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:

Gibbin mesodermal regulation patterns epithelial development

Abstract

Proper ectodermal patterning during human development requires previously identified transcription factors such as GATA3 and p63, as well as positional signalling from regional mesoderm1,2,3,4,5,6. However, the mechanism by which ectoderm and mesoderm factors act to stably pattern gene expression and lineage commitment remains unclear. Here we identify the protein Gibbin, encoded by the Xia–Gibbs AT-hook DNA-binding-motif-containing 1 (AHDC1) disease gene7,8,9, as a key regulator of early epithelial morphogenesis. We find that enhancer- or promoter-bound Gibbin interacts with dozens of sequence-specific zinc-finger transcription factors and methyl-CpG-binding proteins to regulate the expression of mesoderm genes. The loss of Gibbin causes an increase in DNA methylation at GATA3-dependent mesodermal genes, resulting in a loss of signalling between developing dermal and epidermal cell types. Notably, Gibbin-mutant human embryonic stem-cell-derived skin organoids lack dermal maturation, resulting in p63-expressing basal cells that possess defective keratinocyte stratification. In vivo chimeric CRISPR mouse mutants reveal a spectrum of Gibbin-dependent developmental patterning defects affecting craniofacial structure, abdominal wall closure and epidermal stratification that mirror patient phenotypes. Our results indicate that the patterning phenotypes seen in Xia–Gibbs and related syndromes derive from abnormal mesoderm maturation as a result of gene-specific DNA methylation decisions.

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: Gibbin regulates mesoderm gene expression during development.
Fig. 2: Gibbin interacts with zinc-finger transcription factors and DNA methylation regulators.
Fig. 3: Loss of Gibbin causes DNA hypermethylation and decreased CTCF deposition.
Fig. 4: Gibbin regulates mesoderm maturation to pattern skin development.
Fig. 5: Loss of Gibbin in vivo causes global development defects.

Similar content being viewed by others

Data availability

Deep-sequencing and array data generated in this paper have been deposited at the Gene Expression Omnibus (GEO: GSE180495). Previously published datasets analysed in this study are available at the GEO (GSE114846) or through ENCODE (https://www.encodeproject.org/experiments/ENCSR168AUX/). Raw data from BASU experiments are provided in Supplementary Table 1. Uncropped immunoblots and FACS gating controls are provided in Supplementary Figs. 1 and 2. The human reference genome hg38 was downloaded from the UCSC genome browser. The GRCh38 reference genome for single-cell mapping can be downloaded from 10x genomics (https://support.10xgenomics.com/single-cell-gene-expression/software/downloads/latest). There are no restrictions on data availability.

Code availability

The manifest and annotation files for methylation analysis are available at GitHub (https://github.com/achilleasNP/IlluminaHumanMethylationEPICanno.ilm10b5.hg38). Previously published custom code used in this study is available at GitHub (https://github.com/OroLabStanford).

References

  1. Abe, M. et al. GATA3 is essential for separating patterning domains during facial morphogenesis. Development 148, dev199534 (2021).

    Article  CAS  PubMed  Google Scholar 

  2. Tsarovina, K. et al. Essential role of Gata transcription factors in sympathetic neuron development. Development 131, 4775–4786 (2004).

    Article  CAS  PubMed  Google Scholar 

  3. Ralston, A. et al. Gata3 regulates trophoblast development downstream of Tead4 and in parallel to Cdx2. Development 137, 395–403 (2010).

    Article  CAS  PubMed  Google Scholar 

  4. Romano, R. A. et al. ΔNp63 knockout mice reveal its indispensable role as a master regulator of epithelial development and differentiation. Development 139, 772–782 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Pattison, J. M. et al. Retinoic acid and BMP4 cooperate with p63 to alter chromatin dynamics during surface epithelial commitment. Nat. Genet. 50, 1658–1665 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Chikh, A. et al. Expression of GATA-3 in epidermis and hair follicle: relationship to p63. Biochem. Biophys. Res. Commun. 361, 1–6 (2007).

