+
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:

SNORD113–114 cluster maintains haematopoietic stem cell self-renewal via orchestrating the translation machinery

Abstract

Haematopoietic stem cells (HSCs) self-renew and differentiate to replenish the pool of blood cells, which require a low but finely tuned protein synthesis rate. Nonetheless, the translatome landscape in HSCs and how the translation machinery orchestrates HSC self-renewal remain largely elusive. Here we perform ultra-low-input Ribo-seq in HSCs, progenitor and lineage cells, and reveal HSC-specific translated genes involved in rRNA processing. We systematically profile small nucleolar RNAs (snoRNAs) and uncover an indispensable role of the SNORD113–114 cluster in regulating HSC self-renewal. Maternal knockout (Mat-KO) of this cluster substantially impairs HSC self-renewal, whereas loss of the paternal allele shows no obvious phenotype. Mechanistically, Mat-KO results in dysregulation of translation machinery (rRNA 2′-O-Me modifications, pre-rRNA processing, 60S ribosome assembly and translation) and induces nucleolar stress in HSCs, which exempts p53 from Mdm2-mediated proteasomal degradation and leads to apoptosis. Collectively, our study provides a promising facet to our understanding of snoRNA-mediated regulation in HSC homeostasis.

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: Ultra-low-input Ribo-seq identifies specific translatome profile in HSCs.
Fig. 2: The SNORD113–114 cluster is specifically expressed in LT-HSCs and critical for HSC self-renewal.
Fig. 3: SNORD113–114 Mat-KO impairs HSC reconstitution capacity.
Fig. 4: Transcriptomic profiling reveals dysregulation of translation pathway in SNORD113–114 Mat-KO HSCs.
Fig. 5: Loss of SNORD113–114 cluster disrupts translation machinery.
Fig. 6: SNORD113–114 Mat-KO results in dysregulation of HSC-specific translatome.
Fig. 7: SNORD113–114 Mat-KO induces nucleolar stress and apoptosis in HSCs.
Fig. 8: SNORD113–114 Mat-KO mediated HSC deficiency is rescued by ectopic expression of snoRNAs or l-leucine treatment.

Similar content being viewed by others

Data availability

All sequencing data have been deposited in the Gene Expression Omnibus under the following accession codes: GSE166514, GSE166515, GSE166516, GSE166517, GSE166518, GSE267887, GSE269874 and GSE270936. The public mouse genome assembly GRCm38 was used for alignment. Previously published data by Spevak et al.3 that were re-analysed here (Fig. 1e) are available under accession code GSE113886. Data supporting the findings of this study are available from the corresponding authors on reasonable request. Source data are provided with this paper.

References

  1. Orkin, S. H. & Zon, L. I. Hematopoiesis: an evolving paradigm for stem cell biology. Cell 132, 631–644 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Signer, R. A. J., Magee, J. A., Salic, A. & Morrison, S. J. Haematopoietic stem cells require a highly regulated protein synthesis rate. Nature 509, 49–54 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. 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.e7 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Kruta, M. et al. Hsf1 promotes hematopoietic stem cell fitness and proteostasis in response to ex vivo culture stress and aging. Cell Stem Cell 28, 1950–1965.e6 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Lv, K. et al. HectD1 controls hematopoietic stem cell regeneration by coordinating ribosome assembly and protein synthesis. Cell Stem Cell 28, 1275–1290.e9 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Keyvani Chahi, A. et al. PLAG1 dampens protein synthesis to promote human hematopoietic stem cell self-renewal. Blood 140, 992–1008 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Li, C. et al. Amino acid catabolism regulates hematopoietic stem cell proteostasis via a GCN2–eIF2α axis. Cell Stem Cell 29, 1119–1134.e7 (2022).

    Article  CAS  PubMed  Google Scholar 

  8. Cai, X. et al. Runx1 deficiency decreases ribosome biogenesis and confers stress resistance to hematopoietic stem and progenitor cells. Cell Stem Cell 17, 165–177 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Signer, R. A. J. et al. The rate of protein synthesis in hematopoietic stem cells is limited partly by 4E-BPs. Genes Dev. 30, 1698–1703 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. van Galen, P. et al. The unfolded protein response governs integrity of the haematopoietic stem-cell pool during stress. Nature 510, 268–272 (2014).

