Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Nuclear corepressors NCOR1/NCOR2 regulate B cell development, maintain genomic integrity and prevent transformation

Abstract

The nuclear corepressors NCOR1 and NCOR2 interact with transcription factors involved in B cell development and potentially link these factors to alterations in chromatin structure and gene expression. Herein, we demonstrate that Ncor1/2 deletion limits B cell differentiation via impaired recombination, attenuates pre-BCR signaling and enhances STAT5-dependent transcription. Furthermore, NCOR1/2-deficient B cells exhibited derepression of EZH2-repressed gene modules, including the p53 pathway. These alterations resulted in aberrant Rag1 and Rag2 expression and accessibility. Whole-genome sequencing of Ncor1/2 DKO B cells identified increased number of structural variants with cryptic recombination signal sequences. Finally, deletion of Ncor1 alleles in mice facilitated leukemic transformation, whereas human leukemias with less NCOR1 correlated with worse survival. NCOR1/2 mutations in human leukemia correlated with increased RAG expression and number of structural variants. These studies illuminate how the corepressors NCOR1/2 regulate B cell differentiation and provide insights into how NCOR1/2 mutations may promote B cell transformation.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: B cell development requires nuclear corepressors NCOR1 and NCOR2 and is sensitive to their gene dosage.
Fig. 2: Nuclear corepressors regulate both proliferation and differentiation during B cell development.
Fig. 3: Absence of nuclear corepressors results in defective pre-BCR signaling and in turn promotes STAT5-mediated transcription.
Fig. 4: scATAC-seq reveals the epigenetic landscape regulated by nuclear corepressors.
Fig. 5: Nuclear corepressors segregate RAG expression from proliferating cells and protect the genome integrity.
Fig. 6: NCOR1 deficiency promotes leukemic transformation with phenotypic heterogeneity.
Fig. 7: Human B-ALL patients with nuclear corepressor mutations harbor structural variants with high RAG expression.

Similar content being viewed by others

Data availability

The data supporting this study are available within the paper and supplementary information file. A reporting summary is also available. Mouse genome mm10 was used as reference sequence (https://www.ncbi.nlm.nih.gov/assembly/GCF_000001635.20/). scRNA-seq and scATAC-seq data were deposited at Gene Expression Omnibus with the accession code GSE208656. Whole-genome sequencing data were deposited at SRA with the accession code PRJNA860179. STAT5 ChIP-seq peak data were obtained from GSE86878, and H3K27ac ChIP-seq peak data were obtained from http://www.cistrome.org via project GSM1463433. IMMGEN RNA-seq count data were from GSE124829. Human B-ALL NCOR1 and NCOR2 mutation and RNA-seq data were obtained from St Jude ProteinPaint (https://pecan.stjude.cloud/proteinpaint). Source data are provided with this paper.

Code availability

Only publicly available software tools were used for analysis (for example, CellRanger, Seurat, Signac, whole-genome sequencing and GSEA). No custom software was used or developed in this study, but analysis scripts will be provided upon request.

References

  1. Melchers, F. Checkpoints that control B cell development. J. Clin. Invest. 125, 2203–2210 (2015).

    Article  Google Scholar 

  2. Katerndahl, C. D. S. et al. Antagonism of B cell enhancer networks by STAT5 drives leukemia and poor patient survival. Nat. Immunol. 18, 694–704 (2017).

    Article  CAS  Google Scholar 

  3. Mullighan, C. G. et al. Deletion of IKZF1 and prognosis in acute lymphoblastic leukemia. N. Engl. J. Med. 360, 470–480 (2009).

    Article  CAS  Google Scholar 

  4. Heltemes-Harris, L. M. et al. Identification of mutations that cooperate with defects in B cell transcription factors to initiate leukemia. Oncogene 40, 6166–6179 (2021).

    Article  CAS  Google Scholar 

  5. Heltemes-Harris, L. M. et al. Ebf1 or Pax5 haploinsufficiency synergizes with STAT5 activation to initiate acutelymphoblastic leukemia. J. Exp. Med. 208, 1135–1149 (2011).