    Article  CAS  PubMed  Google Scholar 

  7. Ellis, C., Pai, G. S. & Wine Lee, L. Atypical aplasia cutis in association with Xia Gibbs syndrome. Pediatr. Dermatol. 38, 533–535 (2021).

    Article  PubMed  Google Scholar 

  8. Jiang, Y. et al. The phenotypic spectrum of Xia-Gibbs syndrome. Am. J. Med. Genet. A 176, 1315–1326 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Ritter, A. L. et al. Variable clinical manifestations of Xia-Gibbs syndrome: findings of consecutively identified cases at a single children’s hospital. Am. J. Med. Genet. A 176, 1890–1896 (2018).

    Article  CAS  PubMed  Google Scholar 

  10. Tchieu, J. et al. A modular platform for differentiation of human PSCs into all major ectodermal lineages. Cell Stem Cell 21, 399–410 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Liang, Y. C. et al. Folding keratin gene clusters during skin regional specification. Dev. Cell 53, 561–576 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Kelly, O. G. & Melton, D. A. Induction and patterning of the vertebrate nervous system. Trends Genet. 11, 273–278 (1995).

    Article  CAS  PubMed  Google Scholar 

  13. Liem, K. F., Tremml, G., Roelink, H. & Jessell, T. M. Dorsal differentiation of neural plate cells induced by BMP-mediated signals from epidermal ectoderm. Cell 82, 969–979 (1995).

    Article  CAS  PubMed  Google Scholar 

  14. Larsen, W. J. & Sherman, L. S. in Human Embryology 3rd edn 85–102; 126–130 (Churchill Livingstone, 2002).

  15. Hota, S. K. & Bruneau, B. G. ATP-dependent chromatin remodeling during mammalian development. Development 143, 2882–2897 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Pauli, A., Rinn, J. L. & Schier, A. F. Non-coding RNAs as regulators of embryogenesis. Nat. Rev. Genet. 12, 136–149 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Liu, J. et al. Transcriptional dysregulation in NIPBL and cohesin mutant human cells. PLoS Biol. 7, e1000119 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Li, L. et al. TFAP2C- and p63-dependent networks sequentially rearrange chromatin landscapes to drive human epidermal lineage commitment. Cell Stem Cell 24, 271–284 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Li, W. & Cornell, R. A. Redundant activities of Tfap2a and Tfap2c are required for neural crest induction and development of other non-neural ectoderm derivatives in zebrafish embryos. Dev. Biol. 304, 338–354 (2007).

    Article  CAS  PubMed  Google Scholar 

  20. Sebastiano, V. et al. Human COL7A1-corrected induced pluripotent stem cells for the treatment of recessive dystrophic epidermolysis bullosa. Sci. Transl. Med. 6, 264ra163 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Chahrour, M. & Zoghbi, H. Y. The story of Rett syndrome: from clinic to neurobiology. Neuron 56, 422–437 (2007).

    Article  CAS  PubMed  Google Scholar 

  22. Sarogni, P., Pallotta, M. M. & Musio, A. Cornelia de Lange syndrome: from molecular diagnosis to therapeutic approach. J. Med. Genet. 57, 289–295 (2020).

    Article  CAS  PubMed  Google Scholar 

  23. Ostapcuk, V. et al. Activity-dependent neuroprotective protein recruits HP1 and CHD4 to control lineage-specifying genes. Nature 557, 739–743 (2018).

    Article  CAS  PubMed  ADS  Google Scholar 

  24. Xia, F. et al. De novo truncating mutations in AHDC1 in individuals with syndromic expressive language delay, hypotonia, and sleep apnea. Am. J. Hum. Genet. 94, 784–789 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Díaz-Ordoñez, L., Ramirez-Montaño, D., Candelo, E., Cruz, S. & Pachajoa, H. Syndromic intellectual disability caused by a novel truncating variant in AHDC1: a case report. Iran. J. Med. Sci. 44, 257–261 (2019).

