Originally identified as a cancer susceptibility gene, Wilms’ Tumor 1 gene (WT1) is overexpressed or mutated in a wide variety of malignancies, including acute myeloid leukemia (AML). WT1 is a zinc-finger transcription factor comprised of C-terminal zinc-finger DNA binding domains and an N-terminal transactivation domain thought to regulate interactions with partner proteins. Germline WT1 mutations consist primarily of nonsense mutations that truncate the C-terminal domains, or missense mutations that disrupt DNA binding, and these mutations result in both developmental abnormalities and predisposition to Wilms’ tumor.1
In normal human CD34+ hematopoietic stem/progenitor cells (HSPC), wild-type WT1 is expressed at a low level, but it is highly expressed in nearly all cases of AML. Among all AML subtypes, WT1 expression is generally highest in acute promyelocytic leukemia (APL/M3 AML), the AML subtype initiated by the PML-RARA fusion gene (Online Supplementary Figure S1A and D).2,3 In addition, we and others4 have identified recurrent WT1 mutations in APL cases (11/42 [26%] in this study) (Online Supplementary Figure S1C). In seven of 11 of these cases, WT1 mutations occurred at a significantly lower variant allele frequency (VAF) than PML-RARA, suggesting they are co-operating events in subclones (data not shown). WT1 mutations have been associated with worse prognosis in non-M3 AML, although no such association has been shown in APL. The spectrum of WT1 mutations is similar in APL versus other AML cases (Online Supplementary Figure S1B), suggesting that WT1 mutations may have similar biologic activities across all AML subtypes.
These observations frame a well-known - but unsolved - paradox that we attempt to address here: does a high level of wild type WT1 expression contribute to the initiation or progression of AML/APL, or conversely, does it reflect a tumor suppressor activity, since inactivating mutations appear to contribute to disease progression?
In order to explore these questions, we first tested the ability of Wt1 mutations to co-operate with PML-RARA in a well-characterized murine APL model. Ctsg-PMLRARA mice express the PML-RARA fusion cDNA in immature hematopoietic progenitor cells, and succumb to an APL-like disease with a latency of about 1 year in C57Bl/6J mice.5 In order to test whether Wt1 mutations can co-operate with PML-RARA in this model, we used CRISPR/Cas9 to generate indels in Wt1 exon 8 or, as a control, the Rosa26 locus. Since murine Wt1 is highly homologous to the human protein, these mutations should mimic those commonly found in APL patients. Despite efficient mutation generation in Ctsg-PML-RARA bone marrow cells (Online Supplementary Figure S2A), there was no survival difference between mice transplanted with Ctsg-PML-RARA cells with Wt1 mutations versus Rosa26 mutations (Online Supplementary Figure S2C). We therefore evaluated APL tumors arising in these mice, and observed that tumors could arise either from wild-type or mutant Wt1/Rosa26 progenitors (Online Supplementary Figure S2B). Surprisingly, we did not detect Wt1 protein in these tumors by western blotting (data not shown). Similarly, in a panel of 16 previously banked murine APL tumors from Ctsg-PML-RARA mice, RNA sequencing revealed virtually undetectable levels of Wt1 mRNA (Online Supplementary Figure S2D), in contrast to the high expression seen in human APL samples. Together, these data suggest that important interspecies differences in Wt1 regulation and function are important for the lack of a phenotype in this mouse model.
