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:

RNA sequestration in P-bodies sustains myeloid leukaemia

Abstract

Post-transcriptional mechanisms are fundamental safeguards of progenitor cell identity and are often dysregulated in cancer. Here, we identified regulators of P-bodies as crucial vulnerabilities in acute myeloid leukaemia (AML) through genome-wide CRISPR screens in normal and malignant haematopoietic progenitors. We found that leukaemia cells harbour aberrantly elevated numbers of P-bodies and show that P-body assembly is crucial for initiation and maintenance of AML. Notably, P-body loss had little effect upon homoeostatic haematopoiesis but impacted regenerative haematopoiesis. Molecular characterization of P-bodies purified from human AML cells unveiled their critical role in sequestering messenger RNAs encoding potent tumour suppressors from the translational machinery. P-body dissolution promoted translation of these mRNAs, which in turn rewired gene expression and chromatin architecture in leukaemia cells. Collectively, our findings highlight the contrasting and unique roles of RNA sequestration in P-bodies during tissue homoeostasis and oncogenesis. These insights open potential avenues for understanding myeloid leukaemia and future therapeutic interventions.

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: P-body regulators are AML dependencies.
Fig. 2: DDX6 is crucial for human and mouse AML progression in vivo.
Fig. 3: DDX6 plays a minor role during homoeostatic haematopoiesis but is important for regenerative haematopoiesis.
Fig. 4: P-bodies sequester translationally repressed mRNAs encoding key tumour suppressors.
Fig. 5: Loss of DDX6 impacts the chromatin architecture of AML cells.

Similar content being viewed by others

Data availability

RNA-seq, ATAC-seq, eCLIP-seq, polysome profiling, CUT&RUN and CUT&Tag data that support the findings of this study have been deposited in the Gene Expression Omnibus under accession codes GSE260919, GSE224858, GSE261265 and GSE224643. Raw data and the MaxQuant output for the proteomics have been deposited to the MassIVE database under the accession number MSV000090973. Previously published ChIP-seq data and polysome profiling data that were re-analysed here are available under accession code GSM1003586 (ref. 22) and GSE202227 (ref. 76). The human AML data were derived from the TCGA Research Network at http://cancergenome.nih.gov/. All other data supporting the findings of this study are available from the corresponding author on reasonable request. Source data are provided with this paper.

References

  1. van Galen, P. et al. Single-cell RNA-seq reveals AML hierarchies relevant to disease progression and immunity. Cell 176, 1265–1281 e1224 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Fabbri, L., Chakraborty, A., Robert, C. & Vagner, S. The plasticity of mRNA translation during cancer progression and therapy resistance. Nat. Rev. Cancer 21, 558–577 (2021).

    Article  CAS  PubMed  Google Scholar 

  3. Buxbaum, A. R., Haimovich, G. & Singer, R. H. In the right place at the right time: visualizing and understanding mRNA localization. Nat. Rev. Mol. Cell Biol. 16, 95–109 (2015).

    Article  CAS  PubMed  Google Scholar 

  4. Roden, C. & Gladfelter, A. S. RNA contributions to the form and function of biomolecular condensates. Nat. Rev. Mol. Cell Biol. 22, 183–195 (2021).

    Article  CAS  PubMed  Google Scholar 

  5. Sheth, U. & Parker, R. Decapping and decay of messenger RNA occur in cytoplasmic processing bodies. Science 300, 805–808 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Courel, M. et al. GC content shapes mRNA storage and decay in human cells. eLife 8, e49708 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Brengues, M., Teixeira, D. & Parker, R. Movement of eukaryotic mRNAs between polysomes and cytoplasmic processing bodies. Science 310, 486–489 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Di Stefano, B. et al. The RNA helicase DDX6 controls cellular plasticity by modulating P-body homeostasis. Cell Stem Cell 25, 622–638 e613 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Hubstenberger, A. et al. P-body purification reveals the condensation of repressed mRNA regulons. Mol. Cell 68, 144–157 e145 (2017).

    Article  CAS  PubMed  Google Scholar 

  10. Teixeira, D., Sheth, U., Valencia-Sanchez, M. A., Brengues, M. & Parker, R. Processing bodies require RNA for assembly and contain nontranslating mRNAs. RNA 11, 371–382 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Ayache, J. et al. P-body assembly requires DDX6 repression complexes rather than decay or Ataxin2/2L complexes. Mol. Biol. Cell 26, 2579–2595 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Wilbertz, J. H. et al. Single-molecule imaging of mRNA localization and regulation during the integrated stress response. Mol. Cell 73, 946–958 e947 (2019).

    Article  CAS  PubMed  Google Scholar 

  13. Cardona, A. H. et al. Self-demixing of mRNA copies buffers mRNA:mRNA and mRNA:regulator stoichiometries. Cell 186, 4310–4324 e4323 (2023).

    Article  CAS  PubMed  Google Scholar 

  14. Hallacli, E. et al. The Parkinson’s disease protein alpha-synuclein is a modulator of processing bodies and mRNA stability. Cell 185, 2035–2056 e2033 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Lavalee, M., Curdy, N., Laurent, C., Fournie, J. J. & Franchini, D. M. Cancer cell adaptability: turning ribonucleoprotein granules into targets. Trends Cancer 7, 902–915 (2021).

    Article  CAS  PubMed  Google Scholar 

  16. Nsengimana, B. et al. Processing body (P-body) and its mediators in cancer. Mol. Cell. Biochem. 477, 1217–1238 (2022).

    Article  CAS  PubMed  Google Scholar 

  17. Ghashghaei, M. et al. miR-148a-3p and DDX6 functional link promotes survival of myeloid leukemia cells. Blood Adv. 7, 3846–3861 (2023).

    Article  CAS  PubMed  Google Scholar 

  18. Hernandez, G. et al. Decapping protein EDC4 regulates DNA repair and phenocopies BRCA1. Nat. Commun. 9, 967 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Heyes, E. et al. Identification of gene targets of mutant C/EBPα reveals a critical role for MSI2 in CEBPA-mutated AML. Leukemia 35, 2526–2538 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Brandstoetter, T. et al. SBNO2 is a critical mediator of STAT3-driven hematological malignancies. Blood https://doi.org/10.1182/blood.2022018494 (2023).

  21. Luo, Y., Na, Z. & Slavoff, S. A. P-bodies: composition, properties, and functions. Biochemistry 57, 2424–2431 (2018).

    Article  CAS  PubMed  Google Scholar 

  22. Consortium, E. P. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

    Article  Google Scholar 

  23. Rasch, F., Weber, R., Izaurralde, E. & Igreja, C. 4E-T-bound mRNAs are stored in a silenced and deadenylated form. Genes Dev. 34, 847–860 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Kamenska, A. et al. The DDX6-4E-T interaction mediates translational repression and P-body assembly. Nucleic Acids Res. 44, 6318–6334 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Zong, H. et al. A hyperactive signalosome in acute myeloid leukemia drives addiction to a tumor-specific Hsp90 species. Cell Rep. 13, 2159–2173 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Nakagawa, T. et al. Establishment of a leukaemic cell line from a patient with acquisition of chromosomal abnormalities during disease progression in myelodysplastic syndrome. Br. J. Haematol. 85, 469–476 (1993).