    PubMed  Google Scholar 

  26. Savic, D. et al. CETCh-seq: CRISPR epitope tagging ChIP-seq of DNA-binding proteins. Genome Res. 25, 1581–1589 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Mumbach, M. R. et al. HiChIP: efficient and sensitive analysis of protein-directed genome architecture. Nat. Methods 13, 919–922 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Ramanathan, M. et al. RNA-protein interaction detection in living cells. Nat. Methods 15, 207–212 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Roux, K. J., Kim, D. I., Raida, M. & Burke, B. A promiscuous biotin ligase fusion protein identifies proximal and interacting proteins in mammalian cells. J. Cell Biol. 196, 801–810 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Roux, K. J., Kim, D. I. & Burke, B. BioID: a screen for protein-protein interactions. Curr. Protoc. Protein Sci. 2013, 19.23.1–19.23.14 (2013).

    Google Scholar 

  31. Villaseñor, R. et al. ChromID identifies the protein interactome at chromatin marks. Nat. Biotechnol. 38, 728–736 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Saksouk, N. et al. Redundant mechanisms to form silent chromatin at pericentromeric regions rely on BEND3 and DNA methylation. Mol. Cell 56, 580–594 (2014).

    Article  CAS  PubMed  Google Scholar 

  33. Kaaij, L. J. T., Mohn, F., van der Weide, R. H., de Wit, E. & Bühler, M. The ChAHP complex counteracts chromatin looping at CTCF sites that emerged from SINE expansions in mouse. Cell 178, 1437–1451 (2019).

    Article  CAS  PubMed  Google Scholar 

  34. Maurano, M. T. et al. Role of DNA methylation in modulating transcription factor occupancy. Cell Rep. 12, 1184–1195 (2015).

    Article  CAS  PubMed  Google Scholar 

  35. Wang, H. et al. Widespread plasticity in CTCF occupancy linked to DNA methylation. Genome Res. 22, 1680–1688 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Lu, M. F., Pressman, C., Dyer, R., Johnson, R. L. & Martin, J. F. Function of Rieger syndrome gene in left-right asymmetry and craniofacial development. Nature 401, 276–278 (1999).

    Article  CAS  PubMed  ADS  Google Scholar 

  37. Günschmann, C. et al. Insulin/IGF-1 controls epidermal morphogenesis via regulation of FoxO-mediated p63 inhibition. Dev. Cell 26, 176–187 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Li, A. et al. Deciphering principles of morphogenesis from temporal and spatial patterns on the integument. Dev. Dyn. 244, 905–920 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Wolpert, L. Positional information and the spatial pattern of cellular differentiation. J. Theor. Biol. 25, 1–47 (1969).

    Article  CAS  PubMed  ADS  Google Scholar 

  40. Jin, S. et al. Inference and analysis of cell-cell communication using CellChat. Nat. Commun. 12, 1088 (2021).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  41. Niessen, M. T., Iden, S. & Niessen, C. M. The in vivo function of mammalian cell and tissue polarity regulators—how to shape and maintain the epidermal barrier. J. Cell Sci. 125, 3501–3510 (2012).

    Article  CAS  PubMed  Google Scholar 

  42. Delaney, C. et al. Combinatorial prediction of marker panels from single‐cell transcriptomic data. Mol. Syst. Biol. 15, e9005 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Kurek, D., Garinis, G. A., van Doorninck, J. H., van der Wees, J. & Grosveld, F. G. Transcriptome and phenotypic analysis reveals Gata3-dependent signalling pathways in murine hair follicles. Development 134, 261–272 (2007).

    Article  CAS  PubMed  Google Scholar 

  44. Kaufman, C. K. et al. GATA-3: An unexpected regulator of cell lineage determination in skin. Genes Dev. 17, 2108–2122 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Bardhan, T. et al. Gata3 is required for the functional maturation of inner hair cells and their innervation in the mouse cochlea. J. Physiol. 597, 3389–3406 (2019).

