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:

Identification of RIOK2 as a master regulator of human blood cell development

Abstract

Anemia is a major comorbidity in aging, chronic kidney and inflammatory diseases, and hematologic malignancies. However, the transcriptomic networks governing hematopoietic differentiation in blood cell development remain incompletely defined. Here we report that the atypical kinase RIOK2 (right open reading frame kinase 2) is a master transcription factor (TF) that not only drives erythroid differentiation, but also simultaneously suppresses megakaryopoiesis and myelopoiesis in primary human stem and progenitor cells. Our study reveals the previously uncharacterized winged helix-turn-helix DNA-binding domain and two transactivation domains of RIOK2 that are critical to regulate key hematopoietic TFs GATA1, GATA2, SPI1, RUNX3 and KLF1. This establishes RIOK2 as an integral component of the transcriptional regulatory network governing human hematopoietic differentiation. Importantly, RIOK2 mRNA expression significantly correlates with these TFs and other hematopoietic genes in myelodysplastic syndromes, acute myeloid leukemia and chronic kidney disease. Further investigation of RIOK2-mediated transcriptional pathways should yield therapeutic approaches to correct defective hematopoiesis in hematologic disorders.

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: RIOK2 drives erythropoiesis and suppresses megakaryopoiesis and myelopoiesis.
Fig. 2: RIOK2 and GATA1 function in a positive feedback loop to regulate hematopoietic differentiation.
Fig. 3: RIOK2 regulates the expression of key transcription factors in hematopoiesis.
Fig. 4: RIOK2 binds to the promoter regions of its targets and regulates chromatin accessibility.
Fig. 5: RIOK2 binds to a specific de novo nucleotide motif at the promoter regions of its targets to regulate transcription.
Fig. 6: The wHTH domain of RIOK2 harbors a DNA-binding domain that is critical in hematopoiesis.
Fig. 7: The two transactivation domains of RIOK2 facilitate binding with a core transcriptional complex required for hematopoiesis.
Fig. 8: mRNA expression of RIOK2 correlates with its targets and other hematopoietic genes in individuals with hematologic disorders.

Similar content being viewed by others

Data availability

Raw data generated from proteomic analysis are deposited in MassIVE under accession code MSV000088238. Data generated from RNA-seq are deposited in the Gene Expression Omnibus (GEO) database under accession code GSE185922. ATAC–seq and ChIP–seq datasets are deposited in the GEO database under accession numbers GSE174714 and GSE174719, respectively. The mRNA correlations in individuals with MDS, AML and CKD have been drawn using publicly available GEO datasets under accession codes GSE19429, GSE131184 and GSE37171. This study did not generate new unique reagents. Requests for any materials (plasmids generated in this study, crRNAs) and reagents should be directed to and will be fulfilled by L.H.G. with appropriate material transfer agreements. Source data are provided with this paper.

References

  1. Palapar, L. et al. Anaemia and physical and mental health in the very old: an individual participant data meta-analysis of four longitudinal studies of ageing. Age Ageing 50, 113–119 (2021).

    Article  PubMed  Google Scholar 

  2. Lopes, M. B. et al. A real-world longitudinal study of anemia management in non-dialysis-dependent chronic kidney disease patients: a multinational analysis of CKDopps. Sci. Rep. 11, 1784 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Becktell, K. et al. Aplastic Anemia & MDS International Foundation: Bone Marrow Failure Disease Scientific Symposium 2018. Leuk. Res. 80, 19–25 (2019).

    Article  PubMed  Google Scholar 

  4. Saygin, C. & Carraway, H. E. Current and emerging strategies for management of myelodysplastic syndromes. Blood Rev. 48, 100791 (2020).

  5. Hong, S. et al. Survival following relapse after allogeneic hematopoietic cell transplantation for acute leukemia and myelodysplastic syndromes in the contemporary era. Hematol. Oncol. Stem Cell Ther. S1658-3876, 30178–3 (2020).