    Article  CAS  Google Scholar 

  6. Mottis, A., Mouchiroud, L. & Auwerx, J. Emerging roles of the corepressors NCoR1 and SMRT in homeostasis. Genes Dev. 27, 819–835 (2013).

    Article  CAS  Google Scholar 

  7. You, S. H. et al. Nuclear receptor co-repressors are required for the histone-deacetylase activity of HDAC3 in vivo. Nat. Struct. Mol. Biol. 20, 182–187 (2013).

    Article  CAS  Google Scholar 

  8. Mullighan, C. G. et al. CREBBP mutations in relapsed acute lymphoblastic leukaemia. Nature 471, 235–241 (2011).

    Article  CAS  Google Scholar 

  9. Reddy, A. et al. Genetic and functional drivers of diffuse large B cell lymphoma. Cell 171, 481 (2017).

    Article  CAS  Google Scholar 

  10. Jepsen, K. et al. Combinatorial roles of the nuclear receptor corepressor in transcription and development. Cell 102, 753–763 (2000).

    Article  CAS  Google Scholar 

  11. Jepsen, K. et al. SMRT-mediated repression of an H3K27 demethylase in progression from neural stem cell to neuron. Nature 450, 415–419 (2007).

    Article  CAS  Google Scholar 

  12. Yamamoto, H. et al. NCoR1 is a conserved physiological modulator of muscle mass and oxidative function. Cell 147, 827–839 (2011).

    Article  CAS  Google Scholar 

  13. Schnütgen, F. et al. A directional strategy for monitoring Cre-mediated recombination at the cellular level in the mouse. Nat. Biotechnol. 21, 562–565 (2003).

    Article  Google Scholar 

  14. Hardy, R. R., Carmack, C. E., Shinton, S. A., Kemp, J. D. & Hayakawa, K. Resolution and characterization of pro-B and pre-pro-B cell stages in normal mouse bone marrow. J. Exp. Med. 173, 1213–1225 (1991).

    Article  CAS  Google Scholar 

  15. Heng, T. S. P. et al. The immunological genome project: Networks of gene expression in immune cells. Nat. Immunol. 9, 1091–1094 (2008).

    Article  CAS  Google Scholar 

  16. Goodnow, C. C. et al. Altered immunoglobulin expression and functional silencing of self-reactive B lymphocytes in transgenic mice. Nature 334, 676–682 (1988).

    Article  CAS  Google Scholar 

  17. Lee, R. D. et al. Single-cell analysis identifies dynamic gene expression networks that govern B cell development and transformation. Nat. Commun. 12, 6843 (2021).

    Article  CAS  Google Scholar 

  18. Heizmann, B., Kastner, P. & Chan, S. Ikaros is absolutely required for pre-B cell differentiation by attenuating IL-7 signals. J. Exp. Med. 210, 2823–2832 (2013).

    Article  CAS  Google Scholar 

  19. Chan, L. N. et al. Signalling input from divergent pathways subverts B cell transformation. Nature 583, 845–851 (2020).

    Article  CAS  Google Scholar 

  20. Goetz, C. A., Harmon, I. R., O’Neil, J. J., Burchill, M. A. & Farrar, M. A. STAT5 activation underlies IL7 receptor-dependent B cell development. J. Immunol. 172, 4770–4778 (2004).

    Article  CAS  Google Scholar 

  21. Stengel, K. R. et al. Histone deacetylase 3 controls a transcriptional network required for B cell maturation. Nucleic Acids Res. 47, 10612–10627 (2019).

    Article  CAS  Google Scholar 

  22. Hatzi, K. et al. A hybrid mechanism of action for BCL6 in B cells defined by formation of functionally distinct complexes at enhancers and promoters. Cell Rep. 4, 578–588 (2013).

    Article  CAS  Google Scholar 

  23. Su, I. H. et al. Ezh2 controls B cell development through histone H3 methylation and Igh rearrangement. Nat. Immunol. 4, 124–131 (2002).