    Article  CAS  PubMed  Google Scholar 

  46. Koch, P. J. et al. Targeted disruption of the pemphigus vulgaris antigen (desmoglein 3) gene in mice causes loss of keratinocyte cell adhesion with a phenotype similar to pemphigus vulgaris. J. Cell Biol. 137, 1091–1102 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Cheng, X. et al. Two Chinese Xia-Gibbs syndrome patients with partial growth hormone deficiency. Mol. Genet. Genomic Med. 7, e00596 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Yang, S. et al. Rare mutations in AHDC1 in patients with obstructive sleep apnea. Biomed. Res. Int. 2019, 5907361 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  49. García-Acero, M. & Acosta, J. Whole-exome sequencing identifies a de novo AHDC1 mutation in a Colombian patient with Xia-Gibbs syndrome. Mol. Syndromol. 8, 308–312 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Qin, Y., Yang, S., Li, K. & Wei, Y. Extreme trait next generation sequencing identifies AHDC1 as a novel candidate gene in obstructive sleep apnea. Sleep 41, A8–A9 (2018).

    Article  Google Scholar 

  51. Cardoso-Dos-Santos, A. C. et al. Novel AHDC1 gene mutation in a Brazilian individual: implications of Xia-Gibbs syndrome. Mol. Syndromol. 11, 24–29 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Billingham, R. E. & Silvers, W. K. Studies on the conservation of epidermal specificies of skin and certain mucosas in adult mammals. J. Exp. Med. 125, 429–446 (1967).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Dhouailly, D., Prin, F., Kanzler, B., Viallet, J. P. & Chuong, C. Molecular Basis of Epithelial Appendage Morphogenesis (Landes Biosciences, 1998).

  54. Wu, H. J. et al. Estrogen modulates mesenchyme-epidermis interactions in the adult nipple. Development 144, 1498–1509 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Helsmoortel, C. et al. A SWI/SNF-related autism syndrome caused by de novo mutations in ADNP. Nat. Genet. 46, 380–384 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Khayat, M. M. et al. AHDC1 missense mutations in Xia-Gibbs syndrome. Hum. Genet. Genomics Adv. 2, 100049 (2021).

    Article  Google Scholar 

  57. 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. 2015, 21.29.1–21.29.9 (2015).

    Google Scholar 

  58. Chen, E. Y. et al. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinform. 14, 128 (2013).

    Article  Google Scholar 

  59. 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  PubMed Central  Google Scholar 

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

    Article  MathSciNet  MATH  Google Scholar 

  61. Whyte, W. A. et al. Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 153, 307–319 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Mellacheruvu, D. et al. The CRAPome: a contaminant repository for affinity purification-mass spectrometry data. Nat. Methods 10, 730–736 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Choi, H. et al. SAINT: probabilistic scoring of affinity purificationg-mass spectrometry data. Nat. Methods 8, 70–73 (2011).

    Article  CAS  PubMed  Google Scholar 

  64. 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 

  65. Haarhuis, J. H. I. et al. The cohesin release factor WAPL restricts chromatin loop extension. Cell 169, 693–707 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Bhattacharyya, S., Chandra, V., Vijayanand, P. & Ay, F. Identification of significant chromatin contacts from HiChIP data by FitHiChIP. Nat. Commun. 10, 4221 (2019).