    Article  PubMed  Google Scholar 

  11. Xu, L. et al. Protein quality control through endoplasmic reticulum-associated degradation maintains haematopoietic stem cell identity and niche interactions. Nat. Cell Biol. 22, 1162–1169 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Hidalgo San Jose, L. et al. Modest declines in proteome quality impair hematopoietic stem cell self-renewal. Cell Rep. 30, 69–80.e6 (2020).

    Article  CAS  PubMed  Google Scholar 

  13. Kumar, S. et al. Repolarization of HSC attenuates HSCs failure in Shwachman-Diamond syndrome. Leukemia 35, 1751–1762 (2021).

    Article  CAS  PubMed  Google Scholar 

  14. Warren, A. J. Molecular basis of the human ribosomopathy Shwachman-Diamond syndrome. Adv. Biol. Regul. 67, 109–127 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Woloszynek, J. R. et al. Mutations of the SBDS gene are present in most patients with Shwachman-Diamond syndrome. Blood 104, 3588–3590 (2004).

    Article  CAS  PubMed  Google Scholar 

  16. Finch, A. J. et al. Uncoupling of GTP hydrolysis from eIF6 release on the ribosome causes Shwachman-Diamond syndrome. Genes Dev. 25, 917–929 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Wong, C. C., Traynor, D., Basse, N., Kay, R. R. & Warren, A. J. Defective ribosome assembly in Shwachman-Diamond syndrome. Blood 118, 4305–4312 (2011).

    Article  CAS  PubMed  Google Scholar 

  18. Qiu, Y., Xu, M. & Huang, S. Long noncoding RNAs: emerging regulators of normal and malignant hematopoiesis. Blood 138, 2327–2336 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Zhu, B. et al. Loss of miR-31-5p drives hematopoietic stem cell malignant transformation and restoration eliminates leukemia stem cells in mice. Sci. Transl. Med. 14, eabh2548 (2022).

    Article  CAS  PubMed  Google Scholar 

  20. Krivdova, G. et al. Identification of the global miR-130a targetome reveals a role for TBL1XR1 in hematopoietic stem cell self-renewal and t(8;21) AML. Cell Rep. 38, 110481 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Yang, S. et al. WDR82-binding long noncoding RNA lncEry controls mouse erythroid differentiation and maturation. J. Exp. Med. 219, e20211688 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Qian, P. et al. The Dlk1-Gtl2 locus preserves LT-HSC function by inhibiting the PI3K-mTOR pathway to restrict mitochondrial metabolism. Cell Stem Cell 18, 214–228 (2016).

    Article  CAS  PubMed  Google Scholar 

  23. Watkins, N. J. & Bohnsack, M. T. The box C/D and H/ACA snoRNPs: key players in the modification, processing and the dynamic folding of ribosomal RNA. Wiley Interdiscip. Rev. RNA 3, 397–414 (2012).

    Article  CAS  PubMed  Google Scholar 

  24. McMahon, M., Contreras, A. & Ruggero, D. Small RNAs with big implications: new insights into H/ACA snoRNA function and their role in human disease. Wiley Interdiscip. Rev. RNA 6, 173–189 (2015).

    Article  CAS  PubMed  Google Scholar 

  25. Piekna-Przybylska, D., Decatur, W. A. & Fournier, M. J. The 3D rRNA modification maps database: with interactive tools for ribosome analysis. Nucleic Acids Res. 36, D178–D183 (2008).

    Article  CAS  PubMed  Google Scholar 

  26. Meier, U. T. The many facets of H/ACA ribonucleoproteins. Chromosoma 114, 1–14 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Warner, W. A. et al. Expression profiling of snoRNAs in normal hematopoiesis and AML. Blood Adv. 2, 151–163 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Valleron, W. et al. Specific small nucleolar RNA expression profiles in acute leukemia. Leukemia 26, 2052–2060 (2012).

    Article  CAS  PubMed  Google Scholar 

  29. Cohen, Y. et al. The increased expression of 14q32 small nucleolar RNA transcripts in promyelocytic leukemia cells is not dependent on PML-RARA fusion gene. Blood Cancer J. 2, e92 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Teittinen, K. J. et al. Expression of small nucleolar RNAs in leukemic cells. Cell. Oncol. 36, 55–63 (2013).

    Article  CAS  Google Scholar 

  31. Ronchetti, D. et al. Small nucleolar RNAs as new biomarkers in chronic lymphocytic leukemia. BMC Med. Genet. 6, 27 (2013).

