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:

Pioneer factor Pax7 initiates two-step cell-cycle-dependent chromatin opening

Abstract

Pioneer transcription factors direct cell differentiation by deploying new enhancer repertoires through their unique ability to target and initiate remodelling of closed chromatin. The initial steps of their action remain undefined, although pioneers have been shown to interact with nucleosomal target DNA and with some chromatin-remodeling complexes. We now define the sequence of events that enables the pioneer Pax7 with its unique abilities. Chromatin condensation exerted by linker histone H1 is the first constraint on Pax7 recruitment, and this establishes the initial speed of chromatin remodeling. The first step of pioneer action involves recruitment of the KDM1A (LSD1) H3K9me2 demethylase for removal of this repressive mark, as well as recruitment of the MLL complex for deposition of the activating H3K4me1 mark. Further progression of pioneer action requires passage through cell division, and this involves dissociation of pioneer targets from perinuclear lamin B. Only then are the SWI–SNF remodeling complex and the coactivator p300 recruited, leading to nucleosome displacement and enhancer activation. Thus, the unique features of pioneer actions are those occurring in the lamin-associated compartment of the nucleus. This model is consistent with previous work that showed a dependence on cell division for establishment of new cell fates.

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: Pax7 primes enhancers for activation.
Fig. 2: Pax7-dependent chromatin opening occurs in two steps.
Fig. 3: Initiation of Pax7 recruitment is delayed by histone-H1-dependent chromatin compaction.
Fig. 4: Activation of Pax7-pioneered enhancers requires cell division.
Fig. 5: Pax7-dependent H3K9me2 demethylation and dissociation from lamin B.
Fig. 6: Pax7 initiates domain-wide lamin B dissociation.
Fig. 7: Model of Pax7-initiated chromatin opening and enhancer activation.

Similar content being viewed by others

Data availability

Sequencing data are available on the GEO as GSE87185, GSE125671, GSE225231 and GSE240090. Source data are provided with this paper.

References

  1. Balsalobre, A. & Drouin, J. Pioneer factors as master regulators of the epigenome and cell fate. Nat. Rev. Mol. Cell Biol. 23, 449–464 (2022).

    Article  CAS  PubMed  Google Scholar 

  2. Soufi, A. et al. Pioneer transcription factors target partial DNA motifs on nucleosomes to initiate reprogramming. Cell 161, 555–568 (2015).

    Article  CAS  PubMed  Google Scholar 

  3. Mayran, A. et al. Pioneer factor Pax7 deploys a stable enhancer repertoire for specification of cell fate. Nat. Genet. 50, 259–269 (2018).

    Article  CAS  PubMed  Google Scholar 

  4. Pelletier, A. et al. Pax7 pioneer factor action requires both paired and homeo DNA binding domains. Nucleic Acids Res. 49, 7424–7436 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Minderjahn, J. et al. Mechanisms governing the pioneering and redistribution capabilities of the non-classical pioneer PU.1. Nat. Commun. 11, 402 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Cirillo, L. A. et al. Binding of the winged-helix transcription factor HNF3 to a linker histone site on the nucleosome. EMBO J. 17, 244–254 (1998).

    Article  CAS  PubMed  Google Scholar 

  7. Dodonova, S. O., Zhu, F., Dienemann, C., Taipale, J. & Cramer, P. Nucleosome-bound SOX2 and SOX11 structures elucidate pioneer factor function. Nature 580, 669–672 (2020).

    Article  CAS  PubMed  Google Scholar 

  8. Michael, A. K. et al. Mechanisms of OCT4–SOX2 motif readout on nucleosomes. Science 368, 1460–1465 (2020).

    Article  CAS  PubMed  Google Scholar 

  9. Tanaka, H. et al. Interaction of the pioneer transcription factor GATA3 with nucleosomes. Nat. Commun. 11, 4136 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Iwafuchi-Doi, M. et al. The pioneer transcription factor FoxA maintains an accessible nucleosome configuration at enhancers for tissue-specific gene activation. Mol. Cell 62, 79–91 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Soufi, A., Donahue, G. & Zaret, K. S. Facilitators and impediments of the pluripotency reprogramming factors’ initial engagement with the genome. Cell 151, 994–1004 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Nicetto, D. et al. H3K9me3-heterochromatin loss at protein-coding genes enables developmental lineage specification. Science 363, 294–297 (2019).

    Article  CAS  PubMed  Google Scholar 

  13. Jozwik, K. M., Chernukhin, I., Serandour, A. A., Nagarajan, S. & Carroll, J. S. FOXA1 directs H3K4 monomethylation at enhancers via recruitment of the methyltransferase MLL3. Cell Rep. 17, 2715–2723 (2016).

