Ribosome subunit attrition and activation of the p53–MDM4 axis dominate the response of MLL-rearranged cancer cells to WDR5 WIN site inhibition

  1. Gregory Caleb Howard
  2. Jing Wang
  3. Kristie L Rose
  4. Camden Jones
  5. Purvi Patel
  6. Tina Tsui
  7. Andrea C Florian
  8. Logan Vlach
  9. Shelly L Lorey
  10. Brian C Grieb
  11. Brianna N Smith
  12. Macey J Slota
  13. Elizabeth M Reynolds
  14. Soumita Goswami
  15. Michael R Savona
  16. Frank M Mason
  17. Taekyu Lee
  18. Stephen Fesik
  19. Qi Liu
  20. William P Tansey  Is a corresponding author
  1. Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, United States
  2. Department of Biostatistics, Vanderbilt University Medical Center, United States
  3. Center for Quantitative Sciences, Vanderbilt University Medical Center, United States
  4. Mass Spectrometry Research Center, Vanderbilt University School of Medicine, United States
  5. Department of Biochemistry, Vanderbilt University School of Medicine, United States
  6. Department of Medicine, Vanderbilt University Medical Center, United States
  7. Department of Pharmacology, Vanderbilt University School of Medicine, United States
  8. Department of Chemistry, Vanderbilt University, United States
6 figures, 1 table and 1 additional file

Figures

Figure 1 with 2 supplements
Impact of WIN site inhibitors (WINi) on the transcriptome of MLLr cancer cells.

(A) Chemical structures of C6 and C16. (B) Crystal structures of C6 or C16 bound to the WIN site of WDR5 with electrostatic surfaces mapped (PDB IDs: 6E23 [Aho et al., 2019a]; 6UCS [Tian et al., 2020]). The image shows a close-up view of the WIN site. (C) Superimposed WIN site-binding conformations of C6 (green) and C16 (blue). (D) Transcript levels as determined by QuantiGene analysis of representative WDR5-bound (color) or non-bound (grayscale) ribosomal protein genes in MV4;11 cells treated with a serial dilution range of either C6 (left) or C16 (right) and relative to DMSO-treated cells (n = 2–3; mean ± SEM). Vertical dashed line indicates either 2 µM C6 (left) or 100 nM C16 (right). (E) Number of genes with significantly (false discovery rate [FDR] < 0.05) altered transcript levels following treatment of MV4;11 cells with C6 (2 µM) or C16 (100 nM) for 48 hr, as determined by RNA-Seq (n = 3). See Figure 1—source data 1 for complete output of RNA-seq analysis. (F) Comparison of gene expression changes elicited by C6 (x-axis) and C16 (y-axis), represented as Log2 fold change (FC) compared to DMSO. WDR5-bound genes are colored red. Locations of RPL22L1 and ZMAT3 are indicated. (G) Overlap of genes with decreased (left) or increased (right) transcript levels in MV4;11 cells treated with C6 or C16. (H) Gene set enrichment analysis (GSEA) showing the distribution of genes suppressed in MV4;11 cells in response to C6 (left) or C16 (right) against the list of all genes bound by WDR5 in those cells (Aho et al., 2019a). NES, normalized enrichment score. (I) Enrichment analysis of genes suppressed (left) or induced (right) by C6 or C16 in MV4;11 cells. KEGG and Hallmark.MSigDB pathways are shown. Fold enrichment of indicated pathways is presented on the x-axis, the number of genes is shown in italics in each bar, and colors represent -Log10 FDR. See Figure 1—source data 2 for additional GSEA (Hallmark) and over-representation analysis (ORA) (Hallmark) analyses of differentially expressed genes. (J) Transcript level changes in WDR5-bound (left) and non-bound (right) RPGs elicited by C6 (top) or C16 (bottom).

Figure 1—source data 1

Output of RNA-seq analysis of MV4;11 cells treated with C6/C16.

https://cdn.elifesciences.org/articles/90683/elife-90683-fig1-data1-v1.xlsx
Figure 1—source data 2

GSEA Hallmark and over-representation analysis (ORA) Hallmark enrichment analysis of differentially expressed genes in RNA-seq.

https://cdn.elifesciences.org/articles/90683/elife-90683-fig1-data2-v1.xlsx
Figure 1—figure supplement 1
Transcript changes elicited by WIN site inhibitors (WINi) in MLLr cancer cells.

(A) Crystal structures of C6 or C16 bound to the WIN Site of WDR5 with electrostatic surfaces mapped (PDB IDs: 6E23 [Aho et al., 2019a]; 6UCS [Tian et al., 2020]). (B) Viabilities of MV4;11 cells treated with a serial dilution range of either C6 (left) or C16 (right) for 72 hr, relative to viability of DMSO-treated cells (n = 3; mean ± SEM). (C) As in (B) but for MOLM13 cells. (D) Transcript levels as determined by QuantiGene analysis of representative WDR5-bound (color) or non-bound (grayscale) ribosomal protein genes in MOLM13 cells treated with a serial dilution range of either C6 (left) or C16 (right) and relative to DMSO-treated cells (n = 3; mean ± SEM). Vertical dashed line indicates either 2 µM C6 (left) or 100 nM C16 (right). (E) Transformed z-scores of genes with significantly altered transcript levels (RNA-seq) in MV4;11 cells treated with either C6 (2 µM) or C16 (100 nM) for 48 hr, compared to DMSO treatment. (F) Volcano plots, showing transcript level alterations in MV4;11 cells treated 48 hr with 2 µM C6 (left) or 100 nM C16 (right) compared to DMSO (n = 3; red indicates false discovery rate [FDR] < 0.05). (G) Dispersion plot describing the variance in gene expression for the RNA-seq data in a previous study (left) and this study (right).

Figure 1—figure supplement 2
Impact of WIN site inhibitors (WINi) on RPL22L1 and p53 target gene expression.

(A) Venn diagram, showing the overlap of consensus p53 target genes (Fischer, 2017) with genes significantly induced by C6 or C16 in MV4;11 cells. (B) Graph showing the changes in expression of the 91 common genes in (A) elicited by WINi C6 (red) or C16 (blue) in MV4;11 cells, compared to DMSO. (C) Changes in expression (and false discovery rate [FDR]) of RPL22L1 elicited in response to C6 (red) or C16 (blue) treatment of K562 leukemia cells (Aho et al., 2019a) or five rhabdoid tumor cell lines (TTC642, KYM-1, G401, TM87-16, and TTC549; Florian et al., 2022).

Figure 2 with 3 supplements
Impact of WIN site inhibitors (WINi) on the translatome of MLLr cancer cells.