    Google Scholar 

  32. Sloan, K. E. et al. Tuning the ribosome: The influence of rRNA modification on eukaryotic ribosome biogenesis and function. RNA Biol. 14, 1138–1152 (2017).

    Article  PubMed  Google Scholar 

  33. Decatur, W. A. & Fournier, M. J. rRNA modifications and ribosome function. Trends Biochem. Sci. 27, 344–351 (2002).

    Article  CAS  PubMed  Google Scholar 

  34. Bellodi, C. et al. Loss of function of the tumor suppressor DKC1 perturbs p27 translation control and contributes to pituitary tumorigenesis. Cancer Res. 70, 6026–6035 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Bellodi, C., Kopmar, N. & Ruggero, D. Deregulation of oncogene-induced senescence and p53 translational control in X-linked dyskeratosis congenita. EMBO J. 29, 1865–1876 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Bellodi, C. et al. H/ACA small RNA dysfunctions in disease reveal key roles for noncoding RNA modifications in hematopoietic stem cell differentiation. Cell Rep. 3, 1493–1502 (2013).

    Article  CAS  PubMed  Google Scholar 

  37. Guzzi, N. et al. Pseudouridylation of tRNA-derived fragments steers translational control in stem cells. Cell 173, 1204–1216.e26 (2018).

    Article  CAS  PubMed  Google Scholar 

  38. Guzzi, N. et al. Pseudouridine-modified tRNA fragments repress aberrant protein synthesis and predict leukaemic progression in myelodysplastic syndrome. Nat. Cell Biol. 24, 299–306 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Grozdanov, P. N., Fernandez-Fuentes, N., Fiser, A. & Meier, U. T. Pathogenic NAP57 mutations decrease ribonucleoprotein assembly in dyskeratosis congenita. Hum. Mol. Genet. 18, 4546–4551 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Erales, J. et al. Evidence for rRNA 2′-O-methylation plasticity: control of intrinsic translational capabilities of human ribosomes. Proc. Natl Acad. Sci. USA 114, 12934–12939 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Zhou, F. et al. AML1-ETO requires enhanced C/D box snoRNA/RNP formation to induce self-renewal and leukaemia. Nat. Cell Biol. 19, 844–855 (2017).

    Article  CAS  PubMed  Google Scholar 

  42. Lawo, S. et al. HAUS, the 8-subunit human Augmin complex, regulates centrosome and spindle integrity. Curr. Biol. 19, 816–826 (2009).

    Article  CAS  PubMed  Google Scholar 

  43. Corrigan, P. M., Dobbin, E., Freeburn, R. W. & Wheadon, H. Patterns of Wnt/Fzd/LRP gene expression during embryonic hematopoiesis. Stem Cells Dev. 18, 759–772 (2009).

    Article  CAS  PubMed  Google Scholar 

  44. Qian, P. et al. Retinoid-sensitive epigenetic regulation of the Hoxb cluster maintains normal hematopoiesis and inhibits leukemogenesis. Cell Stem Cell 22, 740–754.e7 (2018).

    Article  CAS  PubMed  Google Scholar 

  45. Carpenter, K. A., Thurlow, K. E., Craig, S. E. L. & Grainger, S. Wnt regulation of hematopoietic stem cell development and disease. Curr. Top. Dev. Biol. 153, 255–279 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Singh, R. P. et al. HIF prolyl hydroxylase 2 (PHD2) is a critical regulator of hematopoietic stem cell maintenance during steady-state and stress. Blood 121, 5158–5166 (2013).

    Article  CAS  PubMed  Google Scholar 

  47. Theilgaard-Mönch, K. et al. Transcription factor-driven coordination of cell cycle exit and lineage-specification in vivo during granulocytic differentiation: in memoriam Professor Niels Borregaard. Nat. Commun. 13, 3595 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  48. Simsek, T. et al. The distinct metabolic profile of hematopoietic stem cells reflects their location in a hypoxic niche. Cell Stem Cell 7, 380–390 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Pinho, S. et al. VCAM1 confers innate immune tolerance on haematopoietic and leukaemic stem cells. Nat. Cell Biol. 24, 290–298 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Rho, S.-S. et al. Rap1b promotes notch-signal-mediated hematopoietic stem cell development by enhancing integrin-mediated cell adhesion. Dev. Cell 49, 681–696.e6 (2019).

