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:

CRISPR screening uncovers a central requirement for HHEX in pancreatic lineage commitment and plasticity restriction

Abstract

The pancreas and liver arise from a common pool of progenitors. However, the underlying mechanisms that drive their lineage diversification from the foregut endoderm are not fully understood. To tackle this question, we undertook a multifactorial approach that integrated human pluripotent-stem-cell-guided differentiation, genome-scale CRISPR–Cas9 screening, single-cell analysis, genomics and proteomics. We discovered that HHEX, a transcription factor (TF) widely recognized as a key regulator of liver development, acts as a gatekeeper of pancreatic lineage specification. HHEX deletion impaired pancreatic commitment and unleashed an unexpected degree of cellular plasticity towards the liver and duodenum fates. Mechanistically, HHEX cooperates with the pioneer TFs FOXA1, FOXA2 and GATA4, shared by both pancreas and liver differentiation programmes, to promote pancreas commitment, and this cooperation restrains the shared TFs from activating alternative lineages. These findings provide a generalizable model for how gatekeeper TFs like HHEX orchestrate lineage commitment and plasticity restriction in broad developmental contexts.

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: CRISPR–Cas9 screens identify regulators of pancreatic differentiation.
Fig. 2: HHEX expression in pancreas development.
Fig. 3: Deletion of HHEX impairs human pancreatic differentiation.
Fig. 4: HHEX deletion results in induction of a liver-like transcriptional programme.
Fig. 5: HHEX KO cells acquire a duodenum-like cell state following inhibition of liver differentiation.
Fig. 6: scRNA-seq reveals differentiation trajectories of WT and HHEX KO cells.
Fig. 7: HHEX cooperates with FOXA2 to safeguard pancreatic differentiation.
Fig. 8: HHEX protects FOXA2 binding to pancreatic regulatory regions.

Similar content being viewed by others

Data availability

Sequencing data are available at the GEO under accession code GSE181480. Previously published data that were re-analysed here are available at the GEO under accession codes GSE136689 and GSE86225. Source data are provided with this paper. All other data supporting the findings of this study are available from the corresponding authors upon reasonable request.

References

  1. Zaret, K. S. & Grompe, M. Generation and regeneration of cells of the liver and pancreas. Science 322, 1490–1494 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Willnow, D. et al. Quantitative lineage analysis identifies a hepato-pancreato-biliary progenitor niche. Nature 597, 87–91 (2021).

    Article  CAS  PubMed  Google Scholar 

  3. Puri, S., Folias, A. E. & Hebrok, M. Plasticity and dedifferentiation within the pancreas: development, homeostasis, and disease. Cell Stem Cell 16, 18–31 (2015).

    Article  CAS  PubMed  Google Scholar 

  4. Yuan, S., Norgard, R. J. & Stanger, B. Z. Cellular plasticity in cancer. Cancer Discov. 9, 837–851 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Tata, P. R. et al. Developmental history provides a roadmap for the emergence of tumor plasticity. Dev. Cell 44, 679–693.e5 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Zhu, Z. et al. Genome editing of lineage determinants in human pluripotent stem cells reveals mechanisms of pancreatic development and diabetes. Cell Stem Cell 18, 755–768 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Wang, X. et al. Point mutations in the PDX1 transactivation domain impair human β-cell development and function. Mol. Metab. 24, 80–97 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Jonsson, J., Carlsson, L., Edlund, T. & Edlund, H. Insulin-promoter-factor 1 is required for pancreas development in mice. Nature 371, 606–609 (1994).

    Article  CAS  PubMed  Google Scholar 

  9. Offield, M. F. et al. PDX-1 is required for pancreatic outgrowth and differentiation of the rostral duodenum. Development 122, 983–995 (1996).

    Article  CAS  PubMed  Google Scholar 

  10. Stoffers, D. A., Zinkin, N. T., Stanojevic, V., Clarke, W. L. & Habener, J. F. Pancreatic agenesis attributable to a single nucleotide deletion in the human IPF1 gene coding sequence. Nat. Genet. 15, 106–110 (1997).