    Article  Google Scholar 

  24. Jacobsen, J. A. et al. EZH2 regulates the developmental timing of effectors of the pre–antigen receptor checkpoints. J. Immunol. 198, 4682–4691 (2017).

    Article  CAS  Google Scholar 

  25. Rouault, J. P. et al. Identification of BTG2, an antiproliferative p53-dependent component of the DNA damage cellular response pathway. Nat. Genet. 14, 482–486 (1996).

    Article  CAS  Google Scholar 

  26. Tomasini, R. et al. p53-dependent expression of the stress-induced protein (SIP). Eur. J. Cell Biol. 81, 294–301 (2002).

    Article  CAS  Google Scholar 

  27. Han, J., Goldstein, L. A., Hou, W., Gastman, B. R. & Rabinowich, H. Regulation of mitochondrial apoptotic events by p53-mediated disruption of complexes between antiapoptotic bcl-2 members and bim. J. Biol. Chem. 285, 22473–22483 (2010).

    Article  CAS  Google Scholar 

  28. Wang, J. et al. NCoR1 restrains thymic negative selection by repressing Bim expression to spare thymocytes undergoing positive selection. Nat. Commun. 8, 959 (2017).

    Article  Google Scholar 

  29. Müller, L. et al. The corepressor NCOR1 regulates the survival of single-positive thymocytes. Sci. Rep. 7, 15928 (2017).

    Article  Google Scholar 

  30. Carlson, C. M. et al. Kruppel-like factor 2 regulates thymocyte and T-cell migration. Nature 442, 299–302 (2006).

    Article  CAS  Google Scholar 

  31. Hart, G. T., Wang, X., Hogquist, K. A. & Jameson, S. C. Krüppel-like factor 2 (KLF2) regulates B-cell reactivity, subset differentiation, and trafficking molecule expression. Proc. Natl Acad. Sci. USA 108, 716–721 (2011).

    Article  CAS  Google Scholar 

  32. Skon, C. N. et al. Transcriptional downregulation of S1pr1 is required for the establishment of resident memory CD8+ T cells. Nat. Immunol. 14, 1285–1293 (2013).

    Article  CAS  Google Scholar 

  33. Teng, G. et al. RAG represents a widespread threat to the lymphocyte genome. Cell 162, 751–765 (2015).

    Article  CAS  Google Scholar 

  34. Burchill, M. A. et al. Distinct effects of STAT5 activation on CD4+ and CD8+ T cell homeostasis: Development of CD4+CD25+ regulatory T cells versus CD8+ memory T cells. J. Immunol. 171, 5853–5864 (2003).

    Article  CAS  Google Scholar 

  35. Nakayama, J. et al. BLNK suppresses pre–B-cell leukemogenesis through inhibition of JAK3. Blood 113, 1483–1492 (2009).

    Article  CAS  Google Scholar 

  36. Heltemes-Harris, L. M. et al. Sleeping Beauty transposon screen identifies signaling modules that cooperate with STAT5 activation to induce B-cell acute lymphoblastic leukemia. Oncogene 35, 3454–3464 (2015).

    Article  Google Scholar 

  37. Gu, Z. et al. PAX5-driven subtypes of B-progenitor acute lymphoblastic leukemia. Nat. Genet. 51, 296–307 (2019).

    Article  CAS  Google Scholar 

  38. Tsai, A. G. et al. Human chromosomal translocations at CpG Sites and a theoretical basis for their lineage and stage specificity. Cell 135, 1130–1142 (2008).

    Article  CAS  Google Scholar 

  39. Fischer, U. et al. Genomics and drug profiling of fatal TCF3-HLF-positive acute lymphoblastic leukemia identifies recurrent mutation patterns and therapeutic options. Nat. Genet. 47, 1020 (2015).

    Article  CAS  Google Scholar 

  40. Papaemmanuil, E. et al. RAG-mediated recombination is the predominant driver of oncogenic rearrangement in ETV6-RUNX1 acute lymphoblastic leukemia. Nat. Genet. 46, 116–125 (2014).