    Article  CAS  PubMed  Google Scholar 

  27. Erb, M. A. et al. Transcription control by the ENL YEATS domain in acute leukaemia. Nature 543, 270–274 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Yan, M. et al. A previously unidentified alternatively spliced isoform of t(8;21) transcript promotes leukemogenesis. Nat. Med. 12, 945–949 (2006).

    Article  CAS  PubMed  Google Scholar 

  29. Kim, J. et al. The RNA helicase DDX6 controls early mouse embryogenesis by repressing aberrant inhibition of BMP signaling through miRNA-mediated gene silencing. PLoS Genet. 18, e1009967 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Doulatov, S. et al. Induction of multipotential hematopoietic progenitors from human pluripotent stem cells via respecification of lineage-restricted precursors. Cell Stem Cell 13, 459–470 (2013).

    Article  CAS  PubMed  Google Scholar 

  31. Brzezinka, K. et al. Functional diversity of inhibitors tackling the differentiation blockage of MLL-rearranged leukemia. J. Hematol. Oncol. 12, 66 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Hong, Y., Martin, J. F., Vainchenker, W. & Erusalimsky, J. D. Inhibition of protein kinase C suppresses megakaryocytic differentiation and stimulates erythroid differentiation in HEL cells. Blood 87, 123–131 (1996).

    Article  CAS  PubMed  Google Scholar 

  33. Stein, E. M. et al. The DOT1L inhibitor pinometostat reduces H3K79 methylation and has modest clinical activity in adult acute leukemia. Blood 131, 2661–2669 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Jangra, R. K., Yi, M. & Lemon, S. M. DDX6 (Rck/p54) is required for efficient hepatitis C virus replication but not for internal ribosome entry site-directed translation. J. Virol. 84, 6810–6824 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Brandmann, T. et al. Molecular architecture of LSM14 interactions involved in the assembly of mRNA silencing complexes. EMBO J. 37, e97869 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Brothers, W. R., Fakim, H., Kajjo, S. & Fabian, M. R. P-bodies directly regulate MARF1-mediated mRNA decay in human cells. Nucleic Acids Res. 50, 7623–7636 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Na, Z. et al. The NBDY microprotein regulates cellular RNA decapping. Biochemistry 59, 4131–4142 (2020).

    Article  CAS  PubMed  Google Scholar 

  38. Na, Z. et al. Phosphorylation of a human microprotein promotes dissociation of biomolecular condensates. J. Am. Chem. Soc. 143, 12675–12687 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. D’Lima, N. G. et al. A human microprotein that interacts with the mRNA decapping complex. Nat. Chem. Biol. 13, 174–180 (2017).

    Article  PubMed  Google Scholar 

  40. Ren, Z. et al. A PRC2-Kdm5b axis sustains tumorigenicity of acute myeloid leukemia. PNAS 119, e2122940119 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Wong, S. H. et al. The H3K4-methyl epigenome regulates leukemia stem cell oncogenic potential. Cancer Cell 28, 198–209 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Muto, T. et al. TRAF6 functions as a tumor suppressor in myeloid malignancies by directly targeting MYC oncogenic activity. Cell Stem Cell 29, 298–314 e299 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Gudgeon, C. J. et al. High expression of neutrophil elastase predicts improved survival in pediatric acute myeloid leukemia: a report from the Children’s Oncology Group. Leuk. Lymphoma 54, 202–204 (2013).

    Article  PubMed  Google Scholar 

  44. Perner, F. et al. YBX1 mediates translation of oncogenic transcripts to control cell competition in AML. Leukemia 36, 426–437 (2022).

    Article  CAS  PubMed  Google Scholar 

  45. Jiang, L. et al. Multidimensional study of the heterogeneity of leukemia cells in t(8;21) acute myelogenous leukemia identifies the subtype with poor outcome. PNAS 117, 20117–20126 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Van Nostrand, E. L. et al. Robust transcriptome-wide discovery of RNA-binding protein binding sites with enhanced CLIP (eCLIP). Nat. Methods 13, 508–514 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Sloan, K. E. & Bohnsack, M. T. Unravelling the mechanisms of RNA helicase regulation. Trends Biochem. Sci. 43, 237–250 (2018).

    Article  CAS  PubMed  Google Scholar 

  48. Chan, S. P. & Slack, F. J. microRNA-mediated silencing inside P-bodies. RNA Biol. 3, 97–100 (2006).

    Article  CAS  PubMed  Google Scholar 

  49. Freimer, J. W., Hu, T. J. & Blelloch, R. Decoupling the impact of microRNAs on translational repression versus RNA degradation in embryonic stem cells. eLife 7, e38014 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Kulkarni, M., Ozgur, S. & Stoecklin, G. On track with P-bodies. Biochem. Soc. Trans. 38, 242–251 (2010).

    Article  CAS  PubMed  Google Scholar 

  51. Xue, S. et al. Histone lysine demethylase KDM5B maintains chronic myeloid leukemia via multiple epigenetic actions. Exp. Hematol. 82, 53–65 (2020).

    Article  CAS  PubMed  Google Scholar 

  52. Patel, K. P. et al. Acute myeloid leukemia with IDH1 or IDH2 mutation: frequency and clinicopathologic features. Am. J. Clin. Pathol. 135, 35–45 (2011).

    Article  CAS  PubMed  Google Scholar 

  53. Bluemn, T. et al. Differential roles of BAF and PBAF subunits, Arid1b and Arid2, in MLL-AF9 leukemogenesis. Leukemia 36, 946–955 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Fog, C. K. et al. Loss of PRDM11 promotes MYC-driven lymphomagenesis. Blood 125, 1272–1281 (2015).

    Article  CAS  PubMed  Google Scholar 

  55. Horak, P. et al. TUSC3 loss alters the ER stress response and accelerates prostate cancer growth in vivo. Sci. Rep. 4, 3739 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Katerndahl, C. D. S. et al. Tumor suppressor function of Gata2 in acute promyelocytic leukemia. Blood 138, 1148–1161 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Yu, J. S. et al. PCDH8, the human homolog of PAPC, is a candidate tumor suppressor of breast cancer. Oncogene 27, 4657–4665 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Konopleva, M. & Letai, A. BCL-2 inhibition in AML: an unexpected bonus? Blood 132, 1007–1012 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Park, S. M. et al. IKZF2 drives leukemia stem cell self-renewal and inhibits myeloid differentiation. Cell Stem Cell 24, 153–165 e157 (2019).

    Article  CAS  PubMed  Google Scholar 

  60. Roe, J. S. & Vakoc, C. R. The essential transcriptional function of BRD4 in acute myeloid leukemia. Cold Spring Harb. Symp. Quant. Biol. 81, 61–66 (2016).