    Article  CAS  PubMed  Google Scholar 

  14. Kawabe, Y., Wang, Y. X., McKinnell, I. W., Bedford, M. T. & Rudnicki, M. A. Carm1 regulates Pax7 transcriptional activity through MLL1/2 recruitment during asymmetric satellite stem cell divisions. Cell Stem Cell 11, 333–345 (2012).

    Article  CAS  PubMed  Google Scholar 

  15. King, H. W. & Klose, R. J. The pioneer factor OCT4 requires the chromatin remodeller BRG1 to support gene regulatory element function in mouse embryonic stem cells. eLife 6, e22631 (2017).

  16. Takaku, M. et al. GATA3-dependent cellular reprogramming requires activation-domain dependent recruitment of a chromatin remodeler. Genome Biol. 17, 36 (2016).

    Article  PubMed  Google Scholar 

  17. Gao, R. et al. Pioneering function of Isl1 in the epigenetic control of cardiomyocyte cell fate. Cell Res. 29, 486–501 (2019).

    Article  CAS  PubMed  Google Scholar 

  18. Mayran, A., Pelletier, A. & Drouin, J. Pax factors in transcription and epigenetic remodelling. Semin. Cell Dev. Biol. 44, 135–144 (2015).

    Article  CAS  PubMed  Google Scholar 

  19. Budry, L. et al. The selector gene Pax7 dictates alternate pituitary cell fates through its pioneer action on chromatin remodeling. Genes Dev. 26, 2299–2310 (2012).

    Article  CAS  PubMed  Google Scholar 

  20. Mayran, A. et al. Pioneer and nonpioneer factor cooperation drives lineage specific chromatin opening. Nat. Commun. 10, 3807 (2019).

    Article  PubMed  Google Scholar 

  21. Mohammed, H. et al. Rapid immunoprecipitation mass spectrometry of endogenous proteins (RIME) for analysis of chromatin complexes. Nat. Protoc. 11, 316–326 (2016).

    Article  CAS  PubMed  Google Scholar 

  22. Poleshko, A. et al. H3K9me2 orchestrates inheritance of spatial positioning of peripheral heterochromatin through mitosis. eLife 8, e49278 (2019).

  23. Kind, J. et al. Single-cell dynamics of genome-nuclear lamina interactions. Cell 153, 178–192 (2013).

    Article  CAS  PubMed  Google Scholar 

  24. Reyes, J. C., Muchardt, C. & Yaniv, M. Components of the human SWI/SNF complex are enriched in active chromatin and are associated with the nuclear matrix. J. Cell Biol. 137, 263–274 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Shah, P. P. et al. An atlas of lamina-associated chromatin across twelve human cell types reveals an intermediate chromatin subtype. Genome Biol. 24, 16 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Hansen, J. L., Loell, K. J. & Cohen, B. A. A test of the pioneer factor hypothesis using ectopic liver gene activation. eLife 11, e73358 (2022).

  27. Blassberg, R. et al. Sox2 levels regulate the chromatin occupancy of WNT mediators in epiblast progenitors responsible for vertebrate body formation. Nat. Cell Biol. 24, 633–644 (2022).

    Article  CAS  PubMed  Google Scholar 

  28. Bascunana, V. et al. Chromatin opening ability of pioneer factor Pax7 depends on unique isoform and C-terminal domain. Nucleic Acids Res 51, 7254–7268 (2023).

    Article  CAS  PubMed  Google Scholar 

  29. Brennan, K. J. et al. Chromatin accessibility is a two-tier process regulated by transcription factor pioneering and enhancer activation. Preprint at bioRxiv https://doi.org/10.1101/2022.12.20.520743 (2022).

  30. Frederick, M. A. et al. A pioneer factor locally opens compacted chromatin to enable targeted ATP-dependent nucleosome remodeling. Nat. Struct. Mol. Biol. 30, 31–37 (2023).

    Article  CAS  PubMed  Google Scholar 

  31. Heslop, J. A., Pournasr, B., Liu, J. T. & Duncan, S. A. GATA6 defines endoderm fate by controlling chromatin accessibility during differentiation of human-induced pluripotent stem cells. Cell Rep. 35, 109145 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Ismail, T. et al. KDM1A microenvironment, its oncogenic potential, and therapeutic significance. Epigenetics Chromatin 11, 33 (2018).

    Article  PubMed  Google Scholar 

  33. Poleshko, A. et al. Genome-nuclear lamina interactions regulate cardiac stem cell lineage restriction. Cell 171, 573–587(2017).

    Article  CAS  PubMed  Google Scholar 

  34. Donaghey, J. et al. Genetic determinants and epigenetic effects of pioneer-factor occupancy. Nat. Genet. 50, 250–258 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Sardina, J. L. et al. Transcription factors drive Tet2-mediated enhancer demethylation to reprogram cell fate. Cell Stem Cell 23, 727–741(2018).

    Article  CAS  PubMed  Google Scholar 

  36. Stadhouders, R. et al. Transcription factors orchestrate dynamic interplay between genome topology and gene regulation during cell reprogramming. Nat. Genet. 50, 238–249 (2018).

    Article  CAS  PubMed  Google Scholar 

  37. Jiang, Y. et al. The methyltransferase SETDB1 regulates a large neuron-specific topological chromatin domain. Nat. Genet. 49, 1239–1250 (2017).

    Article  CAS  PubMed  Google Scholar 

  38. Langlais, D., Couture, C. & Drouin, J. The Stat3/GR interaction code: predictive value of direct/indirect DNA recruitment for transcription outcome. Mol. Cell 47, 38–49 (2012).

    Article  CAS  PubMed  Google Scholar 

  39. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S. L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  41. Ramírez, F. et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 44, W160–W165 (2016).

    Article  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  43. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are very grateful to A. Shilatifard (Northwestern University) for the MLL3 and MLL4 antibodies, to O. Neyret and S. Boisset for next-generation sequencing analyses, and to V. Magoon for her expert secretarial assistance. Data analyses were possible thanks to the support of Compute Canada. This work was supported by Foundation grant FDN-154297 to J.D. from the Canadian Institutes of Health Research. The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

A.G. and J.D. conceived the study; A.G., A.B. and J.D. conceived and designed the experiments; A.G., J.D.-G., V.L.-R. and J.H. performed experiments; A.G., V.L.-R. and J.D. wrote the paper.

Corresponding author

Correspondence to Jacques Drouin.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Structural & Molecular Biology thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling Editors: Carolina Perdigoto and Dimitris Typas, in collaboration with the Nature Structural & Molecular Biology team.

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 Genome-wide effect of Pax7.

a, Pax7 western blot analysis of total cell extracts from neurointermediate lobe (IL) pituitary cells (~50 % melanotropes), AtT20 cells and AtT20-ERPax7 cells showing ectopic expression of ERPax7 at physiological levels (n = 3). b, H3K4me1 deposition (ChIP-seq) and DNA accessibility (ATAC-seq) at Pax7 binding sites before (0 h) and after 48 h of Pax7 activation. Bottom plots show selection threshold for constitutively active (n = 20,775), pioneered (n = 248) and primed (n = 2,023) enhancers bound by Pax7. c, Boxplots of Pax7 recruitment, H3K4me1 deposition and ATAC signals at constitutively active and pioneered enhancers as defined in (b) as well as at random sites from no to 72 h of Pax7 activation and with stable Pax7 expression. ChIP-seq and ATAC-seq data, p-values from two-tailed t-tests. d, Frequency per bin of Pax7 recruitment (ChIP-seq) at pioneered enhancers binned according to signal strength. The randomly selected n = 14 pioneered enhancers used in the qPCR panel (Fig. 1d) are indicated by vertical dashed bars. e, t1/2 of H3K4me1 deposition at the n = 14 pioneered enhancers computed from time courses in Fig. 1e. For each site, H3K4me1 was measured either at the Pax7 binding site (centered) or shifted towards one of the lateral H3K4me1 deposition summits. ChIP-qPCR data from n ≥ 2 biological replicates.

Source data

Extended Data Fig. 2 Marker enrichment at pioneered enhancers.

Enrichment of chromatin and enhancer markers at the n = 14 pioneered enhancers (Pio, Fig. 1d) as well as constitutively active enhancers (Con) and random sites (Rnd) used as controls at no (blue) or 72 h (red) of Pax7 activation. Enrichments are expressed relative to mean enrichment at random loci (for Pax7 data) or constitutively active enhancers (all others). ChIP-qPCR data expressed as median ± min and max values of n ≥ 2 technical replicates per site. One of n ≥ 2 biological replicates is shown for each marker. Boxplots at right show the enrichment (1-2.5-5-7.5-9 deciles) at the n = 14 pioneered enhancers and the p-values (two-tailed paired t-test) compare enrichments at no vs. 72 h of Pax7 activation.