We next evaluated WT1 expression in human CD34+ cells by transducing umbilical cord blood-derived CD34+ cells with retroviruses encoding PML-RARA, RUNX1-RUNX1T1, or MYC; RUNX1-RUNX1T1 and MYC have previously been shown to confer self-renewal and expansion of human HSPCs in vitro and in xenograft models, and therefore act as positive controls.6,7 Consistent with previously reported results, we found that human HSPC transduced with RUNX1-RUNX1T1 and MYC expand robustly over 2 weeks in culture, while HSPC transduced with PML-RARA expand more slowly (data not shown). In order to test whether WT1 expression is affected by transduction with these retroviral constructs, GFP+ cells were sorted 7 days after transduction, RNA was isolated, and WT1 expression was measured by quantitative reverse transcription polymerase chain reaction (RT-PCR). Figure 1A shows upregulation of WT1 mRNA in sorted human HSPC transduced with PML-RARA, RUNX1-RUNX1T1, and MYC (6 to 18-fold increase, P<0.05 for PML-RARA and MYC). WT1 protein abundance also increased dramatically in the same cells during this timeframe (Figure 1B). In order to identify other genes dysregulated by PML-RARA transduction, we transduced both mouse and human HSPC with GFPtagged retroviruses encoding PML-RARA or an empty vector, as has been previously reported (n=2 and n=3 separate experiments for human and mouse cells respectively). 8,9 Seven days after transduction, GFP+ cells were sorted and bulk RNA sequencing was performed to identify differentially expressed genes (DEG, 5,347 identified for mouse samples, and 1,885 identified for human samples, Figure 1C). There was significant overlap between orthologous mouse and human DEG after transduction with PML-RARA (Figure 1D, P=9.6x10-118 based on the hypergeometric test); further, of 867 overlapping orthologues, 82% were coordinately regulated. However, while WT1 was ~13-fold upregulated in human cells transduced with PML-RARA, Wt1 expression was extremely low and did not increase in murine cells transduced with PML-RARA (Figure 1E), validating the interspecies difference in WT1/Wt1 regulation noted above.
GFP+ PML-RARA-expressing human cord blood cells expanded modestly during the first weeks of culture, but increased dramatically 3-4 weeks after transduction (Online Supplementary Figure S3A). After 6 weeks in culture, they resembled primary APL cells morphologically and immunophenotypically (Online Supplementary Figure S3B and C). In addition, they were sensitive to treatment with all-trans retinoic acid (ATRA), a hallmark of APL cells (Online Supplementary Figure S3D). Transduced cells were not immortalized, as they stopped proliferating around 8-9 weeks after initiation, and they failed to engraft immunodeficient mice (data not shown). Given its reported role as a tumor suppressor in other cancer types, we hypothesized that this upregulation may reflect an attempt of WT1 to suppress the proliferative response induced by PML-RARA, similar to the increased TP53 activity observed in cells responding to genotoxic stressors.10 However, as noted above, it is also possible that high WT1 expression actively promotes the growth or survival of APL cells. In order to distinguish between these possibilities, we performed WT1 overexpression versus loss-of-function experiments in PML-RARA-transduced cord blood cells.
First, we used lentiviral vectors to overexpress the two most common isoforms of WT1 (KTS+ and KTS-) in human CD34 cells in culture. In the absence of other cooperating oncogenes, WT1 overexpression led to the rapid disappearance of transduced cells (Figure 2A), consistent with previous reports that WT1 overexpression causes differentiation and death of CD34+ cells.11 Next, we asked whether inactivating mutations in WT1 could enhance expansion of CD34+ cells expressing PMLRARA. We transduced CD34+ cells with PML-RARA or empty vector, and 2 days after transduction used CRISPR/Cas9 to generate inactivating indels in WT1, or as a control, AAVS1. After 4-8 weeks in culture, GFP+ cells were sorted and the frequency of WT1 or AAVS1 indels in GFP+ cells at the end of the culture period was compared to the frequency at the beginning. WT1 mutations were selected for over time, and were significantly increased in PML-RARA-transduced cells 4-8 weeks after the WT1 mutations were introduced (Figure 2B, bottom panels). In contrast, cells containing mutations in AAVS1 did not increase in frequency (Figure 2B, top panels). Overall cell numbers significantly increased in cells bearing both PML-RARA and WT1 mutations, compared to cells with PML-RARA and AAVS1 mutations, or cells transduced with an empty vector with WT1 mutations (Figure 2C). Together, these findings suggest that WT1 inactivation enhances the growth of PML-RARAexpressing hematopoietic cells, strongly suggesting that WT1 acts as a tumor suppressor in this context.