    Google Scholar 

  6. Garcia-Manero, G., Chien, K. S. & Montalban-Bravo, G. Myelodysplastic syndromes: 2021 update on diagnosis, risk stratification and management. Am. J. Hematol. 95, 1399–1420 (2020).

    Article  PubMed  Google Scholar 

  7. Feld, J., Navada, S. C. & Silverman, L. R. Myelo-deception: luspatercept & TGF-beta ligand traps in myeloid diseases & anemia. Leuk. Res. 97, 106430 (2020).

    Article  CAS  PubMed  Google Scholar 

  8. Bhatt, V. R. & Steensma, D. P. Hematopoietic cell transplantation for myelodysplastic syndromes. J. Oncol. Pract. 12, 786–792 (2016).

    Article  PubMed  Google Scholar 

  9. Ferreira-Cerca, S. et al. ATPase-dependent role of the atypical kinase Rio2 on the evolving pre-40S ribosomal subunit. Nat. Struct. Mol. Biol. 19, 1316–1323 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Zemp, I. et al. Distinct cytoplasmic maturation steps of 40S ribosomal subunit precursors require hRio2. J. Cell Biol. 185, 1167–1180 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Ameismeier, M., Cheng, J., Berninghausen, O. & Beckmann, R. Visualizing late states of human 40S ribosomal subunit maturation. Nature 558, 249–253 (2018).

    Article  CAS  PubMed  Google Scholar 

  12. Raundhal, M. et al. Blockade of IL-22 signaling reverses erythroid dysfunction in stress-induced anemias. Nat. Immunol. 22, 520–529 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Pellagatti, A. & Boultwood, J. The molecular pathogenesis of the myelodysplastic syndromes. Eur. J. Haematol. 95, 3–15 (2015).

    Article  CAS  PubMed  Google Scholar 

  14. Pellagatti, A. et al. Deregulated gene expression pathways in myelodysplastic syndrome hematopoietic stem cells. Leukemia 24, 756–764 (2010).

    Article  CAS  PubMed  Google Scholar 

  15. Brennan, R. G. The winged-helix DNA-binding motif: another helix-turn-helix takeoff. Cell 74, 773–776 (1993).

    Article  CAS  PubMed  Google Scholar 

  16. LaRonde-LeBlanc, N. & Wlodawer, A. A family portrait of the RIO kinases. J. Biol. Chem. 280, 37297–37300 (2005).

    Article  CAS  PubMed  Google Scholar 

  17. Teichmann, M., Dumay-Odelot, H. & Fribourg, S. Structural and functional aspects of winged-helix domains at the core of transcription initiation complexes. Transcription 3, 2–7 (2012).

    Article  PubMed  Google Scholar 

  18. Nakahata, T. & Okumura, N. Cell surface antigen expression in human erythroid progenitors: erythroid and megakaryocytic markers. Leuk. Lymphoma 13, 401–409 (1994).

    Article  CAS  PubMed  Google Scholar 

  19. Tothova, Z. et al. Multiplex CRISPR–Cas9-based genome editing in human hematopoietic stem cells models clonal hematopoiesis and myeloid neoplasia. Cell Stem Cell 21, 547–555 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Khajuria, R. K. et al. Ribosome levels selectively regulate translation and lineage commitment in human hematopoiesis. Cell 173, 90–103 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Gutierrez, L., Caballero, N., Fernandez-Calleja, L., Karkoulia, E. & Strouboulis, J. Regulation of GATA1 levels in erythropoiesis. IUBMB Life 72, 89–105 (2020).

    Article  CAS  PubMed  Google Scholar 

  22. Gutierrez, L. et al. Ablation of Gata1 in adult mice results in aplastic crisis, revealing its essential role in steady-state and stress erythropoiesis. Blood 111, 4375–4385 (2008).

    Article  CAS  PubMed  Google Scholar 

  23. Fujiwara, T. GATA transcription factors: basic principles and related human disorders. Tohoku J. Exp. Med. 242, 83–91 (2017).

    Article  CAS  PubMed  Google Scholar 

  24. Ling, T. & Crispino, J. D. GATA1 mutations in red cell disorders. IUBMB Life 72, 106–118 (2020).

    Article  CAS  PubMed  Google Scholar 

  25. Gnanapragasam, M. N. & Bieker, J. J. Orchestration of late events in erythropoiesis by KLF1/EKLF. Curr. Opin. Hematol. 24, 183–190 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Zhang, D. E. et al. Function of PU.1 (Spi-1), C/EBP and AML1 in early myelopoiesis: regulation of multiple myeloid CSF receptor promoters. Curr. Top. Microbiol. Immunol. 211, 137–147 (1996).

    CAS  PubMed  Google Scholar 

  27. Yokomizo-Nakano, T. et al. Overexpression of RUNX3 represses RUNX1 to drive transformation of myelodysplastic syndrome. Cancer Res. 80, 2523–2536 (2020).

    Article  CAS  PubMed  Google Scholar 

  28. Daw, S. & Law, S. The functional interplay of transcription factors and cell adhesion molecules in experimental myelodysplasia including hematopoietic stem progenitor compartment. Mol. Cell. Biochem. 476, 535–551 (2020).

    Article  PubMed  Google Scholar 

  29. Nerlov, C. & Graf, T. PU.1 induces myeloid lineage commitment in multipotent hematopoietic progenitors. Genes Dev. 12, 2403–2412 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Rekhtman, N. et al. PU.1 and pRB interact and cooperate to repress GATA-1 and block erythroid differentiation. Mol. Cell. Biol. 23, 7460–7474 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Shimizu, R. & Yamamoto, M. Quantitative and qualitative impairments in GATA2 and myeloid neoplasms. IUBMB Life 72, 142–150 (2020).

    Article  CAS  PubMed  Google Scholar 

  32. Huang, Z. et al. GATA-2 reinforces megakaryocyte development in the absence of GATA-1. Mol. Cell. Biol. 29, 5168–5180 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Aravind, L., Anantharaman, V., Balaji, S., Babu, M. M. & Iyer, L. M. The many faces of the helix-turn-helix domain: transcription regulation and beyond. FEMS Microbiol. Rev. 29, 231–262 (2005).

    Article  CAS  PubMed  Google Scholar 

  34. Zhong, X. et al. HoxA9 transforms murine myeloid cells by a feedback loop driving expression of key oncogenes and cell cycle control genes. Blood Adv. 2, 3137–3148 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Xu, J. et al. Transcriptional silencing of γ-globin by BCL11A involves long-range interactions and cooperation with SOX6. Genes Dev. 24, 783–798 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Chen, T. W. et al. ChIPseek, a web-based analysis tool for ChIP data. BMC Genomics 15, 539 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Harish, B., Swapna, G. V., Kornhaber, G. J., Montelione, G. T. & Carey, J. Multiple helical conformations of the helix-turn-helix region revealed by NOE-restrained MD simulations of tryptophan aporepressor, TrpR. Proteins 85, 731–740 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Bi, X. et al. RNA targets ribogenesis factor WDR43 to chromatin for transcription and pluripotency control. Mol. Cell 75, 102–116 (2019).

    Article  CAS  Google Scholar 

  39. Chaparro, C. M. & Suchdev, P. S. Anemia epidemiology, pathophysiology, and etiology in low- and middle-income countries. Ann. N. Y. Acad. Sci. 1450, 15–31 (2019).

    PubMed  PubMed Central  Google Scholar 

  40. Feld, J., Belasen, A. & Navada, S. C. Myelodysplastic syndromes: a review of therapeutic progress over the past 10 years. Expert Rev. Anticancer Ther. 20, 465–482 (2020).

    Article  CAS  PubMed  Google Scholar 

  41. Groopman, J. E. & Itri, L. M. Chemotherapy-induced anemia in adults: incidence and treatment. J. Natl Cancer Inst. 91, 1616–1634 (1999).

    Article  CAS  PubMed  Google Scholar 

  42. Edginton-White, B. & Bonifer, C. The transcriptional regulation of normal and malignant blood cell development. FEBS J https://doi.org/10.1111/febs.15735 (2021).

  43. Suzuki, M. et al. GATA factor switching from GATA2 to GATA1 contributes to erythroid differentiation. Genes Cells 18, 921–933 (2013).

    Article  CAS  PubMed  Google Scholar 

  44. Moriguchi, T. & Yamamoto, M. A regulatory network governing Gata1 and Gata2 gene transcription orchestrates erythroid lineage differentiation. Int. J. Hematol. 100, 417–424 (2014).

    Article  CAS  PubMed  Google Scholar 

  45. Szabo, S. J. et al. A novel transcription factor, T-bet, directs Th1 lineage commitment. Cell 100, 655–669 (2000).

    Article  CAS  PubMed  Google Scholar 

  46. Li, J. et al. Isolation and transcriptome analyses of human erythroid progenitors: BFU-E and CFU-E. Blood 124, 3636–3645 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Kaufman, D. S., Hanson, E. T., Lewis, R. L., Auerbach, R. & Thomson, J. A. Hematopoietic colony-forming cells derived from human embryonic stem cells. Proc. Natl Acad. Sci. USA 98, 10716–10721 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Myers, S. A. et al. Streamlined protocol for deep proteomic profiling of FAC-sorted cells and its application to freshly isolated murine immune cells. Mol. Cell Proteomics 18, 995–1009 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Wang, J., Varin, T., Vieth, M. & Elkins, J. M. Crystal structure of human RIOK2 bound to a specific inhibitor. Open Biol. 9, 190037 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Chen, C. S., White, A., Love, J., Murphy, J. R. & Ringe, D. Methyl groups of thymine bases are important for nucleic acid recognition by DtxR. Biochemistry 39, 10397–10407 (2000).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank V. Sankaran and R. Voit (Boston Children’s hospital) for their valuable input in primary human HSPC culture techniques and for providing HMD-empty vector and HMD-GATA1 expression plasmids; S. Badrinath, N. Mathewson and K. Wucherpfennig for their kind assistance in CRISPR–Cas9 genome editing and for providing EF1α-MCS-IRES-ZsGreen lentiviral expression vector; and M. Katsuyama (Kyoto Prefectural University of Medicine) for generously providing GATA2 and GATA3 overexpression plasmids. We also thank A. Wight for assistance in analyzing RNA-seq results. This work was funed by a Discovery Research Grant, the Edward P. Evans Foundation (to L.H.G.), the National Institutes of Health (NIH) small grant program (1R03HL156574-01 to L.H.G.), institutional funding, the Dana-Farber Cancer Institute (to L.H.G.), The Pussycat Foundation Helen Gurley Brown Presidential Initiative (to S.G.), National Cancer Institute (NCI) Clinical Proteomic Tumor Analysis Consortium grants (NIH/NCI U24-CA210986 and U01 CA214125 to S.A.C.) and NIH grants (R01HL146642 and R37CA228304 to X.C.).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, S.G. and L.H.G.; design, S.G., G.A.P. and L.H.G.; methodology, S.G., S.A.M., S.A.C., X.C. and G.A.P.; data analysis, S.G., M.R., G.A.P. and L.H.G.; funding acquisition, S.G., M.R. and L.H.G.; writing–original draft, S.G., G.A.P. and L.H.G.; supervision, G.A.P. and L.H.G.

Corresponding authors

Correspondence to Gregory A. Petsko or Laurie H. Glimcher.

Ethics declarations

Competing interests

L.H.G. is a former Director of Bristol-Myers Squibb and the Waters Corporation and currently serves on the Board of Directors of GlaxoSmithKline Pharmaceuticals and Analog Devices and on the scientific advisory boards of Repare Therapeutics, Abpro Therapeutics and Kaleido Therapeutics. G.A.P. is on the Scientific Advisory Boards of Amicus Therapeutics, MeiraGTx, Annovis Bio, Retromer Therapeutics and Proclara Biosciences and holds equity or stock options in Denali Therapeutics, MeiraGTx, Annovis Bio, Retromer Therapeutics and Proclara Biosciences, companies that are developing therapies for neurodegenerative diseases. M.R. is currently an employee at Jnana Therapeutics. The other authors declare no competing interests.

Additional information

Peer review information Nature Immunology thanks Kazuhiko Igarashi and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. L. A. Dempsey was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 RIOK2 drives erythropoiesis and concomitantly suppresses megakaryopoiesis and myelopoiesis.

(a,b) Frequency of CD71 + CD235a- and CD71 + CD235a + population in control vs RIOK2 depleted TF-1 and K562 cells, respectively. (c,d) FACS plots and frequency of megakaryocytes (CD41/CD61 + ) after RIOK2 knockdown in TF-1 and K562 cells, respectively; sh#1 and 2: shRIOK2 #1 and #2. (e) Histogram plots depicting erythroblasts (CD235a), megakaryocytes (CD41/61) and myeloblasts (CD11b) in differentiating HSPCs after KD and KO of RIOK2. (f) Quantification of data presented in e, n = 4 independent donors. n = 3 technical replicates in a-d. ** p < 0.01, *** p < 0.001, **** p < 0.0001, one-way ANOVA with Tukey’s or Dunnett’s correction in c,d,f; unpaired two-tailed Student’s t-test in a,b. Data represented as mean ± s.e.m.

Source data

Extended Data Fig. 2 RIOK2 drives erythropoiesis and inhibits megakaryopoiesis and myelopoiesis.

(a,b) Total no. of myeloid (CD11b + ) and megakaryocytic (CD41/61 + ) cells after selective differentiation of control (Ctrl) vs RIOK2 KD and KO HSPCs to myeloid and megakaryocytic lineages, respectively. (c,d) Myeloid (CD13 + CD14 + ) and megakaryocytic (CD41 + CD42b + ) cells after selective differentiation of control (Ctrl) vs RIOK2 KD and KO HSPCs to myeloid and megakaryocytic lineages, respectively. (e) Images of blast forming unit-erythroid (BFU-E) and colony forming unit-granulocyte macrophage progenitors (CFU-GM) after KD and KO of RIOK2 in HSPCs, scale bar 750 µm. (f) Quantification of BFU-E, CFU-E and CFU-GM colonies in control vs RIOK2 KD and KO HSPCs. (g) Principal component analysis (PCA) of quantitative proteomic dataset showing control vs RIOK2 KD and KO primary human HSPCs, n = 3 donors. (h) Volcano plot showing differentially expressed proteins in control vs RIOK2 KD HSPCs. (i) Gene set enrichment analysis (GSEA) plot showing defective ribosome biogenesis in RIOK2 KO vs control HSPCs. n = 4 independent donors in a-d, f. * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001, one-way ANOVA with Tukey’s or Dunnett’s correction. Data represented as mean ± s.e.m. Fig. e representative of 4 independent experiments.

Source data

Extended Data Fig. 3 RIOK2 and GATA1 form a positive feedback loop to regulate hematopoietic differentiation.

(a) GATA binding motif (yellow) in the promoter region of RIOK2 (black) followed by the first exon (blue) and ATG start codon (red). (b) RIOK2 promoter-driven luciferase activity in response to increasing GATA1, GATA2 and GATA3 expression in HEK293 cells. (c) mRNA levels of RIOK2 (normalized to actin) after GATA1 overexpression (OE) and adjoining co-relation plot; EV: empty vector; Pearson’s correlation coefficient (r) and P value are shown. (d) RIOK2 mRNA expression (normalized to actin) in RIOK2 KD vs control cells. (e) GATA1 mRNA expression (normalized to actin) in GATA1 KD vs control cells. (f) HBB, ITGAM and ITGB3 mRNA expression (normalized to actin) in RIOK2 KD vs GATA1 KD HSPCs, compared to control. (g) Schema showing positive feedback loop between GATA1 and RIOK2 regulating hematopoietic differentiation. n = 3 independent donors in d-f; n = 4 and 6 technical replicates in b and c respectively. * p < 0.05, ** p < 0.01, *** p < 0.001, one-way ANOVA with Tukey’s correction in f, Unpaired two-tailed Student’s t-test in fig. c-e. Data represented as mean ± s.e.m.

Source data

Extended Data Fig. 4 RIOK2 regulates expression of transcription factors involved in hematopoietic lineage commitment.

(a) Volcano plots showing differentially expressed genes (cut off: adjusted P-Value<0.05) in control vs RIOK2 KD and control vs RIOK2 KO HSPCs; red: upregulated genes, blue: downregulated genes. (b) GATA2 mRNA level (normalized to actin) in control vs RIOK2 KD cells after suppression of GATA2 with 2 different crRNAs. (c) Erythroid progression (CD235a) in scrambled vs RIOK2 KD cells with or without suppression of GATA2 with 2 different crRNAs. (d) Megakaryocytic progression (CD41/61+ cells) in scrambled vs RIOK2 KD cells with or without knockdown of SPI1/RUNX3/GATA2 using 2 crRNAs against each. (e) Graph showing comparable mapped reads in control and RIOK2 KO HSPCs in ATAC-sequencing. n = 4 technical replicates in b-d. * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001, one-way ANOVA with Tukey’s or Dunnett’s correction. Data represented as mean ± s.e.m.

Source data

Extended Data Fig. 5 RIOK2 binds to a specific de novo nucleotide motif in the human genome.

(a) Relative binding intensity of RIOK2 at the transcription start site (TSS) in RIOK2 immunoprecipitated (RIOK2 IP) vs input sample. (b) Chromosome view plots depicting binding of RIOK2 at the promoters of GATA2 and SPI1. (c) De novo DNA binding sequences identified in the entire genome via ChIP sequencing using monoclonal antibodies of RIOK2. The sequence highlighted in red was identified in the promoters of RIOK2’s target genes. (d) Presence of de novo nucleotide binding motif specific for RIOK2 in the promoter regions of its putative target genes: GATA1, GATA2, RUNX3, SPI1, and KLF1.

Extended Data Fig. 6 RIOK2 binds DNA in vitro.

(a) Electrophoretic mobility shift assay (EMSA) showing WT (central CCC) or mutant (MUT: central CCC mutated to TTT) DNA migration in the presence of recombinant human RIOK2. (b) Quantitative band shift represented in a. (c,d) EMSA and adjoining quantification showing increasing DNA binding ability of RIOK2 over a time course; mins: minutes. (e,f) EMSA and adjoining quantification showing dose-dependent increase in DNA binding ability of RIOK2 with increasing protein concentration. (g) mRNA levels of early erythroid genes (HBB, HBA1, FECH, TFRC, KEL: normalized to actin) in RIOK2-KO HSPCs ectopically expressing EV, WT, DBM or NTE RIOK2, SCR: scrambled. n = 3 and 4 per group in d and f respectively. n = 3 experimental replicates in b,g. ** p < 0.01, **** p < 0.0001, ns: not significant, Unpaired two-tailed Student’s t-test in b, one-way ANOVA with Dunnett’s correction in g. Data represented as mean ± s.e.m. Fig. a,c representative of 3 independent experiments and fig. e representative of 4 independent experiments.

Source data

Extended Data Fig. 7 Characterization of the transactivation (TAD) and transrepressor (TRD) domains of RIOK2.

(a) ChIP of the promoter regions of GATA1, GATA2, RUNX3 and KLF1 by empty vector (EV) or HA-tagged wild-type (WT), ΔTAD1 (transactivation domain 1 deleted) and ΔTAD2 (transactivation domain 2 deleted) RIOK2 using anti-HA antibodies. (b) mRNA levels of early erythroid genes (HBB, HBA1, FECH, TFRC, KEL: normalized to actin) in RIOK2-KO HSPCs ectopically expressing EV, WT, ΔTAD1 or ΔTAD2 RIOK2, SCR: scrambled. (c) Erythroid progression (CD235a) in RIOK2-KO HSPCs ectopically expressing EV, WT or ΔTRD (deletion of transrepressor domain) RIOK2. (d) mRNA levels of early erythroid genes (HBA1, HBA2, SPTA1, TFRC: normalized to actin) in RIOK2-KO HSPCs ectopically expressing EV, WT or ΔTRD RIOK2. All comparisons done with respect to KO + EV group in fig. b-d. (e) Schema showing the 3 known domains of human RIOK2: N-terminal wHTH domain, central RIO domain and C-terminal domain. Positions of the DNA binding domain (DBD) and the transactivation domains (TAD1 and TAD2) of RIOK2 are shown. (f) Diagram illustrating RIOK2 as a master transcriptional regulator of key transcription factors (GATA1, KLF1, RUNX3, SPI1 and GATA2) in hematopoiesis. n = 2 per group in a; n = 3 independent donors in b-d. Data represented as mean ± SEM. * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001, ns: not significant, one-way ANOVA with Tukey’s or Dunnett’s correction in b-d. Data represented as mean ± s.e.m.

Source data

Extended Data Fig. 8 The kinase domain of RIOK2 is involved in erythropoiesis but does not affect RIOK2’s transcriptional activities.

(a) GATA1, SPI1 and RUNX3 promoter-driven luciferase reporter activity after expression of EV, WT, K123A, DBM or ΔTAD1 RIOK2; all comparisons done with respect to EV group (b) EMSA showing DNA-binding affinities of EV, WT, K123A, DBM, ΔTAD1 and ΔTAD2 RIOK2. (c) Western blot showing expression of ectopically expressed HA-tagged WT, DBM, K123A, ΔTAD1 RIOK2 or EV in RIOK2 KO HSPCs. (d) O-propargyl-puromycin (OPP) incorporation in control (Ctrl) vs RIOK2 KO cells after reconstitution of WT, DBM, K123A RIOK2 or EV. (e-g) Selective progression of erythroid (CD235a), myeloid (CD11b), and megakaryocytic (CD41/61) differentiation in HSPCs ectopically expressing EV, WT, DBM, ΔTAD1 or K123A RIOK2 in RIOK2-KO setting, respectively. All comparisons done with respect to EV in a, and KO + EV group in d-g. n = 3 technical replicates in a, d-g. * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001, ns: not significant, one-way ANOVA with Tukey’s or Dunnett’s correction. Data represented as mean ± s.e.m. Fig. b and c representative of 2 independent experiments.

Source data

Extended Data Fig. 9 Graphical illustration of RIOK2 functioning as a master transcription factor governing hematopoietic differentiation.

RIOK2 transcriptionally regulates key TFs in hematopoietic differentiation.

Extended Data Fig. 10 Flow cytometry plots showing gating strategies.

(a) erythroid, myeloid and megakaryocytic lineages (b) BFU-E and CFU-E formation (c) CFU-GM and CFU-Mk formation.

Supplementary information

Supplementary Information

Supplementary Tables 1 and 2

Reporting Summary

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 1

Unprocessed immunoblots.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 5

Unprocessed immunoblots.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 6

Unprocessed immunoblots.

Source Data Fig. 7

Statistical source data.

Source Data Fig. 7

Unprocessed immunoblots

Source Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 6

Unprocessed immunoblots.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 8

Unprocessed immunoblots and gels.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ghosh, S., Raundhal, M., Myers, S.A. et al. Identification of RIOK2 as a master regulator of human blood cell development. Nat Immunol 23, 109–121 (2022). https://doi.org/10.1038/s41590-021-01079-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-021-01079-w

This article is cited by

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