(A) Volcano plots depicting alterations in translation efficiency (TE) induced by 48 hr treatment of MV4;11 cells with either 2 µM C6 (left) or 100 nM C16 (right) compared to DMSO (n = 2; red indicates false discovery rate [FDR] < 0.05 and Log2 FC > 0.25), as determined by Ribo-seq. (B) Number of mRNAs with significantly (FDR < 0.05 and Log2 FC > 0.25) altered TE levels following treatment of MV4;11 cells with C6 (2 µM) or C16 (100 nM) for 48 hr. See Figure 2—source data 1 for complete output of Ribo-seq analysis. (C) Overlap of mRNAs with significantly decreased TE in response to C6 or C16 treatment. (D) TE of mRNAs in DMSO-treated MV4;11 cells plotted against translation efficiencies of mRNAs in cells treated with either C6 (left) or C16 (right). Red indicates mRNAs with significantly altered translation efficiencies following inhibitor treatment (FDR < 0.05 and Log2 FC > 0.25). (E) Numbers of differentially translated mRNAs (∆TE) in each quartile of genes (stratified by TE in DMSO) in cells treated with C6 (left) or C16 (right). (F) Enrichment analysis of common mRNAs suppressed by C6/C16 at the mRNA (blue) and translational (red; TE) level in MV4;11 cells. Hallmark.MSigDB pathways are shown. The x-axis indicates the number of suppressed genes in each category; the italic numbers are the corresponding FDR. See Figure 2—source data 2 for the full Hallmark.MSigDB analysis, as well as for Reactome and KEGG pathways. (G) Enrichment analysis of mRNAs suppressed translationally by C6/C16 but with no significant changes in mRNA levels. Gene Ontology (GO) Biological Process (BP) and Molecular Function (MF) categories are shown, as well as KEGG pathways. The x-axis displays -Log10 FDR; the number of mRNAs is shown in italics in each bar. See Figure 2—source data 3 for extended enrichment analyses, broken down by TE and mRNA direction changes. (H) TE changes in WDR5-bound (left) and non-bound (right) RPGs elicited by C6 (top) or C16 (bottom).

Figure 2—source data 1

Output of Ribo-seq analysis of MV4;11 cells treated with C6/C16.

https://cdn.elifesciences.org/articles/90683/elife-90683-fig2-data1-v1.xlsx
Figure 2—source data 2

Hallmark, Reactome, and KEGG enrichment analysis of differentially translated genes in Ribo-seq.

https://cdn.elifesciences.org/articles/90683/elife-90683-fig2-data2-v1.xlsx
Figure 2—source data 3

Enrichment analysis of differentially translated genes, broken down by mRNA level change direction.

https://cdn.elifesciences.org/articles/90683/elife-90683-fig2-data3-v1.xlsx
Figure 2—figure supplement 1
WIN site inhibitors (WINi) suppress bulk protein synthesis.

(A) Representative histograms from protein synthesis assays in MV4;11 cells treated 24, 48, or 96 hr with either 0.1% DMSO (blue), 2 µM C6 (red), or 100 nM C16 (orange). Cells were pulsed with O-propargyl-puromycin (OPP) to label nascent proteins, Alexa Fluor 647 linked to incorporated OPP in Click chemistry reactions, and fluorescence measured by flow cytometry analysis. MV4;11 cells treated 30 min with 100 µg/mL cycloheximide (‘CHX’; green) serve as a positive control for inhibited protein synthesis. MV4;11 cells pulsed with DMSO (‘No OPP’; black) serve as a control for background fluorescence. (B) Quantification of protein synthesis assays. Fluorescence from CHX-treated cells was set as the baseline, and fluorescence presented relative to DMSO-treated (DM) cells at each time point (n = 3; normalized geometric mean ± SEM). p-Values calculated by Student’s t-tests are represented by asterisks: *<0.05, **<0.01, ***<0.001.

Figure 2—figure supplement 2
WIN site inhibitors (WINi) suppress translation.

(A) Distribution of ribosome-protected fragment (RPF) lengths in each Ribo-seq sample/replicate. The length distribution of RPFs in mammalian Ribo-seq experiments typically peaks at 30–31 nucleotides. (B) Proportion of RPFs mapping to the coding sequence (CDS) or 5′ or 3′ untranslated regions (UTR) of transcripts. Color of dots is the same as in (A). (C) Proportion of RPFs mapping to each reading frame in the 5′ UTR (left), the CDS (middle), and the 3′ UTR (right). Color of dots is the same as in (A). (D) Magnitudes of significant translation efficiency (TE) alterations of mRNAs in each quartile (stratified by TE in DMSO) in cells treated with C6 (left) or C16 (right). Color dots represent individual genes. Bottom, middle, and top horizontal lines of each box represent first, second, and third quartiles, respectively. Vertical lines extend to data points within 1.5-fold of the interquartile range. Black dots represent values beyond 1.5-fold of the interquartile range. (E) Changes in TE induced by C6 (left) or C16 (right) in MV4;11 cells, binned according to mRNA TOPscores (Philippe et al., 2020). Dashed lines represent the median; dotted lines indicate quartiles. Significance by t-test is indicated compared to group with TOPscore 0–1 (*≤0.05, **≤0.0001). (F) UpSet plot, showing the breakdown of genes encoding PRMT5 substrates Radzisheuskaya et al., 2019 whose transcript levels and/or translation efficiencies decrease following WIN site inhibition (p-value calculated by hypergeometric test for over-representation of genes encoding PRMT5 substrates in genes with decreased translation efficiencies). (G) Overlap of C6/C16 common mRNAs with decreased abundance (RNA; blue) and those with decreased TE (salmon), grouped according to the indicated Hallmark.MSigDB categories. (H) Overlap of all C6/C16 common mRNAs with altered abundance and decreased TE. (I) The top row of the heatmap displays the codon stability coefficient (CSC) for each codon (Wu et al., 2019) ranked from lowest ('non-optimal') to highest ('optimal'). The middle row displays enrichment of each codon in mRNAs that are decreased at both the TE and mRNA levels (RNA + TE) versus those that show a decrease in TE without an accompanying decrease in mRNA abundance (TE only). Bottom row is -Log10 false discovery rate (FDR).

Figure 2—figure supplement 3
WIN site inhibitors (WINi) impair translation of mitochondrial ribosomal proteins.

(A) Top: transcript level changes in mitochondrial ribosomal protein genes elicited by C6 or C16, as indicated. Bottom: translational efficiency (TE) changes in mitochondrial ribosomal protein genes elicited by C6 or C16. All of the mitochondrial RPGs are nuclear-encoded; none have detectable binding of WDR5.

Figure 3 with 4 supplements
Impact of WIN site inhibitors (WINi) on the ribosome inventory of MLLr cancer cells.