    Article  CAS  Google Scholar 

  41. Zhang, J. et al. Deregulation of DUX4 and ERG in acute lymphoblastic leukemia. Nat. Genet. 48, 1481–1489 (2016).

    Article  CAS  Google Scholar 

  42. Abdelrasoul, H. et al. Synergism between IL7R and CXCR4 drives BCR-ABL induced transformation in Philadelphia chromosome-positive acute lymphoblastic leukemia. Nat. Commun. 11, 3194 (2020).

    Article  CAS  Google Scholar 

  43. Waanders, E. et al. Mutational landscape and patterns of clonal evolution in relapsed pediatric acute lymphoblastic leukemia. Blood Cancer Discov. 1, 96–111 (2020).

    Article  CAS  Google Scholar 

  44. Bhaskara, S. et al. Hdac3 is essential for the maintenance of chromatin structure and genome stability. Cancer Cell 18, 436–447 (2010).

    Article  CAS  Google Scholar 

  45. Swaminathan, S. et al. Mechanisms of clonal evolution in childhood acute lymphoblastic leukemia. Nat. Immunol. 16, 766–774 (2015).

    Article  CAS  Google Scholar 

  46. Mori, H. et al. Chromosome translocations and covert leukemic clones are generated during normal fetal development. Proc. Natl Acad. Sci. 99, 8242–8247 (2002).

    Article  CAS  Google Scholar 

  47. Narendra, V. et al. CTCF establishes discrete functional chromatin domains at the Hox clusters during differentiation. Science 347, 1017–1021 (2015).

    Article  CAS  Google Scholar 

  48. Khoury, A. et al. Constitutively bound CTCF sites maintain 3D chromatin architecture and long-range epigenetically regulated domains. Nat. Commun. 11, 54 (2020).

    Article  CAS  Google Scholar 

  49. Degner, S. C. et al. CCCTC-binding factor (CTCF) and cohesin influence the genomic architecture of the Igh locus and antisense transcription in pro-B cells. Proc. Natl Acad. Sci. USA 108, 9566–9571 (2011).

    Article  CAS  Google Scholar 

  50. Ba, Z. et al. CTCF orchestrates long-range cohesin-driven V(D)J recombinational scanning. Nature 586, 305–310 (2020).

    Article  CAS  Google Scholar 

  51. Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587.e29 (2021).

    Article  CAS  Google Scholar 

  52. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902.e21 (2019).

    Article  CAS  Google Scholar 

  53. Hafemeister, C. & Satija, R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 20, 296 (2019).

    Article  CAS  Google Scholar 

  54. Qin, Q. et al. Lisa: inferring transcriptional regulators through integrative modeling of public chromatin accessibility and ChIP-seq data. Genome Biol. 21, 32 (2020).

    Article  Google Scholar 

  55. Subramanian, A. et al. Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. 102, 15545–15550 (2005).

    Article  CAS  Google Scholar 

  56. Liberzon, A. et al. The molecular signatures database hallmark gene set collection. Cell Syst. 1, 417–425 (2015).

    Article  CAS  Google Scholar 

  57. Stuart, T., Srivastava, A., Madad, S., Lareau, C. A. & Satija, R. Single-cell chromatin state analysis with Signac. Nat. Methods 18, 1333–1341 (2021).

    Article  CAS  Google Scholar 

  58. Schep, A. N., Wu, B., Buenrostro, J. D. & Greenleaf, W. J. chromVAR: inferring transcription-factor-associated accessibility from single-cell epigenomic data. Nat. Methods 14, 975–978 (2017).

    Article  CAS  Google Scholar 

  59. Fornes, O. et al. JASPAR 2020: update of the open-access database of transcription factor binding profiles. Nucleic Acids Res. 48, D87–D92 (2020).

    CAS  Google Scholar 

  60. Layer, R. M., Chiang, C., Quinlan, A. R. & Hall, I. M. LUMPY: A probabilistic framework for structural variant discovery. Genome Biol. 15, 1–19 (2014).