    Article  PubMed  Google Scholar 

  61. Shi, X. et al. Nuclear NAD(+) homeostasis governed by NMNAT1 prevents apoptosis of acute myeloid leukemia stem cells. Sci. Adv. 7, eabf3895 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Vegi, N. M. et al. MEIS2 is an oncogenic partner in AML1-ETO-positive AML. Cell Rep. 16, 498–507 (2016).

    Article  CAS  PubMed  Google Scholar 

  63. Buenrostro, J. D. et al. Integrated single-cell analysis maps the continuous regulatory landscape of human hematopoietic differentiation. Cell 173, 1535–1548 e1516 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Itoh-Nakadai, A. et al. The transcription repressors Bach2 and Bach1 promote B cell development by repressing the myeloid program. Nat. Immunol. 15, 1171–1180 (2014).

    Article  CAS  PubMed  Google Scholar 

  65. Iwasaki, H. et al. Distinctive and indispensable roles of PU.1 in maintenance of hematopoietic stem cells and their differentiation. Blood 106, 1590–1600 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Santaguida, M. et al. JunB protects against myeloid malignancies by limiting hematopoietic stem cell proliferation and differentiation without affecting self-renewal. Cancer Cell 15, 341–352 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Gaillard, C. et al. Identification of IRF8 as a potent tumor suppressor in murine acute promyelocytic leukemia. Blood Adv. 2, 2462–2466 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Xhabija, B. & Kidder, B. L. KDM5B is a master regulator of the H3K4-methylome in stem cells, development and cancer. Semin. Cancer Biol. 57, 79–85 (2019).

    Article  CAS  PubMed  Google Scholar 

  69. Hinge, A. et al. Asymmetrically segregated mitochondria provide cellular memory of hematopoietic stem cell replicative history and drive HSC attrition. Cell Stem Cell 26, 420–430 e426 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Ho, T. T. et al. Autophagy maintains the metabolism and function of young and old stem cells. Nature 543, 205–210 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Spevak, C. C. et al. Hematopoietic stem and progenitor cells exhibit stage-specific translational programs via mTOR- and CDK1-dependent mechanisms. Cell Stem Cell 26, 755–765 e757 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Herrejon Chavez, F. et al. RNA binding protein SYNCRIP maintains proteostasis and self-renewal of hematopoietic stem and progenitor cells. Nat. Commun. 14, 2290 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Hidalgo San Jose, L. et al. Modest declines in proteome quality impair hematopoietic stem cell self-renewal. Cell Rep. 30, 69–80 e66 (2020).

    Article  CAS  PubMed  Google Scholar 

  74. Liu, J., Valencia-Sanchez, M. A., Hannon, G. J. & Parker, R. MicroRNA-dependent localization of targeted mRNAs to mammalian P-bodies. Nat. Cell Biol. 7, 719–723 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Zhao, M., Kim, P., Mitra, R., Zhao, J. & Zhao, Z. TSGene 2.0: an updated literature-based knowledgebase for tumor suppressor genes. Nucleic Acids Res. 44, D1023–D1031 (2016).

    Article  CAS  PubMed  Google Scholar 

  76. Park, H. J. et al. Therapeutic resistance in acute myeloid leukemia cells is mediated by a novel ATM/mTOR pathway regulating oxidative phosphorylation. eLife 11, e79940 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Pinto do, O. P., Kolterud, A. & Carlsson, L. Expression of the LIM-homeobox gene LH2 generates immortalized steel factor-dependent multipotent hematopoietic precursors. EMBO J. 17, 5744–5756 (1998).

    Article  CAS  Google Scholar 

  78. Michlits, G. et al. Multilayered VBC score predicts sgRNAs that efficiently generate loss-of-function alleles. Nat. Methods 17, 708–716 (2020).

    Article  CAS  PubMed  Google Scholar 

  79. Schmoellerl, J. et al. EVI1 drives leukemogenesis through aberrant ERG activation. Blood https://doi.org/10.1182/blood.2022016592 (2022).

  80. de Almeida, M. et al. AKIRIN2 controls the nuclear import of proteasomes in vertebrates. Nature 599, 491–496 (2021).

    Article  PubMed  Google Scholar 

  81. Maeda, R. et al. RNA decay in processing bodies is indispensable for adipogenesis. Cell Death Dis. 12, 285 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Chen, B. et al. Dynamic imaging of genomic loci in living human cells by an optimized CRISPR/Cas system. Cell 155, 1479–1491 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Tsanov, N. et al. smiFISH and FISH-quant - a flexible single RNA detection approach with super-resolution capability. Nucleic Acids Res. 44, e165 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  84. Mueller, F. et al. FISH-quant: automatic counting of transcripts in 3D FISH images. Nat. Methods 10, 277–278 (2013).

    Article  CAS  PubMed  Google Scholar 

  85. Buenrostro, J. D., Giresi, P. G., Zaba, L. C., Chang, H. Y. & Greenleaf, W. J. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods 10, 1213–1218 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Blue, S. M. et al. Transcriptome-wide identification of RNA-binding protein binding sites using seCLIP-seq. Nat. Protoc. 17, 1223–1265 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Conway, A. E. et al. Enhanced CLIP uncovers IMP protein-RNA targets in human pluripotent stem cells important for cell adhesion and survival. Cell Rep. 15, 666–679 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Tomas-Daza, L. et al. Low input capture Hi-C (liCHi-C) identifies promoter-enhancer interactions at high-resolution. Nat. Commun. 14, 268 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Di Tommaso, P. et al. Nextflow enables reproducible computational workflows. Nat. Biotechnol. 35, 316–319 (2017).

    Article  PubMed  Google Scholar 

  90. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    Article  CAS  PubMed  Google Scholar 

  92. Li, W. et al. MAGeCK enables robust identification of essential genes from genome-scale CRISPR/Cas9 knockout screens. Genome Biol. 15, 554 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  93. Wang, B. et al. Integrative analysis of pooled CRISPR genetic screens using MAGeCKFlute. Nat. Protoc. 14, 756–780 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Zhang, Y. et al. Model-based analysis of ChIP-seq (MACS). Genome Biol. 9, R137 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  95. Li, Q. & Zhang, F. A regression framework for assessing covariate effects on the reproducibility of high-throughput experiments. Biometrics 74, 803–813 (2018).

    Article  PubMed  Google Scholar 

  96. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  97. Yu, G., Wang, L. G. & He, Q. Y. ChIPseeker: an R/Bioconductor package for ChIP peak annotation, comparison and visualization. Bioinformatics 31, 2382–2383 (2015).

    Article  CAS  PubMed  Google Scholar 

  98. Nystrom, S. L. & McKay, D. J. Memes: a motif analysis environment in R using tools from the MEME Suite. PLoS Comput. Biol. 17, e1008991 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Castro-Mondragon, J. A. et al. JASPAR 2022: the 9th release of the open-access database of transcription factor binding profiles. Nucleic Acids Res. 50, D165–D173 (2022).

