the RBTN1 and RBTN2 LIM- domain genes. Oncogene. 1992;7(12):2389- 2397.
13. Neale GA, Rehg JE, Goorha RM. Ectopic expression of rhombotin-2 causes selective expansion of CD4-CD8- lymphocytes in the
thymus and T-cell tumors in transgenic
mice. Blood. 1995;86(8):3060-3071.
14. Smith S, Tripathi R, Goodings C, et al. LIM domain only-2 (LMO2) induces T-cell leukemia by two distinct pathways. PloS
One. 2014;9(1):e85883.
15. McCormack MP, Young LF, Vasudevan S, et
al. The Lmo2 oncogene initiates leukemia in mice by inducing thymocyte self-renewal. Science. 2010;327(5967):879-883.
16. Cleveland SM, Smith S, Tripathi R, et al. Lmo2 induces hematopoietic stem cell-like features in T-cell progenitor cells prior to leukemia. Stem Cells. 2013;31(5):882-894.
17. McCormack MP, Shields BJ, Jackson JT, et al. Requirement for Lyl1 in a model of Lmo2- driven early T-cell precursor ALL. Blood. 2013;122(12):2093-2103.
18. Comijn J, Berx G, Vermassen P, et al. The two-handed E box binding zinc finger pro- tein SIP1 downregulates E-cadherin and induces invasion. Mol Cell. 2001;7(6):1267- 1278.
19. Remacle JE, Kraft H, Lerchner W, et al. New mode of DNA binding of multi-zinc finger transcription factors: delta EF1 family mem- bers bind with two hands to two target sites. EMBO J. 1999;18(18):5073-5084.
20. Wellner U, Schubert J, Burk UC, et al. The EMT-activator ZEB1 promotes tumorigenic-
haematologica | 2019; 104(8)
1615


































































































   121   122   123   124   125