Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2023 Apr 28;14(1):2439.
doi: 10.1038/s41467-023-38132-1.

Dependency of NELF-E-SLUG-KAT2B epigenetic axis in breast cancer carcinogenesis

Affiliations

Dependency of NELF-E-SLUG-KAT2B epigenetic axis in breast cancer carcinogenesis

Jieqiong Zhang et al. Nat Commun. .

Abstract

Cancer cells undergo transcriptional reprogramming to drive tumor progression and metastasis. Using cancer cell lines and patient-derived tumor organoids, we demonstrate that loss of the negative elongation factor (NELF) complex inhibits breast cancer development through downregulating epithelial-mesenchymal transition (EMT) and stemness-associated genes. Quantitative multiplexed Rapid Immunoprecipitation Mass spectrometry of Endogenous proteins (qPLEX-RIME) further reveals a significant rewiring of NELF-E-associated chromatin partners as a function of EMT and a co-option of NELF-E with the key EMT transcription factor SLUG. Accordingly, loss of NELF-E leads to impaired SLUG binding on chromatin. Through integrative transcriptomic and genomic analyses, we identify the histone acetyltransferase, KAT2B, as a key functional target of NELF-E-SLUG. Genetic and pharmacological inactivation of KAT2B ameliorate the expression of EMT markers, phenocopying NELF ablation. Elevated expression of NELF-E and KAT2B is associated with poorer prognosis in breast cancer patients, highlighting the clinical relevance of our findings. Taken together, we uncover a crucial role of the NELF-E-SLUG-KAT2B epigenetic axis in breast cancer carcinogenesis.

PubMed Disclaimer

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1
Fig. 1. Abolishment of the NELF complex impairs tumorigenic properties.
a Western blot analysis of NELF-A and NELF-E in non-tumorigenic breast epithelial cell line MCF10A, as well as in different breast cancer cell lines T-47D (ER+, PR+), SK-BR-3 (HER2+), MCF7 (ER+), BT-474 (HER2+), BT-549 (triple-negative), MDA-MB-231 (triple-negative), and SUM159 (triple-negative). β-actin was used as the loading control. b, c Left: MCF7 cells (n = 3) and BT-549 cells (n = 4) were transfected with non-targeting siRNA (‘scrambled, siSCR’) and siRNAs targeting NELF-A and NELF-E, respectively, followed by western blot analysis. GAPDH was used as the loading control. Right: Representative images and quantification of soft agar assays. d Left: Western blot analysis of NELF-A, NELF-E, NELF-B, and NELF-C/D in WT and NELF-A/NELF-E KO SUM159 cells. β-actin was used as the loading control. Right: Representative images and quantification of soft agar assays (n = 4). e 5 × 106 WT, NELF-A KO, and NELF-E KO SUM159 cells were injected into the mammary fat pads of NOD/SCID mice, respectively. Tumor volume was measured every 3 days after the tumor was palpable; mean ± SEM, n = 4/group. f Left: Tumors harvested from WT, NELF-A KO, and NELF-E KO groups; Right: Quantification of tumor weights; mean ± SEM, n = 4/group. Blots and images are representative of at least three independent experiments. Data in (bd) are presented as mean ± SD. p-values are determined by a two-tailed Student’s t-test. Source data are provided as a Source Data file.
Fig. 2
Fig. 2. NELF depletion leads to reduced stemness-like traits and downregulated EMT pathway.
a MA plot showing gene expression changes between NELF-E KO and WT SUM159 cells. Red and blue points indicate significantly upregulated (n = 388) and downregulated (n = 540) genes, respectively. b GSEA enrichment plot for stemness and epithelial-mesenchymal transition pathways in NELF-E KO SUM159 cells. c Western blot analysis of NELF-E and NELF-A in WT, NELF-E KO, and NELF-E rescue SUM159 cells. β-actin was used as the loading control. d Quantification of wound healing assay in WT, NELF-E KO, and NELF-E rescue SUM159 cells. Scale bar = 100 μm. e Quantification of mammosphere formation assay in WT, NELF-E KO, and NELF-E rescue SUM159 cells (n = 4). Scale bar = 100 μm. f Quantification of invasion assay in WT, NELF-E KO, and NELF-E rescue SUM159 cells (n = 3). Scale bar = 100 μm. g Body weight (mean ± SEM) of mice injected with 1.25 × 106 WT and NELF-E KO SUM159 cells, respectively (n = 8, each group). h Lungs inflated and fixed with 10% neutral buffer formalin from mice in WT (n = 8) and NELF-E KO group (n = 8). i Representative images of H&E staining in lung tissues at three magnifications (mf: metastatic foci). From top to bottom, Bar = 2 mm, 500 μm, and 100 μm, respectively. j Quantitative analysis of lung metastasis presented as % of occupation by metastasis and the number of metastatic sites per lung (n = 8/group). k Graph showing quantification of mammospheres in WT, NELF-E KO, and NELF-E rescue MCF7 cells at primary (n = 3), secondary (n = 4), and tertiary passages (n = 4). l MCF7 cells from tertiary spheroids were seeded at a density of 1000 cells per well and incubated for 2 weeks. The colonies were then fixed and stained with crystal violet. m Flow cytometry analysis and quantification of the CD24low/CD44high population in MCF7 tertiary mammospheres (n = 3). Blots and images are representative of at least three independent experiments. Data in (d, e, f, j, k, m) are presented as mean ± SD. p-values are determined by a two-tailed Student’s t-test. Source data are provided as a Source Data file.
Fig. 3
Fig. 3. Loss of NELF-E suppresses EMT in a SOX9-SLUG overexpression MCF7 cell line model.
a Barplot showing the top perturbed pathways from Molecular Signatures Database (MSigDB) in Doxycycline (Dox)-treated MCF7ras+SS cells compared to the vehicle control. b Volcano plot depicting the significantly upregulated genes (n = 1750) and downregulated genes (n = 2223). Select mesenchymal/stemness-related genes are labeled in red, while epithelial genes are labeled in blue. adj. p-value ≤ 0.05; |fold change| ≥ 1.5. p-value was calculated using DESeq2 package (see details in “Methods”) c Western blot analysis for different markers in Dox-induced MCF7ras+SS cells transduced with scrambled or two independent shRNAs targeting NELF-E. GAPDH was used as the loading control. d GSEA plot of shNELF-E+Dox vs SCR+Dox cells. p-value was calculated from gene set enrichment analysis. (see detail in “Methods”). e Representative images and quantification of the migration and invasion assays. MCF7ras+SS cells were transduced with scrambled or NELF-E shRNA and treated with Dox to induce SOX9 and SLUG expression for 72 h prior to the assays (n = 3). Scale Bar = 100 μm. f Representative images and quantification of the sphere-formation assay. MCF7ras+SS cells were transduced with scrambled or NELF-E shRNA, followed by 72-h Dox treatment. Cells were seeded at a density of 10K per well, and the number of spheres was counted 5–7 days later (n = 4). Scale Bar = 100 μm. g Representative images and quantification of the soft agar assays for SCR+Dox and shNELF-E+Dox cells (n = 3). h Flow cytometry analysis and quantification of the CD24low/CD44high population in NELF-E KD+Dox cells compared to SCR+Dox cells (n = 3). Blots and images are representative of at least three independent experiments. p-values in (eh) are determined by a two-tailed Student’s t-test. Mean ± SD is represented by bar graphs. Source data are provided as a Source Data file.
Fig. 4
Fig. 4. Loss of NELF-E attenuates gene activation and RNAPII binding.
a Log2 fold change of UP-genes (n = 1750), DOWN-genes (n = 2223), and unchanged genes (n = 1581) in the two different conditions. Gene categorization was based on the condition of SCR+Dox vs SCR-Con with the cutoffs of adj. p-value ≤ 0.05 and |fold change| ≥ 1.5. Center lines show median values, box limits represent the upper and lower quartiles, and whiskers show 1.5× the interquartile range. Two-tailed Student’s t-tests were used for all comparisons. p-values were not adjusted for multiple tests, and t-statistics are provided in source data. b Genomic distribution of NELF-E binding sites in Dox-treated MCF7ras+SS cells. c Metagene plots of NELF-E ChIP-seq signals in SCR-Con and SCR+Dox cells for UP-genes (top panel) and DOWN-genes (bottom panel). d Metagene plots of RNAPII ChIP-seq signals in SCR-Con, SCR+Dox, and shNELF-E+Dox cells for UP-genes and DOWN-genes. e Genome browser tracks showing NELF-E and RNAPII occupancies at the promoters of mesenchymal gene FN1 (left) and stemness-related gene CD44 (right). Source data are provided as a Source Data file.
Fig. 5
Fig. 5. NELF-E interacts with and alters the genomic occupancy of SLUG.
a Volcano plot of NELF-E qPLEX-RIME analysis of WT+Dox vs WT-Con. Proteins that satisfy the significance threshold of |log2(fold change)| ≥0.5 and adj. p-value <0.05 are labeled with their gene names and colored red. The p-value was adjusted by Benjamini–Hochberg multiple hypothesis correction. b Interaction network plot of proteins enriched in Dox induction by qPLEX-RIME, superimposed on the STRING interaction network. Interactions detected by qPLEX-RIME are colored in blue, while interactions from the STRING database are colored in gray. Proteins/nodes are colored based on log2(fold change) value. c Western blot analysis of SOX9, SLUG, and NELF-E following NELF-E immunoprecipitation (IP), performed on nuclear extracts from Dox-treated MCF7ras+SS cells. Blots are representative of three independent experiments. d Genomic distribution of SLUG binding sites in MCF7ras+SS cells transduced with scrambled shRNA and treated with Dox (SCR+Dox). e Metaplots of SLUG binding across the three different categories in SCR+Dox and shNELF-E+Dox MCF7ras+SS cells. f, g Pie chart and heatmap depiction of SLUG and NELF-E co-bound peaks at TSS and distal regions. Source data are provided as a Source Data file.
Fig. 6
Fig. 6. KAT2B is a key target of NELF-E and SLUG in response to EMT.
a Dot plot showing KAT2B as one of the top genes (n = 41) whose SLUG-mediated activation was most significantly attenuated by NELF-E KD. Expressions of corresponding genes in the two comparisons are connected by gray lines. b Genome browser tracks showing the occupancies of NELF-E, SLUG, and RNAPII on the KAT2B locus. c RT-qPCR analysis (left) and western blot analysis (right) of KAT2B in MCF7ras+SS cells treated with Dox for different time points (n = 3). d Western blot analysis of KAT2B in MCF7ras+SS cells transduced with scrambled shRNA or two independent NELF-E shRNAs and treated with or without Dox. GAPDH was used as the loading control. e Heatmap depiction of SLUG/NELF-E/KAT2B/RNAPII signals at NELF-E-alone, SLUG-alone and NELF-E/SLUG co-bound regions. f Metaplot of KAT2B ChIP-Seq signals over the different cohorts of SLUG-bound regions. g Venn diagram showing the overlap between SLUG-bound UP-genes and NELF-E/KAT2B targets. The percentage overlap of SLUG-bound UP-genes compared to other categories is also indicated. h Genome browser tracks showing the occupancies of NELF-E, SLUG, and KAT2B on LAMC1 and ZEB1 loci. Blots are representative of three independent experiments. p-values are determined by a two-tailed Student’s t-test. Mean ± SD is represented by bar graphs. Source data are provided as a Source Data file.
Fig. 7
Fig. 7. KAT2B activates EMT progression.
a GSEA plot showing significantly enriched pathways in siKAT2B+Dox vs siSCR+Dox MCF7ras+SS cells. p-value was calculated from gene set enrichment analysis (see details in “Methods”). b Western blot analysis of ECM1 and CD44 in MCF7ras+SS cells treated with or without GA and AA. GAPDH was used as the loading control. Images and blots are representative of three independent experiments. c qRT-PCR analysis of FN1 and ECM1 in GA- and AA-treated Dox-induced MCF7ras+SS cells (n = 3). d Flow cytometry analysis and quantification of the CD24low/CD44high population in vehicle control, GA-, and AA-treated MCF7ras+SS cells, as a function of Dox induction. e Flow cytometry analysis and quantification of the CD24low/CD44high population following KAT2B overexpression in MCF7ras+SS cells. Vector control (GFP only; pCAGIG) and KAT2B-overexpression (KAT2B+GFP; pCAGIG-KAT2B) plasmids were transfected into MCF7ras+SS cells, respectively. The GFP and GFP+ populations were isolated and analyzed for the CD24low/CD44high population (n = 3). f, g Patient-derived breast cancer organoids were transduced with scrambled shRNA or two independent KAT2B or NELF-E shRNAs, followed by western blot and sphere-formation assay (n = 3). β-actin was used as the loading control. h Patient-derived breast cancer organoids were transduced with vector control or KAT2B overexpressing plasmid, followed by western blot and sphere-formation assay (n = 4). GAPDH was used as the loading control. Blots and images are representative of at least three independent experiments. p-values in (ch) are determined by a two-tailed Student’s t-test. Mean ± SD is represented by bar graphs. Source data are provided as a Source Data file.

References

    1. Lambert SA, et al. The human transcription factors. Cell. 2018;172:650–665. doi: 10.1016/j.cell.2018.01.029. - DOI - PubMed
    1. Jones PA, Issa JP, Baylin S. Targeting the cancer epigenome for therapy. Nat. Rev. Genet. 2016;17:630–641. doi: 10.1038/nrg.2016.93. - DOI - PubMed
    1. Bradner JE, Hnisz D, Young RA. Transcriptional addiction in cancer. Cell. 2017;168:629–643. doi: 10.1016/j.cell.2016.12.013. - DOI - PMC - PubMed
    1. Whyte WA, et al. Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell. 2013;153:307–319. doi: 10.1016/j.cell.2013.03.035. - DOI - PMC - PubMed
    1. Marazzi I, Greenbaum BD, Low DHP, Guccione E. Chromatin dependencies in cancer and inflammation. Nat. Rev. Mol. Cell Biol. 2018;19:245–261. doi: 10.1038/nrm.2017.113. - DOI - PubMed

Publication types

MeSH terms