    Article  PubMed  PubMed Central  ADS  CAS  Google Scholar 

  67. Ernst, J. & Kellis, M. Chromatin-state discovery and genome annotation with ChromHMM. Nat. Protoc. 12, 2478–2492 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the members of the Oro laboratory for comments on this manuscript; S. Yamanaka for sharing the PiggyBac inducible expression plasmid and P. Khavari for sharing the BASU cDNA. This work was supported by R. L. Kirchstein NRSA (F32AR074221 to A.C.), the Stanford Dean’s Fellowship (to A.C.), the Stanford Maternal and Childhood Health Research Institute (to A.C.), NIH (R01ARO73170 to A.O.) and the California Institute of Regenerative Medicine (RT3-07796 to A.O.). Mass spectrometry was performed at the Vincent Coates Foundation Mass Spectrometry Laboratory, Stanford University Mass Spectrometry and supported in part by NIH P30 CA124435 using the Stanford Cancer Institute Proteomics/Mass Spectrometry Shared Resource. FACS analysis was performed at the Stanford Shared FACS Facility using an instrument obtained with an NIH S10 Shared Instrument Grant (S10RR025518-01).

Author information

Authors and Affiliations

Authors

Contributions

A.C. and A.O. conceptualized the study and prepared the manuscript. A.C. generated all GKO and GATA3-knockout lines, performed single-cell and bulk RNA-seq, cohesin HiChIP, ATAC-seq, BASU, FACS, IF staining, immunoblotting, qPCR and methylation arrays. A.L. performed Gibbin ChIP–seq and FACS. J.T. assisted with the creation of BASU lines and performed keratinocyte sorting with K.M. J.P. generated the AP2a knockout line and performed RNA-seq. H.Z. and T.P. performed mouse grafting and assisted with embryo extraction and phenotype assessment. H.Z. performed 3D organoid and imaging experiments. H.G. performed DNA methylation colorimetric and enzyme assays. A.C. performed all bioinformatic analyses with assistance from S.G. and A.L.

Corresponding author

Correspondence to Anthony E. Oro.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature thanks Ya-Chieh Hsu and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 Gibbin regulates mesoderm gene expression in response to RA/BMP4.

(a) Genotyping evidence from two different GKO hES cell clones. (b) Gene expression comparing WT to each GKO clone. Clone 2 was used for the remainder of the study. Error bars are mean +/− s.d., n = 4 biologically independent samples averaged from 2 technical replicates repeated across 2 different runs. * Indicates p-value < 0.05 calculated by two-tailed t-test. (c) Proportions of Gibbin-regulated genes that are also altered by RA/BMP4. (d) Representative RPKM values from RNA-seq differential expression after 7 days RA/BMP4. Error bars are mean +/− s.d., n = 2 biologically independent samples, adjusted p-values were measured by DESeq2. (e) Expression patterns of ectoderm (purple, red) or mesoderm (blue) transcripts after RA/BMP4 treatment. (f) UMAP of PDGFRA or (g) EPCAM expression at day 7, marking the mesoderm or ectoderm clusters, respectively. (h) Representative expression plots and adjusted p-values from differential expression testing of scRNA-seq subsets as measured by DESeq2. (i) UMAP of hESCs after 3 days of RA/BMP4. (j) Integrated UMAP and (k) cluster quantification comparing WT and GKO after 3 days of RA/BMP4.

Extended Data Fig. 2 Gibbin ChIP–seq QC.

(a) Western blot of doxycycline inducible HA-Gibbin used for ChIP–seq. Immunoblotting was repeated experimentally twice and GAPDH was run on the same gel as a loading control. For gel source data, see Supplementary Figure 1. (b) Gibbin ChIP–seq binding profile in hESCs or after 7 days of RA/BMP4 at Gibbin binding sites or (c) at all gene transcription start sites. n = 2 biologically independent samples. (d) Representative Gibbin ChIP–seq tracks at example target genes. (e) Differential expression analysis from untreated hESC RNA-seq between WT and GKO cells. Gene expression is either unchanged (grey), increased (blue, >2-fold), or decreased (red, <2-fold), with an adjusted p-value cutoff < 0.05 as measured by DESeq2. n = 2 biologically independent samples. (f) Number of Gibbin-regulated genes with a nearby Gibbin ChIP–seq peak and number of mesoderm, ectoderm, or neuroectoderm genes (identified by scRNA-seq) that are bound by Gibbin. (g) Expression of Gibbin-bound genes across scRNA-seq clusters. (h) ChromHMM analysis of Gibbin binding sites in HEPG2 cells (from ENCODE26), showing enrichment of various chromatin states in relation to Gibbin ChIP–seq peaks. (i) Numbers of peaks and (j) overlap of genes identified in our Gibbin ChIP–seq dataset or the ENCODE HEPG2 dataset. (k) GO term enrichment for direct, Gibbin-bound target genes as measured using a two-sided Fisher’s Exact Test (via EnrichR).