(A) Lysates from MV4;11 cells treated 24 or 72 hr with either 0.1% DMSO or 250 nM C16 were subjected to liquid chromatography coupled with tandem mass spectrometry and analyzed by label-free quantification (LFQMS). The table shows the number of proteins detected in DMSO and C16 samples and those with significantly altered levels at each time point (n = 4; adj. p-value<0.05). See Figure 3—source data 1 for complete output of LFQMS analysis. (B) Volcano plot, showing protein level alterations in cells treated with C16 for 24 hr (red indicates adj. p-value<0.05). The location of RPL22L1 is indicated. (C) As in (B) but for 72 hr treatment with C16. (D) Overlap of proteins significantly increased (top) or decreased (bottom) following 24 or 72 hr C16 treatment. (E) Protein level alterations induced by C16 in consensus p53 target proteins (Fischer, 2017) at the 24 and 72 hr treatment timepoints. Those proteins only altered in abundance at 24 hr are represented as blue dots; proteins only altered at 72 hr are red; proteins altered at both timepoints are gray. (F) As in (E) but for ribosomal proteins. (G) Changes in expression of proteins encoded by WDR5-bound (left) and non-bound (right) RPGs elicited by 24 (top) or 72 (bottom) hr treatment with C16. Note that, due to the magnitude of change, Log2(FC) for RPL22L1 is presented on a separate scale.

Figure 3—source data 1

Output of label-free quantitative mass spectrometry (LFQMS) analysis of MV4;11 cells treated with C16.

https://cdn.elifesciences.org/articles/90683/elife-90683-fig3-data1-v1.xlsx
Figure 3—source data 2

Enrichment analysis of proteins altered in abundance by 24 or 72 hr of C16 treatment.

https://cdn.elifesciences.org/articles/90683/elife-90683-fig3-data2-v1.xlsx
Figure 3—figure supplement 1
Distribution of peptide/protein intensities in label-free quantitative mass spectrometry (LFQMS) analysis.

(A) Peptide intensities of all proteins detected in each mass spectrometry run before (left) and after normalization (right). (B) Magnitudes of significant protein level alterations within each decile (stratified by protein intensity in DMSO samples) in MV4;11 cells treated 24 hr with 250 nM C16. Red points represent ribosome proteins. (C) Box plot representation of data presented in (B). Bottom, middle, and top horizontal line of each box represents first, second, and third quartiles, respectively. Vertical lines extend to data points within 1.5-fold of the interquartile range. Black dots represent values beyond 1.5-fold of the interquartile range. (D) Number of proteins increased or decreased within each decile.

Figure 3—figure supplement 2
Enrichment analysis of proteins with altered expression in response to C16 treatment.

(A) Graphs showing enrichment of proteins in GO BP (top) and Hallmark.MSigDB (bottom) pathways that are induced by C16 treatment at 24 (blue) or 72 (green) hr. The x-axis displays -Log10 (FDR); the number of proteins in each category is given in italics. (B) Graphs showing enrichment of proteins in Gene Ontology (GO) Biological Process (BP) (top) and Hallmark.MSigDB (bottom) pathways that are suppressed by C16 treatment at 24 (red) or 72 (brown) hr. The x-axis displays -Log10 (FDR); the number of proteins in each category is given in italics. See Figure 3—source data 2 for output of the full enrichment analyses.

Figure 3—figure supplement 3
WIN site inhibitors suppress rRNA levels.

(A) In-gel fluorescence assay detecting metabolically labeled rRNA (top) isolated from MV4;11 cells treated 24, 48, or 96 hr with either DMSO (0.1%), C6 (2 µM), or C16 (100 nM), and pulsed with 2′-azido-2′-cytidine (AzCyd). As a positive control for inhibited rRNA synthesis, MV4;11 cells were treated 1 hr with 5 nM actinomycin D (‘ActD’). As a control for background labeling, MV4;11 cells were pulsed with DMSO (‘No AzCyd’). Fluorescent probes were covalently linked to incorporated AzCyd in Click chemistry reactions. Total RNA (bottom) was detected by SYBR stain. (B) Quantification of metabolic rRNA labeling (n = 3; mean ± SEM). p-Values calculated by Student’s t-tests are indicated. Raw unprocessed gel images are presented in Figure 3—figure supplement 3—source data 1.

Figure 3—figure supplement 4
C16 induces redistribution of nucleophosmin from the nucleolus to the nucleoplasm.

(A) Representative immunofluorescent images of MV4;11 cells treated with vehicle (DMSO control), C16 (100 nM), or ActD (5 nM) for the times indicated and stained for nucleophosmin (NPM1, green), gH2A.X pSer139 (double-stranded break marker, magenta) and Hoechst (blue). DNA damage arises upon cell death following drug treatment. Scale bars are 5 μm. (B) Quantification of the ratio of nucleolar to total NPM1 in the cells described in (A). C6, C16, and ActD treatment disrupted NPM1 localization. p-Values calculated by Student’s t-tests are represented by asterisks: **0.0012, ****<0.0001.

Figure 4 with 1 supplement
A two-tier loss-of-function screen for modulators of the response to WIN site inhibitors (WINi).

(A) Two-tier screen design. In the first tier, Cas9-expressing MV4;11 cells were transduced with a genome-wide sgRNA library and treated with 2 µM C6 until a resistant cell population emerged. sgRNA representation in the pretreatment population was compared to the post-treatment population (n = 2). In the second tier, cells were transduced with a custom library of distinct sgRNAs targeting non-pan-essential ‘hits’ from the first tier, cultured in the presence of DMSO, C6, or C16, and sgRNA representation in C6/C16-treated cultures compared to that from DMSO-treated cultures (n = 2). (B) Volcano plot, showing gene-level changes in sgRNA representation from the first tier (orange indicates false discovery rate [FDR] < 0.05). Datapoints corresponding to TP53, RPL22, and CDKN2A are indicated. See Figure 4—source data 1 for full output of the tier 1 screen. (C) Comparison of gene-level changes in sgRNA representation in C6- and C16-treated populations in the second tier screen, each compared to DMSO-treated populations (red indicates FDR < 0.05; black indicates non-targeting control sgRNAs). See Figure 4—source data 2 for full output of the tier 2 screen. (D) Top: overlap of genes from the tier 2 screen with enriched (left) or depleted (right) sgRNAs in C6- and C16-treated MV4;11 populations, compared to the DMSO control. Bottom: overlap of genes with enriched (left) or depleted (right) sgRNAs in the first versus second tiers of the screen. ‘Tier 1’ contains only those genes targeted in the tier 2 screen. ‘Tier 2’ contains the intersection of genes with altered sgRNAs in both the C6 and C16 treatments. (E) Ranked heatmap, representing the mean gene-level Log2 fold change (FC) of sgRNAs from the C6 and C16 treatments in the tier 2 screen, as well as gene enrichment analysis outputs. Note that ‘Signal transduction by p53 class mediator’ is a GO:BP term (orange); ‘p53’ assignments (yellow) were added by manual curation.

