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:

Activation of γ-globin gene expression by GATA1 and NF-Y in hereditary persistence of fetal hemoglobin

Abstract

Hereditary persistence of fetal hemoglobin (HPFH) ameliorates β-hemoglobinopathies by inhibiting the developmental switch from γ-globin (HBG1/HBG2) to β-globin (HBB) gene expression. Some forms of HPFH are associated with γ-globin promoter variants that either disrupt binding motifs for transcriptional repressors or create new motifs for transcriptional activators. How these variants sustain γ-globin gene expression postnatally remains undefined. We mapped γ-globin promoter sequences functionally in erythroid cells harboring different HPFH variants. Those that disrupt a BCL11A repressor binding element induce γ-globin expression by facilitating the recruitment of nuclear transcription factor Y (NF-Y) to a nearby proximal CCAAT box and GATA1 to an upstream motif. The proximal CCAAT element becomes dispensable for HPFH variants that generate new binding motifs for activators NF-Y or KLF1, but GATA1 recruitment remains essential. Our findings define distinct mechanisms through which transcription factors and their cis-regulatory elements activate γ-globin expression in different forms of HPFH, some of which are being recreated by therapeutic genome editing.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: HPFH variants in the γ-globin promoter distal CCAAT box facilitate recruitment of GATA1 to an upstream motif.
Fig. 2: The −189 GATA motif facilitates γ-globin gene activation in HPFH.
Fig. 3: Disruption of the −189 GATA motif inhibits HbF expression in primary erythroblasts.
Fig. 4: Distal CCAAT box HPFH variants recruit NF-Y to the γ-globin promoter.
Fig. 5: GATA1 and NF-Y cooperate to activate γ-globin gene expression.
Fig. 6: NF-Y binding to the −85 proximal CCAAT box is dispensable for HPFH mutations that create de novo transcription factor binding sites.
Fig. 7: Competition between transcriptional repressors and activators for the γ-globin promoter in HPFH.

Similar content being viewed by others

Data availability

Datasets used in this study are listed in Supplementary Table 7. Raw and processed sequencing data generated in this study are available from the NCBI Gene Expression Omnibus under accession GSE152338. Source data are provided with this paper.

Code availability

The code used to perform ATAC–seq (HemTools atac_seq), ChIP–seq (HemTools chip_seq_pair) and Hi-C analyses (hicpro_batch.py) is available at https://github.com/YichaoOU/HemTools/ and at https://doi.org/10.5281/zenodo.4783657. Pipeline documentation is available at https://hemtools.readthedocs.io/en/latest/. The code used to perform motif analysis is available at https://github.com/YichaoOU/HPFH_code and at https://doi.org/10.5281/zenodo.4784805.

References

  1. Dean, A. On a chromosome far, far away: LCRs and gene expression. Trends Genet. 22, 38–45 (2006).

    Article  CAS  PubMed  Google Scholar 

  2. Palstra, R., de Laat, W. & Grosveld, F. Beta-globin regulation and long-range interactions. Adv. Genet. 61, 107–142 (2008).

    Article  CAS  PubMed  Google Scholar 

  3. Platt, O. S. et al. Mortality in sickle cell disease. Life expectancy and risk factors for early death. N. Engl. J. Med. 330, 1639–1644 (1994).

    Article  CAS  PubMed  Google Scholar 

  4. Musallam, K. M. et al. Fetal hemoglobin levels and morbidity in untransfused patients with β-thalassemia intermedia. Blood 119, 364–367 (2012).

    Article  CAS  PubMed  Google Scholar 

  5. Lettre, G. et al. DNA polymorphisms at the BCL11A, HBS1L-MYB and beta-globin loci associate with fetal hemoglobin levels and pain crises in sickle cell disease. Proc. Natl Acad. Sci. USA 105, 11869–11874 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Uda, M. et al. Genome-wide association study shows BCL11A associated with persistent fetal hemoglobin and amelioration of the phenotype of beta-thalassemia. Proc. Natl Acad. Sci. USA 105, 1620–1625 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Wonkam, A. et al. Association of variants at BCL11A and HBS1L-MYB with hemoglobin F and hospitalization rates among sickle cell patients in Cameroon. PLoS ONE 9, e92506 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Lettre, G. & Bauer, D. E. Fetal haemoglobin in sickle cell disease: from genetic epidemiology to new therapeutic strategies. Lancet 387, 2554–2564 (2016).