Extended Data Fig. 3 Gibbin and GATA3 are co-expressed.

(a) Principal component analysis (PCA) of RNA-seq from WT, GKO, TFAP2A KO, GATA3 KO, and p63 KO cells after 7 days of RA/BMP4 treatment. n = 2 biologically independent samples. (b) Violin plot of AHDC1/Gibbin or (b) GATA3 expression in each scRNA-seq cluster after 7 days of RA/BMP4. (c) Percent of cells after 7 days of RA/BMP4 which express AHDC1/Gibbin and/or GATA3. (d) qPCR of transcripts induced by GATA3 overexpression in WT or GKO backgrounds. Error bars are mean +/− s.d., n = 3 biologically independent samples averaged from 2 technical replicates. * indicates p-value < 0.05 by two-tailed Student’s t-test. (e) ChromHMM analysis of GATA3 binding sites showing enrichment of various chromatin states in relation to GATA3 ChIP–seq peaks. (f) Average distances between Gibbin and GATA3 ChIP–seq peaks.

Extended Data Fig. 4 Gibbin does not alter chromatin accessibility or GATA3 DNA binding.

(a) ATAC-seq signal heatmaps in WT or GKO cells at all gene transcription start sites, or those subset by mesoderm, ectoderm, or neuroectoderm. n = 2 biologically independent samples. (b) ATAC-seq differential expression analysis comparing WT to Gibbin or (c) GATA3 KO cells after 7 days of RA/BMP4. Differential chromatin accessibility is either unchanged (grey), increased (blue, >2-fold), or decreased (red, <2-fold), with an adjusted p-value cutoff <0.05 measured by DESeq2. (d) Chromatin accessibility at GATA3 binding sites in WT or GKO cells. (e) RPKM values for GATA3 or AHDC1/Gibbin in reciprocal mutant cell lines. Error bars are mean +/− s.d., n = 2 biologically independent samples, adjusted p-value <0.05 as measured by DESeq2. (f) Known motif enrichment at GATA3 ChIP–seq peaks in WT and GKO cells, calculated by Homer. (g) GATA3 ChIP–seq differential binding analysis comparing WT and GKO cells. GATA3 DNA binding is either unchanged (grey), increased (blue, >2-fold), or decreased (red, <2-fold), with an adjusted p-value cutoff <0.05 as measured by DESeq2.

Extended Data Fig. 5 Gibbin regulates gene expression by maintaining local enhancer–promoter chromatin architecture.

(a) Sequencing depth, PCR duplicates, and (b) valid interaction characterization of cohesin HiChIP libraries. (c) Correlation of insulation scores between WT and GKO. (d) A and B compartment scores for chromosome 1, which are representative of the entire genome. (e) Contact probability relative to contact distance for each cell type. (f) Length of contacts called by FitHiChIP in each cell type. (g) Chromatin contact matrix comparing WT and GKO cohesin HiChIP, overlaid with domains called by insulation score. (h) Contact matrix displaying the difference between GKO and WT cohesin HiChIP signal. Blue indicates greater contact in the WT, while red suggests more contact in the GKO. All matrices display chr4:110,000,000-120,000,000. (i) Overlap of contact coordinates called by FitHiChiP in either the WT and GKO. (j) Number of Gibbin-dependent chromatin contacts that are anchored in Gibbin target gene TSS’s. (k) Number of cell-type specific contacts that have one end anchored in a gene TSS (l) APA showing contact strength difference between WT and GKO cohesin Hi-ChIP at Gibbin-independent genes.

Extended Data Fig. 6 Validation of the BASU proximal proteomics system.

(a) Validation of BASU system from whole cell lysate western blots of hESCs stably expressing doxycycline-inducible, HA-tagged BASU control or Gibbin-BASU fusion proteins. The N-terminal version is shown here. Biotinylated protein was measured using dye-labelled streptavidin (Strep). Signal was only detected in the presence of both doxycycline (DOX) and biotin (BIO). (b) SAINT scores plotted against fold change over the BASU control for all proteins detected by Gibbin-BASU N-terminal or C-terminal mass spectrometry. SAINT scores > 0.9 are indicated in blue. (d) Representative immunoblot of biotinylated extracts from Gibbin-BASU and negative control cells after 24 h DOX and 2 h BIO. (d) GO term or Pfam domain enrichment of Gibbin interacting proteins as measured using a two-sided Fisher’s Exact Test (via EnrichR). (e) Overlap of N and C terminal Gibbin interacting proteins identified by BASU. (f) Expression of Gibbin interacting transcription factors across clusters identified by scRNA-seq. (g) Validation of GATA3-BASU system by western blot. (h) SAINT scores and fold change over negative BASU control for all proteins detected by GATA3-BASU mass spectrometry. (i) Overlap of Gibbin and GATA3 interacting proteins from BASU datasets. (j) Differential binding analysis from H3K9me3 ChIP–seq between WT and GKO cells. Signal is either unchanged (grey), increased (blue, >2-fold), or decreased (red, <2-fold), with an adjusted p-value cutoff < 0.05 as measured by DESeq2, n = 2 biologically independent samples. (k) Number, size and intensity measurements of HP1α foci in WT vs GKO cells, quantified from four representative immunofluorescent images for each cell type. All immunoblots were repeated experimentally twice. GAPDH was used as a processing control. For gel source data, see Supplementary Figure 1.

Extended Data Fig. 7 Gibbin loss causes DNA hypermethylation independent of DNMT expression.

(a) DNA methylation signal in WT and GKO cells as measured by a colorimetric assay. Error bars are mean +/− s.d., n = 4 biologically independent samples averaged from 2 technical replicates repeated across 2 different runs. * indicates p-value <0.001 as calculated by two-tailed t-test (p-value = 0.0003). (b) Overlap of RA/BMP4-dependent demethylation with Gibbin-dependent hypermethylation. (c) Number of Gibbin-dependent methylation sites that are demethylated during differentiation with RA/BMP4. (d) Volcano plot of differentially methylated probes (DMPs) between WT and GKO hESCs. Signal at each probe is either unchanged (grey), hypermethylated (red, >2-fold), or hypomethylated (blue, <2-fold), with an adjusted p-value cutoff <0.01 as measured by limma. n = 2 biologically independent samples. (e) ChromHMM analysis of the DMPs. (f) RPKM and adjusted p-values values for known DNMTs and effectors as measured by differential RNA-seq. Error bars are mean +/− s.d., n = 2 biologically independent samples, and adjusted p-values were calculated with DESeq2. (g) Differential binding analysis from CTCF ChIP–seq between WT and GKO cells. CTCF DNA binding is either unchanged (grey), increased (blue, >2-fold), or decreased (red, <2-fold), with an adjusted p-value cutoff <0.05 as measured by DESeq2. n = 2 biologically independent samples. (h) Overlap of genes anchored in WT chromatin contacts, GKO chromatin contacts, and sites of differential CTCF binding. (i) Overlap of genes that are bound by Gibbin and have Gibbin-dependent methylated DNA or CTCF binding sites. (j) Virtual 4C plot of normalized cohesin HiChIP signal for the normally repressed ATF3 locus, overlayed with a WashU Genome Browser screenshot depicting Gibbin, GATA3, and WT/GKO CTCF ChIP–seq peaks, DMP pileups, and FitHiChIP contacts. The ATF3 TSS was used as an anchor point in constructing the 4C plot.