    Article  CAS  PubMed  Google Scholar 

  100. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows–Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Ramirez, F., Dundar, F., Diehl, S., Gruning, B. A. & Manke, T. deepTools: a flexible platform for exploring deep-sequencing data. Nucleic Acids Res. 42, W187–W191 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Robinson, J. T. et al. Integrative genomics viewer. Nat. Biotechnol. 29, 24–26 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  104. Tarasov, A., Vilella, A. J., Cuppen, E., Nijman, I. J. & Prins, P. Sambamba: fast processing of NGS alignment formats. Bioinformatics 31, 2032–2034 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Xie, Z. et al. Gene set knowledge discovery with Enrichr. Curr. Protoc. 1, e90 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Huber, W. et al. Orchestrating high-throughput genomic analysis with Bioconductor. Nat. Methods 12, 115–121 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Lawrence, M. et al. Software for computing and annotating genomic ranges. PLoS Comput. Biol. 9, e1003118 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Shishkova, E., Hebert, A. S., Westphall, M. S. & Coon, J. J. Ultra-high pressure (>30,000 psi) packing of capillary columns enhancing depth of shotgun proteomic analyses. Anal. Chem. 90, 11503–11508 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Cox, J. & Mann, M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat. Biotechnol. 26, 1367–1372 (2008).

    Article  CAS  PubMed  Google Scholar 

  110. Brademan, D. R. et al. Argonaut: a web platform for collaborative multi-omic data visualization and exploration. Patterns 1, 100122 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  111. Xiao, Z., Zou, Q., Liu, Y. & Yang, X. Genome-wide assessment of differential translations with ribosome profiling data. Nat. Commun. 7, 11194 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank all members of the Sardina, Grebien and Di Stefano groups for stimulating scientific discussions. We thank S. Doulatov for the human iPS cell-derived CD34+ cells, D. Lacorazza for the THP-1 cells, D. Nakada for the MOLM-13-Cas9 cells, L. Brunetti for advice on generating the DDX6 degron AML cells, J. Yustein and M. Mamonkin for the NSG mice, M. Fabian for the LSM14 mutant plasmids, F. Stossi and A. Tostado of the BCM Integrated Microscopy Core for help with the smFISH experiments, the BCM GEMC for the generation of Ddx6 transgenic mice and Z. Ianniello for help with polysome profiling experiments. B.D.S. is a Cancer Prevention and Research Institute of Texas (CPRIT) Scholar in Cancer Research. B.D.S. is supported by the CPRIT Recruitment of First-Time, Tenure-Track Faculty Member Award RR200079, the American Society of Hematology Scholar Award, the B+ Foundation, the Worldwide Cancer Research Foundation, the Milky Way Research Foundation Investigator Award, the Nancy Chang, PhD Award for Research Excellence and the National Institutes of Health (NIH) MIRA award 1R35GM147126-01. S.K. is supported by NIH 5T32DK060445-19 and NIH 1F32CA288043-01. Research in the F.G. laboratory was supported by the European Union’s Horizon 2020 research and innovation programme (Marie Sklodowska-Curie grant agreement no. 813091) and the Austrian Science Fund (projects P-35628, P35298 to F.G.). Research in the J.L.S. laboratory was funded by Worldwide Cancer Research (20-0269) and the grant PID2019-111243RA-I00 funded by MICIU/AEI/10.13039/501100011033. J.L.S. is supported by Instituto de Salud Carlos III (CP19/00176). Research in the B.M.J. laboratory was supported by grant PID2021-125277OB-I00 funded by MICIU/AEI/10.13039/501100011033 and FEDER/EU. B.M.J. is supported by Instituto de Salud Carlos III (CP22/00127). We thank CERCA Programme/Generalitat de Catalunya for institutional support. E.V.N. is a CPRIT Scholar in Cancer Research (RR200040). This project was supported by the Cytometry and Cell Sorting Core at Baylor College of Medicine with funding from the CPRIT Core Facility Support Award (CPRIT-RP180672), the NIH (CA125123 and RR024574) and the assistance of J. M. Sederstrom. We are grateful for support on this project from NIH grant P41 GM108538 (National Center for Quantitative Biology of Complex Systems) to J.J.C.

Author information

Authors and Affiliations

Authors

Contributions

S.K., L.P., F.G. and B.D.S. conceived the study and wrote the manuscript with input from J.L.S.; S.K., L.P., N.L., P.P., T.E., Y.C., C.S., B.A. and G.M. performed experiments and analysed the data; A.V.L., G.V. and J.L.S. performed and analysed the AML patient-derived xenograft and primary CD34+ experiments; A.P.-R., L.T.-D. and B.M.J. performed liHi-C and CUT&RUN experiments in collaboration with G.V., A.V.L. and J.L.S.; J.S. and Q.C. performed and analysed the small RNA-seq experiments; A.J., E.S. and J.C. performed and analysed the proteomic data; A.S., M.B. and E.V.N. performed and analysed the eCLIP-seq; A.M., T.V. and P.M. helped with the AML patient-derived xenograft and the primary CD34+ experiments; A.F. supervised the polysome profiling experiments; I.M.M. and K.K. helped with HPC7 experiments; R.R. helped with the primary patient AML and CD34+ cells.

Corresponding authors

Correspondence to Jose L. Sardina, Florian Grebien or Bruno Di Stefano.

Ethics declarations

Competing interests

E.V.N. is co-founder, member of the Board of Directors, on the SAB, equity holder and paid consultant for Eclipse BioInnovations, on the SAB of RNAConnect and is inventor of intellectual property owned by University of California San Diego. The interests of E.V.N. have been reviewed and approved by the Baylor College of Medicine in accordance with its conflict of interest policies. P.M. is a co-founder, member of the Board of Directors, equity holder and paid consultant for OneChain Immunotherapeutics (Barcelona, Spain). This work has no connection with and is not related to the scientific interests of OneChain Immunotherapeutics. J.J.C. is a consultant for Thermo Fisher Scientific, 908 Devices and Seer. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Cell Biology thanks Anthony Khong and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. 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 CRISPR screens identify P-body regulators as a selective dependency in AML.