    Article  CAS  PubMed  Google Scholar 

  9. Natta, C. L., Niazi, G. A., Ford, S. & Bank, A. Balanced globin chain synthesis in hereditary persistence of fetal hemoglobin. J. Clin. Invest. 54, 433–438 (1974).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Stamatoyannopoulos, G., Wood, W. G., Papayannopoulou, T. & Nute, P. E. A new form of hereditary persistence of fetal hemoglobin in blacks and its association with sickle cell trait. Blood 46, 683–692 (1975).

    Article  CAS  PubMed  Google Scholar 

  11. Martyn, G. E. et al. Natural regulatory mutations elevate the fetal globin gene via disruption of BCL11A or ZBTB7A binding. Nat. Genet. 50, 498–503 (2018).

    Article  CAS  PubMed  Google Scholar 

  12. Liu, N. et al. Direct promoter repression by BCL11A controls the fetal to adult hemoglobin switch. Cell 173, 430–442 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Fessas, P. & Stamatoyannopoulos, G. Hereditary persistence of fetal hemoglobin in Greece: a study and a comparison. Blood 24, 223–240 (1964).

    Article  CAS  PubMed  Google Scholar 

  14. Collins, F. S. et al. A point mutation in the Aγ-globin gene promoter in Greek hereditary persistence of fetal haemoglobin. Nature 313, 325–326 (1985).

    Article  CAS  PubMed  Google Scholar 

  15. Oner, R., Kutlar, F., Gu, L. H. & Huisman, T. H. The Georgia type of nondeletional hereditary persistence of fetal hemoglobin has a C–T mutation at nucleotide-114 of the Aγ-globin gene. Blood 77, 1124–1125 (1991).

    Article  CAS  PubMed  Google Scholar 

  16. Fucharoen, S., Shimizu, K. & Fukumaki, Y. A novel C-T transition within the distal CCAAT motif of the Gγ-globin gene in the Japanese HPFH: implication of factor binding in elevated fetal globin expression. Nucleic Acids Res. 18, 5245–5253 (1990).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Motum, P. I., Deng, Z. M., Huong, L. & Trent, R. J. The Australian type of nondeletional Gγ-HPFH has a C–>G substitution at nucleotide −114 of the Gγ gene. Br. J. Haematol. 86, 219–221 (1994).

    Article  CAS  PubMed  Google Scholar 

  18. Zertal-Zidani, S. et al. A novel C–>A transversion within the distal CCAAT motif of the Gγ-globin gene in the Algerian Gγβ+-hereditary persistence of fetal hemoglobin. Hemoglobin 23, 159–169 (1999).

    Article  CAS  PubMed  Google Scholar 

  19. Gilman, J. G. et al. Distal CCAAT box deletion in the Aγ-globin gene of two black adolescents with elevated fetal Aγ globin. Nucleic Acids Res. 16, 10635–10642 (1988).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Indrak, K. et al. Compound heterozygosity for a beta zero-thalassemia (frameshift codons 38/39; -C) and a nondeletional Swiss type of HPFH (A–C at NT −110, Gγ) in a Czechoslovakian family. Ann. Hematol. 63, 111–115 (1991).

    Article  CAS  PubMed  Google Scholar 

  21. Amato, A. et al. Interpreting elevated fetal hemoglobin in pathology and health at the basic laboratory level: new and known γ-gene mutations associated with hereditary persistence of fetal hemoglobin. Int. J. Lab. Hematol. 36, 13–19 (2014).

    Article  CAS  PubMed  Google Scholar 

  22. Wienert, B. et al. KLF1 drives the expression of fetal hemoglobin in British HPFH. Blood 130, 803–807 (2017).

    Article  CAS  PubMed  Google Scholar 

  23. Wienert, B. et al. Editing the genome to introduce a beneficial naturally occurring mutation associated with increased fetal globin. Nat. Commun. 6, 7085 (2015).

    Article  CAS  PubMed  Google Scholar 

  24. Martyn, G. E. et al. A natural regulatory mutation in the proximal promoter elevates fetal globin expression by creating a de novo GATA1 site. Blood 133, 852–856 (2019).

    Article  CAS  PubMed  Google Scholar 

  25. Liberati, C., Ronchi, A., Lievens, P., Ottolenghi, S. & Mantovani, R. NF-Y organizes the gamma-globin CCAAT boxes region. J. Biol. Chem. 273, 16880–16889 (1998).