    Article  CAS  PubMed  Google Scholar 

  11. Carrasco, M., Delgado, I., Soria, B., Martin, F. & Rojas, A. GATA4 and GATA6 control mouse pancreas organogenesis. J. Clin. Invest. 122, 3504–3515 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Shi, Z. D. et al. Genome editing in hPSCs reveals GATA6 haploinsufficiency and a genetic interaction with GATA4 in human pancreatic development. Cell Stem Cell 20, 675–688.e6 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Tiyaboonchai, A. et al. GATA6 plays an important role in the induction of human definitive endoderm, development of the pancreas, and functionality of pancreatic β cells. Stem Cell Reports 8, 589–604 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Xuan, S. et al. Pancreas-specific deletion of mouse Gata4 and Gata6 causes pancreatic agenesis. J. Clin. Invest. 122, 3516–3528 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Gao, N. et al. Dynamic regulation of Pdx1 enhancers by Foxa1 and Foxa2 is essential for pancreas development. Genes Dev. 22, 3435–3448 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Lee, K. et al. FOXA2 is required for enhancer priming during pancreatic differentiation. Cell Rep. 28, 382–393.e7 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Geusz, R. J. et al. Sequence logic at enhancers governs a dual mechanism of endodermal organ fate induction by FOXA pioneer factors. Nat. Commun. 12, 6636 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Genga, R. M. J. et al. Single-cell RNA-sequencing-based CRISPRi screening resolves molecular drivers of early human endoderm development. Cell Rep. 27, 708–718.e10 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Lee, C. S., Friedman, J. R., Fulmer, J. T. & Kaestner, K. H. The initiation of liver development is dependent on Foxa transcription factors. Nature 435, 944–947 (2005).

    Article  CAS  PubMed  Google Scholar 

  20. Watt, A. J., Zhao, R., Li, J. & Duncan, S. A. Development of the mammalian liver and ventral pancreas is dependent on GATA4. BMC Dev. Biol. 7, 37 (2007).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  21. Zhao, R. et al. GATA6 is essential for embryonic development of the liver but dispensable for early heart formation. Mol. Cell. Biol. 25, 2622–2631 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Keng, V. W. et al. Homeobox gene Hex is essential for onset of mouse embryonic liver development and differentiation of the monocyte lineage. Biochem. Biophys. Res. Commun. 276, 1155–1161 (2000).

    Article  CAS  PubMed  Google Scholar 

  23. Hunter, M. P. et al. The homeobox gene Hhex is essential for proper hepatoblast differentiation and bile duct morphogenesis. Dev. Biol. 308, 355–367 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Martinez Barbera, J. P. et al. The homeobox gene Hex is required in definitive endodermal tissues for normal forebrain, liver and thyroid formation. Development 127, 2433–2445 (2000).

    Article  CAS  PubMed  Google Scholar 

  25. Bort, R., Martinez-Barbera, J. P., Beddington, R. S. & Zaret, K. S. Hex homeobox gene-dependent tissue positioning is required for organogenesis of the ventral pancreas. Development 131, 797–806 (2004).

    Article  CAS  PubMed  Google Scholar 

  26. Zaret, K. S. et al. Pioneer factors, genetic competence, and inductive signaling: programming liver and pancreas progenitors from the endoderm. Cold Spring Harb. Symp. Quant. Biol. 73, 119–126 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Rezania, A. et al. Reversal of diabetes with insulin-producing cells derived in vitro from human pluripotent stem cells. Nat. Biotechnol. 32, 1121–1133 (2014).