    Article  Google Scholar 

  61. Chikina, M. D. & Troyanskaya, O. G. An effective statistical evaluation of ChIPseq dataset similarity. Bioinformatics 28, 607–613 (2012).

    Article  CAS  Google Scholar 

  62. Livak, K. J. & Schmittgen, T. D. Analysis of relative gene expression data using real-time quantitative PCR and the 2 − ΔΔCT method. Methods 25, 402–408 (2001).

    Article  CAS  Google Scholar 

  63. Zhou, X. et al. Exploring genomic alteration in pediatric cancer using ProteinPaint. Nat. Genet. 48, 4–6 (2015).

    Article  Google Scholar 

  64. Ou, J. & Zhu, L. J. trackViewer: A Bioconductor package for interactive and integrative visualization of multi-omics data. Nat. Methods 16, 453–454 (2019).

    Article  CAS  Google Scholar 

  65. Gentles, A. J. et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat. Med. 21, 938–945 (2015).

    Article  CAS  Google Scholar 

  66. Bunis, D. G., Andrews, J., Fragiadakis, G. K., Burt, T. D. & Sirota, M. dittoSeq: universal user-friendly single-cell and bulk RNA sequencing visualization toolkit. Bioinformatics 36, 5535–5536 (2021).

    Article  Google Scholar 

Download references

Acknowledgements

We thank G. Hubbard, A. Rost and N. Keller for technical assistance and mouse husbandry; E. Stanley, J. Daniels and K. Beckman and the University of Minnesota Genomics Center for 10x Genomics single-cell capture and sequencing; J. Motl, R. Arora and P. Champoux for cell sorting and Flow Cytometry Core Facility maintenance at University of Minnesota (5P01AI035296); and S. Dehm and K. Schwertfeger (University of Minnesota) for comments on the manuscript. We thank E. Olson (UT-Southwestern), W. Ellmeier (Medical University of Vienna) and J. Auwerx (École Polytechnique Fédérale, Lausanne, Switzerland) for providing the Ncor1FL/FL mice. The Minnesota Supercomputing Institute at the University of Minnesota provided bioinformatic support and resources that contributed to the research results reported within this paper. We are thankful for support from the German Gene Trap Consortium, which assisted in the generation of the Ncor2FL/FL mice. This work was supported by an individual predoctoral F30 fellowship from the National Institutes of Health (NIH) (F30CA232399) and a T32 training grant (T32 GM008244) to R.D.L.; St Jude Children’s Research Hospital Cancer Center Core Grant CA021765 and NIH grant R35CA197695 to C.G.M.; and NIH grants R01AI124512, R01AI147540 and R01CA232317 to M.A.F. M.A.F. was supported by the Virginia and David Utz endowed chair in fundamental immunobiology.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, R.D.L. and M.A.F.; Methodology, R.D.L. and M.A.F.; Formal Analysis, R.D.L., S.A.M., T.P.K., J.M., L.H.H. and M.A.F.; Investigation, R.D.L. and L.H.H.; Resources, C.G.M., K.J. and M.A.F.; Data Curation, S.A.M., T.P.K. and L.S.; Writing – Original Draft, R.D.L. and M.A.F.; Writing – Review & Editing, R.D.L, S.A.M., T.P.K., C.G.M., K.J. and M.A.F.; Visualization, R.D.L., L.H.H., S.A.M., T.P.K. and J.M.; Supervision, M.A.F.; Project Administration, M.A.F.; Funding Acquisition, R.D.L. and M.A.F.

Corresponding author

Correspondence to Michael A. Farrar.