(a) Cytospins of HPC7 and CebpaN-mutant/C-mutant (CNC) cells. n = 3 independent experiments. (b) Schematic illustration of pooled genome-wide CRISPR/Cas9 dropout screening strategy. (c) Venn diagram showing filtering strategy for the identification of 308 leukaemia-specific genetic dependencies shown in Fig. 1a. (d, e) Competition-based proliferation assay in (d) CNC Cas9 cells or (e) HPC7 Cas9 cells, illustrated as colour-coded percentage of iRFP670+ cells transduced with indicated sgRNAs over 19 d. The non-targeting sgCTRL is used as a negative control, sgRpa3, targeting essential gene Rpa3, is used as a positive control. Results are normalized to day 5 post-infection. Two-way ANOVA with Dunnett’s post-hoc test, n = 3 biological replicates per group, mean ± s.e.m. (f) Boxplots showing the expression levels (log2 mRNA) of P-body related genes (displayed in Fig. 1c) in AML, normal HSPCs, and MDS samples. AML normal karyotype (n = 28), AML t(15;17) (n = 28), AML t(8;21) (n = 28), AML t(11q23)/MLL (n = 28), HSC (haematopoietic stem cell) (n = 6), GMP (granulocyte monocyte progenitor) (n = 7), MDS (myelodysplastic syndromes) (n = 28). Box centre line indicates median, box limits indicate upper (Q3) and lower quartiles (Q1), lower whisker is Q1 – 1.5 × interquartile range (IQR) and upper whisker is Q3 + 1.5 × IQR. Two-tailed t-test with Welch’s correction. (g) Boxplots showing normalized H3K27ac ChIP-seq signal (RPKM) in MOLM-13 cells (GSM4685439 and GSM4685440) and CD34+ HSPCs at enhancers (n = 26 per group) (left panel) and promoter (n = 214 per group) (right panel) regions (GSM772885 and GSM772894) of P-body genes. Box centre line indicates median, box limits indicate upper (Q3) and lower quartiles (Q1), lower whisker is Q1 – 1.5 × IQR and upper whisker is Q3 + 1.5 × IQR. Two-tailed t-test with Welch’s correction. (h) Significant correlation between the expression of DDX6 and EIF4ENIF1 in the TCGA-LAML dataset. Pearson’s correlation coefficients and corresponding p-values (two-tailed one-sample Student’s t-test) are indicated. (i, j) Kaplan–Meier survival analysis plots of TCGA data for AML patients with low vs high expression of (i) EIF4ENIF1 (n = 27 patients per group) or (j) DDX6 (n = 53 patients per group). (k) DDX6 mRNA expression in AML (red) compared to other cancers, based on data from TCGA database. Data are presented as mean log2 expression with range. Black dots: expression levels in normal cells; Blue dots: expression levels in cancer cells. (l) DDX6 expression in AML patient samples compared to normal HSPCs. AML: n = 48, AML t(15;17): n = 54, AML inv(16)/t(16;16): n = 47, AML t(8;21): n = 60, AML t(11q23)/MLL: n = 43 patients. HSC (haematopoietic stem cell): n = 6, GMP (granulocyte monocyte progenitor): n = 7 healthy individuals (BloodSpot data of DDX6 probe 204909_at). Unpaired two-tailed Student’s t-test. (m) qRT–PCR for DDX6 in human primary CD34+ cells (n = 7 healthy individuals) and AML patient cells (n = 10 patients). Unpaired two-tailed Student’s t-test, mean.

Extended Data Fig. 2 DDX6 is essential for AML cell proliferation and survival in vitro.

(a) Representative IF imaging of EDC4 punctae (green) in control and DDX6 overexpressing HPC7 cells. Nuclei were counterstained with DAPI (blue). Scale: 2.5 µm. (b) Quantification of EDC4 punctae in HPC7 cells overexpressing DDX6 by IF. n = 45-55 cells per group, unpaired two-tailed Student’s t-test, mean ± s.e.m. (c) Representative Western blot image of DDX6 in the nuclear and cytoplasmic fractions of MOLM-13 cells. n = 3 independent experiments. (d) Representative IF imaging of LSM14A (green) and DDX6 (red) punctae in control and DDX6 KD MOLM-13 cells. Nuclei were counterstained with DAPI. Scale: 10 µm. (e) Quantification of LSM14A+DDX6+ punctae in control and DDX6 KD MOLM-13 cells by IF. Unpaired two-tailed Student’s t-test, n = 24–30 cells per group, mean ± s.e.m. (f) Representative intracellular flow cytometry plots showing DDX6 levels in shCTRL and shDDX6 MOLM-13 cells. (g) qRT–PCR validation of DDX6 KD in MOLM-13 cells. Unpaired two-tailed Student’s t-test, n = 3 biologically independent samples per group, mean ± s.e.m. (h) Proliferation assay for shCTRL and shDDX6 AML cell lines at the indicated time points after transduction. Two-way ANOVA with Dunnett’s post-hoc test, n = 3 biological replicates per group, mean ± s.e.m. (i) Competition-based proliferation assays upon DDX6 knockdown performed in indicated cell lines. shRPL17, targeting the essential gene RPL17, is used as a positive control. Two-way ANOVA with Dunnett’s post-hoc test, n = 3 biologically independent samples per group, mean ± s.e.m. (j) Heatmap summarizing the competition-based proliferation assays upon DDX6 knockout in indicated Cas9-expressing cell lines. Data are illustrated as colour-coded percentage of iRFP670+ cells transduced with indicated sgRNAs over 17 d. sgAAVS1 is used as negative control, sgRPL17 is used as positive control. Results are normalized to day 3 post-infection, (n = 3 biologically independent samples per group, mean). (k) Proliferation assay for CTRL and DDX6 KO human AML cell lines at the indicated time points after transduction. Unpaired two-tailed Student’s t-test, n = 3 biologically independent samples per group, mean ± s.e.m. (l) Schematic of dCas9-KRAB and sgRNA vectors (upper panel). Representative intracellular flow cytometry plots for DDX6 in CRISPRi HEL cells, either untreated (UT) or treated with doxycycline (DOX) for 7 d (lower panel). (m) Representative IF imaging of EDC4 punctae (green) in CRISPRi HEL cells, either untreated (UT) or treated with doxycycline (DOX) for 4 d. Nuclei were counterstained with DAPI (blue). Scale: 2.5 µm. (n) Proliferation assay for UT or DOX-treated CRISPRi HEL cells at the indicated time points after transduction. Unpaired two-tailed Student’s t-test, n = 3 biologically independent samples per group, mean ± s.e.m. (o) Representative images of UT or DOX-treated CRISPRi HEL cells. Scale: 10 µm. (p) Megakaryocytic differentiation in CRISPRi HEL cells 7 d after DDX6 silencing was quantified by flow cytometry, using CD41 and CD61 as markers. Unpaired two-tailed Student’s t-test, n = 3 biologically independent samples per group, mean ± s.e.m.

Source data

Extended Data Fig. 3 DDX6 is essential for the proliferation and gene expression programme of AML cells.