    Article  CAS  PubMed  Google Scholar 

  28. Nostro, M. C. et al. Efficient generation of NKX6-1+ pancreatic progenitors from multiple human pluripotent stem cell lines. Stem Cell Reports 4, 591–604 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Pagliuca, F. W. et al. Generation of functional human pancreatic β cells in vitro. Cell 159, 428–439 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Russ, H. A. et al. Controlled induction of human pancreatic progenitors produces functional β-like cells in vitro. EMBO J. 34, 1759–1772 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Hogrebe, N. J., Augsornworawat, P., Maxwell, K. G., Velazco-Cruz, L. & Millman, J. R. Targeting the cytoskeleton to direct pancreatic differentiation of human pluripotent stem cells. Nat. Biotechnol. 38, 460–470 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Pan, F. C. & Wright, C. Pancreas organogenesis: from bud to plexus to gland. Dev. Dyn. 240, 530–565 (2011).

    Article  CAS  PubMed  Google Scholar 

  33. Zhu, Z., Verma, N., Gonzalez, F., Shi, Z. D. & Huangfu, D. A CRISPR/Cas-mediated selection-free knockin strategy in human embryonic stem cells. Stem Cell Reports 4, 1103–1111 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Gonzalez, F. et al. An iCRISPR platform for rapid, multiplexable, and inducible genome editing in human pluripotent stem cells. Cell Stem Cell 15, 215–226 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Sanjana, N. E., Shalem, O. & Zhang, F. Improved vectors and genome-wide libraries for CRISPR screening. Nat. Methods 11, 783–784 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Li, Q. V. et al. Genome-scale screens identify JNK–JUN signaling as a barrier for pluripotency exit and endoderm differentiation. Nat. Genet. 51, 999–1010 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Dixon, G. et al. QSER1 protects DNA methylation valleys from de novo methylation. Science 372, eabd0875 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Jennings, R. E. et al. Laser capture and deep sequencing reveals the transcriptomic programmes regulating the onset of pancreas and liver differentiation in human embryos. Stem Cell Reports 9, 1387–1394 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Xu, Y. et al. A single-cell transcriptome atlas of human early embryogenesis. Preprint at bioRxiv https://doi.org/10.1101/2021.11.30.470583 (2021).

  40. Zhang, J., McKenna, L. B., Bogue, C. W. & Kaestner, K. H. The diabetes gene Hhex maintains δ-cell differentiation and islet function. Genes Dev. 28, 829–834 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Sugiyama, T., Rodriguez, R. T., McLean, G. W. & Kim, S. K. Conserved markers of fetal pancreatic epithelium permit prospective isolation of islet progenitor cells by FACS. Proc. Natl Acad. Sci. USA 104, 175–180 (2007).

    Article  CAS  PubMed  Google Scholar 

  42. Oshima, Y. et al. Isolation of mouse pancreatic ductal progenitor cells expressing CD133 and c-Met by flow cytometric cell sorting. Gastroenterology 132, 720–732 (2007).

    Article  CAS  PubMed  Google Scholar 

  43. Li, L.-C. et al. Single-cell transcriptomic analyses reveal distinct dorsal/ventral pancreatic programs. EMBO Rep, 19, e46148 (2018).

    Article  CAS  Google Scholar 

  44. Odom, D. T. et al. Control of pancreas and liver gene expression by HNF transcription factors. Science 303, 1378–1381 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Rossi, J. M., Dunn, N. R., Hogan, B. L. & Zaret, K. S. Distinct mesodermal signals, including BMPs from the septum transversum mesenchyme, are required in combination for hepatogenesis from the endoderm. Genes Dev. 15, 1998–2009 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Shin, D. et al. Bmp and Fgf signaling are essential for liver specification in zebrafish. Development 134, 2041–2050 (2007).

    Article  CAS  PubMed  Google Scholar 

  47. Ang, L. T. et al. A roadmap for human liver differentiation from pluripotent stem cells. Cell Rep. 22, 2190–2205 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Gouon-Evans, V. et al. BMP-4 is required for hepatic specification of mouse embryonic stem cell-derived definitive endoderm. Nat. Biotechnol. 24, 1402–1411 (2006).