Ethics declarations

Competing interests

C.G.M. provides consultation and advise for Illumina (compensated), Faze Medicines (compensated) and Beam Therapeutics (compensated) and receives research funding from Pfizer and Abbvie. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks the anonymous reviewers for their contribution to the peer review of this work. Editor recognition statement (if applicable to your journal) L. A. Dempsey was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team. Peer reviewer reports are available.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Ncor2 conditional knockout mouse model and flow cytometry gating scheme.

a. Schematic for generating Ncor2 flox mice using the flip-excision system (FlEx). A retrovirus vector containing a strong splice acceptor site followed by a lacZ reporter gene was flanked by FRT and loxP sites. This gene trap was targeted in the intronic region between exon 1 and 2. Mice harboring the gene trap were bred to FLPe constitutive expressing mice, inverting the gene trap and allowing for normal splicing of Ncor2. Subsequent breeding with Cd79a-Cre mice then allows for conditional deletion of Ncor2 in developing B cells. b. Representative NCOR1 protein expression in CD19 + B220 + bone marrow B cells with isotype controls (left) and quantification of NCOR1-positive B cells (right). Each genotype represents n = 3, from three independent experiments. Center of measure indicates mean and error bars indicate standard deviation. A one-way ANOVA was used to calculate statistical significance. c. Representative NCOR1 protein expression in CD19 + B220 + splenic B cells with isotype controls (left) and quantification of NCOR1-positive B cells (right). Each genotype represents n = 3, from three independent experiments. Center of measure indicates mean and error bars indicate standard deviation. A one-way ANOVA was used to calculate statistical significance. d. Quantification of Ncor1 and Ncor2 expression via qPCR in wildtype and NCOR1/2 double-knockout B cells from the bone marrow and the spleen. Each genotype represents n = 3, from two independent experiments. Center of measure indicates mean and error bars indicate standard deviation. A two-tailed t-test was performed to calculate statistical significance. e. Whole-genome sequencing read depth coverage across the Ncor1 gene for wildtype and NCOR1/2 DKO B cells. f. Whole-genome sequencing mapping of reads across the Ncor2 gene trap for wildtype (Cre- littermate) and NCOR1/2 DKO B cells; left and right panels indicate 5’ and 3’ inversion breakpoints, respectively. g. Lymphocytes are gated using FSC-A, SSC-A, and then subsequently on SSC-A and SSC-W for singlets. Dead cells are excluded using the GhostRed780 Live/dead dye and gated on B220 and CD43. CD43+ and CD43- cells are gated on to assess Hardy fractions A-C and D-F, respectively.

Source data

Extended Data Fig. 2 Single-cell RNA-sequencing multiplet removal and cluster identification.

a. Multiplet identification using HTOdemux at a resolution of k = 22 (left). Multiplet and singlet classification based on genotype is demonstrated on the right. b. Feature plots of temporally regulated genes during B cell development. c. Feature plot of CITE-Seq antibodies, B220, CD19, CD25, CD43, CD93, and IgM (top) and violin plot of the CITE-seq antibodies (bottom) are shown. These gene expression profiles and CITE-seq antibody expression patterns were used to characterize different subsets of B cell development.

Extended Data Fig. 3 Overlap between scRNA-seq and scATACseq differentially upregulated expression and accessibility genes.

a. Cluster comparison between scRNAseq and scATACseq genes that are differentially upregulated in expression and accessibility in the NCOR1/2-knockout (DKO) B cells.

Extended Data Fig. 4 Nuclear corepressors regulate the p53 pathway and repress BIM expression.

a. Landscape in silico analysis (LISA)-based prediction of transcriptional regulators of significantly upregulated genes in either WT (left) or NCOR1/2 DKO (right) of cycling pro B cells (top) or pre-BCR-dependent cells (bottom). Number in parentheses indicates rank. A two-tailed wilcoxon rank-sum test was used to calculated statistical significance. b. GSEA of p53 pathway and senescence pathway in NCOR1/2 DKO cells in both cycling pro B (top) and pre-BCR-dependent cells (bottom). c. Changes in expression of p53 pathway genes, Cdkn1a, Btg2, and Trp53inp1, between WT, NCOR1 KO and NCOR1/2 DKO B cell clusters. d. Pro-apoptosis factor, BIM expression of WT and NCOR1/2 DKO in pre-pro B, pro B, pro B/pre B, small pre B, immature B and mature B cells. Plot represents n = 3 of each genotype from two independent experiments. Error bar indicates standard deviation. A one-way ANOVA was used to calculate statistical significance. e. BIM (Bcl2l11) gene expression and accessibility differences in wildtype, NCOR1 KO and NCOR1/2 DKO B cells. Asterisk indicates comparisons with at least a log2 fold change of 0.25 that have a p-value <0.05 when compared to wildtype. RNA expression of Bcl2l11 (left) represents WT (n = 7454 cells), NCOR1 KO (n = 7806 cells), NCOR1/2 DKO (n = 8637 cells) from two biological replicates for each genotype from a single capture. Accessibility of Bcl2l11 (right) represents WT (n = 8552 cells), NCOR1 KO (n = 6008 cells), and NCOR1/2 DKO (n = 8574 cells) from a single biological sample derived from three separate captures. A two-tailed Wilcoxon rank-sum test was used to calculate statistical significance. All measure of centers indicate mean.

Source data

Extended Data Fig. 5 KLF2 and its target genes are aberrantly expressed in NCOR1/2-deficient B cells.

a. RNA expression of Klf2 and its target genes Cd69 and Sell (CD62L; L-selectin) in wildtype (WT), NCOR1-knockout (KO) and NCOR1/2-knockout (DKO) B cells. Asterisk (*; P < 0.05) indicates statistically significant expression changes compared to wildtype cells for Klf2 in PreBCRi-II (DKO; P = 4.24E-15), preBCRi-III (DKO; P = 1.43E-9), and immature B (DKO; P = 1.34 E-8), for Cd69 in PreBCRi-II (DKO; P = 1.86E-39), PreBCRi-III (DKO; P = 2.74E-8), Kappa Pre B (DKO; P = 1.47E-37), Lambda Pre B-I (DKO; P = 1.69E-12), Lambda Pre B-III (DKO; P = 9.14E-14), and Immature B (DKO; P = 1.89E-7). b. Gene activity of Klf2 and its target genes Cd69 and Sell (CD62L; L-selectin)in WT, KO and DKO B cells. Asterisk (*; P < 0.05) indicates statistically significant accessibility changes compared to wildtype cells for Klf2 in Pro B VDJ (KO; P = 1.23E-9), Cycling Pre B (KO; P = 2.94E-36, DKO; P = 4.97E-24), Kappa/Lambda Pre B (KO; P = 5.31E-33), Immature B (KO; P = 1.22E-14, DKO; P = 6.40E-11), mature B (KO; P = 4.94E-13), plasma cells (KO; P = 4.94E-13, DKO; P = 5.39E-7) and for Cd69 in Cycling Pro B (KO; P = 1.74E-6, DKO; P = 2.12E-12), Pro B VDJ (KO; P = 4.20E-13, DKO; P = 2.47E-30), Cycling Pre B (KO; P = 1.29E-27, DKO; P = 6.87E-84), Kappa/Lambda Pre B(KO; P = 1.96E-57, DKO; P = 6.92E-46), Immature B (KO; P = 3.72E-27, DKO; P = 3.13E-22), mature B (KO; P = 1.68E-20, DKO;P = 5.39E-7), plasma cells (KO; P = 1.68E-20, DKO; P = 5.39E-7) and for Sell in Kappa/Lambda Pre B (DKO; P = 4.27E-7), and immature B (DKO;P = 1.47E-16). c. CD62L expression of different B cell development stages in wildtype (WT) and NCOR1/2-knockout (DKO) B cells. Top represents representative flow cytometry plots for CD62L expression and bottom represents summarized frequency of CD62L + cells for prepro B (B220 + CD43 + CD24CD19-, n = 4), proB/preB (B220 ((eB (eB (for 220 ((e, n = 4), small pre B (B220 + CD43 + CD19 + IgM-, n gM4), immature B (B220lowCD43immgM M4, n n4), and mature B cells (B220highCD43andgM M4, n n4) from three independent experiments. A two-tailed t-test was used to calculate statistical significance. Center of measure indicates mean and error bars indicate standard deviation.