© 2024, BioRender Inc. Figure 4A was created using BioRender, and is published under a CC BY-NC-ND license. Further reproductions must adhere to the terms of this license.

Figure 4—figure supplement 1
Genome-wide CRISPR screen identifies genes that influence response to C6/C16.

(A) Tier 1 screen: daily cell counts of MV4;11 Cas9 and MV4;11 Cas9+GeCKOv2 (Library) populations treated with either DMSO or 2 µM C6. The two replicates of this screen are shown separately. (B) Normalized counts of each sgRNA (x-axis) in the GeCKOv.2 library targeting TP53 in the initial transduced cells (red; not visible on this scale) and the C6-treated population (blue). Data represents means of replicates; * indicates false discovery rate (FDR) < 0.05. (C) As in (B) but for sgRNAs targeting CDKN2A. (D) Schematic of the CDKN2A gene locus with indicated sites complementary to tier 1 and tier 2 screen sgRNAs. Red sgRNAs increase in representation in CRISPR screens. (E) miRNet 2.0 (Chang and Xia, 2023) analysis of the 27 miRNAs enriched in the tier 1 screen produced a single significant hit corresponding to the KEGG p53 signaling pathway. The miRNAs are represented as blue boxes and target genes as red circles; the connections between them are indicated. (F) As in (B) but for sgRNAs targeting RPL22. (G) Volcano plots, showing gene-level changes from the tier 2 screen in sgRNA representation in C6- (left) and C16- (right) treated populations compared to DMSO control cultures (orange indicates FDR < 0.05). (H) Graph depicting gene-level Log2 FC and FDR values for genes that were flagged as C6- (squares) or C16- (circles) specific in the tier 2 screen. (I) GO enrichment analysis of the 57 C6/C16 common genes emerging from tier 2 of the screen. Italics represent the number of genes in each category.

Figure 5 with 3 supplements
Identification of agents that synergize with WIN site inhibitors (WINi) in MLLr cells.

(A) Peak synergy (>0) and antagonism (<0) zero interaction potency (ZIP) delta (δ) scores from synergy assays in which MV4;11 cells were treated for 3 d with 49 unique dose combinations of C16 and the indicated compound of interest (n = 4). See Figure 5—source data 1 for numerical ZIP delta analysis output. (B) Heatmaps of MV4;11 cell growth inhibition at each dose of C16 and the indicated six compounds. The remaining five combinations tested are shown in Figure 5—figure supplement 1. (C) As in (A) but for MOLM13 cells. See Figure 5—source data 1 for numerical ZIP delta analysis output. (D) As in (B) but for MOLM13 cells. The remaining five combinations tested are shown in Figure 5—figure supplement 2. (E) Number of genes with significantly (false discovery rate [FDR] < 0.05) altered transcript levels following treatment of MV4;11 cells with C16 (100 nM), mivebresib (Mibv; 2.5 nM), or the combination for 48 hr, as determined by RNA-seq (n = 3). See Figure 5—source data 2 for complete output of RNA-seq analysis. (F) UpSet plot, showing the overlap of genes suppressed (left) or induced (right) in response to C16, mivebresib, or the combination. (G) UpSet plot, showing the breakdown of Reactome ‘Translation’ pathway genes suppressed in response to C16, mivebresib, or the combination. (H) Enrichment of Reactome Pathways in genes with increased transcripts following treatment of MV4;11 cells with C16, mivebresib, or the combination. See Figure 5—source data 3 for complete output of enrichment analyses.

Figure 5—source data 1

Peak synergy and antagonism scores for MV4:11 and MOLM13 cells treated with C16 in combination with 11 agents.

https://cdn.elifesciences.org/articles/90683/elife-90683-fig5-data1-v1.xlsx
Figure 5—source data 2

Output of RNA-seq analysis of MV4;11 cells treated with C16, mivebresib, or both.

https://cdn.elifesciences.org/articles/90683/elife-90683-fig5-data2-v1.xlsx
Figure 5—source data 3

Enrichment analysis of differentially expressed genes in RNA-seq of MV4;11 cells treated with C16, mivebresib, or both.

https://cdn.elifesciences.org/articles/90683/elife-90683-fig5-data3-v1.xlsx
Figure 5—figure supplement 1
C16 is synergistic with multiple agents in MV4;11 cells.

(A) Heatmaps of MV4;11 cell growth inhibition at each dose of C16 and the indicated five compounds. (B) Heatmaps of δ scores from MV4;11 cells at each dose combination of C16 and the indicated agents.

Figure 5—figure supplement 2
C16 is synergistic with multiple agents in MOLM13 cells.

(A) Heatmaps of MOLM13 cell growth inhibition at each dose of C16 and the indicated five compounds. (B) Heatmaps of δ scores from MOLM13 cells at each dose combination of C16 and the indicated agents.

Figure 5—figure supplement 3
Impact of C16 and mivebresib on RPG and p53 target gene expression.

(A) Transcript level changes in WDR5-bound (left) and non-bound (right) RPGs elicited by C16 (top), mivebresib (Mivb; middle), or the combination (bottom). (B) Heatmap, showing significant changes in the expression of consensus p53 target genes (Fischer, 2017) induced by C16, mivebresib (Mivb; middle) or the combination (bottom) in MV4;11 cells.

Figure 6 with 3 supplements
WIN site inhibitors (WINi) inactivate MDM4 in an RPL22-dependent manner.

(A) Differential alternative splicing events affected by C6/C16 treatment of MV4;11 cells were quantified by rMATS. The types of alternative splicing events are cartooned at left, and the number of significantly different events (>5% ∆ψ; false discovery rate [FDR] < 0.05) common to C6/C16 depicted in the graph. See Figure 6—source data 2 for output of rMATS analysis. (B) Sashimi plot quantifying read junctions that span exons 5–7 of MDM4 in MV4;11 cells treated with DMSO (green) or C16 (blue). Numbers in the arcs display junction depth. The location of exons 5, 6, and 7 is depicted at the bottom; skipped exon 6 is highlighted in orange. (C) Viabilities of control (non-targeting: NT) and RPL22 knock out (KO) MV4;11, MOLM13, and K562 cells treated with a serial dilution range of C16 for 72 hr, relative to viability of DMSO-treated cells (n = 3; mean ± SEM). (D) Western blot analysis of p53 levels in control (NT) and RPL22 knockout (KO) MV4;11 and MOLM13 cells treated with either 0.1% DMSO or C16 (MV4;11, 200 nM; MOLM13, 400 nM) for 72 hr. α-Actinin is loading control. Representative images from three biological replicates shown. Raw unprocessed gel images are presented in Figure 6—source data 5. (E) Heatmap, showing significant changes in the expression of consensus p53 target genes (Fischer, 2017) between the indicated pairwise comparisons of RNA-seq datasets. Note that only consensus p53 target genes altered in expression by C16 in control (NT) cells are represented. (F) Sashimi plot quantifying read junctions that span exons 5–7 of MDM4 in RPL22KO MV4;11 cells treated with DMSO or C16. Numbers in the arcs display junction depth. The location of exons 5, 6, and 7 is depicted at the bottom; skipped exon 6 is highlighted in orange. Corresponding NT images are presented alongside RPL22KO images in Figure 6—figure supplement 3B. (G) Western blots, comparing the effects of 72 hr of DMSO (DM) or C16 treatment (MV4;11, 200 nM; MOLM13, 400 nM) of control (NT) or RPL22 knockout (KO) MV4;11 (left) or MOLM13 (right) cells on levels of MDM4, p21, RPL22L1, RPL22, and GAPDH (loading control). Representative images from three biological replicates are shown. Raw unprocessed gel images are presented in Figure 6—source data 9.