    Article  CAS  PubMed  Google Scholar 

  49. Han, L. et al. Single cell transcriptomics identifies a signaling network coordinating endoderm and mesoderm diversification during foregut organogenesis. Nat. Commun. 11, 4158 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Setty, M. et al. Characterization of cell fate probabilities in single-cell data with Palantir. Nat. Biotechnol. 37, 451–460 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Li, Q. V., Rosen, B. P. & Huangfu, D. Decoding pluripotency: genetic screens to interrogate the acquisition, maintenance, and exit of pluripotency. Wiley Interdiscip. Rev. Syst. Biol. Med. 12, e1464 (2020).

    Article  PubMed  Google Scholar 

  52. Yilmaz, A., Braverman-Gross, C., Bialer-Tsypin, A., Peretz, M. & Benvenisty, N. Mapping gene circuits essential for germ layer differentiation via loss-of-function screens in haploid human embryonic stem cells. Cell Stem Cell 27, 679–691.e6 (2020).

    Article  CAS  PubMed  Google Scholar 

  53. Naxerova, K. et al. Integrated loss- and gain-of-function screens define a core network governing human embryonic stem cell behavior. Genes Dev. 35, 1527–1547 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Cai, Y., Yi, J., Ma, Y. & Fu, D. Meta-analysis of the effect of HHEX gene polymorphism on the risk of type 2 diabetes. Mutagenesis 26, 309–314 (2011).

    Article  CAS  PubMed  Google Scholar 

  55. Bort, R., Signore, M., Tremblay, K., Martinez Barbera, J. P. & Zaret, K. S. Hex homeobox gene controls the transition of the endoderm to a pseudostratified, cell emergent epithelium for liver bud development. Dev. Biol. 290, 44–56 (2006).

    Article  CAS  PubMed  Google Scholar 

  56. Xu, C. R. et al. Chromatin “prepattern” and histone modifiers in a fate choice for liver and pancreas. Science 332, 963–966 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Trizzino, M. et al. EGR1 is a gatekeeper of inflammatory enhancers in human macrophages. Sci. Adv. 7, eaaz8836 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Shalom-Feuerstein, R. et al. ΔNp63 is an ectodermal gatekeeper of epidermal morphogenesis. Cell Death Differ. 18, 887–896 (2011).

    Article  CAS  PubMed  Google Scholar 

  59. Markov, G. J. et al. AP-1 is a temporally regulated dual gatekeeper of reprogramming to pluripotency. Proc. Natl Acad. Sci. USA 118, e2104841118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Mall, M. et al. Myt1l safeguards neuronal identity by actively repressing many non-neuronal fates. Nature 544, 245–249 (2017).

    Article  CAS  PubMed  Google Scholar 

  61. 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 

  62. Doench, J. G. et al. Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR–Cas9. Nat. Biotechnol. 34, 184–191 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  CAS  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  65. 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 

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

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  67. Babicki, S. et al. Heatmapper: web-enabled heat mapping for all. Nucleic Acids Res. 44, W147–W153 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Zheng, G. X. et al. Massively parallel digital transcriptional profiling of single cells. Nat. Commun. 8, 14049 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Kowalczyk, M. S. et al. Single-cell RNA-seq reveals changes in cell cycle and differentiation programs upon aging of hematopoietic stem cells. Genome Res. 25, 1860–1872 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 352, 189–196 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Finak, G. et al. MAST: a flexible statistical framework for assessing transcriptional changes and characterizing heterogeneity in single-cell RNA sequencing data. Genome Biol. 16, 278 (2015).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  74. Raudvere, U. et al. g:Profiler: a web server for functional enrichment analysis and conversions of gene lists (2019 update). Nucleic Acids Res. 47, W191–W198 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. van Dijk, D. et al. Recovering gene interactions from single-cell data using data diffusion. Cell 174, 716–729.e27 (2018).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  76. Jacomy, M., Venturini, T., Heymann, S. & Bastian, M. ForceAtlas2, a continuous graph layout algorithm for handy network visualization designed for the Gephi software. PLoS ONE 9, e98679 (2014).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  77. Buenrostro, J. D., Wu, B., Chang, H. Y. & Greenleaf, W. J. ATAC-seq: a method for assaying chromatin accessibility genome-wide. Curr. Protoc. Mol. Biol. 109, 21.29.1–21.29.9 (2015).

    Article  Google Scholar 

  78. Li, H. & Durbin, R. Fast and accurate long-read alignment with Burrows–Wheeler transform. Bioinformatics 26, 589–595 (2010).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  79. 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 

  80. van der Veeken, J. et al. Memory of inflammation in regulatory T cells. Cell 166, 977–990 (2016).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

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

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  82. Li, Q. H., Brown, J. B., Huang, H. Y. & Bickel, P. J. Measuring reproducibility of high-throughput experiments. Ann. Appl. Stat. 5, 1752–1779 (2011).

    Google Scholar 

  83. Liu, T. Use model-based analysis of ChIP-seq (MACS) to analyze short reads generated by sequencing protein–DNA interactions in embryonic stem cells. Methods Mol. Biol. 1150, 81–95 (2014).

    Article  CAS  PubMed  Google Scholar 

  84. Aguilan, J. T., Kulej, K. & Sidoli, S. Guide for protein fold change and p-value calculation for non-experts in proteomics. Mol. Omics 16, 573–582 (2020).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank H. Lickert, K. Yang, S. S. Kim, J. Kazakov and N. Zhang for assisting with additional experiments not included in the manuscript; Z. Bao, S. Chen and L. Studer for insightful discussions; N. Verma and F. C. Pan for critical reading and editing of the manuscript; and M. Ray and B. Jarvis for technical support. This study was supported in part by grants to D.H. from DoD PRMRP (W81XWH-20-1-0298), the NIH (R01DK096239, U01HG012051) and the American Diabetes Association (1-19-IBS-125); grants to C.V.W. from the NIH (UC4DK104211, UC4DK108120) and the Leona M. and Harry B. Helmsley Charitable Trust; grants to J.S.O. and D.H. from JDRF (3-SRA-2021-1060-S-B) and DoD PRMRP (W81XWH-20-1-0670); and the University of Wisconsin-Madison, Office of the Vice Chancellor for Research and Graduate Education with funding from the Wisconsin Alumni Research Foundation. S. Stransky and S. Sidoli were supported by the Leukemia Research Foundation (Hollis Brownstein New Investigator Research Grant), AFAR (Sagol Network GerOmics award), Deerfield (Xseed award) and the NIH Office of the Director (1S10OD030286-01). This study was also supported by a MSKCC Cancer Center Support Grant from the NIH (P30CA008748), a postdoctoral fellowship from a NYSTEM training grant (DOH01-TRAIN3-2015-2016-00006 to D.Y.), NIH T32 Training Grants (T32HD060600 to G.D. and T32GM008539 to S.J.K. and B.P.R.), and a Frank Lappin Horsfall Jr Fellowship (to G.D.).

Author information

Authors and Affiliations

Authors

Contributions

D.Y. and D.H. designed experiments, analysed and interpreted results. D.Y. performed most of the experiments. C.S.L. supervised and H.C., Z.T. and R.K. performed the computational analyses. A.S., Q.V.L., R.L. and Z.-D.S. assisted with the screens. R.L., G.D., B.P.R., S. Stransky and S. Sidoli assisted with scRNA-seq, ChIP-seq and ChIP–MS experiments. C.V.W. and J.S.O. supervised and V.U., D.M.T. and S.D.S. performed immunostaining on human and mouse fetal pancreas tissues. G.D., S.J.K., Z.Z. and B.P.R. assisted with the generation of hESCs lines. J. Park, J. Pulecio, Y.B. and R.E.S. assisted with additional experiments. D.Y. and D.H. wrote the manuscript. All authors provided editorial advice.

Corresponding authors

Correspondence to Christina S. Leslie or Danwei Huangfu.

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. Peer reviewer reports are available.

Additional information

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

Extended data

Extended Data Fig. 1 Analysis of HHEX expression and examination of HHEX KO cells through flow cytometry and RNA-seq.

a, Bar plots for HHEX, PDX1, NKX6-1, and AFP expression in human CS12-14 stages dorsal pancreatic bud (DP), hepatobiliary primordium (HBP), and hepatic cords (HC) based on a published study38. n = 2 independent experiments. The y-axis represents the expression levels RPKM. b, Box plots for Hhex expression in Pdx1 cells sorted from ventral and dorsal pancreatic buds in E9.5 and E10.5 Pdx1-GFP mouse embryos based on a published study43. The y-axis of the box plots represents the expression levels (log2(RPKM + 1)). In each boxplot, the rectangle shows the inter-quartile range (IQR), with the bottom and top hinges representing the 25 and 75 percentiles, respectively. The middle line represents the median. The whiskers extend to the most extreme value within 1.5*IQR above or below the hinges. E9.5 dorsal Pdx1+ cells, n = 31; E10.5 dorsal Pdx1+ cells, n = 27; E9.5 ventral Pdx1+ cells, n = 44; E10.5 ventral Pdx1+ cells, n = 59. E9.5 and E10.5 data are generated from two and three independent mice, respectively. c,d, Flow cytometry gating strategy for HHEX expression. The SSC-A/FSC-A gate identifies cells based on the size and granularity. The FSC-H/FSC-W and SSC-H/SSC-W gates identify single cells. Live-dead staining distinguishes live cells from dead cells (c). HHEX KO cells were used as negative control for WT HHEX expression at the PP1 stage (d). e,f, Flow cytometry analysis of SOX17 and CXCR4 expression at the DE stage (e) and HNF1B expression at the GT stage (f). g,h, Quantification of flow cytometry analysis for SOX17, CXCR4 expression at the DE stage (g) and HNF1B expression at the GT stage (h). n = 3 independent experiments and data are presented as mean ± s.d. i,j, Flow cytometry analysis (i) and quantification (j) of cleaved caspase-3 (C-CSP3) expression at the DE, GT and PP1 stage. n = 3 independent experiments and data are presented as mean ± s.d. k, PCA based on PeakNorm normalization for all WT and KO samples during pancreatic differentiation. Two independent experiments were performed at each stage. Statistical analysis of g, h and j was performed using one-way ANOVA followed by Dunnett multiple comparisons test versus WT control.

Source data

Extended Data Fig. 2 Chromatin accessibility and transcriptional changes upon HHEX deletion.

a,b, Bar graph and volcano plot showing the number (a) and adjusted P value distribution (b) of differential peaks in KO cells compared to WT. Differential ATAC peaks were identified by DESeq2 using default parameters. FDR < 0.05 are counted as one significant peak. Less accessible peaks in KO are marked in blue and more accessible peaks in KO in orange. The number of differential peaks is indicated. c,d, TF motif enrichment in cluster I (c) and II (d) regions. One-sided hypergeometric test was used to compare the enrichment of proportions of TF motifs for each cluster (foreground ratio) versus those for total atlas (background ratio). The horizontal axis shows the binomial Z-score, representing the number of standard deviations between the observed count of each cluster peaks containing a TF motif and the expected count based on the background ratio. The P values are provided in Supplementary Table 3. e, IGV tracks (average of two independent experiments) show chromatin accessibility at representative liver genes loci identified in cluster III. Scale bar, 5 kb. f, Top 7 mouse phenotypes associated with the regulatory regions identified in cluster III. The term of mouse phenotypes was selected based on the rank of binomial test and cut-offs of region fold enrichment >1.4 and observed regions >80. The P values are indicated. g,h, Flow cytometry analysis (g) and quantification (h) of HNF4A+ cells at DE, GT, and PP1 stages. Each symbol represents one independent experiment (n = 4 independent experiments) and data are presented as mean ± s.d. Statistical analysis was performed using unpaired two-tailed Student’s t-test. Data shown in a-f are from two independent experiments.

Source data

Extended Data Fig. 3 The effects of inhibiting liver differentiation on WT and HHEX KO cells.

a,b, Flow cytometry analysis of AFP, PDX1 and CDX2 expression at the PP1 and PP2 stage WT/KO cells using differentiation Condition 1 (a) and Condition 2 (b). c, Quantification of flow cytometry analysis of AFP+, PDX1+ and CDX2 + cells at the PP1 and PP2 stages in both conditions. Each symbol represents one independent experiment (n = 8 independent experiments, except for CDX2 staining, where n = 4 independent experiments) and data are presented as mean ± s.d. Statistical analysis was performed using two-way ANOVA followed by multiple comparisons with Tukey correction. d, Immunostaining images for PDX1, AFP and CDX2 expression at the PP2 stage WT/KO cells using differentiation Condition 2. Images shown represent three independent experiments. Scale bar, 50 μm.

Source data

Extended Data Fig. 4 Investigation of differentiation trajectories in WT and HHEX KO cells through scRNA-seq analysis.

a, UMAP visualization of all Seurat clusters from experiment 2, shown with distinct colors. Clusters 1-15 were annotated as in Fig. 6b. b, UMAP visualization of the integrated data from all samples of both experiments at the DE, GT, PP1, and PP2 stages. The overlapping embedding of DE cells shows that the batch effect was removed. c, WT and KO lineages visualized by forced-directed layouts of the integrated data from the DE, GT, PP1, and PP2 stages. Cells at DE and GT stages are shown here. Data shown in a-c represent one independent experiment.

Extended Data Fig. 5 HHEX and FOXA2 ChIP-MS and ChIP-seq analysis.

a, Venn diagram showing significantly enriched proteins at the GT and PP1 stages for HHEX ChIP-MS. b, GREAT Gene Ontology showing the top 7 biological processes associated with the FOXA2-down regions with the P values indicated. The terms were selected based on the rank from the binomial test and cut-offs of region fold enrichment >1.5 and observed regions >80. c, IGV tracks (average of two independent experiments) showing chromatin accessibility and TFs binding activities at the PDX1 (left) and SOX9 (right) loci in WT and KO cells. Tracks are generated from two independent experiments. Scale bars are indicated. Regions that showed significantly decreased FOXA2 binding upon HHEX deletion are indicated by dashed boxes. d, MA plot showing differential FOXA2 binding (blue) at the PP2 stage following HHEX deletion. The number of significantly increased and decreased FOXA2 binding sites is indicated. e, TF motifs enriched in regions with differential FOXA2 binding upon HHEX deletion. Regions with significantly increased/decreased FOXA2 binding were compared with the FOXA2 ChIP-seq atlas to examine TF motif enrichments using one-sided KS test. The KS test effect size is shown on the y axis, and the proportion of peaks associated with the TF motif is plotted on the x axis. The size of each circle represents the odds ratio, which was defined as the frequency of a given TF motif in regions with increased or decreased FOXA2 binding divided by its frequency in the FOXA2 ChIP-seq atlas. TF motifs with a KS test effect size ≥ 0.1 (indicated by the dashed lines) and odds ratio ≥ 1.2 are shown. f, Volcano plot showing enriched proteins (purple) for FOXA2 ChIP-MS at the PP2 stage in WT versus KO cells. Dotted lines indicate the fold change and significance cut-offs (|log2(FC)| > 1, –log2(P) > 4.32). CDX2 (log2 (FC) = –2.01, P = 0.067) is also indicated. Data shown in a-e are from two independent experiments, and data shown in f represent three independent experiments.