    Article  CAS  PubMed  Google Scholar 

  26. Duan, Z., Stamatoyannopoulos, G. & Li, Q. Role of NF-Y in in vivo regulation of the gamma-globin gene. Mol. Cell. Biol. 21, 3083–3095 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Martyn, G. E., Quinlan, K. G. R. & Crossley, M. The regulation of human globin promoters by CCAAT box elements and the recruitment of NF-Y. Biochim. Biophys. Acta 1860, 525–536 (2017).

    Article  CAS  Google Scholar 

  28. Xu, J. et al. Corepressor-dependent silencing of fetal hemoglobin expression by BCL11A. Proc. Natl Acad. Sci. USA 110, 6518–6523 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Sankaran, V. G. et al. Human fetal hemoglobin expression is regulated by the developmental stage-specific repressor BCL11A. Science 322, 1839–1842 (2008).

    Article  CAS  PubMed  Google Scholar 

  30. Liu, N. et al. Transcription factor competition at the γ-globin promoters controls hemoglobin switching. Nat. Genet. https://doi.org/10.1038/s41588-021-00798-y (2021).

  31. Martin, D. I., Tsai, S. F. & Orkin, S. H. Increased γ-globin expression in a nondeletion HPFH mediated by an erythroid-specific DNA-binding factor. Nature 338, 435–438 (1989).

    Article  CAS  PubMed  Google Scholar 

  32. Traxler, E. A. et al. A genome-editing strategy to treat β-hemoglobinopathies that recapitulates a mutation associated with a benign genetic condition. Nat. Med. 22, 987–990 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Masuda, T. et al. Transcription factors LRF and BCL11A independently repress expression of fetal hemoglobin. Science 351, 285–289 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Métais, J.-Y. et al. Genome editing of HBG1 and HBG2 to induce fetal hemoglobin. Blood Adv. 3, 3379–3392 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Weber, L. et al. Editing a γ-globin repressor binding site restores fetal hemoglobin synthesis and corrects the sickle cell disease phenotype. Sci. Adv. 6, eaay9392 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Kurita, R. et al. Establishment of immortalized human erythroid progenitor cell lines able to produce enucleated red blood cells. PLoS ONE 8, e59890 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Huisman, T. H. et al. The occurrence of different levels of G gamma chain and of the A gamma T variant of fetal hemoglobin in newborn babies from several countries. Am. J. Hematol. 14, 133–148 (1983).

    Article  CAS  PubMed  Google Scholar 

  38. Sukumaran, P. K. et al. Gamma thalassemia resulting from the deletion of a gamma-globin gene. Nucleic Acids Res. 11, 4635–4643 (1983).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Zeng, Y. T., Huang, S. Z., Nakatsuji, T. & Huisman, T. H. -GγAγ-thalassemia and γ-chain variants in Chinese newborn babies. Am. J. Hematol. 18, 235–242 (1985).

    Article  CAS  PubMed  Google Scholar 

  40. 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 

  41. Xiang, G. et al. S3norm: simultaneous normalization of sequencing depth and signal-to-noise ratio in epigenomic data. Nucleic Acids Res. 48, e43 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Gaudelli, N. M. et al. Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature 551, 464–471 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Koblan, L. W. et al. Improving cytidine and adenine base editors by expression optimization and ancestral reconstruction. Nat. Biotechnol. 36, 843–846 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Zhu, X. et al. NF-Y recruits both transcription activator and repressor to modulate tissue- and developmental stage-specific expression of human γ-globin gene. PLoS One 7, e47175 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Superti-Furga, G., Barberis, A., Schaffner, G. & Busslinger, M. The −117 mutation in Greek HPFH affects the binding of three nuclear factors to the CCAAT region of the gamma-globin gene. EMBO J. 7, 3099–3107 (1988).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Grant, C. E., Bailey, T. L. & Noble, W. S. FIMO: scanning for occurrences of a given motif. Bioinformatics 27, 1017–1018 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Forget, B. G. Molecular basis of hereditary persistence of fetal hemoglobin. Ann. N. Y. Acad. Sci. 850, 38–44 (1998).

    Article  CAS  PubMed  Google Scholar 

  48. Wienert, B., Martyn, G. E., Funnell, A. P. W., Quinlan, K. G. R. & Crossley, M. Wake-up sleepy gene: reactivating fetal globin for β-hemoglobinopathies. Trends Genet. 34, 927–940 (2018).

    Article  CAS  PubMed  Google Scholar 

  49. Fang, X., Han, H., Stamatoyannopoulos, G. & Li, Q. Developmentally specific role of the CCAAT box in regulation of human gamma-globin gene expression. J. Biol. Chem. 279, 5444–5449 (2004).

    Article  CAS  PubMed  Google Scholar 

  50. Ronchi, A. et al. Role of the duplicated CCAAT box region in gamma-globin gene regulation and hereditary persistence of fetal haemoglobin. EMBO J. 15, 143–149 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Katsube, T. & Fukumaki, Y. A role for the distal CCAAT box of the gamma-globin gene in Hb switching. J. Biochem. 117, 68–76 (1995).

    Article  CAS  PubMed  Google Scholar 

  52. Huang, D.-Y. et al. GATA-1 and NF-Y cooperate to mediate erythroid-specific transcription of Gfi-1B gene. Nucleic Acids Res. 32, 3935–3946 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Torchy, M. P., Hamiche, A. & Klaholz, B. P. Structure and function insights into the NuRD chromatin remodeling complex. Cell. Mol. Life Sci. 72, 2491–2507 (2015).

    Article  CAS  PubMed  Google Scholar 

  54. Yang, Y. et al. Structural insights into the recognition of γ-globin gene promoter by BCL11A. Cell Res. 29, 960–963 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Jolma, A. et al. DNA-dependent formation of transcription factor pairs alters their binding specificity. Nature 527, 384–388 (2015).

    Article  CAS  PubMed  Google Scholar 

  56. Pique-Regi, R. et al. Accurate inference of transcription factor binding from DNA sequence and chromatin accessibility data. Genome Res. 21, 447–455 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Vökel, S. et al. Zinc finger independent genome-wide binding of SP2 potentiates recruitment of histone-fold protein NF-Y distinguishing it from SP1 and SP3. PLoS Genet. 11, 1–25 (2015).

    Google Scholar 

  58. Suske, G. NF-Y and SP transcription factors—new insights in a long-standing liaison. Biochim. Biophys. Acta 1860, 590–597 (2017).

    Article  CAS  Google Scholar 

  59. Yang, L. et al. Methylation of a CGATA element inhibits binding and regulation by GATA-1. Nat. Commun. 11, 2560 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Vakoc, C. R. et al. Proximity among distant regulatory elements at the beta-globin locus requires GATA-1 and FOG-1. Mol. Cell 17, 453–462 (2005).

    Article  CAS  PubMed  Google Scholar 

  61. Downes, D. J. et al. High-resolution targeted 3C interrogation of cis-regulatory element organization at genome-wide scale. Nat. Commun. 12, 531 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Norton, L. J. et al. KLF1 directly activates expression of the novel fetal globin repressor ZBTB7A/LRF in erythroid cells. Blood Adv. 1, 685–692 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Zhou, D., Liu, K., Sun, C.-W., Pawlik, K. M. & Townes, T. M. KLF1 regulates BCL11A expression and γ- to β-globin gene switching. Nat. Genet. 42, 742–744 (2010).

    Article  CAS  PubMed  Google Scholar 

  64. Humbert, O. et al. Therapeutically relevant engraftment of a CRISPR–Cas9-edited HSC-enriched population with HbF reactivation in nonhuman primates. Sci. Transl. Med. 11, eaaw3768 (2019).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  65. Lux, C. T. et al. TALEN-mediated gene editing of HBG in human hematopoietic stem cells leads to therapeutic fetal hemoglobin induction. Mol. Ther. Methods Clin. Dev. 12, 175–183 (2019).

    Article  CAS  PubMed  Google Scholar 

  66. Anzalone, A. V., Koblan, L. W. & Liu, D. R. Genome editing with CRISPR–Cas nucleases, base editors, transposases and prime editors. Nat. Biotechnol. 38, 824–844 (2020).

    Article  CAS  PubMed  Google Scholar 

  67. Connelly, J. P. & Pruett-Miller, S. M. CRIS.py: a versatile and high-throughput analysis program for CRISPR-based genome editing. Sci. Rep. 9, 4194 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Landt, S. G. et al. ChIP–seq guidelines and practices of the ENCODE and modENCODE consortia. Genome Res. 22, 1813–1831 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. 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 

  70. 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 

  71. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Ramírez, F., Dündar, F., Diehl, S., Grüning, B. A. & Manke, T. deepTools: a flexible platform for exploring deep-sequencing data. Nucleic Acids Res. 42, W187–W191 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  74. Jiang, H., Lei, R., Ding, S.-W. & Zhu, S. Skewer: a fast and accurate adapter trimmer for next-generation sequencing paired-end reads. BMC Bioinformatics 15, 182 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  75. Rao, S. S. P. et al. A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell 159, 1665–1680 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Servant, N. et al. HiC-Pro: an optimized and flexible pipeline for Hi-C data processing. Genome Biol. 16, 259 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  77. Ramírez, F. et al. High-resolution TADs reveal DNA sequences underlying genome organization in flies. Nat. Commun. 9, 189 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Andrews, N. C. & Faller, D. V. A rapid micropreparation technique for extraction of DNA-binding proteins from limiting numbers of mammalian cells. Nucleic Acids Res. 19, 2499 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Xu, J. et al. Combinatorial assembly of developmental stage-specific enhancers controls gene expression programs during human erythropoiesis. Dev. Cell 23, 796–811 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Cheng, L. et al. Single-nucleotide-level mapping of DNA regulatory elements that control fetal hemoglobin expression. Nat. Genet. https://doi.org/10.1038/s41588-021-00861-8 (2021).

Download references

Acknowledgements

We are grateful to A. Impagliazzo for illustration assistance as well as R. Hardison, G. Xiang, M. Osborn, G. Newby and D. Liu for valuable discussion and technical expertise. HUDEP-2 cells were a gift from R. Kurita and Y. Nakamura (RIKEN BioResource Center). ABE7.10 was a gift from M. Osborn (University of Minnesota) and D. Liu (Harvard University/HHMI). APC-conjugated anti-Band3 was a gift from X. An (New York Blood Center). This work was supported by National Institutes of Health (NIH) grants P01HL053749 (to S.M.P.-M. and M.J.W.), R01HL156647 (to M.J.W.), R35GM133614 (to Y.C.), R24106766 (to Y.C and M.J.W.) and F32DK118822 (to P.A.D.); The Assisi Foundation of Memphis (to M.J.W.); Doris Duke Charitable Foundation grant 2017093 (to M.J.W.); an Australian Government Research Training Program Scholarship (to H.W.B.); the Australian National Health and Medical Research Council grant APP1164920 (to M.C.); The St. Jude Collaborative Research Consortium for Sickle Cell Disease and St. Jude/ALSAC. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH. The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript. We thank the members of the St. Jude Children’s Research Hospital Cytogenetics, Hartwell Center, Center for Advanced Genome Engineering and Flow Cytometry core facilities. The St. Jude Cytogenetics, Hartwell Center, Center for Advanced Genome Engineering and Flow Cytometry Shared Resource Laboratories are supported by NIH grant P30CA21765 and by St. Jude/ALSAC.

Author information

Authors and Affiliations

Authors

Contributions

P.A.D., R.F., S.N.P., H.W.B., M.C., S.M.P.-M., Y.C. and M.J.W designed experiments. P.A.D., R.F. and H.W.B. performed experiments and analyzed data. P.A.D., S.N.P., Y.L. and L.E.P. performed sequencing and associated analyses. M.C., S.M.P.-M. and Y.C. provided conceptual advice and technical expertise. M.J.W. supervised the study. P.A.D. and M.J.W. wrote the manuscript. All authors discussed the results and assisted in the preparation of the manuscript.

Corresponding author

Correspondence to Mitchell J. Weiss.

Ethics declarations

Competing interests

M.J.W. is on advisory boards for Cellarity, Graphite Bio, Novartis and Forma Therapeutics, and is an equity owner of Beam Therapeutics. All other authors declare no competing interests.

Additional information

Peer review information Nature Genetics thanks Sjaak Philipsen and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 Derivation of HUDEP-2 cells containing a single γ-globin gene.

Genome browser view of deletions introduced into HUDEP-2 cells to generate the HUDEP-2Δεγδβ/GγΑγ line, which contains a single γ-globin gene. The region of the β-like globin gene cluster that was deleted on one chromosome is shown in blue. Chromatin immunoprecipitation (ChIP-seq) analysis for CTCF occupancy and ATAC-seq analysis of HUDEP-2 cells were derived from publicly available data. b, Generation of a single HBG2-HBG1 fusion gene on the remaining β-like globin gene locus. Positions of the gRNAs targeting intron 2 of HBG1 and HBG2 are in shown with arrows. c, Fluorescence in situ hybridization analysis of wild-type (WT) HUDEP-2, HUDEP-2Δεγδβ, and HUDEP-2Δεγδβ/GγΑγ cells using a 117.8 kb probe for chr11 (green, shown in panel a) and a 5.2 kb probe from the deleted region between HBG1 and HBG2 (HBG; red, shown in panel b) in wild-type HUDEP-2, HUDEP-2Δεγδβ, and HUDEP-2Δεγδβ/GγΑγ cells. Results are from a single experiment. Scale bar = 5 μm.

Extended Data Fig. 2 Characterization of the HUDEP-2Δεγδβ/GγAγ cell line.

a, Next generation sequencing (NGS) analysis of the indicated HUDEP-2 lines showing percentages of reads corresponding to HBG1 or HBG2 exon 3. (mean ± SD; n = 6 independent clones for each genotype). b, %HbF in WT and HUDEP-2Δεγδβ/GγAγ clones, measured by ion-exchange high-performance liquid chromatography (IE-HPLC) after 7 days of erythroid differentiation. Box and whisker plots show minimum, maximum, median, and interquartile ranges. n = 6 independent clones for each genotype. *p = 0.0156 uncorrected two-tailed unpaired t-test. c, Kinetics of erythroid maturation of WT and HUDEP-2Δεγδβ/GγAγ cells determined by flow cytometry for CD49d and Band3 in the CD235a+ population at the indicated timepoints after culture in erythroid differentiation medium. Mean ± SD is shown in each quadrant. n = 6 independent clones analyzed for each genotype.

Source data

Extended Data Fig. 3 Hi-C analysis showing chromatin structure of the extended β-globin locus in HUDEP-2 cells containing a single γ-globin gene.

a, Heat maps comparing chromatin interactions of the extended β-like globin locus in WT HUDEP-2 cells (red) and in HUDEP-2Δεγδβ/GγAγ cells, which contain a single, modified β-like globin locus (blue). Tracks below show transcriptionally open or closed compartments as positive (blue) or negative (magenta) according to Hi-C analysis. CTCF ChIP-seq analysis and ATAC seq analysis are shown for WT HUDEP-2 cells. The 91.5 kb deletion of the extended β-globin locus (Δεγδβ) is shown as a blue rectangle; the 5.4 kb deletion generating a single HBG2-HBG1 fusion gene (GγAγ) is designated in grey. Genes are designated as black vertical bars in the bottom track. b, The topologically associated domain (TAD) separation score correlation comparing the frequency of the union (top) and intersection (bottom) between HUDEP-2 and HUDEP-2Δεγδβ/GγAγ cells. The TAD scores identify the degree of separation between the left and right boundaries based on the Hi-C interaction matrix. A TAD will be called at local minima. The union indicates the left and right boundaries across HUDEP-2 and HUDEP-2Δεγδβ/GγAγ cells. The intersection represents the fraction of shared boundaries between the HUDEP-2 and HUDEP-2Δεγδβ/GγAγ TAD sets. The Spearman correlation coefficients (ρ) are shown. Results were generated from merged reads derived from two independent experiments.

Extended Data Fig. 4 Effects of HPFH variants on HbF expression in HUDEP-2 cells.

a, HPLC tracings showing hemoglobin analysis of WT HUDEP-2, HUDEP-2Δεγδβ/GγAγ and HUDEP-2Δεγδβ/GγAγ cells with the −110 A > C HPFH variant after 7 days of erythroid differentiation. b, ATAC-seq tracks showing open chromatin at the β-like globin gene cluster in single clones with distal CCAAT box HPFH variants −117 G > A and −114 C > A and control mutations −110 A > G and −110 A > T. The shaded area highlighting the HBG promoter is shown in higher resolution on the right. The reference genes are shown at the bottom and the dotted lines indicate the region deleted to create the single in-frame HBG fusion gene.

Extended Data Fig. 5 GATA1 occupancy at the γ-globin promoters is associated with HbF expression.

a, ChIP-seq analysis showing GATA1 occupancy at the β-like globin gene cluster in primary fetal and adult proerythroblasts79. The shaded areas highlighting the HBG1 and HBG2 promoters are shown in higher resolution below. b, ChIP seq analysis for GATA1 in HUDEP-1 and HUDEP-2 cells, which express predominantly γ-globin and β-globin respectively, shown as described for panel a. GATA1 occupancy in HUDEP-2 cells was derived from publicly available data80.

Extended Data Fig. 6 Disruption of the bipartite GATA motif via −186 C > T impairs HbF expression associated with HPFH variants.

a, Representative ion-exchange high-performance liquid chromatography (HPLC) traces showing reduced fetal hemoglobin (HbF) peak intensity in HPFH clones without (top) and with −186 C > T (bottom). Cells were grown in culture for 7 days under erythroid differentiation conditions. b, Representative F-cell staining flow cytometry plots in undifferentiated HPFH clones without (top) and with −186 C > T (bottom). c, Replicate ChIP-seq analysis showing GATA1 occupancy at the β-like globin gene cluster in clones harboring distal CCAAT box HPFH variants ± the −186 C > T GATA motif mutation (related to Fig. 2d). The shaded area highlighting the HBG promoter is shown in higher resolution on the right. The reference genes are shown at the bottom and the dotted lines indicate the region deleted to create the single in-frame HBG fusion gene.

Extended Data Fig. 7 Disruption of the −189 GATA motif in HUDEP-2Δεγδβ/GγAγ cells does not induce γ-globin expression.

a, Sequence of the HBG promoter showing the bipartite GATA motif (blue), BCL11A binding motif (grey) and the distal CCAAT box (dotted rectangle; hg19 – chr11:5,276,112-5,276,201). The −186 C > T mutation (lower case bold), disrupts GATA1 binding. b, %HbF (left) and %F-cells (right) after 7 days of erythroid differentiation in HUDEP-2Δεγδβ/GγAγ cells ± the −186 C > T mutation (lower case bold). Each dot represents an individual clone (n = 12 per genotype). Box and whisker plots show minimum, maximum, median, and interquartile ranges. **p = 0.0017, uncorrected two-tailed unpaired t-test. An uncorrected two-tailed unpaired t-test indicated no significant effect of the −186T mutation on %HbF in WT CCAAT box clones, p > 0.9999.

Source data

Extended Data Fig. 8 −110 A > C at the distal CCAAT box enhances NF-Y binding.

a, Electrophoretic mobility shift assay (EMSA) for NF-Y binding to WT or mutant γ-globin promoter distal CCAAT box oligonucleotides in K562 cell nuclear extracts. Mutations are indicated in lower case bold. Bound probe is indicated by the closed triangle and supershift product of the NF-Y:probe complex is indicated by the open triangle. Graph on the right shows densitometry analysis of NF-Y band intensity relative to WT signal. b, Motif analysis showing the predicted effects of single nucleotide alterations on BCL11A binding to the −115 distal CCAAT box. The −110 A > C HPFH variant (asterisk) is predicted to decrease BCL11A affinity for the motif. c, Competitive EMSA assay for BCL11A binding to distal CCAAT box probes. The autoradiogram shows competition of cold WT or −110 A > C probes (1X, 5X, 10X, 25X, and 50X molar excess) with radiolabeled WT probe for binding to BCL11A zinc fingers 4-6 expressed in COS-7 cells. Bound probe is indicated by a closed triangle. The graph shows densitometry analysis of this band after incubation with cold competitor oligonucleotides, normalized to intensity with no competitor.

Source data

Extended Data Fig. 9 Flow cytometry gating strategies.

a, Gating strategy for monitoring the differentiation of HUDEP-2 cells (see Extended Data Fig. 2c). b, Gating strategy for F-cell determination in undifferentiated HUDEP-2 cells (see Extended Data Fig. 6b). Antibodies used are listed in the methods section.

Supplementary information

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data and unprocessed gels.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data and unprocessed gels.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Doerfler, P.A., Feng, R., Li, Y. et al. Activation of γ-globin gene expression by GATA1 and NF-Y in hereditary persistence of fetal hemoglobin. Nat Genet 53, 1177–1186 (2021). https://doi.org/10.1038/s41588-021-00904-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-021-00904-0

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