Figure 6—source data 1

Raw unprocessed gel images corresponding to Figure 6—figure supplement 1A.

https://cdn.elifesciences.org/articles/90683/elife-90683-fig6-data1-v1.pdf
Figure 6—source data 2

Output of rMATS analysis of MV4;11 cells treated with C6/C16.

https://cdn.elifesciences.org/articles/90683/elife-90683-fig6-data2-v1.xlsx
Figure 6—source data 3

Raw unprocessed gel images corresponding to Figure 6—figure supplement 1F.

https://cdn.elifesciences.org/articles/90683/elife-90683-fig6-data3-v1.pdf
Figure 6—source data 4

Raw unprocessed gel images corresponding to Figure 6—figure supplement 2A.

https://cdn.elifesciences.org/articles/90683/elife-90683-fig6-data4-v1.pdf
Figure 6—source data 5

Raw unprocessed gel images corresponding to Figure 6D.

https://cdn.elifesciences.org/articles/90683/elife-90683-fig6-data5-v1.pdf
Figure 6—source data 6

Output of RNA-seq analysis of NT and RPL22KO MV4;11 cells treated with C16.

https://cdn.elifesciences.org/articles/90683/elife-90683-fig6-data6-v1.xlsx
Figure 6—source data 7

GSEA Hallmark and GOBP enrichment analysis of differentially expressed genes in RNA-seq of NT and RPL22KO MV4;11 cells treated with C16.

https://cdn.elifesciences.org/articles/90683/elife-90683-fig6-data7-v1.xlsx
Figure 6—source data 8

Output of rMATS analysis of NT and RPL22KO MV4;11 cells treated with C16.

https://cdn.elifesciences.org/articles/90683/elife-90683-fig6-data8-v1.xlsx
Figure 6—source data 9

Raw unprocessed gel images corresponding to Figure 6G.

https://cdn.elifesciences.org/articles/90683/elife-90683-fig6-data9-v1.pdf
Figure 6—figure supplement 1
WIN site inhibitors (WINi) alter the abundance of alternatively spliced mRNA isoforms.

(A) Western blots comparing the effects of 72 hr DMSO (DM) or C16 treatment of MV4;11 (top) or MOLM13 (bottom) cells on levels of p53 and GAPDH (loading control). Representative images from three biological replicates are shown. Raw unprocessed gel images are presented in Figure 6—source data 1. (B) Differential alternative splicing events impacted by C6 (red) or C16 (blue) treatment of MV4;11 cells were quantified by rMATS. The number of significantly different events (>5% ∆ψ; false discovery rate [FDR] < 0.05) for each WINi are depicted in the graph. ‘RI’ is retained intron; ‘MEX’ is mutually exclusive exons; ‘A3SS’ is alternative 3' splice site; ‘A5SS’ is alternative 5' splice site; ‘SE’ is skipped exon. See Figure 6—source data 2 for output of rMATS analysis. (C) Sashimi plot quantifying read junctions that span exons 12–17 of KRBA1 in MV4;11 cells treated with DMSO (green) or C16 (blue). Numbers in the arcs display junction depth. Skipped exon 15 is highlighted in orange. (D) As in (C) but for read junctions that span exons 20–22 of TTF2. Skipped exon 21 is highlighted in orange. (E) As in (C), but for read junctions that span exons 2 and 3 of RPL22L1. The location of exons 2 and 3 is depicted at the bottom. Splicing of exon 2 to the distal acceptor site in exon 3 results in an mRNA encoding RPL22L1a (orange); splicing to the proximal acceptor site in exon 3 results in an mRNA encoding RPL22L1b (yellow). (F) Left: representation of amplicons used to discriminate between different MDM4 (top) and RPL22L1 (bottom) isoforms via semi-quantitative PCR. Right: results of semi-quantitative PCR analysis for the various isoforms of MDM4 and RPL22L1, and a GAPDH control, in MV4;11 or MOLM13 cells treated for 48 hr with DMSO or C16 (MV4;11, 100 nM; MOLM13, 250 nM; n = 3). All three biological replicates for DMSO and C16 are shown. Raw unprocessed gel images for the data in (F) are presented in Figure 6—source data 3. (G) Left: representation of amplicons used to discriminate between MDM4 (top) and RPL22L1 (bottom) isoforms via RT-qPCR. Right: results of RT-qPCR analysis for the various isoforms of MDM4 (top) and RPL22L1 (bottom) in MV4;11 or MOLM13 cells treated for 48 hr with DMSO or C16 (MV4;11, 100 nM; MOLM13, 250 nM; n = 3; mean ± SEM). For each amplicon, isoform levels are expressed relative to the DMSO control. p-Values calculated by Student’s t-tests are represented by asterisks: *≤0.05, **≤0.01, ***≤0.001.

Figure 6—figure supplement 2
Impact of RPL22 loss on the response of MLLr cells to WIN site inhibitors (WINi).

(A) Western blot analysis of RPL22 expression in MV4;11, MOLM13, and K562 cells electroporated with Cas9 and either scrambled non-targeting (NT) control or RPL22-targeting sgRNAs. GAPDH and α-actinin are loading controls. Representative images from three biological replicates shown. Raw unprocessed gel images are presented in Figure 6—source data 4. (B) GI50 values of C16 in non-targeted (NT) and RPL22 knock out (KO) MV4;11, MOLM13, and K562 cells in a 72 hr assay (n = 3; mean ± SEM). (C) Number of genes with significantly (false discovery rate [FDR] < 0.05) altered transcript levels following treatment of RPL22KO or control (NT) cells treated with DMSO or 100 nM C16 for 48 hr, as determined by RNA-seq (n = 4). See Figure 6—source data 6 for complete output of RNA-seq analysis. (D) Volcano plots, showing pairwise transcript level alterations in NT (control) and RPL22KO MV4;11 cells treated 48 hr with DMSO or 100 nM C16 (red indicates FDR < 0.05). The locations of transcripts from ZMAT3 and RPL22L1 are indicated. (E) Transcript level changes in WDR5-bound (left) and non-bound (right) RPGs in each of the indicated pairwise comparisons of RNA-seq datasets. (F) Enrichment analysis of genes differentially induced by C16 in RPL22KO cells compared to control (NT) cells. KEGG and Hallmark.MSigDB pathways are shown. Fold enrichment of indicated pathways is presented on the x-axis, the number of genes is shown in italics in each bar, and colors represent -Log10 FDR. See Figure 6—source data 7 for complete enrichment analyses. (G) As in (F) but for suppressed genes. (H) Transcript level changes in mitochondrial ribosomal protein genes elicited by C16 in NT or RPL22KO cells.

Figure 6—figure supplement 3
Impact of RPL22 loss on the abundance of alternatively-spliced mRNA isoforms in MV4;11 cells.

(A) Differential alternative splicing events affected by C16 treatment of control (NT) or RPL22 knockout (KO) MV4;11 cells were quantified by rMATS. The types of alternative splicing events are cartooned at left, and the number of significantly different events (>5% ∆ψ; false discovery rate [FDR] < 0.05) depicted in the graph. See Figure 6—source data 8 for output of rMATS analysis. (B) Sashimi plot quantifying read junctions that span exons 5–7 of MDM4 in NT or RPL22KO MV4;11 cells treated with DMSO or C16. Numbers in the arcs display junction depth. The location of exons 5, 6, and 7 is depicted at the bottom; skipped exon 6 is highlighted in orange. Note that RPL22KO images are also represented in Figure 6F. (C) As in (B) but for read junctions that span exons 2 and 3 of RPL22L1. The location of exons 2 and 3 is depicted at the bottom. Splicing of exon 2 to the distal acceptor site in exon 3 results in an mRNA encoding RPL22L1a (orange); splicing to the proximal acceptor site in exon 3 results in an mRNA encoding RPL22L1b (yellow).

Tables

Appendix 1—key resources table
Reagent type (species) or resourceDesignationSource or referenceIdentifiersAdditional information
Gene (Homo sapiens)RPL22NAENSEMBL:ENSG00000116251
Gene (H. sapiens)RPL22L1NAENSEMBL:ENSG00000163584
Gene (H. sapiens)MDM4NAENSEMBL:ENSG00000198625
Strain, strain background (Escherichia coli)Endura ElectroCompetent CellsLucigenCat# 60242-2
Cell line (H. sapiens)‘MV4;11’ATCCCat# CRL-9591; RRID:CVCL_0064
Cell line (H. sapiens)‘MV4;11 NT’This studyNASee ‘Generation of RPL22-null cell lines’
Cell line (H. sapiens)‘MV4;11 RPL22 KO’This studyNASee ’Generation of RPL22-null cell lines’
Cell line (H. sapiens)‘MV4;11 Cas9’This studyNASee ‘Generation of Cas9-expressing MV4;11 cells’
Cell line (H. sapiens)MOLM13DMSZCat# ACC554; RRID:CVCL_2119
Cell line (H. sapiens)MOLM13 NTThis studyNASee ‘Generation of RPL22-null cell lines’
Cell line (H. sapiens)MOLM13 RPL22 KOThis studyNASee ‘Generation of RPL22-null cell lines’
Cell line (H. sapiens)K562ATCCCat# CCL-243; RRID:CVCL_0004
Cell line (H. sapiens)K562 NTThis studyNASee ‘Generation of RPL22-null cell lines’
Cell line (H. sapiens)K562 RPL22 KOThis studyNASee ‘Generation of RPL22-null cell lines’
Cell line (H. sapiens)HEK293TATCCCat# CRL-11268; RRID:CVCL_1926
AntibodyAnti-p53 (DO-1) (mouse monoclonal)Santa Cruz BiotechnologyCat# sc-126; RRID:AB_628082(1:2000)
AntibodyAnti-RPL22 (52) (mouse monoclonal)Santa Cruz BiotechnologyCat# sc-136413; RRID:AB_10658965(1:1000)
AntibodyAnti-RPL22L1 (rabbit polyclonal)Thermo Fisher ScientificCat# PA5-63266; RRID:AB_2646731(1:1000)
AntibodyAnti-MDMX (mouse monoclonal)Sigma-AldrichCat# M0445; RRID:AB_532256(1:1000)
AntibodyAnti-p21 Waf1/Cip1 (12D1) (rabbit monoclonal)Cell Signaling TechnologyCat# 2947; RRID:AB_823586(1:1000)
AntibodyAnti-⍺-actinin (HRP) (rabbit monoclonal)Cell Signaling TechnologyCat# 12413; RRID:AB_2797903(1:1000)
AntibodyAnti-GAPDH (HRP) (rabbit monoclonal)Cell Signaling TechnologyCat# 8884; RRID:AB_11129865(1:2000)
AntibodyAnti-nucleophosmin (mouse monoclonal)AbcamCat# ab10530; RRID:AB_297271(1:500)
AntibodyAnti-Phospho-Histone H2A.X (Ser139) (20E3) (rabbit monoclonal)Cell Signaling TechnologyCat# 9718; RRID:AB_2118009(1:250)
AntibodyGoat anti-mouse IgG (H+L) Cross-Adsorbed Secondary Antibody, Alexa Fluor 488Thermo Fisher ScientificCat# A-11001; RRID:AB_2534069(1:500)
AntibodyGoat anti-rabbit IgG (H+L) Highly Cross-Adsorbed Secondary Antibody, Alexa Fluor 594Thermo Fisher ScientificCat# A-11037; RRID:AB_2534095(1:500)
AntibodyAnti-GAPDH (HRP) (rabbit monoclonal)Cell Signaling TechnologyCat# 8884; RRID:AB_11129865(1:2000)
AntibodyGoat anti-mouse IgG, Light chain specific (HRP)Jackson ImmunoResearch Laboratories, IncCat# 115-035-174; RRID:AB_2338512(1:5000)
AntibodyGoat anti-rabbit IgG, HRP-linked antibodyCell Signaling TechnologyCat# 7074; RRID:AB_2099233(1:5000)
Recombinant DNA reagentlentiCas9-BlastPMID:25075903Addgene plasmid# 52962; RRID:Addgene_52962
Recombinant DNA reagentpsPAX2AddgeneAddgene plasmid# 12260; RRID:Addgene_12260
Recombinant DNA reagentpMD2.GAddgeneAddgene plasmid# 12259; RRID:Addgene_12259
Recombinant DNA reagentHuman GeCKOv2 CRISPR
Knockout Pooled Library
(A+B) in lentiGuide-PURO
PMID:25075903Addgene plasmid# 1000000048
Recombinant DNA reagentlentiGuide-PUROPMID:25075903Addgene plasmid# 52963; RRID:Addgene_52963
Commercial assay or kitcOmplete, EDTA-free, Protease Inhibitor CocktailRocheCat# 11873580001
Commercial assay or kitPhosSTOPRocheCat# 4906837001
Commercial assay or kitPefabloc SCRocheCat# 11429868001
Commercial assay or kitTURBO DNase (2 U/µL)InvitrogenCat# AM2238
Commercial assay or kitRNase I, E. coliLucigenCat# N6901K
Commercial assay or kitSUPERaseIn RNase InhibitorInvitrogenCat# AM2694
Commercial assay or kitTRIzol ReagentInvitrogenCat# 15596018
Commercial assay or kitSYBR Gold Nucleic Acid Gel StainInvitrogenCat# S11494
Commercial assay or kitT4 Polynucleotide KinaseNew England BioLabsCat# M0201S
Commercial assay or kitT4 RNA Ligase 2, truncated K227QNew England BioLabsCat# M0351S
Commercial assay or kitSuperScript III Reverse TranscriptaseInvitrogenCat# 18080085
Commercial assay or kitRandom HexamersInvitrogenCat# N8080127
Commercial assay or kitCircLigase II ssDNA LigaseLucigenCat# CL9021K
Commercial assay or kitLD-DithiothreitolMilliporeSigmaCat# D9779
Commercial assay or kitIodoacetamideMilliporeSigmaCat# I1149
Commercial assay or kito-Phosphoric acid, 85%Fisher ScientificCat# A260-500
Commercial assay or kitWater, Optima LC/MS GradeFisher ScientificCat# W6-4
Commercial assay or kitMethanol, Optima LC/MS GradeFisher ScientificCat# A456
Commercial assay or kitTriethylammonium bicarbonate bufferMilliporeSigmaCat# T7408
Commercial assay or kitTrypsin Gold, Mass Spectrometry GradePromegaCat# V5280
Commercial assay or kitFormic Acid, LC/MS GradeThermo Scientific PierceCat# 28905
Commercial assay or kitAcetonitrile, Optima LC/MS GradeFisher ScientificCat# A955-1
Commercial assay or kitPhusion High Fidelity DNA polymeraseNew England BioLabsCat# M0530S
Commercial assay or kitBsmBIv2New England BioLabsCat# R0739S
Commercial assay or kitDNase I (RNase-free)New England BioLabsCat# M0303S
Commercial assay or kitRNA Clean and Concentrator-25Zymo ResearchCat# R1017
Commercial assay or kitRNasin Ribonuclease InhibitorPromegaCat# N2515
Commercial assay or kitSYBR Safe DNA Gel StainInvitrogenCat# S33102
Commercial assay or kitClick-iT Cell Reaction Buffer KitInvitrogenCat# C10269
Commercial assay or kitNeon Transfection System 10 µL KitThermo ScientificCat# MPK1096
Commercial assay or kitGene Knockout Kit v2 – human – RPL22SynthegoNA
Commercial assay or kitNegative Control, Scrambled
sgRNA#1, mod-sgRNA
SynthegoNA
Commercial assay or kitProLong Gold Antifade MountantThermo Fisher ScientificCat# P36934
Commercial assay or kitLipofectamine 3000 Transfection ReagentInvitrogenCat# L3000075
Commercial assay or kitQuantiGene Plex panelThermo Fisher ScientificNA
Commercial assay or kitQuantiGene Sample Processing Kit for cultured cellsThermo Fisher ScientificCat# QS0100
Commercial assay or kitQuantiGene Plex Assay kitsThermo Fisher ScientificCat# QP1013
Commercial assay or kitPierce BCA Protein Assay KitThermo ScientificCat# PI23225
Commercial assay or kitClarity Western ECL SubstrateBio-RadCat# 1705061
Commercial assay or kitQubit RNA High Sensitivity Assay KitInvitrogenCat# Q32852
Commercial assay or kitDirect-zol RNA MiniprepZymo ResearchCat# R2050
Commercial assay or kitRiboCop rRNA Depletion Kit V1.2LexogenCat# 037.24
Commercial assay or kitQuick-DNA MidiPrep Plus KitZymo ResearchCat# D4075
Commercial assay or kitNEBNext High Fidelity
2X PCR Master Mix
New England BioLabsCat# M0541L
Commercial assay or kitZymo-Spin V Columns with ReservoirZymo ResearchCat# C1016-25
Commercial assay or kitNucleoSpin Gel and PCR Clean-upMacherey-NagelCat# 740609.250