(a) Flow cytometric analysis of cell death (Annexin V+) in shCTRL and shDDX6 AML cell lines. Unpaired two-tailed Student’s t-test, n = 3 biologically independent samples per group, mean ± s.e.m. (b) Correlation heatmap showing the correlation (r) values between MOLM-13 RNA-seq samples. Scale bar represents the range of the correlation coefficients (r) displayed. (c) Heatmap of RNA-seq data for shCTRL and shDDX6 HL-60 cells (n = 2, FC > 1.5; p < 0.05). Upregulated genes are depicted in red, while in blue are downregulated genes. (d) Correlation heatmap showing the correlation (r) values between HL-60 RNA-seq samples. Scale bar represents the range of the correlation coefficients (r) displayed. (e) GO enrichment analysis of differentially expressed genes in control vs. DDX6 KD HL-60 cells. (f) qRT–PCR validation of (left) EIF4ENIF1 KD and (right) LSM14A KD in MOLM-13 cells. Unpaired two-tailed Student’s t-test, n = 3 biologically independent samples per group, mean ± s.e.m. (g) Quantification of LSM14A+DDX6+ punctae in control and (left) EIF4ENIF1 KD or (right) LSM14A KD MOLM-13 cells by IF. Unpaired two-tailed Student’s t-test, n = 17-31 cells per group, mean ± s.e.m. (h) Representative Western blot showing HA-tagged endogenous DDX6 protein levels in DDX6-FKBP12F36V MOLM-13 cells after 2 d of dTAG-13 treatment, followed by washout and culture for 5 d. n = 3 independent experiments. (i) Representative IF imaging of EDC4 punctae (green) and DDX6 punctae (red) in DDX6-FKBP12F36V MOLM-13 cells after 2 d of dTAG-13 treatment, followed by washout and culture for 5 d. Nuclei were counterstained with DAPI (blue). Scale: 10 µm. (j) Quantification of EDC4+DDX6+ punctae in the indicated cells by IF (n = 47-87 cells per group, mean ± s.e.m.). (k) Representative Western blot showing HA-tagged endogenous DDX6 protein levels in DDX6-FKBP12F36V HL-60 cells after 2 d of dTAG-13 treatment, followed by washout and culture for 5 d. n = 3 independent experiments. (l) Proliferation of DDX6-FKBP12F36V HL-60 cells, either untreated (UT), continuously treated with dTAG-13, or treated with dTAG-13 for 2, 5, or 6 days, followed by washout and culture (n = 3 biological replicates per group, mean ± s.e.m.). (m) Percentages of control or DDX6 KD MOLM-13 cells in the bone marrow and spleens of NSG mice (25 d post-transplant) quantified by flow cytometry. Unpaired two-tailed Student’s t-test, n = 3 mice per group, mean ± s.e.m. (n) Representative intracellular flow cytometry plot for DDX6 in shCTRL and shDDX6 MOLM-13 cells in the bone marrow of NSG mice (45 d post-transplant). (o) Genotyping PCR demonstrating poly(I:C)-induced Ddx6 deletion in c-Kit+ haematopoietic progenitor cells isolated from the bone marrow of Mx1-Cre/Ddx6fl/fl mice. n = 3 independent experiments. (p) Left: Representative IF imaging of EDC4 punctae (green) and DDX6 punctae (red) in Ddx6WT and Ddx6KO c-Kit+ haematopoietic progenitor cells. Nuclei were counterstained with DAPI (blue), scale: 10 µm. Right: Quantification of EDC4+DDX6+ punctae by IF in Ddx6WT and Ddx6KO c-Kit+ haematopoietic progenitor cells. Unpaired two-tailed Student’s t-test, n = 60-63 cells per group, mean ± s.e.m. (q) Left: Representative IF imaging of LSM14A punctae (green) and DDX6 punctae (red) in Ddx6WT and Ddx6KO c-Kit+ haematopoietic progenitor cells. Nuclei were counterstained with DAPI (blue), scale: 10 µm. Right: Quantification of LSM14A+DDX6+ punctae by IF in Ddx6WT and Ddx6KO c-Kit+ haematopoietic progenitor cells. Unpaired two-tailed Student’s t-test, n = 32-33 cells per group, mean ± s.e.m.

Source data

Extended Data Fig. 4 DDX6 loss has little effect on steady-state haematopoiesis.

(a) Representative image of spleens from mice 90 d after transplantation with MLL-AF9-transduced Ddx6WT and Ddx6KO c-Kit+ cells. (b) Weights of spleens isolated from mice 90 days after transplantation with MLL-AF9-transduced Ddx6WT and Ddx6KO c-Kit+ cells. Unpaired two-tailed Student’s t-test, Ddx6WT n = 4 mice, Ddx6KO n = 6 mice, mean ± s.e.m. (c) Representative flow cytometry plots of leukaemia (GFP+) cells in bone marrow from mice 90 d after transplantation with MLL-AF9-transduced Ddx6WT and Ddx6KO c-Kit+ cells. (d) Representative intracellular flow cytometry plot for DDX6 in shCTRL and shDDX6 primary bone marrow human CD34+ cells. (e) Representative flow cytometry plots showing cell death (Annexin V+/Sytox AAD+) in shCTRL and shDDX6 primary human CD34+ cells. (f) qRT–PCR analysis to validate DDX6 KD in human iPSC-derived CD34+ HSPCs. Unpaired two-tailed t-test with Welch’s correction, n = 3 biologically independent samples per group, mean ± s.e.m. (g) Proliferation assay for shCTRL and shDDX6 human iPSC-derived HSPCs at the indicated timepoints after transduction. Unpaired two-tailed Student’s t-test, n = 3 biologically independent samples per group, mean ± s.e.m. (h) Flow cytometric analysis for myeloid differentiation (CD11b median fluorescence intensity (MFI)) in shCTRL vs shDDX6 human iPSC-derived HSPCs. Unpaired two-tailed Student’s t-test, n = 3 biologically independent samples per group. (i) Flow cytometric analysis of cell death (Annexin V+) in human iPSC-derived HPCs. Unpaired two-tailed Student’s t-test, n = 3 biologically independent samples per group, mean ± s.e.m. (j, k) Representative Western blots validating (j) LSM14A KD and (k) EIF4ENIF1 KD in primary human CD34+ cells. n = 3 independent experiments. (l) Representative flow cytometry plots and quantification for CMP, GMP, and MEP populations (gated on LK cells) in the bone marrow of Mx1-Cre and Mx1-Cre/Ddx6fl/fl mice 110 d after Ddx6 deletion. Unpaired two-tailed Student’s t-test, Ddx6WT n = 5 mice, Ddx6KO n = 5 mice, mean ± s.e.m. (m, n) Quantification of myeloid cells, T cells, and B cells as a percentage of CD45+ cells in the (m) bone marrow and (n) spleens of Mx1-Cre and Mx1-Cre/Ddx6fl/fl mice 110 d after Ddx6 deletion. Unpaired two-tailed Student’s t-test, Ddx6WT n = 5 mice, Ddx6KO n = 5 mice, mean ± s.e.m. (o, p) Frequency of erythroid cells in the (o) bone marrow and (p) peripheral blood Mx1-Cre and Mx1-Cre/Ddx6fl/fl mice 110 d after Ddx6 deletion. Unpaired two-tailed Student’s t-test, Ddx6WT n = 5 mice, Ddx6KO n = 5 mice, mean ± s.e.m. (q) Representative Western blot analysis for DDX6 in bone marrow cells of Rosa26-Cre and Rosa26-Cre/Ddx6fl/fl mice. n = 3 independent experiments. (r) Quantification of myeloid cells, T cells, and B cells as a percentage of CD45+ cells in the peripheral blood of Rosa26-Cre-ERT2 and Rosa26-Cre-ERT2/Ddx6fl/fl mice 80 d after tamoxifen treatment. Unpaired two-tailed Student’s t-test, Ddx6WT n = 3 mice, Ddx6KO n = 3 mice, mean ± s.e.m. (s) Frequency of erythroid cells in the peripheral blood of Rosa26-Cre-ERT2 and Rosa26-Cre-ERT2/Ddx6fl/fl mice 80 d after tamoxifen treatment. Unpaired two-tailed Student’s t-test, Ddx6WT n = 3 mice, Ddx6KO n = 3 mice, mean ± s.e.m. (t) Representative flow cytometry plots showing percentages of HSC, MPP1, MPP2, and MPP4 populations, gated on LSK cells, in the bone marrow of Rosa26-Cre-ERT2 and Rosa26-Cre-ERT2/Ddx6fl/fl mice after Ddx6 deletion. (u) Quantification of HSC, MPP1, MPP2, and MPP4 populations, as a percentage of LSK cells, in the bone marrow of Rosa26-Cre-ERT2 and Rosa26-Cre-ERT2/Ddx6fl/fl mice after Ddx6 deletion. Unpaired two-tailed Student’s t-test, Ddx6WT n = 5 mice, Ddx6KO n = 5 mice, mean ± s.e.m. (v) Kaplan–Meier survival curves of Rosa26-Cre-ERT2 and Rosa26-Cre-ERT2/Ddx6fl/fl mice 4 months after tamoxifen treatment. Mantel–Cox test, Ddx6WT n = 3 mice, Ddx6KO n = 3 mice.

