The DBD-α4 helix of EWS::FLI is required for GGAA microsatellite binding that underlies genome regulation in Ewing sarcoma

Curation statements for this article:
  • Curated by eLife

    eLife logo

    eLife assessment

    This paper investigates how the EWS::FLI1 fusion protein organizes chromatin topology and regulates gene expression in an aggressive pediatric bone cancer known as Ewing sarcoma. The authors used the most recent genomics methodologies to provide a solid base of evidence for the role of a short alpha helix in the DNA binding domain of FLI1 in modulating binding to GGAA microsatellites and promoting enhancer activity. The study provides valuable insight into the underlying oncogenic mechanisms in Ewing sarcoma, but is limited to a single cell line and would benefit from consolidation of the main conclusions using additional techniques.

This article has been Reviewed by the following groups

Read the full article See related articles

Abstract

Ewing sarcoma is the second most common bone cancer in children and young adults. In 85% of patients, a translocation between chromosomes 11 and 22 results in a potent fusion oncoprotein, EWS::FLI. EWS::FLI is the only genetic alteration in an otherwise unaltered genome of Ewing sarcoma tumors. The EWS portion of the protein is an intrinsically disordered domain involved in transcriptional regulation by EWS::FLI. The FLI portion of the fusion contains a DNA binding domain shown to bind core GGAA motifs and GGAA repeats. A small alpha-helix in the DNA binding domain of FLI, DBD- α 4 helix, is critical for the transcription function of EWS::FLI. In this study, we aimed to understand the mechanism by which the DBD- α 4 helix promotes transcription, and therefore oncogenic transformation. We utilized a multi-omics approach to assess chromatin organization, active chromatin marks, genome binding, and gene expression in cells expressing EWS::FLI constructs with and without DBD- α 4 helix. Our studies revealed DBD- α 4 helix is crucial for cooperative binding of EWS::FLI at GGAA microsatellites. This binding underlies many aspects of genome regulation by EWS::FLI such as formation of TADs, chromatin loops, enhancers and productive transcription hubs.

Article activity feed

  1. eLife assessment

    This paper investigates how the EWS::FLI1 fusion protein organizes chromatin topology and regulates gene expression in an aggressive pediatric bone cancer known as Ewing sarcoma. The authors used the most recent genomics methodologies to provide a solid base of evidence for the role of a short alpha helix in the DNA binding domain of FLI1 in modulating binding to GGAA microsatellites and promoting enhancer activity. The study provides valuable insight into the underlying oncogenic mechanisms in Ewing sarcoma, but is limited to a single cell line and would benefit from consolidation of the main conclusions using additional techniques.

  2. Reviewer #1 (Public Review):

    Summary:

    Ewing sarcoma is an aggressive pediatric cancer driven by the EWS-FLI oncogene. Ewing sarcoma cells are addicted to this chimeric transcription factor, which represents a strong therapeutic vulnerability. Unfortunately, targeting EWS-FLI has proven to be very difficult, and a better understanding of how this chimeric transcription factor works is critical to achieving this goal. Towards this perspective, the group had previously identified a DBD-𝛼4 helix (DBD) in FLI that appears to be necessary to mediate EWS-FLI transcriptomic activity. Here, the authors used multi-omic approaches, including CUT&tag, RNAseq, and MicroC to investigate the impact of this DBD domain. Importantly, these experiments were performed in the A673 Ewing sarcoma model where endogenous EWS-FLI was silenced, and EWS-FLI-DBD proficient or deficient isoforms were re-expressed (isogenic context). They found that the DBD domain is key to mediating EWS-FLI cis activity (at msat) and to generating the formation of specific TADs. Furthermore, cells expressing DBD-deficient EWS-FLI display very poor colony-forming capacity, highlighting that targeting this domain may lead to therapeutic perspectives.

    Strengths:

    The group has strong expertise in Ewing sarcoma genetics and epigenetics and also in using and analyzing this model (Theisen et al., 2019; Boone et al., 2021; Showpnil et al., 2022).

    They aim at better understanding how EWS-FLI mediated its oncogenic activity, which is critical to eventually identifying novel therapies against this aggressive cancer.

    They use the most recent state-of-the-art omics methods to investigate transcriptome, epigenetics, and genome conformation methods. In particular, Micro-C enables achieving up to 1kb resolved 3D chromatin structures, making it possible to investigate a large number of TADs and sub-TADs structures where EWS-FLI1 mediates its oncogenic activity.

    They performed all their experiments in an Ewing sarcoma genetic background (A673 cells) which circumvents bias from previously reported approaches when working in non-orthologous cell models using similar approaches.