Commercial assay or kitGibson Assembly Master MixNew England BioLabsCat# E2611S
Commercial assay or kitNucleoBond Xtra Maxi EFMacherey-NagelCat# 740424.10
Commercial assay or kitQ5 DNA PolymeraseNew England BioLabsCat# M0491S
Commercial assay or kitTaq DNA Polymerase
with Standard Taq Buffer
New England BioLabsCat# M0273S
Commercial assay or kitSYBR Safe DNA Gel StainInvitrogenCat# S33102
Commercial assay or kitKAPA SYBR Fast qPCR
Master Mix (2×)
RocheCat# 07959397001
Commercial assay or kitCellTiter-Glo Luminescent
Cell Viability Assay
PromegaCat# G7572
Chemical compound, drugBlasticidin S Hydrochloride PowderResearch Products InternationalCat# B12200-0.05
Chemical compound, drugDMSOSigmaCat# D2650
Chemical compound, drugC6PMID:30865883N/A
Chemical compound, drugC16PMID:31858797N/A
Chemical compound, drug2’-Azido-2’-deoxycytidineBiosynthCat# NA05412
Chemical compound, drugActinomycin DCayman Chemical CompanyCat# 11421-10mg
Chemical compound, drugMB 680R DBCOVector LaboratoriesCat# CCT-1462
Chemical compound, drugCycloheximideResearch Products InternationalCat# C81040-1.0
Chemical compound, drugOPP (O-propargyl-puromycin)InvitrogenCat# C10459
Chemical compound, drugHoechst 33342Thermo Fisher ScientificCat# H3570
Chemical compound, drugAlexa Fluor 647 Azide,
Triethylammonium Salt
InvitrogenCat# A10277
Chemical compound, drugCycloheximideSigmaCat# C4859-1ML
Chemical compound, drugNutlin-3aCayman Chemical CompanyCat# 18585
Chemical compound, drugRapamycinMedChem ExpressCat# HY-10219
Chemical compound, drugPinometostatCayman Chemical CompanyCat# 16175
Chemical compound, drugHarmineSigma-AldrichCat# 286044
Chemical compound, drugMivebresibCayman Chemical CompanyCat# 21033
Chemical compound, drugVenetoclaxCayman Chemical CompanyCat# 16233
Chemical compound, drugEtoposideCayman Chemical CompanyCat# 12092
Chemical compound, drugOlaparibCayman Chemical CompanyCat# 10621
Chemical compound, drugVE-821Cayman Chemical CompanyCat# 17587
Chemical compound, drugPemrametostatSelleck ChemicalsCat# S8664
Chemical compound, drugAlvespimycinCayman Chemical CompanyCat# 11036
Software, algorithmRThe R FoundationRRID:SCR_001905https://www.r-project.org
Software, algorithmdrcRitz et al., 2015; Ritz and Streibig, 2021NAhttps://github.com/DoseResponse/drc
Software, algorithmCHOPCHOPLabun et al., 2019RRID:SCR_015723https://chopchop.cbu.uib.no
Software, algorithmcount_spacers.pyJoung et al., 2017; Joung, 2017NAhttps://github.com/fengzhanglab/Screening_Protocols_manuscript/blob/master/design_targeted_library.py
Software, algorithmSynergyFinder PlusZheng et al., 2022RRID:SCR_019318https://synergyfinder.org/
Software, algorithmPASWRUgarte et al., 2015; Arnholt, 2022NAhttps://github.com/cran/PASWR
Software, algorithmcutadaptMartin, 2011RRID:SCR_011841https://github.com/marcelm/cutadapt/
Software, algorithmsabreNARRID:SCR_011843https://github.com/najoshi/sabre
Software, algorithmbowtie2Langmead and Salzberg, 2012RRID:SCR_016368https://github.com/BenLangmead/bowtie2
Software, algorithmSTARDobin et al., 2013RRID:SCR_004463https://github.com/alexdobin/STAR
Software, algorithmUMI-toolsSmith et al., 2017RRID:SCR_017048https://github.com/CGATOxford/UMI-tools
Software, algorithmBEDToolsQuinlan and Hall, 2010RRID:SCR_006646https://github.com/arq5x/bedtools2
Software, algorithmXtailXiao et al., 2016; xryanglab, 2016NAhttps://github.com/xryanglab/xtail
Software, algorithmriboWaltzLauria et al., 2018RRID:SCR_016948https://github.com/LabTranslationalArchitectomics/riboWaltz
Software, algorithmfeatureCountsLiao et al., 2014RRID:SCR_012919http://bioconductor.org/packages/release/bioc/html/Rsubread.html
Software, algorithmDESeq2Love et al., 2014RRID:SCR_015687https://bioconductor.org/packages/release/bioc/html/DESeq2.html
Software, algorithmMAGeCKLi et al., 2014; Li and Song, 2022NAhttps://sourceforge.net/p/mageck/wiki/Home/
Software, algorithmMaxquantCox and Mann, 2008RRID:SCR_014485https://cox-labs.github.io/coxdocs/maxquant_instructions.html
Software, algorithmAndromedaCox et al., 2011NAhttps://cox-labs.github.io/coxdocs/andromeda_instructions.html
Software, algorithmMsstatsChoi et al., 2014RRID:SCR_014353https://msstats.org/
Software, algorithmfgseaKorotkevich et al., 2021RRID:SCR_020938https://bioconductor.org/packages/release/bioc/html/fgsea.html
Software, algorithmBiorenderNARRID:SCR_018361https://biorender.com
Software, algorithmPyMOLNARRID:SCR_000305https://pymol.org/2/
Software, algorithmrMATSShen et al., 2014RRID:SCR_013049https://rnaseq-mats.sourceforge.net
Software, algorithmMolecular Signatures DatabaseLiberzon et al., 2011RRID:SCR_016863https://www.gsea-msigdb.org/gsea/msigdb/index.jsp
Software, algorithmUniversal Protein ResourceBateman et al., 2021RRID:SCR_002380https://www.uniprot.org
Software, algorithmFlowJoNARRID:SRC_008520https://www.flowjo.com/solutions/flowjo
Software, algorithmEmpiria StudioLI-CORRRID:SCR_022512https://www.licor.com/bio/empiria-studio/
Software, algorithmNIS-Elements AR 5.42.03 64-bitNikon InstrumentsRRID:SCR_014329https://www.nikoninstruments.com/Products/Software
Software, algorithmFijiFiji/ImageJRRID:SCR_002285http://fiji.sc
OtherS-Trap Micro ColumnsProtiFiCat# C02-micro-80
OtherJupiter 3 um C18 300A, Bulk packagingPhenomenexCat# 04A-4263
OtherMolex Polymicro Capillary
100 um × 363 um
Fisher ScientificCat# 50-110-8623
OtherNeon Transfection SystemThermo Fisher ScientificCat# MPK5000
OtherLuminex FLEXMAP 3D SystemInvitrogenCat# APX1342
Other4–20% Mini-PROTEAN
TGX Precast Gel
Bio-RadCat# 4561096
OtherAmersham Protran Western Blotting Membranes, NitrocelluloseCytivaCat# GE10600001
OtherC1000 Touch Thermal Cycler ChassisBio-RadCat# 1841100
OtherCFX96 Optical Reaction Module
for Real-Time PCR System
Bio-RadCat# 1845097
OtherChemiDoc Imaging SystemBio-RadCat# 17001401
OtherThick-wall Polycarbonate Tubes,
13 × 51 mm
Beckman-CoulterCat# 349622
OtherNovex TBE-Urea Gels 15%, 12 wellInvitrogenCat# EC68852BOX
OtherCostar Spin-X Centrifuge Tube FiltersCorningCat# CLS8162
OtherNovex TBE Gels, 8%, 15 wellInvitrogenCat# EC62155BOX
OtherGloMax Explorer Multimode Microplate ReaderPromegaCat# GM3500
OtherOrbitrap Exploris 480 Mass SpectrometerThermo ScientificCat# BRE725533

Additional files

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Gregory Caleb Howard
  2. Jing Wang
  3. Kristie L Rose
  4. Camden Jones
  5. Purvi Patel
  6. Tina Tsui
  7. Andrea C Florian
  8. Logan Vlach
  9. Shelly L Lorey
  10. Brian C Grieb
  11. Brianna N Smith
  12. Macey J Slota
  13. Elizabeth M Reynolds
  14. Soumita Goswami
  15. Michael R Savona
  16. Frank M Mason
  17. Taekyu Lee
  18. Stephen Fesik
  19. Qi Liu
  20. William P Tansey
(2024)
Ribosome subunit attrition and activation of the p53–MDM4 axis dominate the response of MLL-rearranged cancer cells to WDR5 WIN site inhibition
eLife 12:RP90683.
https://doi.org/10.7554/eLife.90683.3