Supplementary information

Reporting Summary

Peer Review File

Supplementary Table 1

MAGeCK analysis of the DE and PP1 screens. Sheet 1, results from MAGeCK analysis of the DE and PP1 screens. Data were analysed with MAGeCK 0.5.9.4 default RRA parameters. Sheet 2, DE screen gRNA raw counts. Sheet 3, PP1 screen gRNA raw counts.

Supplementary Table 2

Results from transcriptome analysis. Sheet 1, all normalized counts. Sheet 2, significant hits at the DE stage. Sheet 3, significant hits at the GT stage. Sheet 4, significant hits at the PP1 stage. A two-tailed Wald test was used for statistical significance and a Benjamini–Hochberg procedure was used to correct for multiple hypothesis testing.

Supplementary Table 3

Results from chromatin-accessibility analysis. Sheet 1, differential peak analysis in KO cells versus WT cells at the GT and PP1 stage. Differential peaks were identified by DESeq2 using default parameters. Sheets 2–4, clusters I, II and III enriched peaks information. Differential peaks were identified by DESeq2 using default parameters, and adjusted P value cut-off is 0.01. Sheets 5–7, hypergeometric test analysis of clusters I, II and III. One-sided hypergeometric test was used to compare the enrichment of proportions of TF motifs for each cluster (foreground ratio) versus those for total atlas (background ratio). Multiple testing correction used Bonferroni. The horizontal axis shows the binomial Z-score, representing the number of standard deviations between the observed count of each cluster peaks containing a TF motif and the expected count based on the background ratio. Analysis is from two independent experiments. BG, background; FG, foreground.

Supplementary Table 4

Results from scRNA-seq analysis. Sheet 1, sample information. Sheet 2, cell meta-data. Sheet 3, differential expression analysis for the annotated PP1/PP2 group cells. Sheet 4, mapping data from mouse endoderm-derived cells to human PP1 and PP2 populations. Differential expression tests, comparing log-normalized gene expression values of each group to the rest, were performed by MAST, and P values were adjusted with Bonferroni multiple testing correction.

Supplementary Table 5

Results from HHEX and FOXA2 ChIP–MS analysis. Sheet 1, HHEX ChIP–MS analysis. HHEX KO cells stained with HHEX antibody was used as negative control. Sheet 2, FOXA2 ChIP–MS analysis on PP1 WT cells. Sheet 3, FOXA2 ChIP–MS analysis on PP1 WT versus KO cells. Sheet 4, FOXA2 ChIP–MS analysis on PP2 WT versus KO cells. Cells stained with IgG was used as negative control for FOXA2 ChIP-MS analysis. Two-tailed heteroscedastic t-test was used for all MS experiments. Sheets 5–7, raw data of HHEX and FOXA2 ChIP–MS experiments.

Supplementary Table 6

Results from FOXA2 and HHEX ChIP-seq analysis. Sheet 1, PP1 FOXA2 atlas analysis. Sheet 2, Identification of FOXA2 and HHEX co-bind ATAC peaks at the GT and PP1 stages. Sheet 3, PP2 FOXA2 atlas analysis. To show co-binding of FOXA2 and HHEX at ATAC-seq peaks, ChIP-seq signal was quantified on the same ATAC-seq-based atlas. Differential ChIP-seq analysis using the default parameters of DESeq2.

Supplementary Table 7

List of gRNA target sequences and primer sequences for PCR genotyping and RT–qPCR.

Supplementary Table 8

List of antibodies.

Source data

Source Data Fig. 3

Statistical source data.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

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.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yang, D., Cho, H., Tayyebi, Z. et al. CRISPR screening uncovers a central requirement for HHEX in pancreatic lineage commitment and plasticity restriction. Nat Cell Biol 24, 1064–1076 (2022). https://doi.org/10.1038/s41556-022-00946-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-022-00946-4

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research