Source data

Extended Data Fig. 5 DDX6 regulates HSC quiescence and response to stress.

(a) Unsupervised clustering of Ddx6WT and Ddx6KO HSC, MPP1, MPP2, and MPP4 populations subjected to RNA-seq. (b) Representative flow cytometry plots of (left) MitoTracker Green FM and (right) TMRM staining in Ddx6WT and Ddx6KO HSCs, 24 d after Ddx6 deletion. (c) Representative flow cytometry plots of Ddx6WT and Ddx6KO chimerism within the donor haematopoietic compartment (CD45+) in the bone marrow 187 d after primary competitive transplantation. (d) HSCs sorted from CD45.1 and Mx1-Cre/Ddx6fl/fl mice were competitively transplanted into lethally irradiated WT recipient mice, followed by poly(I:C) treatment 34 d later. Quantification is shown of Ddx6WT and Ddx6KO chimerism within the donor haematopoietic compartment (CD45+) in peripheral blood, spleen, or bone marrow, at the indicated timepoints after Ddx6 deletion. Ddx6WT n = 8 mice, Ddx6KO n = 8 mice, two-way ANOVA with Bonferroni’s multiple comparisons test, mean ± s.e.m. (e) Flow cytometric analysis for the megakaryocytic differentiation markers CD41 and CD61 in HEL cells 4 d after vehicle (DMSO) or PMA treatment. Unpaired two-tailed Student’s t-test, n = 3 biologically independent samples per group, mean ± s.e.m. (f) Representative IF imaging of EDC4 punctae (green) and DDX6 punctae (red) in vehicle-treated and PMA-treated HEL cells. Nuclei were counterstained with DAPI (blue). Scale: 10 µm. (g) Quantification of EDC4+DDX6+ punctae in vehicle-treated and PMA-treated HEL cells by IF. Unpaired two-tailed Student’s t-test, n = 24-26 cells per group, mean ± s.e.m. (h) Representative IF imaging of LSM14A punctae (green) and DDX6 punctae (red) in vehicle-treated and PMA-treated HEL cells. Nuclei were counterstained with DAPI (blue). Scale: 10 µm. (i) Quantification of LSM14A+DDX6+ punctae in vehicle-treated and PMA-treated HEL cells by IF. Unpaired two-tailed Student’s t-test, n = 25-33 cells per group, mean ± s.e.m. (j) Flow cytometric analysis for myeloid differentiation (% CD11b+) in MOLM-13 cells 5 d after treatment with the anti-leukaemic drug EPZ-5676. Unpaired two-tailed Student’s t-test, n = 3 biologically independent samples per group, mean ± s.e.m. (k) P-body numbers (EDC4+DDX6+ punctae) in MOLM-13 cells treated with EPZ-5676. Unpaired two-tailed Student’s t-test, n = 26-30 cells per group. (l) qRT–PCR analysis of DDX6 expression in DDX6 KD MOLM-13 cells rescued with DDX6 WT or DDX6 EQ. n = 3 biologically independent samples per group, mean ± s.e.m. (m) Representative IF imaging of EDC4 punctae (green) and DDX6 punctae (red) in shCTRL MOLM-13 cells with exogenous DDX6 WT and EQ expression. Nuclei were counterstained with DAPI (blue). Scale: 10 µm. (n) Representative IF imaging of LSM14A punctae (green) and DDX6 punctae (red) in shCTRL MOLM-13 cells with exogenous DDX6 WT and EQ expression. Nuclei were counterstained with DAPI (blue). (o) Quantification of LSM14A+DDX6+ punctae in shCTRL MOLM-13 cells by IF. Unpaired two-tailed Student’s t-test, n = 12-13 cells per group, mean ± s.e.m. (p) Representative IF imaging of LSM14A punctae (green) and DDX6 punctae (red) in shDDX6 MOLM-13 cells with exogenous DDX6 WT and EQ expression. Nuclei were counterstained with DAPI (blue). Scale: 10 µm. (q) Quantification of LSM14A+DDX6+ punctae in shCTRL MOLM-13 cells by IF. Unpaired two-tailed Student’s t-test, n = 13-33 cells per group, mean ± s.e.m.

Extended Data Fig. 6 Disrupting P-body assembly abrogates AML cell proliferation.

(a) Representative intracellular flow cytometry plots showing expression of FLAG-LSM14A WT, FLAG-LSM14A ∆TFG, or FLAG-LSM14A ∆FFD in control and LSM14A KD HEL cells. (b) Representative IF imaging of EDC4 punctae (green) and DDX6 punctae (red) in control HEL cells expressing LSM14A WT, LSM14A ∆TFG, or LSM14A ∆FFD. Nuclei were counterstained with DAPI (blue). (c, d) Representative IF imaging of LSM14A punctae (green) and DDX6 punctae (red) in (c) LSM14A KD and (d) control HEL cells expressing LSM14A WT, LSM14A ∆TFG, or LSM14A ∆FFD. Nuclei were counterstained with DAPI (blue). Scale: 10 µm. (e) Quantification of LSM14A+DDX6+ punctae in the indicated cells by IF. One-way ANOVA with Dunnett’s post-hoc test, n = 16-29 cells per group, mean ± s.e.m. (f) Representative intracellular flow cytometry plot showing NBDY expression (FLAG) in MOLM-13 cells. (g) Left: Representative IF imaging of LSM14A punctae (green) and FLAG (red) in control and NBDY-expressing MOLM-13 cells, scale: 10 µm. Right: quantification of LSM14A+DDX6+ punctae in control and NBDY-expressing MOLM-13 cells by IF. Unpaired two-tailed Student’s t-test, n = 24 cells per group, mean ± s.e.m. (h) HEL cell numbers 13 d after forced expression of NBDY. Unpaired two-tailed Student’s t-test, n = 3 biologically independent samples per group, mean ± s.e.m. (i) Representative flow cytometry plots showing loss of GFP-LSM14A+ P-bodies after DDX6 silencing. (j) Distribution of RNA biotypes within the P-bodies and cytoplasm of HEL and MOLM-13 cells. (k) Distribution of P-body-enriched, cytoplasm-enriched, and non-enriched genes within each expression quartile, which range from Q1 (low expression) to Q4 (high expression). (l) RNA-seq analysis showing counts per million (CPM) values for the indicated transcripts in the cytoplasm and P-bodies of MOLM-13 cells (n = 2 biologically independent samples per group, mean). (m) Representative smFISH images of (left) POLK or (right) RSRC2 mRNA molecules (red) and GFP-LSM14A+ punctae (green). Nuclei were counterstained with DAPI (blue). Scale: 5 µm (n) Quantification of the fraction of POLK or RSCR2 transcripts colocalizing with GFP-LSM14A+ punctae in individual cells (KDM5B: n = 30 cells, mean = 69.53%, RSRC2: n = 31 cells, mean = 55.05%).