    Article  CAS  PubMed  Google Scholar 

  51. Zhou, F. et al. Tracing haematopoietic stem cell formation at single-cell resolution. Nature 533, 487–492 (2016).

    Article  CAS  PubMed  Google Scholar 

  52. Duker, A. L. et al. Paternally inherited microdeletion at 15q11.2 confirms a significant role for the SNORD116 C/D box snoRNA cluster in Prader-Willi syndrome. Eur. J. Hum. Genet 18, 1196–1201 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Basbous, J. et al. Induction of ASAP (MAP9) contributes to p53 stabilization in response to DNA damage. Cell Cycle 11, 2380–2390 (2012).

    Article  CAS  PubMed  Google Scholar 

  54. Zhao, J. et al. Human hematopoietic stem cell vulnerability to ferroptosis. Cell 186, 732–747.e16 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Lowe, T. M. & Eddy, S. R. A computational screen for methylation guide snoRNAs in yeast. Science 283, 1168–1171 (1999).

    Article  CAS  PubMed  Google Scholar 

  56. Domostegui, A. et al. Impaired ribosome biogenesis checkpoint activation induces p53-dependent MCL-1 degradation and MYC-driven lymphoma death. Blood 137, 3351–3364 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Pelletier, J. et al. Nucleotide depletion reveals the impaired ribosome biogenesis checkpoint as a barrier against DNA damage. EMBO J. 39, e103838 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Boulon, S., Westman, B. J., Hutten, S., Boisvert, F.-M. & Lamond, A. I. The nucleolus under stress. Mol. Cell 40, 216–227 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Boisvert, F.-M., van Koningsbruggen, S., Navascués, J. & Lamond, A. I. The multifunctional nucleolus. Nat. Rev. Mol. Cell Biol. 8, 574–585 (2007).

    Article  CAS  PubMed  Google Scholar 

  60. Brooks, C. L. & Gu, W. p53 ubiquitination: Mdm2 and beyond. Mol. Cell 21, 307–315 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Horn, H. F. & Vousden, K. H. Cooperation between the ribosomal proteins L5 and L11 in the p53 pathway. Oncogene 27, 5774–5784 (2008).

    Article  CAS  PubMed  Google Scholar 

  62. Lohrum, M. A. E., Ludwig, R. L., Kubbutat, M. H. G., Hanlon, M. & Vousden, K. H. Regulation of HDM2 activity by the ribosomal protein L11. Cancer Cell 3, 577–587 (2003).

    Article  CAS  PubMed  Google Scholar 

  63. Xu, B., Gogol, M., Gaudenz, K. & Gerton, J. L. Improved transcription and translation with L-leucine stimulation of mTORC1 in Roberts syndrome. BMC Genomics 17, 25 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  64. Ma, H. et al. N6-Methyladenosine methyltransferase ZCCHC4 mediates ribosomal RNA methylation. Nat. Chem. Biol. 15, 88–94 (2019).

    Article  CAS  PubMed  Google Scholar 

  65. van Tran, N. et al. The human 18S rRNA m6A methyltransferase METTL5 is stabilized by TRMT112. Nucleic Acids Res. 47, 7719–7733 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Sergeeva, O. et al. Modification of adenosine196 by Mettl3 methyltransferase in the 5′-external transcribed spacer of 47S pre-rRNA affects rRNA maturation. Cells 9, 1061 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Bowie, M. B. et al. Hematopoietic stem cells proliferate until after birth and show a reversible phase-specific engraftment defect. J. Clin. Invest. 116, 2808–2816 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Habibian, H. K. et al. The fluctuating phenotype of the lymphohematopoietic stem cell with cell cycle transit. J. Exp. Med. 188, 393–398 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Hammond, C. A., Wu, S. W., Wang, F., MacAldaz, M. E. & Eaves, C. J. Aging alters the cell cycle control and mitogenic signaling responses of human hematopoietic stem cells. Blood 141, 1990–2002 (2023).

    Article  CAS  PubMed  Google Scholar 

  70. VanInsberghe, M., van den Berg, J., Andersson-Rolf, A., Clevers, H. & van Oudenaarden, A. Single-cell Ribo-seq reveals cell cycle-dependent translational pausing. Nature 597, 561–565 (2021).

    Article  CAS  PubMed  Google Scholar 

  71. Tang, F. et al. mRNA-seq whole-transcriptome analysis of a single cell. Nat. Methods 6, 377–382 (2009).

    Article  CAS  PubMed  Google Scholar 

  72. Marchand, V., Blanloeil-Oillo, F., Helm, M. & Motorin, Y. Illumina-based RiboMethSeq approach for mapping of 2′-O-Me residues in RNA. Nucleic Acids Res. 44, e135 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  73. Birkedal, U. et al. Profiling of ribose methylations in RNA by high-throughput sequencing. Angew. Chem. Int. Ed. Engl. 54, 451–455 (2015).

    Article  CAS  PubMed  Google Scholar 

  74. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  75. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. 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 

  77. Lê, S., Josse, J. & Husson, F. FactoMineR: an R Package for multivariate analysis. J. Stat. Softw. 25, 1–18 (2008).

    Article  Google Scholar 

  78. Kim, D., Langmead, B. & Salzberg, S. L. HISAT: a fast spliced aligner with low memory requirements. Nat. Methods 12, 357–360 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Anders, S., Pyl, P. T. & Huber, W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  CAS  PubMed  Google Scholar 

  80. Huang, D. W., Sherman, B. T. & Lempicki, R. A. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 37, 1–13 (2009).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  83. Patro, R., Duggal, G., Love, M. I., Irizarry, R. A. & Kingsford, C. Salmon provides fast and bias-aware quantification of transcript expression. Nat. Methods 14, 417–419 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Soneson, C., Love, M. I. & Robinson, M. D. Differential analyses for RNA-seq: transcript-level estimates improve gene-level inferences. F1000Research 4, 1521 (2015).

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Chen, J., Bardes, E. E., Aronow, B. J. & Jegga, A. G. ToppGene suite for gene list enrichment analysis and candidate gene prioritization. Nucleic Acids Res. 37, W305–W311 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Penzo, M., Carnicelli, D., Montanaro, L. & Brigotti, M. A reconstituted cell-free assay for the evaluation of the intrinsic activity of purified human ribosomes. Nat. Protoc. 11, 1309–1325 (2016).

    Article  CAS  PubMed  Google Scholar 

  89. Panthu, B., Décimo, D., Balvay, L. & Ohlmann, T. In vitro translation in a hybrid cell free lysate with exogenous cellular ribosomes. Biochem. J. 467, 387–398 (2015).

    Article  CAS  PubMed  Google Scholar 

  90. Schmidt, E. K., Clavarino, G., Ceppi, M. & Pierre, P. SUnSET, a nonradioactive method to monitor protein synthesis. Nat. Methods 6, 275–277 (2009).

    Article  CAS  PubMed  Google Scholar 

  91. Ge, J. et al. Dyskerin ablation in mouse liver inhibits rRNA processing and cell division. Mol. Cell. Biol. 30, 413–422 (2010).

    Article  CAS  PubMed  Google Scholar 

  92. Wang, H. et al. SARS-CoV-2 N protein potentiates host NPM1-snoRNA translation machinery to enhance viral replication. Signal Transduct. Target Ther. 7, 356 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by grants from the National Key Research and Development Program of China (2022YFA1103500 and 2024YFA1107102 to P.Q., 2021YFA1102400 to F.W. and 2022YFA1106300 and 2024YFA1803500 to J.X.), the National Natural Science Foundation of China (82222003 and 92268117 to P.Q., 92268205 and 82330007 to J.Y., 82470123 to F.W. and 31900815 to H.W.), the Key R&D Program of Zhejiang (2024SSYS0024, 2024SSYS0023 and 2024SSYS0025 to P.Q.), the Zhejiang Provincial Natural Science Foundation of China (Z24H080001 to P.Q.), the Department of Science and Technology of Zhejiang Province (2023R01012 to P.Q.), the Fundamental Research Funds for the Central Universities (226-2024-00007 to P.Q.), Chinese Academy of Medical Sciences Innovation Fund for Medical Sciences (2021-I2M-1-040 to F.W.) and Zhejiang Province Postdoctoral Research Excellence Funding Project (ZJ2022098 to H.W.). Haihe Laboratory of Cell Ecosystem Innovation Fund (22HHXBSS00027 to J.Y.). P.Q. gratefully acknowledges the support of the K.C. Wong Education Foundation. Technical support was provided by the core facilities at the Zhejiang University School of Medicine (special thanks to C. Guo and X. Hong for assistance during experiments), as well as the core facilities at the Zhejiang University Medical Center and Liangzhu Laboratory. We thank State Key Laboratory of Common Mechanism Research of Major Diseases Platform for consultation and instrument availability that supported this work, and High-performance Computing Platform at the Center for Bioinformatics, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences.

Author information

Authors and Affiliations

Contributions

H.W. and P.Q. conceived the project and designed the experiments. H.W. performed the experiments, analysed data and wrote the manuscript. C.H. and J. Li helped perform library construction work of Ribo-seq experiments. P.J., J.X. and S.C. analysed second-generation sequencing data. Z.Z., Y.H. and D.H. helped perform transplantation experiments and undertook mouse breeding work. Z.Z. and J.Z. performed the SMART-Seq2 experiments. Y.L. and J.S. helped perform the northern blot experiments. J.C. and J. Liu helped perform the UHPLC–MS experiments. M.D. helped make the SNORD113–114 cluster knockout mice. P.Q., B.L., J.Y. and F.W. supervised the overall project and co-wrote the manuscript. All authors contributed to reading and editing the manuscript.

Corresponding authors

Correspondence to Bing Liu, Jia Yu, Fang Wang or Pengxu Qian.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cell Biology thanks the anonymous reviewers for their contribution to the peer review of this work.

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 Gating strategies used in flow cytometry.

a, Flow cytometry plots showing gating strategies used for detecting HSPCs, progenitors, mature cells. b, Flow cytometry plots showing gating strategies used for detecting human HSC. c, Flow cytometry plots showing gating strategies used for detecting donor repopulation rate.

Extended Data Fig. 2 Translatome profiling of haematopoietic cells.

a, Barplot showing representative reads distributions along the transcripts. Read position relative to different reading frames (0, 1, 2) are colour-coded. b, Barplot showing representative ratios of reads belonging to different reading frames. c, Reads length distribution frequencies of all cell types. d, Scatterplots showing Pearson’s correlation between two batches of data from Ribo-seq. e, Scatterplots showing Pearson’s correlation between RNA level and RPF level of genes in each cell type.

Source data

Extended Data Fig. 3 HSC-specific SNORD113–114 cluster is indispensable for HSC maintenance.

a, Heatmap of snoRNA expression (Z-score normalized) in progenitors and lineage mature cells. b, c, Raw read counts of different types of ncRNAs got from sncRNA-seq. d, Sequence alignments of snoRNAs belonging to SNORD113–114 cluster between Mus musculus and Homo sapiens. Only identical sequences of specific snoRNAs between human and mouse are presented. e, Electrophoresis showing PCR products patterns got from genotyping of WT and Mat-KO mice. f, Absolute numbers of total BM cells of WT and Mat-KO mice were studied by flow cytometry (n = 4 for WT, n = 3 for Mat-KO biological replicates). Error bars, SEM. g-j, Frequencies and absolute numbers of SLAM HSCs (g, h) and progenitors (i, j) from WT and Mat-KO mice detected by flow cytometry (n = 4 for WT, n = 3 for Mat = -KO biological replicates). Error bars, SEM. k, l, Frequency (l), absolute number (m) of lineage cells collected from WT and Mat-KO mice (n = 4 for WT, n = 3 for Mat-KO biological replicates). Error bars, SEM. All P values were determined by two-tailed unpaired Student’s t-test.

Source data

Extended Data Fig. 4 SNORD113–114 Mat-KO does not affect the spleen haematopoiesis.

a, b, Spleen weight (a) and cellularity (b) were examined and compared between WT and Mat-KO mice. N = 3 for WT, n = 4 for Mat-KO biological replicates in (a). N = 4 for WT, n = 3 for Mat-KO in (b). Error bars, SEM. c-h, Frequency and absolute number of spleen HSC (c, d), progenitors (e, f) and lineage cell (g, h) were analysed and compared between WT and Mat-KO mice (n = 4 for WT, n = 3 for Mat-KO biological replicates). Error bars, SEM. i, Whole blood counts of WT and Mat-KO mice (n = 7 biological replicates). Error bars, SEM. All P values were determined by two-tailed unpaired Student’s t-test.

Source data

Extended Data Fig. 5 SNORD113–114 Pat-KO does not lead to HSC deficiency.

a, BM cellularity of WT and Pat-KO mice (n = 3 biological replicates). Error bars, SEM. b, c, Frequency (b) and absolute number (c) of HSCs were analysed and compared between WT and Pat-KO mice (n = 3 biological replicates). Error bars, SEM. d, e, Apoptosis rate (d) and cell cycle distribution (e) were analysed and compared between WT and Pat-KO mice (n = 3 biological replicates). Different cell cycle phases are colour-coded and presented as a percentage of whole population (n = 6 biological replicates). Error bars, SEM. f, Schematic presentation of workflow of transplantation using sorted HSCs. g, h, Repopulation rate (g) and lineage distribution (h) of WT and Pat-KO mice HSC (n = 3 biological replicates). Error bars, SEM; All P values were determined by two-tailed unpaired Student’s t-test.

Source data

Extended Data Fig. 6 SnoRNAs in SNORD113–114 cluster are specifically important for HSC.

a-f, SNORD116 was not functionally important for HSCs. SNORD116 were knocked out using specific sgRNAs. After 7-day’s culture in 96-well plates, expressions of snoRNAs were examined by RT–qPCR (a), HSPC ratios and absolute numbers were examined by flow cytometry (b-d), and colony formation ability was examined (e, f). n = 3 biological replicates. Scale bar, 200 μm. Error bars, SEM. g, Expressions of specific snoRNAs were examined by RT–qPCR and compared between HSCs from male and female mice. n = 3 biological replicates. Error bars, SEM. h-k, SnoRNAs in SNORD113–114 cluster were also functionally important for human HSCs. CD34+ HSCs were collected from healthy donors, and 3 snoRNAs (SNORD113–6, SNORD113-9, SNORD114-1) were knocked out using specific sgRNAs. After 7-day’s culture in 96-well plates, expressions of snoRNAs were examined by RT–qPCR (h), HSPC ratios were examined by flow cytometry (i), and colony formation ability was examined (j, k). n = 3 biological replicates. Scale bar, 200 μm. Error bars, SEM. Cartoon illustrations were created by Figdraw. All P values were determined by two-tailed unpaired Student’s t-test.

Extended Data Fig. 7 High-throughput sequencing analysis of HSCs of WT/Mat-KO mice.

a, Raw read counts of different types of ncRNAs got from sncRNA-seq of WT and Mat-KO mice. b, Western blot result showing expressions of Dlk1 in WT and Mat-KO HSCs (n = 3 biological replicates). c, PCA analysis of sncRNA-seq of WT/Pat-KO mice LSK cells. d, Volcano-plot showing differentially expressed snoRNAs between WT/Pat-KO LSK cells. e, Raw read counts of different types of ncRNAs got from sncRNA-seq of WT and Pat-KO mice. f, Heatmap showing expression of snoRNAs belonging to SNORD113–114 cluster in WT/Pat-KO LSK cells. Only detected snoRNAs are shown. g, PCA analysis of RNA-seq results of WT/Pat-KO HSCs. h, Volcano-plot showing differentially expressed genes between WT/Pat-KO HSCs. i, Visualization of colour-coded clustering of HSCs via t-SNE based on experimental groups and gene signature, showing distribution of HSCs of WT and Mat-KO mice. j, Distribution of WT/Mat-KO HSCs among different cell populations. k, Violin-plot of different marker genes, showing expression patterns in all clusters. Violin box is colour-coded as for population clusters shown in Fig. 4m. l, GO term analysis of signature genes of different populations.

Source data

Extended Data Fig. 8 Loss of SNORD113–114 cluster dysregulates rRNA modification in HSCs.

a, Read length distributions of samples from RiboMethSeq. b, PCA analysis of RNA fragments got from RiboMethSeq of WT/Mat-KO LSK cells. c, Conservation of all differentially methylated sites detected by RiboMethSeq across E. coli, S. cerevisiae, M. musculus and H. sapiens. Different species are colour-coded, and sites that are not conserved were made vacant in the coloured circle representing according species. d, 2′-O-Me modification levels were examined by RTL-P. Upper panel shows schematic of primers used for reverse transcription and qPCR. Lower panel shows agarose electrophoresis images of semi-quantitative PCR results (n = 3 biological replicates). e, Targets of snoRNAs in SNORD113–114 cluster were predicted using online tool Snoscan. Predicted targets are listed under each snoRNA, and the nucleotide predicted to be 2′-O-methylated are labelled red. f, Relative methylation ratio (from data of RiboMethSeq) of specific sites that were predicted to be targeted by snoRNAs in SNORD113–114 cluster (n = 3 biological replicates). Error bars, SEM. All P values were determined by two-tailed unpaired Student’s t-test.