Based on the above findings, we propose a simple model to explain these paradoxical observations: WT1 expression in HSPC is normally activated as an adaptive and inhibitory response to oncogenic mutations that cause proliferation, a response that is intended to slow their growth. The subsequent development of inactivating WT1 mutations in some cases would then provide a further growth advantage by removing that normal inhibitory response. Supporting this hypothesis, we found that i) retroviral transduction of CD34+ cells with PML-RARA, RUNX1-RUNX1T1, or MYC all led to a robust induction of WT1 expression; ii) forced expression of wild-type WT1 by itself does not promote CD34 cell expansion; and iii) inactivation of WT1 in PMLRARA- expressing CD34+ cells leads to an additional growth advantage. Although the mechanism of WT1 gene activation by oncogenes is not yet clear, high levels of WT1 expression are found in nearly all AML cases, regardless of subtype or mutational landscape. In addition, since the majority of AML/APL cases do not have WT1 mutations, a corollary of this hypothesis is that leukemias with wild-type WT1 must have developed alternative means to circumvent the inhibitory pressure that WT1 induction may exert.
Finally, the downstream mechanisms by which WT1 mutations lead to a growth advantage in AML cells are currently unclear, and may depend on the context of the co-operating mutations. In addition to its welldescribed function as a locus-specific transcription factor, 12 recent studies have suggested that WT1 mutations may cause epigenetic changes via effects on DNA methylation and interactions with TET family methylcytosine deoxygenases.13-16 A better understanding of how WT1 mutations activate these pathways in AML cells will be required to fully exploit their potential therapeutic value.
Footnotes
- Received July 9, 2021
- Accepted October 12, 2021
Correspondence
Disclosures: no conflicts of interest to disclose.
Contributions: MJC performed data analysis, supervised the study, and wrote the manuscript; DHS performed data analysis; EJD, SK, LDW, SRM and CAM performed data analysis; CDSK, HRL, TTE, VB, MG and AJM performed experiments; TJL supervised the study and wrote the manuscript.
Funding
this work was supported by the National Cancer Institute to MJC (K12 program, CA167540, R. Govindan, PI; SPORE Career Enhancement Award, P50 CA171963, D. Link, PI; and K08CA222630). Additional support was provided by NIH grants CA101937 and CA197561 (TJL), CA211782 (CAM), T32 HL007088 (CDSK), K08CA190815 (DS), and the Barnes Jewish Hospital Foundation (TJL).
Acknowledgments
Technical assistance was provided by the Siteman Cancer Center Tissue Procurement and Cell Sorting cores (NCI Cancer Center Support Grant P30CA91842). Umbilical cord blood units were provided by the St. Louis Cord Blood Bank.