Extended Data Fig. 8 Aberrant dermal maturation results in keratinocyte dysfunction.

Violin plot of (a) AHDC1/Gibbin or (b) GATA3 expression after 50 days of differentiation. (c) Monocle pseudotime trajectory of the EPCAM+/KRT5+ ectodermal and epidermal clusters from WT or (d) GKO cells. (e) RNA-seq differential expression analysis comparing WT and GKO day 50 cells. Gene expression is either unchanged (grey), increased (blue, >2-fold), or decreased (red, <2-fold), with an adjusted p-value cutoff <0.05 as measured by DESeq2. n = 2 biologically independent samples. (f) Representative RPKM and adjusted p-values from day 50 RNA-seq. Error bars are mean +/− standard deviation. (g) RNA-seq differential expression analysis between WT and GKO keratinocytes, n = 2 biologically independent samples. (h) CellChat circle plot showing the number of cell signalling interactions between each identity. (i) Number of cell–cell signalling interactions in WT and GKO day 50 cells as measured by CellChat. (j) H&E and immunofluorescent images of 3D skin organoid cultures generated from primary normal human keratinocyte (NHKs). NHKs were nucleofected with control or Gibbin targeting sgRNAs prior to stratification. Organoids were made in biological triplicate and repeated experimentally twice. All imaging was repeated in biological duplicate across 2 independent experiments.

Extended Data Fig. 9 FACS sorting strategy.

(a) Surface marker rankings identified by COMET for the EPCAM+ ectoderm or the (b) PDGFRA+ mesoderm after 7 days of RA/BMP4. Markers used for further analyses are noted in red, and markers that were tested but did not work well by FACS are noted in blue. (c) Expression patterns and fold changes in day 7 scRNA-seq of markers used for cell sorting including VTCN1 (ectoderm), (d) ABCG2 (ectoderm), and (e) PDGFRA (mesoderm). (f) Fluorescence associated cell sorting (FACS) strategy used to separate day 7 cells into ectoderm and mesoderm. Plots show a distinction between ABCG2+ or PDGFRA+ populations or (g) overlapping ABCG2+ and VTCN1+ populations. Populations were gated on unstained control cells, and the gating strategy can be found in the Supplementary Figure 2.

Extended Data Fig. 10 Pronuclear CRISPR Injections Targeting Gibbin Cause a Spectrum of Phenotypes in Mice.

(a) Quantification of three phenotype classes observed following Gibbin depletion in vivo. Mosaic genotypes exhibited CRISPR cutting on at least one allele. (b) Images of mouse embryos collected 18 days following pronuclear CRISPR injection to deplete Gibbin. Embryos were organized into four phenotypic classes. Injections were performed in duplicate across 6 different litters totaling n = 52 collected embryos. (c) Adult mice 5 weeks after receiving grafts from E18 mice. The GKO graft was taken from a more moderate mutant. n = 3 for each genotype, and grafting was repeated in two separate experiments. (d) H&E images depicting hair follicle cyst phenotypes from grafted mice. Images are representative of at least 3 biological and 3 technical replicates.

Supplementary information

Supplementary Information

Supplementary Figs. 1 and 2 and legends for Supplementary Tables 1–3.

Reporting Summary

Supplementary Table 1

A list of differentially expressed genes and adjusted P values from RNA-seq experiments, as well as a list of nearest genes from Gibbin ChIP–seq.

Supplementary Table 2

A list of SAINT scores for all Gibbin and GATA3 BASU experiments.

Supplementary Table 3

PCR primers and sgRNA sequences used in this study.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Collier, A., Liu, A., Torkelson, J. et al. Gibbin mesodermal regulation patterns epithelial development. Nature 606, 188–196 (2022). https://doi.org/10.1038/s41586-022-04727-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-022-04727-9

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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