    Weaknesses:

    The main weakness comes from the poor reproducibility of Micro-C data. Indeed, it appears that the distances/clustering observed between replicates are typically similar or even larger than between biological conditions. For instance, in Figure 1B, I do not see any clustering when considering DBD1, DBD2, DBD+1, DBD+2.

    Lanes 80-83: "KD replicates clustered together with DBD replicate 1 on both axes and with DBD replicate 2 on the y-axis. DBD+ replicates, on the other hand, clustered away from both KD and DBD replicates. These observations suggest that the global chromatin structure of DBD replicates is more similar to KD than DBD+ replicates."

    When replacing DBD replicate 1 with DBD replicate 2, their statement would not be true anymore.

    Additional replicates to clarify this aspect seem absolutely necessary since those data are paving the way for the entire manuscript.

    Similarly:
    - In Figure 1C, how would the result look when comparing DBD2/KD2/DBD+2? Same when comparing DBD 1 with KD1 and DBD+1. Would the difference go in the same direction?
    - Figure 1D-E. How would these plots look like when comparing each replicate to each other's? How much difference would be observed when comparing, for instance, DBD1/DBD2 ? or DBD1/DBD+1?
    - Figure 2: again, how would these analyses look like when performing the analysis with only DBD1/DBD+1/KD1 or DBD2/DBD+2/KD?

    Another major question is the stability of EWS-FLI DBD vs EWS-FLI DBD+ proteins. Indeed, it seems that they have more FLAG (i.e., EWS-FLI) peaks in the DBD+ condition compared to the DBD condition (Figure 2B). In the WB, FLAG intensities seem also higher (2/3 replicates) in DBD+ condition compared to the DBD condition (Figure S1B).

    Would it be possible that DBD+ is just more expressed or more stable than DBD? The higher stability of the re-expressed DBD+ could also partially explain their results independently of the 3D conformational change. In other words, can they exclude that DBD+ and DBD binding are not related to their respective protein stability or their global re-expression levels?

    Surprisingly, WB FLI bands in DBD+ conditions are systematically (3/3 replicates) fainter than in DBD conditions (Figure S1B). How do the authors explain these opposite results between FLI and FALG in the WB?

  3. Reviewer #2 (Public Review):

    Summary:

    The manuscript by Bayanjargal et al. entitled "The DBD-alpha4 helix of EWS::FLI is required for GGAA microsatellite binding that underlies genome regulation in Ewing sarcoma" reports on the critical role of a small alpha helix in the DNA binding domain (DBD) of the FLI1 portion of EWS::FLI1 that is critical for binding to repetitive stretches of GGAA-motifs, i.e. GGAA microsatellites, which serve as potent neoenhancers in Ewing sarcoma.

    Strengths:

    The paper is generally well-written, and easy to follow and the data presented are of high quality, well-described and underpin the conclusions of the authors. The report sheds new light on how EWS::FLI1 mechanistically binds to and activates GGAA microsatellite enhancers, which is of importance to the field.

    Weaknesses:

    While there are no major weaknesses in this paper, there are a few minor issues that the authors may wish to address:

    (1) While the official protein symbol for the gene EWSR1 is indeed EWS, the protein symbol for the gene FLI1 is identical, i.e. FLI1. The authors nominate the fusion oncoprotein EWS::FLI1 (even in the title) but it appears more adequate to use EWS::FLI1.

    (2) The used cell lines should be spelled according to their official nomenclature (e.g. A-673 instead of A673).

    (3) It appears as if the vast majority of results were generated in a single Ewing sarcoma cell line (A-673) which is an atypical Ewing sarcoma cell line harboring an activating BRAF mutation and may be genomically quite unstable as compared to other Ewing sarcoma cell lines (Kasan et al. 2023 preprint at bioRxiv https://www.biorxiv.org/content/10.1101/2023.11.20.567802v1). Hence, it may be supportive for the paper to recapitulate/cross-validate a few key results in other Ewing sarcoma cell lines, e.g. by using EWS::ERG-positive cell lines. Perhaps the authors could make use of available published data.

    (4) Figure 6 and Supplementary Figure 5 are very interesting but focus on two selected target genes of the fusion (FCGRT and CCND1). It would be interesting to see whether these findings also extend to common EWS::ETS transcriptional signatures that have been reported. The authors could explore their data and map established consensus EWS::ETS signatures to investigate which other hubs might be affected at relevant target genes.

    (5) Table 1 is a bit hard to read. In my opinion, it is not necessary to display P-values with up to 8 decimal positions. The gene symbols should be displayed in italic font.