References
- Royer-Pokora B, Beier M Fau - Henzler M. Twenty-four new cases of WT1 germline mutations and review of the literature: genotype/ phenotype correlations for Wilms tumor development. Am J Med Genet A. 2014; 127A(3):249-257. https://doi.org/10.1002/ajmg.a.30015PubMedGoogle Scholar
- Ley TJ, Miller C, Ding L. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med. 2013; 368(22):2059-2074. https://doi.org/10.1056/NEJMoa1301689PubMedPubMed CentralGoogle Scholar
- Cilloni D, Gottardi E, De Micheli D. Quantitative assessment of WT1 expression by real time quantitative PCR may be a useful tool for monitoring minimal residual disease in acute leukemia patients. Leukemia. 2002; 16(10):2115-2121. https://doi.org/10.1038/sj.leu.2402675PubMedGoogle Scholar
- Madan V, Shyamsunder P, Han L. Comprehensive mutational analysis of primary and relapse acute promyelocytic leukemia. Leukemia. 2016; 30(12):2430. https://doi.org/10.1038/leu.2016.237PubMedPubMed CentralGoogle Scholar
- Westervelt P, Lane AA, Pollock JL. High-penetrance mouse model of acute promyelocytic leukemia with very low levels of PML-RARα expression. Blood. 2003; 102(5):1857-1865. https://doi.org/10.1182/blood-2002-12-3779PubMedGoogle Scholar
- Bulaeva E, Pellacani D, Nakamichi N. MYC-induced human acute myeloid leukemia requires a continuing IL3/GM-CSF co-stimulus. Blood. 2020; 136(24):2764-2773. https://doi.org/10.1182/blood.2020006374PubMedGoogle Scholar
- Mulloy JC, Cammenga J, MacKenzie KL, Berguido FJ, Moore MA, Nimer SD. The AML1-ETO fusion protein promotes the expansion of human hematopoietic stem cells. Blood. 2002; 99(1):15-23. https://doi.org/10.1182/blood.V99.1.15PubMedGoogle Scholar
- Minucci S, Monestiroli S, Giavara S. PML-RAR induces promyelocytic leukemias with high efficiency following retroviral gene transfer into purified murine hematopoietic progenitors. Blood. 2002; 100(8):2989-2995. https://doi.org/10.1182/blood-2001-11-0089PubMedGoogle Scholar
- Sternsdorf T, Phan VT, Maunakea ML. Forced retinoic acid receptor alpha homodimers prime mice for APL-like leukemia. Cancer Cell. 2006; 9(2):81-94. https://doi.org/10.1016/j.ccr.2005.12.030PubMedGoogle Scholar
- Karpinich NO, Tafani M, Rothman RJ, Russo MA, Farber JL. The course of etoposide-induced apoptosis from damage to DNA and p53 activation to mitochondrial release of cytochrome c. J Biol Chem. 2002; 277(19):16547-16552. https://doi.org/10.1074/jbc.M110629200PubMedGoogle Scholar
- Ellisen LW, Carlesso N, Cheng T, Scadden DT, Haber DA. The Wilms tumor suppressor WT1 directs stage-specific quiescence and differentiation of human hematopoietic progenitor cells. EMBO J. 2001; 20(8):1897-1909. https://doi.org/10.1093/emboj/20.8.1897PubMedPubMed CentralGoogle Scholar
- Toska E, Roberts SGE. Mechanisms of transcriptional regulation by WT1 (Wilms' tumour 1). Biochem J. 2014; 461(1):15-32. https://doi.org/10.1042/BJ20131587PubMedGoogle Scholar
- Pronier E, Bowman RL, Ahn J. Genetic and epigenetic evolution as a contributor to WT1-mutant leukemogenesis. Blood. 2018; 132(12):1265-1278. https://doi.org/10.1182/blood-2018-03-837468PubMedPubMed CentralGoogle Scholar
- Rampal R, Alkalin A, Madzo J. DNA hydroxymethylation profiling reveals that WT1 mutations result in loss of TET2 function in acute myeloid leukemia. Cell Rep. 2014; 9(5):1841-1855. https://doi.org/10.1016/j.celrep.2014.11.004PubMedPubMed CentralGoogle Scholar
- Sinha S, Thomas D, Yu L. Mutant WT1 is associated with DNA hypermethylation of PRC2 targets in AML and responds to EZH2 inhibition. Blood. 2015; 125(2):316-326. https://doi.org/10.1182/blood-2014-03-566018PubMedPubMed CentralGoogle Scholar
- Wang Y, Xiao M, Chen X. WT1 recruits TET2 to regulate its target gene expression and suppress leukemia cell proliferation. Mol Cell. 2015; 57(4):662-673. https://doi.org/10.1016/j.molcel.2014.12.023PubMedPubMed CentralGoogle Scholar
Data Supplements
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.