Source data

Extended Data Fig. 6 Transcriptional and accessibility changes in genes associated with Rag-mediated genomic changes in human B-ALLs.

a. Transcriptional changes between WT, NCOR1 KO, and NCOR1/2 DKO derived from scRNA-seq. b. Accessibility changes between WT, NCOR1 KO, and NCOR1/2 DKO derived from scATAC-seq. Asterisk (*) indicates comparisons with at least a log2 fold change of 0.25 that have a p-value <0.05 when compared to wildtype. RNA expression (left) data represents WT (n = 7454 cells), NCOR1 KO (n = 7806 cells), NCOR1/2 DKO (n = 8637 cells) from two biological replicates for each genotype from a single capture. ATAC accessibility data (right) represents WT (n = 8552 cells), NCOR1 KO (n = 6008 cells), and NCOR1/2 DKO (n = 8574 cells) from a single biological sample derived from three separate captures. A two-tailed Wilcoxon rank-sum test was used to calculate statistical significance.

Supplementary information

Reporting Summary

Peer Review File

Supplementary Tables 1–5

Supplementary Table 1. Pairwise comparison of differentially expressed genes among WT, NCOR1 KO and NCOR1/2 DKO. A two-tailed Wilcoxon rank-sum test was performed to calculate statistical significance. Cluster labels correspond to the following cell type as following: cluster 0: κ pre-B, cluster 1: Pro B VDJ, cluster 2: PreBCRi-I, cluster 3 – PreBCRi II, cluster 4: pre-pro-B, cluster 5: Immature B, cluster 6: Cycling Pro B, cluster 7: Mature B, cluster 8: PreBCRi-III, cluster 9: Pre B-II, cluster 10: λ Pre B-III, cluster 11: PreBCR-d, cluster 12: High mitochondrial B, cluster 13: λ Pre B-I, cluster 14: Committed Pro B, cluster 15: Plasma Cells, cluster 16: Cycling Immature B. Supplementary Table 2. Pairwise comparison of differential transcription factor motifs present among WT, NCOR1 KO and NCOR1/2 DKO. Transcription factor motifs were derived from the JASPAR database. A two-tailed Wilcoxon rank-sum test was performed to calculate statistical significance. Supplementary Table 3. Pairwise comparison of differentially accessible genes among WT, NCOR1 KO and NCOR1/2 DKO. Accessibility of the gene was defined as peaks found within the gene body and peaks present within the upstream 2,000-bp promoter. A two-tailed Wilcoxon rank-sum test was performed to calculate statistical significance. Supplementary Table 4. Structural variants found in WT and NCOR DKO B cells. Structural variants including BND (break end), DEL (deletion), INS (insertion), TRA (translocation) and INV (inversion) and their genomic position and affected genes are listed. Structural variant calling was performed using Smoove. Supplementary Table 5. Human B-ALL NCOR1 mutation type and the presence of intrachromosomal or interchromosomal structural variants. Types of NCOR1 mutations and structural variants that are present are broken down into intrachromosomal and interchromosomal. Interchromosomal translocations that are B-ALL subtype defining are present. Structural variant information was derived from StJude ProteinPaint

Source data

Source Data Fig. 1

Statistical Source Data.

Source Data Fig. 2

Statistical Source Data.

Source Data Fig. 3

Statistical Source Data.

Source Data Fig. 5

Statistical Source Data.

Source Data Fig. 6

Statistical Source Data.

Source Data Fig. 7

Statistical Source Data

Source Data Extended Data Fig. 1

Statistical Source Data.

Source Data Extended Data Fig. 4

Statistical Source Data.

Source Data Extended Data Fig. 5

Statistical Source Data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lee, R.D., Knutson, T.P., Munro, S.A. et al. Nuclear corepressors NCOR1/NCOR2 regulate B cell development, maintain genomic integrity and prevent transformation. Nat Immunol 23, 1763–1776 (2022). https://doi.org/10.1038/s41590-022-01343-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-022-01343-7

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing