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:

Nucleation of DNA repair factors by FOXA1 links DNA demethylation to transcriptional pioneering

Subjects

Abstract

FOXA1 functions in epigenetic reprogramming and is described as a 'pioneer factor'. However, exactly how FOXA1 achieves these remarkable biological functions is not fully understood. Here we report that FOXA1 associates with DNA repair complexes and is required for genomic targeting of DNA polymerase β (POLB) in human cells. Genome-wide DNA methylomes demonstrate that the FOXA1 DNA repair complex is functionally linked to DNA demethylation in a lineage-specific fashion. Depletion of FOXA1 results in localized reestablishment of methylation in a large portion of FOXA1-bound regions, and the regions with the most consistent hypermethylation exhibit the greatest loss of POLB and are represented by active promoters and enhancers. Consistently, overexpression of FOXA1 commits its binding sites to active DNA demethylation in a POLB-dependent manner. Finally, FOXA1-associated DNA demethylation is tightly coupled with estrogen receptor genomic targeting and estrogen responsiveness. Together, these results link FOXA1-associated DNA demethylation to transcriptional pioneering by FOXA1.

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

Figure 1: Identification of FOXA1-associated proteins.
Figure 2: Characterization of endogenous FOXA1 DNA repair complex.
Figure 3: Hierarchical architecture of the FOXA1 DNA repair complex.
Figure 4: Genomic landscape of the FOXA1 DNA repair complex.
Figure 5: DNA methylation dynamics after FOXA1 depletion.
Figure 6: FOXA1 overexpression evokes POLB-dependent demethylation.
Figure 7: The FOXA1 DNA repair complex in estrogen response.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Referenced accessions

Gene Expression Omnibus

References

  1. Augello, M.A., Hickey, T.E. & Knudsen, K.E. FOXA1: master of steroid receptor function in cancer. EMBO J. 30, 3885–3894 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Katoh, M. & Katoh, M. Human FOX gene family (Review). Int. J. Oncol. 25, 1495–1500 (2004).

    CAS  PubMed  Google Scholar 

  3. Wijchers, P.J., Burbach, J.P. & Smidt, M.P. In control of biology: of mice, men and Foxes. Biochem. J. 397, 233–246 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Carroll, J.S. et al. Chromosome-wide mapping of estrogen receptor binding reveals long-range regulation requiring the forkhead protein FoxA1. Cell 122, 33–43 (2005).

    Article  CAS  PubMed  Google Scholar 

  5. Cirillo, L.A. et al. Opening of compacted chromatin by early developmental transcription factors HNF3 (FoxA) and GATA-4. Mol. Cell 9, 279–289 (2002).

    Article  CAS  PubMed  Google Scholar 

  6. Li, Q. et al. FOXA1 mediates p16INK4a activation during cellular senescence. EMBO J. 32, 858–873 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Zaret, K.S. & Carroll, J.S. Pioneer transcription factors: establishing competence for gene expression. Genes Dev. 25, 2227–2241 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Lupien, M. et al. FoxA1 translates epigenetic signatures into enhancer-driven lineage-specific transcription. Cell 132, 958–970 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Hurtado, A., Holmes, K.A., Ross-Innes, C.S., Schmidt, D. & Carroll, J.S. FOXA1 is a key determinant of estrogen receptor function and endocrine response. Nat. Genet. 43, 27–33 (2011).

    Article  CAS  PubMed  Google Scholar 

  10. Wang, D. et al. Reprogramming transcription by distinct classes of enhancers functionally defined by eRNA. Nature 474, 390–394 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Wang, Q. et al. Androgen receptor regulates a distinct transcription program in androgen-independent prostate cancer. Cell 138, 245–256 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Kong, S.L., Li, G., Loh, S.L., Sung, W.K. & Liu, E.T. Cellular reprogramming by the conjoint action of ERα, FOXA1, and GATA3 to a ligand-inducible growth state. Mol. Syst. Biol. 7, 526 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. 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  PubMed Central  Google Scholar 

  14. Cirillo, L.A. & Zaret, K.S. An early developmental transcription factor complex that is more stable on nucleosome core particles than on free DNA. Mol. Cell 4, 961–969 (1999).

    Article  CAS  PubMed  Google Scholar 

  15. Clark, K.L., Halay, E.D., Lai, E. & Burley, S.K. Co-crystal structure of the HNF-3/fork head DNA-recognition motif resembles histone H5. Nature 364, 412–420 (1993).

    Article  CAS  PubMed  Google Scholar 

  16. Sérandour, A.A. et al. Epigenetic switch involved in activation of pioneer factor FOXA1-dependent enhancers. Genome Res. 21, 555–565 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Bartke, T. et al. Nucleosome-interacting proteins regulated by DNA and histone methylation. Cell 143, 470–484 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Bernstein, B.E., Meissner, A. & Lander, E.S. The mammalian epigenome. Cell 128, 669–681 (2007).

    Article  CAS  PubMed  Google Scholar 

  19. Wu, H. & Zhang, Y. Reversing DNA methylation: mechanisms, genomics, and biological functions. Cell 156, 45–68 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Wu, S.C. & Zhang, Y. Active DNA demethylation: many roads lead to Rome. Nat. Rev. Mol. Cell Biol. 11, 607–620 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Zhu, J.K. Active DNA demethylation mediated by DNA glycosylases. Annu. Rev. Genet. 43, 143–166 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Conticello, S.G. The AID/APOBEC family of nucleic acid mutators. Genome Biol. 9, 229 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Cortellino, S. et al. Thymine DNA glycosylase is essential for active DNA demethylation by linked deamination-base excision repair. Cell 146, 67–79 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Rai, K. et al. DNA demethylation in zebrafish involves the coupling of a deaminase, a glycosylase, and Gadd45. Cell 135, 1201–1212 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Maiti, A. & Drohat, A.C. Thymine DNA glycosylase can rapidly excise 5-formylcytosine and 5-carboxylcytosine: potential implications for active demethylation of CpG sites. J. Biol. Chem. 286, 35334–35338 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Dalton, S.R. & Bellacosa, A. DNA demethylation by TDG. Epigenomics 4, 459–467 (2012).

    Article  CAS  PubMed  Google Scholar 

  27. Shen, L. et al. Genome-wide analysis reveals TET- and TDG-dependent 5-methylcytosine oxidation dynamics. Cell 153, 692–706 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Lloyd, R.S. The initiation of DNA base excision repair of dipyrimidine photoproducts. Prog. Nucleic Acid Res. Mol. Biol. 62, 155–175 (1999).

    Article  CAS  PubMed  Google Scholar 

  29. Schärer, O.D. & Jiricny, J. Recent progress in the biology, chemistry and structural biology of DNA glycosylases. BioEssays 23, 270–281 (2001).

    Article  PubMed  Google Scholar 

  30. Whitehouse, C.J. et al. XRCC1 stimulates human polynucleotide kinase activity at damaged DNA termini and accelerates DNA single-strand break repair. Cell 104, 107–117 (2001).

    Article  CAS  PubMed  Google Scholar 

  31. Ma, Y. et al. A biochemically defined system for mammalian nonhomologous DNA end joining. Mol. Cell 16, 701–713 (2004).

    Article  CAS  PubMed  Google Scholar 

  32. Ma, Y., Pannicke, U., Schwarz, K. & Lieber, M.R. Hairpin opening and overhang processing by an Artemis/DNA-dependent protein kinase complex in nonhomologous end joining and V(D)J recombination. Cell 108, 781–794 (2002).

    Article  CAS  PubMed  Google Scholar 

  33. Burma, S. & Chen, D.J. Role of DNA-PK in the cellular response to DNA double-strand breaks. DNA Repair (Amst.) 3, 909–918 (2004).

    Article  CAS  Google Scholar 

  34. Kubota, Y. et al. Reconstitution of DNA base excision-repair with purified human proteins: interaction between DNA polymerase β and the XRCC1 protein. EMBO J. 15, 6662–6670 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Caldecott, K.W., Aoufouchi, S., Johnson, P. & Shall, S. XRCC1 polypeptide interacts with DNA polymerase β and possibly poly (ADP-ribose) polymerase, and DNA ligase III is a novel molecular 'nick-sensor' in vitro. Nucleic Acids Res. 24, 4387–4394 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Kim, M.Y., Zhang, T. & Kraus, W.L. Poly(ADP-ribosyl)ation by PARP-1: 'PAR-laying' NAD+ into a nuclear signal. Genes Dev. 19, 1951–1967 (2005).

    Article  CAS  PubMed  Google Scholar 

  37. Kim, M.Y., Mauro, S., Gévry, N., Lis, J.T. & Kraus, W.L. NAD+-dependent modulation of chromatin structure and transcription by nucleosome binding properties of PARP-1. Cell 119, 803–814 (2004).

    Article  CAS  PubMed  Google Scholar 

  38. Krishnakumar, R. & Kraus, W.L. PARP-1 regulates chromatin structure and transcription through a KDM5B-dependent pathway. Mol. Cell 39, 736–749 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Ernst, J. & Kellis, M. ChromHMM: automating chromatin-state discovery and characterization. Nat. Methods 9, 215–216 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

  42. Stadler, M.B. et al. DNA-binding factors shape the mouse methylome at distal regulatory regions. Nature 480, 490–495 (2011).

    Article  CAS  PubMed  Google Scholar 

  43. Serandour, A.A., Brown, G.D., Cohen, J.D. & Carroll, J.S. Development of an Illumina-based ChIP–exonuclease method provides insight into FoxA1–DNA binding properties. Genome Biol. 14, R147 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. McLean, C.Y. et al. GREAT improves functional interpretation of cis-regulatory regions. Nat. Biotechnol. 28, 495–501 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Menet, J.S., Pescatore, S. & Rosbash, M. CLOCK:BMAL1 is a pioneer-like transcription factor. Genes Dev. 28, 8–13 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Pihlajamaa, P. et al. Tissue-specific pioneer factors associate with androgen receptor cistromes and transcription programs. EMBO J. 33, 312–326 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Sherwood, R.I. et al. Discovery of directional and nondirectional pioneer transcription factors by modeling DNase profile magnitude and shape. Nat. Biotechnol. 32, 171–178 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Malovannaya, A. et al. Analysis of the human endogenous coregulator complexome. Cell 145, 787–799 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Yu, M. et al. Base-resolution analysis of 5-hydroxymethylcytosine in the mammalian genome. Cell 149, 1368–1380 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Spruijt, C.G. et al. Dynamic readers for 5-(hydroxy)methylcytosine and its oxidized derivatives. Cell 152, 1146–1159 (2013).

    Article  CAS  PubMed  Google Scholar 

  51. Ooi, S.K. & Bestor, T.H. The colorful history of active DNA demethylation. Cell 133, 1145–1148 (2008).

    Article  CAS  PubMed  Google Scholar 

  52. Billaud, M. & Santoro, M. Is co-option a prevailing mechanism during cancer progression? Cancer Res. 71, 6572–6575 (2011).

    Article  CAS  PubMed  Google Scholar 

  53. Ziller, M.J. et al. Charting a dynamic DNA methylation landscape of the human genome. Nature 500, 477–481 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Vizcaíno, J.A. et al. 2016 update of the PRIDE database and its related tools. Nucleic Acids Res. 44, D1, D447–D456 (2016).

    Article  CAS  Google Scholar 

  55. Johnson, W.E. et al. Model-based analysis of tiling-arrays for ChIP-chip. Proc. Natl. Acad. Sci. USA 103, 12457–12462 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Shang, Y., Hu, X., DiRenzo, J., Lazar, M.A. & Brown, M. Cofactor dynamics and sufficiency in estrogen receptor–regulated transcription. Cell 103, 843–852 (2000).

    Article  CAS  PubMed  Google Scholar 

  57. Zhang, H. et al. Differential gene regulation by the SRC family of coactivators. Genes Dev. 18, 1753–1765 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Wang, Y. et al. LSD1 is a subunit of the NuRD complex and targets the metastasis programs in breast cancer. Cell 138, 660–672 (2009).

    Article  CAS  PubMed  Google Scholar 

  59. Frommer, M. et al. A genomic sequencing protocol that yields a positive display of 5-methylcytosine residues in individual DNA strands. Proc. Natl. Acad. Sci. USA 89, 1827–1831 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Schmidt, D. et al. ChIP-seq: using high-throughput sequencing to discover protein–DNA interactions. Methods 48, 240–248 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. 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).

    PubMed  PubMed Central  Google Scholar 

  62. Neph, S. et al. BEDOPS: high-performance genomic feature operations. Bioinformatics 28, 1919–1920 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Xi, Y. & Li, W. BSMAP: whole genome bisulfite sequence MAPping program. BMC Bioinformatics 10, 232 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Lister, R. et al. Human DNA methylomes at base resolution show widespread epigenomic differences. Nature 462, 315–322 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Lawrence, M. et al. Software for computing and annotating genomic ranges. PLoS Comput. Biol. 9, e1003118 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. R Development Core Team. R: A Language and Environment for Statistical Computing (R Foundation for Statistical Computing, 2014).

  67. Wickham, H. ggplot2: Elegant Graphics for Data Analysis (Springer-Verlag, 2009).

  68. Li, W. et al. Functional roles of enhancer RNAs for oestrogen-dependent transcriptional activation. Nature 498, 516–520 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Liu, Z. et al. Enhancer activation requires trans-recruitment of a mega transcription factor complex. Cell 159, 358–373 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by grants (91219201, 81530073, and 81130048 to Y.S. and 81300254 to Y.Z.) from the National Natural Science Foundation of China and a grant (973 Program: 2014CB542004 to J.L.) from the Ministry of Science and Technology of China.

Author information

Authors and Affiliations

Authors

Contributions

Y.S. and Y.Z. conceived the project. Y.S. and Y.Z. conceived and designed the experiments. Y.Z. and D.Z. performed the experiments. Y.Z. performed computational analysis. Y.S. and Y.Z. wrote the manuscript. Y.S., Y.Z., D.Z., J.L., and Q.L. analyzed the data. Y.Z., D.Z., J.L., Q.L., L.S., X.Y., Z.C., R.Y., G.X., S.L., B.X., W.L., L.L., J.Y., and L.H. performed material preparation.

Corresponding author

Correspondence to Yongfeng Shang.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Antibodies used in detection of the components of the FOXA1 DNA repair complex.

Note the lower band in the DNA-PKcs blot possibly resulted from degradation during sample preparation owing to the large size of DNA-PKcs (~460 kDa). Specificity of the major components of the FOXA1 DNA repair complex, including FOXA1, POLB, LIG3, Ku80, and PARP1, was further supported by their specific depletion in the targeting RNAi– transfected cells.

Supplementary Figure 2 Composition of the FOXA1 DNA repair complex.

(a) Left, schematic of FOXA1 mutants. Middle and right the indicated proteins purified from E. coli were analyzed by SDS–PAGE followed by Coomassie blue staining. (b) MCF-7 cells were transfected with siNS or siRNAs targeting FOXA1 DNA repair factors (siFOXA1, siPOLB, siPARP1, siKu80, or siLIG3). The effect of RNAi on the mRNA levels of ERα, FOXA1, and PARP1 was examined by real-time RT–PCR. Data are displayed as means ± s.d. for triplicates (*P < 0.01, one-way ANOVA). (c) FOXA1 has no discernible effects on PARP1 binding. qChIP was performed to examine the binding pattern of FOXA1 and PARP1 at the indicated sites in MCF-7 cells transfected with siNS or siFOXA1. Data are displayed as means ± s.d. for triplicates (*P < 0.01, t test for the qChIP data in siFOXA1-transfected cells versus siNS-transfected cells). enh, enhancer. (d) Whole-cell lysates from MCF-7 cells in the presence or absence of DNase I were subjected to immunoprecipitation with FOXA1 antibodies followed by immunoblotting using antibodies against POLB.

Supplementary Figure 3 The efficiency of RNAi in decreasing the protein expression of the subunits of the FOXA1 DNA repair complex.

MCF-7 cells were transfected with siNS or siRNAs targeting FOXA1 DNA repair factors (siFOXA1, siPOLB, siPARP1, siKu80, or siLIG3). The RNAi effect was examined by immunoblotting.

Supplementary Figure 4 Enrichment of the FOXA1 DNA repair complex at distinct epigenetic states.

Enrichment of the FOXA1 complex at distinct epigenetic states. (a) Chromatin state annotation was produced from ChromHMM using ChIP-seq data on H3K4me1, H3K4me3, H3K9me3, H3K27me3, H3K36me3, and H3K27ac in MCF-7 cells. This model distinguished seven broad classes of chromatin state, referred to as follows: state E1, Polycomb-repressed state enriched for characteristic H3K27me3; state E2, heterochromatin state without enrichment for any of these marks; state E3, repressive state enriched for H3K9me3; state E4, transcribed state enriched with H3K36me3 at gene bodies; state E5, enhancer state strongly enriched for the enhancer markers H3K27ac and H3K4me1; state E6, enhancer state weakly enriched for H3K27ac; state E7, promoter state strongly enriched for H3K4me3 and moderately enriched for H3K27ac. Emission and transition parameters for the multivariate hidden Markov model are displayed as a heat map. (b) The heat map indicates the relative percentage of the genome represented by each chromatin state and the relative fold enrichment of the full set of FOXA1 complex binding sites for the states determined in a. (c) The entire set of FOXA1 complex binding sites were grouped on the basis of their intersection with the epigenomic states determined in a. Average profiles of FOXA1 and POLB in these seven subgroups were plotted.

Supplementary Figure 5 Average ChIP-seq read density profile (log2 transformed) of the indicated proteins at FOXA1-binding sites or p300-associated enhancers without FOXA1 binding under the indicated conditions.

BS, binding sites.

Supplementary Figure 6 Sequential ChIP for the co-binding of the FOXA1 DNA repair complex subunits at control regions.

Data are displayed as means ± s.d. for triplicates.

Supplementary Figure 7 DNA methylation profiles of one locus on chromosome 2 and one locus on chromosome 10.

Tracks of two biological replicates of bisulfate sequencing and ChIP-seq density of FOXA1 are shown.

Supplementary Figure 8 DNA methylation patterns at cell-lineage-specific FOXA1-binding sites.

(a) Cell-lineage-specific FOXA1-binding sites in MCF-7 cells or HepG2 cells or sites shared by the two cell lines are shown. (b) The biolin plot shows the density of average CpG levels at tissue-specific FOXA1-binding sites and control regions. (c) Average profiles of FOXA1 at tissue-specific FOXA1-binding sites and control regions.

Supplementary Figure 9 DNA methylation changes at three clusters.

(ac) The scatterplots of the indicated clusters show mean 5mC abundance in the indicated windows (350 bp) in siNS-transfected MCF-7 cells (x axis) against its counterpart in siFOXA1-transfected cells (y axis). Loci with significant methylation changes in the scatterplot are identified as off-diagonal points. (d) 5mC abundance differences in MCF-7 cells transfected with siFOXA1 versus siNS are measured from each library and plotted as a box plot.

Supplementary Figure 10 DNA methylation changes at the central groups.

(a) The sites from cluster I with a centralized demethylation pattern were further stratified into five subgroups by CpG counts within their LCHs. (b) Average number of CpG dinucleotides in each subgroup of central cluster I. (c) The scatterplots of the indicated subgroups show mean 5mC abundance within the 200-bp window centered on LCH midpoints in siNS-transfected MCF-7 cells (x axis) plotted against its counterpart in siFOXA1-transfected cells (y axis). Loci with significant methylation changes in the scatterplot are identified as off-diagonal points.

Supplementary Figure 11 Multiple-track plots showing profiles of DNA methylation, histone modifications, and ChIP-seq read density of FOXA1 and ERα in the indicated conditions at one locus on chromosome 12 and another locus on chromosome 19.

Methylated CpG differences in MCF-7 cells transfected with siFOXA1 versus siNS were calculated with combined replicated libraries. The densities of H3K4me1, H3K4me3, and H3K27ac signals are shown in a heat map with yellow indicative of strong signals and blue indicative of weak signals.

Supplementary Figure 12 Average profile of the indicated histone modifications at each group.

Groups were determined in Figure 5.

Supplementary Figure 13 Sole knockdown of POLB has no discernible impact on DNA methylation at FOXA1-binding sites in MDA-MB-231 cells.

DNA methylation was examined by bisulfite sequencing of cloned PCR products. The BSP results in MDA-MB-231 cells transfected with siNS or siPOLB together with empty vector are shown as open or closed circles representing unmethylated and methylated CpGs, respectively. The CpG sites within the red boxes highlight the closest CpG dinucleotides surrounding FOXA1 binding motifs as in Figure 6b.

Supplementary Figure 14 DNA methylation and FOXA1 complex binding were examined in FOXA1- or POLB-depleted MCF-7 cells.

(a) DNA methylation levels at the indicated regions (Fig. 7a) were measured by MEDIP-coupled quantitative real-time PCR. Data are displayed as means ± s.d. for triplicates (*P < 0.05, one-way ANOVA). (b) qChIP was performed in MCF-7 cells transfected with siNS, siFOXA1, or siPOLB with antibodies against FOXA1 (top) and POLB (bottom). Data are displayed as means ± s.d. for triplicates (*P < 0.05, one-way ANOVA).

Supplementary Figure 15 Dependency of induced estrogen responsiveness on the FOXA1 DNA repair complex.

MDA-MB-231 cells overexpressing the indicated genes were further challenged with siNS or siPOLB. (a) Expression of these proteins was measured by immunoblotting. (bd) In these cells, we further measured the mRNA levels of pS2 in the presence or absence of estrogen using real-time RT–PCR (b), the time course of ERα and FOXA1 binding at the pS2 promoter using qChIP (c), and the DNA methylation status of a CCGG site in the pS2 promoter using the methods described in Figure 7d (d). Data are displayed as means ± s.d. for triplicates (*P < 0.01, one-way ANOVA).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–15 and Supplementary Tables 1–5. (PDF 2978 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, Y., Zhang, D., Li, Q. et al. Nucleation of DNA repair factors by FOXA1 links DNA demethylation to transcriptional pioneering. Nat Genet 48, 1003–1013 (2016). https://doi.org/10.1038/ng.3635

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3635

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