Source data

Extended Data Fig. 3 Variations in Pax7 pioneering kinetics and initial chromatin state.

a, b, Mean (± standard error of the mean) time courses of mark changes at the stronger and weaker third Pax7 recruitment subsets (n = 4 each) of the n = 14 pioneered enhancers described in Fig. 1d. Each data point was determined by ChIP-qPCR from n ≥ 2 biological replicates. The sequence of events is preserved but a delay of ~6 h in markers t1/2 is observed for the weaker compared to stronger sites. c, H3K4me1 deposition time courses at genome-wide (ChIP-seq) pioneered enhancers (n = 248, Extended Data Fig. 1b) color-coded according to Pax7 recruitment at 1 h and 48 h. d, Correlations between the indicated parameters at the n = 14 pioneered enhancers. Significant correlations are observed between the initial H1 levels, the initial Pax7 recruitment (1 h) up to 48 h and the overall speed of the pioneering process. Theses correlations are lost for long Pax7 activation (> 20 days) or for stably expressed Pax7 suggesting that pioneered enhancers ultimately reach a similar final state. No correlation is observed with the initial levels of H3K9me2 or total histone H3. ChIP-seq data and ChIP-qPCR data from n ≥ 2 biological replicates, p-values from two-tailed t-tests.

Extended Data Fig. 4 Sequential Pax7 recruitment and chromatin opening at pioneered enhancers.

Pax7 ChIP-seq and ATAC-seq profiles at the panel of n = 14 pioneered enhancers specified in Fig. 1d at the indicated times after Pax7 activation or after stable Pax7 expression.

Extended Data Fig. 5 Characterization of mimosine-arrested cells.

a, FACS profiles of AtT20 cells treated with mimosine. Cells arrested in G1 by 24 h mimosine treatment are viable and re-enter cell cycle following release from mimosine-induced cell cycle arrest. One of n = 2 biological replicates is shown. b, p300, Brg1 and Tpit western blots in nuclear extracts of normal and cells treated for 60 h with mimosine. The treatment does not affect p300 or Brg1 levels but results in a two-fold decrease of Tpit expression. n = 3 biological replicates are shown. c, FACS data from control plot in (a) exemplifying the gating strategy used to produce FACS profiles of propidium iodide treated cells.

Source data

Extended Data Fig. 6 Pioneering assessment in mimosine-arrested AtT20 cells.

Enrichment of chromatin and enhancer marks at the n = 14 pioneered enhancers (Pio, Fig. 1d) as well as at constitutively active enhancers (Con) and random sites (Rnd) used as controls at no (blue) or 48 h (red) of Pax7 activation in normal and mimosine-arrested cells. Enrichments are expressed relative to mean enrichment at random loci (for Pax7 data) or constitutively active enhancers (all others). ChIP-qPCR data expressed as median ± min and max values of n ≥ 2 technical replicates per site. One of n ≥ 2 biological replicates is shown for each marker. Boxplots at right show the enrichment (1-2.5-5-7.5-9 deciles) at the n = 14 pioneered enhancers and the p-values (two-tailed paired t-test) compare enrichments at no vs. 48 h of Pax7 activation.

Extended Data Fig. 7 Time course of KDM3A recruitment and further characterization of Pax7-primed enhancers.

a, Mean time courses of KDM3A compared to Pax7 recruitment at the n = 14 pioneered enhancers described in Fig. 1d (± standard error of the mean). Each data point was determined by ChIP-qPCR from n ≥ 2 biological replicates. b, Heatmaps (ChIP-seq) of the indicated markers at multiple subsets of Pax7-targeted sites with or without stable Pax7 expression. 4 kb window centered on Pax7 peak.

Supplementary information

Reporting Summary

Supplementary Tables 1–3

Loci and primers, genome-wide sequencing datasets and antibodies.

Source data

Source Data Fig. 2

Time course qPCR data.

Source Data Fig. 4

Cell cycle arrest qPCR data.

Source Data Fig. 5

KDM1A inhibition qPCR data.

Source Data Fig. 6

Domain-wide qPCR data.

Source Data Extended Data Fig. 1

Unprocessed western blots.

Source Data Extended Data Fig. 2

Markers enrichment qPCR data.

Source Data Extended Data Fig. 5

Unprocessed western blots.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gouhier, A., Dumoulin-Gagnon, J., Lapointe-Roberge, V. et al. Pioneer factor Pax7 initiates two-step cell-cycle-dependent chromatin opening. Nat Struct Mol Biol 31, 92–101 (2024). https://doi.org/10.1038/s41594-023-01152-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-023-01152-y

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