Source data

Extended Data Fig. 9 Loss of SNORD113–114 cluster impairs rRNA processing and translation in HSCs.

a, Schematic representation of primer designing for detecting various pre-rRNA isoforms generated during pre-rRNA processing. Red bars show PCR products of according primers. b–e, Relative expression levels of various pre-rRNA isoforms detected by RT–qPCR (n = 3 biological replicates). Error bars, SEM. f, PCA analysis of RPFs of WT and Mat-KO HSCs from Ribo-seq. g, Reads length distribution frequencies of RPFs from Ribo-seq. h-j, Expressions of representative genes in different sucrose gradients from polysome profiling assay were examined by RT–qPCR (n = 3 biological replicates). Error bars, SEM. k, Expressions of snoRNAs were examined by RT–qPCR after WT/Mat-KO HSCs were infected by according viruses (n = 3 biological replicates). Error bars, SEM. l, m, BM cells were infected by lentivirus carrying vectors expressing snoRNAs or according control empty vectors, and were analysed using flow cytometry after 7-day culture. Frequencies (l) and absolute numbers (m) were analysed in different groups. n = 3 biological replicates. Error bars, SEM. All P values were determined by two-tailed unpaired Student’s t-test.

Source data

Extended Data Fig. 10 SnoRNAs in SNORD113–114 cluster function through guiding 2′-O-Methylation of specific rRNA sites.

a-d, 2′-O-Me modifications of specific sites targeted by the ectopically expressed snoRNAs were also examined by RTL-P. e-g, Reconstitution ability analysis of HSCs from WT and Mat-KO mice after L-leucine treatment. BM cells of WT/Mat-KO mice were treated with L-leucine at dose of 5 μM for 48 h, and then were transplanted (2 e5/sample) into lethally irradiated recipient mice together with CD45.1 competitor cells (2 e5/sample). Repopulation rate (e), frequency of TNC (f) and absolute cell number (g) were analysed and compared between WT and Mat-KO mice at 16 weeks post-transplantation using flow cytometry (n = 7 biological replicates). Error bars, SEM. h-j, Statistical analysis of apoptosis ratio, cell cycle distributions and translation rate (OP-Puro assay) of HSCs after transplantation. N = 7 biological replicates. Error bars, SEM. k-p, 2′-O-Me modifications of specific sites were examined by RTL-P after L-leucine treatment (5 μM for 48 h). q, p53 expressions in WT/Mat-KO HSCs after L-leucine treatment (5 μM for 48 h). r, Expressions of HSC self-renewal related genes in WT/Mat-KO HSCs after L-leucine treatment (5 μM for 48 h). n = 3 biological replicates. Error bars, SEM. All P values were determined by two-tailed unpaired Student’s t-test.

Source data

Supplementary information

Reporting Summary

Supplementary Tables

Supplementary Table 1. RPFs of genes of all haematopoietic cell types by Ribo-seq. Supplementary Table 2. CPM of sncRNAs acquired from sncRNA-seq. Supplementary Table 3. FPKMs of all transcripts and DEGs in LSK cells of WT and Mat-KO mice by RNA-seq. Supplementary Table 4. GO analysis using biological process between Mat-KO/WT group. Supplementary Table 5. DEGs between WT and Mat-KO by single-cell RNA-seq. Supplementary Table 6. DMSs between WT and Mat-KO by RiboMethSeq. Supplementary Table 7. RPFs of genes of Mat-KO/WT HSC by Ribo-seq. Supplementary Table 8. Information of reagents and materials used in this study.

Source data

Source Data Figs. 2–5, 7 and 8, and Extended Data Figs. 2–5 and 7–9

Combined file for statistical source data for all figures.

Source Data Figs. 5g,k, 7d–g and 8k, and Extended Data Figs. 3e, 7b, 8d and 10a,c,k,m,q

Combined file for unprocessed western blots and gels for all figures.

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

Wang, H., Zhang, Z., Han, C. et al. SNORD113–114 cluster maintains haematopoietic stem cell self-renewal via orchestrating the translation machinery. Nat Cell Biol 27, 246–261 (2025). https://doi.org/10.1038/s41556-024-01593-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Version of record:

  • Issue date:

  • DOI: https://doi.org/10.1038/s41556-024-01593-7

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
点击 这是indexloc提供的php浏览器服务,不要输入任何密码和下载