Extended Data Fig. 7 DDX6 sequesters translationally repressed mRNAs in P-bodies.

(a) Venn diagrams showing overlap between (left) cytoplasmic mRNAs or (right) P-body-associated mRNAs (FC > 1.5, p < 0.05) in MOLM-13 cells and HEL cells. Shared P-body-enriched transcripts encoding genes with potential tumour-suppressive activity are listed. (b) GO enrichment analysis for shared P-body-enriched RNAs in MOLM-13 and HEL cells (n = 2042). Two-sided Fisher’s exact test. (c) Histogram of region-based FC for DDX6 eCLIP-seq read density over size-matched input (FC > 2, padj < 0.05). (d) Scatter-plot indicating correlation between region-based fold enrichment of DDX6 eCLIP-seq datasets across biological replicates (n = 2 biologically independent samples per group). (e) Venn diagram showing the overlap between DDX6 eCLIP-seq targets and P-body-enriched RNAs (n = 2 biologically independent samples per group, FC > 2, padj < 0.05, Wald test with Benjamini–Hochberg correction) in MOLM-13 cells. (f) GO pathway enrichment analysis of DDX6-bound, P-body-enriched RNAs (n = 589) identified in (e). Two-tailed Fisher’s exact test. (g, h) Scatter-plots showing lack of correlation between P-body enrichment and expression for transcripts upregulated in DDX6 KD (g) MOLM-13 or (h) HEL cells. (i) GC content and length distribution for P-body-enriched vs cytosolic mRNAs. (j) Cumulative distribution function (CDF) plot showing translation rate (log2 FC) of P-body enriched and P-body-depleted mRNAs for shDDX6 vs. shCTRL cells. Two-sided Mann–Whitney U test. (k) Dynamic changes in small RNA distribution in MOLM-13 cells following DDX6 suppression (n = 2 biologically independent samples per group). (l) Heatmap showing expression levels of selected tsRNAs in control vs DDX6 depleted MOLM-13 cells (n = 2 biologically independent samples per group).

Extended Data Fig. 8 Genome topology rewiring following DDX6 depletion.

(a) Correlation heatmap showing the correlation (r) values between proteomic samples (n = 3). Scale bar represents the range of the correlation coefficients (r) displayed. (b) Heatmap for differentially expressed proteins exhibiting a 1.5-fold or greater difference between control and DDX6 KD MOLM-13 cells (n = 3). (c) Gene ontology analysis of upregulated and downregulated proteins (FC > 1.5; p < 0.05, Wald test with Benjamini–Hochberg correction) in shDDX6 compared to shCTRL MOLM-13 cells. (d) Heatmap showing downregulation of P-body-related proteins in DDX6 KD MOLM-13 cells (n = 3 biologically independent samples per group). (e) Representative Western blots showing loss of P-body-related proteins upon DDX6 loss in DDX6-FKBP12F36V MOLM-13 cells. n = 3 independent experiments. (f, g) qRT–PCR validation of tumour suppressor gene overexpression in MOLM-13 cells. (n = 3). (h) Total number of ATAC-seq peaks detected in control and DDX6 KD (left) MOLM-13 or (right) HEL cells. (i) Genomic distribution of ATAC-seq peaks in control and DDX6 KD HEL and MOLM-13 cells. (j) Scatter-plot showing ATAC-seq data for shCTRL and shDDX6 HEL cells (n = 2 biologically independent samples per group). Blue dots indicate genomic regions showing significantly decreased chromatin accessibility in DDX6-depleted cells (FC > 1.5, p < 0.05, Wald test with Benjamini–Hochberg correction; n = 570); red dots indicate genomic regions showing significantly increased chromatin accessibility in DDX6-depleted cells (FC > 1.5, p < 0.05; n = 1044). (k) TF motif enrichment analysis on shDDX6 gained and lost ATAC-seq peaks in HEL cells. (l) Increased H3K4me1 levels at loci of genes that gained chromatin accessibility and became upregulated after DDX6 KD. Wilcoxon rank-sum test, shCTRL open (n = 2026), shDDX6 open (n = 2045), shCTRL closed (n = 1024), shDDX6 closed (n = 1035). Box centre line indicates median, box limits indicate upper (Q3) and lower quartiles (Q1), lower whisker is Q1 – 1.5 × IQR and upper whisker is Q3 + 1.5 × IQR. (m) Total number of promoter interactions detected by liCHi-C in control (n = 2) and DDX6 KD (n = 2) MOLM-13 cells. (n) Gene tracks of ATAC-seq, RNA-seq, H3K27ac CUT&Tag, and liCHi-C data for the genomic region surrounding AGO4. Blue shadow highlights the gene promoter. Arcs represent significant promoter interactions (CHiCAGO score > 5). (o) Representative intracellular flow cytometry plot for KDM5B in control and KDM5B knockout MOLM-13 cells.

Source data

Supplementary information

Supplementary Information

Supplementary Fig. 1.

Reporting Summary

Peer Review File

Supplementary Tables

Supplementary Table 1: all CRISPR screen data. Supplementary Table 2: top 308 CRISPR screen hits. Supplementary Table 3: AML patient mutations and cytogenetics. Supplementary Table 4: human AML cell line subtypes and mutations. Supplementary Table 5: shRNA and sgRNA oligonucleotide sequences.

Source data

Source Data Fig. 1

Unprocessed western blots and gels.

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Unprocessed western blots and gels.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Extended Data Table 1

Statistical source data.

Source Data Extended Data Fig. 2

Unprocessed western blots and gels.

Source Data Extended Data Table 2

Statistical source data.

Source Data Extended Data Fig. 3

Unprocessed western blots and gels.

Source Data Extended Data Table 3

Statistical source data.

Source Data Extended Data Fig. 4

Unprocessed western blots and gels.

Source Data Extended Data Table 4

Statistical source data.

Source Data Extended Data Table 5

Statistical source data.

Source Data Extended Data Table 6

Statistical source data.

Source Data Extended Data Fig. 8

Unprocessed western blots and gels.

Source Data Extended Data Table 8

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

Kodali, S., Proietti, L., Valcarcel, G. et al. RNA sequestration in P-bodies sustains myeloid leukaemia. Nat Cell Biol (2024). https://doi.org/10.1038/s41556-024-01489-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41556-024-01489-6

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer