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.

  • Letter
  • Published:

Keratin-dependent regulation of Aire and gene expression in skin tumor keratinocytes

Abstract

Expression of the intermediate filament protein keratin 17 (K17) is robustly upregulated in inflammatory skin diseases and in many tumors originating in stratified and pseudostratified epithelia1,2,3. We report that autoimmune regulator (Aire), a transcriptional regulator, is inducibly expressed in human and mouse tumor keratinocytes in a K17-dependent manner and is required for timely onset of Gli2-induced skin tumorigenesis in mice. The induction of Aire mRNA in keratinocytes depends on a functional interaction between K17 and the heterogeneous nuclear ribonucleoprotein hnRNP K4. Further, K17 colocalizes with Aire protein in the nucleus of tumor-prone keratinocytes, and each factor is bound to a specific promoter region featuring an NF-κB consensus sequence in a relevant subset of K17- and Aire-dependent proinflammatory genes. These findings provide radically new insight into keratin intermediate filament and Aire function, along with a molecular basis for the K17-dependent amplification of inflammatory and immune responses in diseased epithelia.

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: Loss of Krt17 attenuates HPV16Tg-induced skin tumorigenesis.
Figure 2: Aire expression, function and regulation in skin tumor keratinocytes.
Figure 3: K17 regulates Aire subnuclear distribution and localizes to the nucleus to promote gene expression.
Figure 4: K17 and Aire associate with target gene promoter regions.

Similar content being viewed by others

References

  1. Moll, R., Franke, W.W., Schiller, D.L., Geiger, B. & Krepler, R. The catalog of human cytokeratins: patterns of expression in normal epithelia, tumors and cultured cells. Cell 31, 11–24 (1982).

    Article  CAS  Google Scholar 

  2. Karantza, V. Keratins in health and cancer: more than mere epithelial cell markers. Oncogene 30, 127–138 (2011).

    Article  CAS  Google Scholar 

  3. Jin, L. & Wang, G. Keratin 17: a critical player in the pathogenesis of psoriasis. Med. Res. Rev. 34, 438–454 (2014).

    Article  CAS  Google Scholar 

  4. Chung, B.M. et al. Regulation of C-X-C chemokine gene expression by keratin 17 and hnRNP K in skin tumor keratinocytes. J. Cell Biol. 208, 613–627 (2015).

    Article  CAS  Google Scholar 

  5. Depianto, D., Kerns, M.L., Dlugosz, A.A. & Coulombe, P.A. Keratin 17 promotes epithelial proliferation and tumor growth by polarizing the immune response in skin. Nat. Genet. 42, 910–914 (2010).

    Article  CAS  Google Scholar 

  6. Sankar, S. et al. A novel role for keratin 17 in coordinating oncogenic transformation and cellular adhesion in Ewing sarcoma. Mol. Cell. Biol. 33, 4448–4460 (2013).

    Article  CAS  Google Scholar 

  7. Kim, S., Wong, P. & Coulombe, P.A. A keratin cytoskeletal protein regulates protein synthesis and epithelial cell growth. Nature 441, 362–365 (2006).

    Article  CAS  Google Scholar 

  8. Chen, R. et al. Rac1 regulates skin tumors by regulation of keratin 17 through recruitment and interaction with CD11b+Gr1+ cells. Oncotarget 5, 4406–4417 (2014).

    PubMed  PubMed Central  Google Scholar 

  9. Arbeit, J.M., Munger, K., Howley, P.M. & Hanahan, D. Progressive squamous epithelial neoplasia in K14–human papillomavirus type 16 transgenic mice. J. Virol. 68, 4358–4368 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. McGowan, K.M. & Coulombe, P.A. Onset of keratin 17 expression coincides with the definition of major epithelial lineages during skin development. J. Cell Biol. 143, 469–486 (1998).

    Article  CAS  Google Scholar 

  11. Woodworth, C.D. et al. Strain-dependent differences in malignant conversion of mouse skin tumors is an inherent property of the epidermal keratinocyte. Carcinogenesis 25, 1771–1778 (2004).

    Article  CAS  Google Scholar 

  12. Mueller, M.M. Inflammation in epithelial skin tumours: old stories and new ideas. Eur. J. Cancer 42, 735–744 (2006).

    Article  CAS  Google Scholar 

  13. Lee, C.H., Kim, M.S., Chung, B.M., Leahy, D.J. & Coulombe, P.A. Structural basis for heteromeric assembly and perinuclear organization of keratin filaments. Nat. Struct. Mol. Biol. 19, 707–715 (2012).

    Article  CAS  Google Scholar 

  14. Tong, X. & Coulombe, P.A. A novel mouse type I intermediate filament gene, keratin 17n (K17n), exhibits preferred expression in nail tissue. J. Invest. Dermatol. 122, 965–970 (2004).

    Article  CAS  Google Scholar 

  15. Laan, M. & Peterson, P. The many faces of Aire in central tolerance. Front. Immunol. 4, 326 (2013).

    Article  Google Scholar 

  16. Mathis, D. & Benoist, C. A decade of AIRE. Nat. Rev. Immunol. 7, 645–650 (2007).

    Article  CAS  Google Scholar 

  17. Metzger, T.C. & Anderson, M.S. Control of central and peripheral tolerance by Aire. Immunol. Rev. 241, 89–103 (2011).

    Article  CAS  Google Scholar 

  18. Kisand, K. & Peterson, P. Autoimmune polyendocrinopathy candidiasis ectodermal dystrophy: known and novel aspects of the syndrome. Ann. NY Acad. Sci. 1246, 77–91 (2011).

    Article  CAS  Google Scholar 

  19. Sillanpää, N. et al. Autoimmune regulator induced changes in the gene expression profile of human monocyte-dendritic cell-lineage. Mol. Immunol. 41, 1185–1198 (2004).

    Article  Google Scholar 

  20. Giraud, M. et al. Aire unleashes stalled RNA polymerase to induce ectopic gene expression in thymic epithelial cells. Proc. Natl. Acad. Sci. USA 109, 535–540 (2012).

    Article  CAS  Google Scholar 

  21. Laan, M. et al. Autoimmune regulator deficiency results in decreased expression of CCR4 and CCR7 ligands and in delayed migration of CD4+ thymocytes. J. Immunol. 183, 7682–7691 (2009).

    Article  CAS  Google Scholar 

  22. Uhlén, M. et al. Proteomics: tissue-based map of the human proteome. Science 347, 1260419 (2015).

    Article  Google Scholar 

  23. Kumar, V. et al. The autoimmune regulator (AIRE), which is defective in autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy patients, is expressed in human epidermal and follicular keratinocytes and associates with the intermediate filament protein cytokeratin 17. Am. J. Pathol. 178, 983–988 (2011).

    Article  CAS  Google Scholar 

  24. Anderson, M.S. et al. Projection of an immunological self shadow within the thymus by the Aire protein. Science 298, 1395–1401 (2002).

    Article  CAS  Google Scholar 

  25. Barboro, P., Ferrari, N. & Balbi, C. Emerging roles of heterogeneous nuclear ribonucleoprotein K (hnRNP K) in cancer progression. Cancer Lett. 352, 152–159 (2014).

    Article  CAS  Google Scholar 

  26. Pitkänen, J., Vähämurto, P., Krohn, K. & Peterson, P. Subcellular localization of the autoimmune regulator protein. Characterization of nuclear targeting and transcriptional activation domain. J. Biol. Chem. 276, 19597–19602 (2001).

    Article  Google Scholar 

  27. Akiyoshi, H. et al. Subcellular expression of autoimmune regulator is organized in a spatiotemporal manner. J. Biol. Chem. 279, 33984–33991 (2004).

    Article  CAS  Google Scholar 

  28. Lallemand-Breitenbach, V. & de The, H. PML nuclear bodies. Cold Spring Harb. Perspect. Biol. 2, a000661 (2010).

    Article  Google Scholar 

  29. Gaetani, M. et al. AIRE-PHD fingers are structural hubs to maintain the integrity of chromatin-associated interactome. Nucleic Acids Res. 40, 11756–11768 (2012).

    Article  CAS  Google Scholar 

  30. Liao, J., Lowthert, L.A., Ghori, N. & Omary, M.B. The 70-kDa heat shock proteins associate with glandular intermediate filaments in an ATP-dependent manner. J. Biol. Chem. 270, 915–922 (1995).

    Article  CAS  Google Scholar 

  31. Kumeta, M., Hirai, Y., Yoshimura, S.H., Horigome, T. & Takeyasu, K. Antibody-based analysis reveals “filamentous vs. non-filamentous” and “cytoplasmic vs. nuclear” crosstalk of cytoskeletal proteins. Exp. Cell Res. 319, 3226–3237 (2013).

    Article  CAS  Google Scholar 

  32. Kosugi, S., Hasebe, M., Tomita, M. & Yanagawa, H. Systematic identification of cell cycle–dependent yeast nucleocytoplasmic shuttling proteins by prediction of composite motifs. Proc. Natl. Acad. Sci. USA 106, 10171–10176 (2009).

    Article  CAS  Google Scholar 

  33. Abramson, J., Giraud, M., Benoist, C. & Mathis, D. Aire's partners in the molecular control of immunological tolerance. Cell 140, 123–135 (2010).

    Article  CAS  Google Scholar 

  34. Lenardo, M.J. & Baltimore, D. NF-κB: a pleiotropic mediator of inducible and tissue-specific gene control. Cell 58, 227–229 (1989).

    Article  CAS  Google Scholar 

  35. Pasparakis, M. Role of NF-κB in epithelial biology. Immunol. Rev. 246, 346–358 (2012).

    Article  Google Scholar 

  36. McGowan, K.M. et al. Keratin 17 null mice exhibit age- and strain-dependent alopecia. Genes Dev. 16, 1412–1422 (2002).

    Article  CAS  Google Scholar 

  37. Reichelt, J. & Haase, I. Establishment of spontaneously immortalized keratinocyte lines from wild-type and mutant mice. Methods Mol. Biol. 585, 59–69 (2010).

    Article  CAS  Google Scholar 

  38. Ran, F.A. et al. Genome engineering using the CRISPR-Cas9 system. Nat. Protoc. 8, 2281–2308 (2013).

    Article  CAS  Google Scholar 

  39. Lessard, J.C. et al. Keratin 16 regulates innate immunity in response to epidermal barrier breach. Proc. Natl. Acad. Sci. USA 110, 19537–19542 (2013).

    Article  CAS  Google Scholar 

  40. Adamson, K.A., Pearce, S.H., Lamb, J.R., Seckl, J.R. & Howie, S.E. A comparative study of mRNA and protein expression of the autoimmune regulator gene (Aire) in embryonic and adult murine tissues. J. Pathol. 202, 180–187 (2004).

    Article  CAS  Google Scholar 

  41. Wan, F. et al. Ribosomal protein S3: a KH domain subunit in NF-κB complexes that mediates selective gene regulation. Cell 131, 927–939 (2007).

    Article  CAS  Google Scholar 

  42. Fu, K. et al. Sam68 modulates the promoter specificity of NF-κB and mediates expression of CD25 in activated T cells. Nat. Commun. 4, 1909 (2013).

    Article  Google Scholar 

Download references

Acknowledgements

We thank members of the Coulombe laboratory for support, P. Peterson (Aire constructs; Tartu University), R. Foisner (subcellular fractionation), M. Vladut-Talor (central tolerance study) and J. Folmer (electron microscopy) for advice and assistance. These studies were supported by research grants CA160255 and AR044232 (to P.A.C.) and GM111682 (to F.W.) and by training grant CA009110 (to R.P.H.), all from the US National Institutes of Health.

Author information

Authors and Affiliations

Authors

Contributions

R.P.H. and D.J.D. jointly led the characterization of skin tumorigenesis in the HPV mouse model, with assistance from M.C.H. and A.S.B. R.P.H. led the studies performed in A431 cells and mouse keratinocytes in culture, with contributions from J.T.J., B.-M.C., B.G.P., Y.G. and J.H. S.O. and D.Č. conducted central tolerance study with assistance from R.P.H. and A.S.B. J.M.T. provided human basal cell carcinoma samples. W.Z. and F.W. performed the flow cytometry analyses. F.W. provided guidance to R.P.H. for EMSA analyses. R.P.H., D.J.D. and P.A.C. contributed to experimental design and data analysis. R.P.H. and P.A.C. jointly wrote the manuscript.

Corresponding author

Correspondence to Pierre A Coulombe.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Characterization of HPVTg/+; Krt17−/− mice.

(a) Macroscopic images of HPV16Tg/+ mouse ears at P40, P70 and P120. (b) Immunostaining of HPV16Tg/+ ear tissue sections for K17 (red) and Hoechst (blue) at P20 and P40. (c) H&E staining of a wild-type P70 ear tissue section (this micrograph complements the data presented in Fig. 1b). (d) Immunostaining and quantification of vasculature in P70 ear tissue sections from wild-type (green bar), HPV16Tg/+ (blue bar) and HPV16Tg/+; Krt17−/− (open bar) mice. The elements stained are PECAM (green), K14 (red) and Hoechst (blue). n = 9 biological replicates per genotype. (e) Flow cytometry analysis of total cells isolated from P70 ear tissue for four distinct genotypes. Cells double-positive for CD45 and CD4, CD207, CD11b or CD11c were analyzed. Number indicates the percentage of double-positive cells per genotype. Data represent one of three biological replicates. (f) Immunoprecipitation of HPV16 E7 from P2 mouse skin keratinocytes. (g) p53 expression levels in P70 ear tissue. Actin was used as a loading control for total protein, and K14 serves as a loading control for epithelial cells. The K17 immunoblot establishes proper genotyping. PIS, preimmune sera. The blots in f and g represent one of six biological replicates. (h) TUNEL staining of P70 ear tissue, denoting apoptotic cells restricted to the suprabasal layers of epidermis, for both genotypes. Images represent one of six biological replicates. (i) Electron micrographs and aspect ratio quantification (box-and-whisker plot) of basal epithelial cells from 8-month-old ear tissue (8,400× magnification). n = 100 basal cells counted across 3 biological replicates per genotype.

Supplementary Figure 2 TPA- and HPV16-induced gene expression in skin tumor keratinocytes.

(a) Normalized expression for target genes in intact A431 keratinocytes stimulated with TPA relative to DMSO (solid red bars) and in TPA-treated A431 cells expressing either shControl or shKrt17 plasmid (open bars). n = 5 biological replicates. (b) Normalized expression for gene transcripts restored upon Krt17 re-expression (n = 7 biological replicates; blue bars) relative to vector control and compared to Krt42 re-expression (n = 5 biological replicates; red bars). (c) Normalized expression for distinct regions of the Aire transcript (exon 2 or 10 or a region spanning exons 13 and 14) in TPA-treated ears (blue bars) or thymus (red bars) relative to liver, in two different genetic backgrounds (C57BL/6, left; FVB/N, right). n = 4 biological replicates. (d,e) Sense-strand RNA in situ hybridization controls in skin tissue sections (complements data reported in Fig. 2c,d). Scale bars, 200 µm. (f) Coimmunoprecipitation of K17 with RFP-Trap pulldown of mCherry-Aire compared to unfused mCherry. GAPDH serves as a loading control for total protein. n = 1 of 7 biological replicates.

Supplementary Figure 3 Loss of Krt17 does not affect thymic Aire expression or function.

(a) Normalized expression for Aire in P10 thymic tissue between wild-type (blue bar), HPV16Tg/+ (red bar) and HPV16Tg/+; Krt17−/− (open bar) mice. *NS, no statistical significance. (b) RNA in situ hybridization for Aire in thymic tissue sections. Arrows denote regions of medullary thymus positive for hybridization. Scale bars, 200 µm. (c) H&E staining of lacrimal glands from aged (>4 month) C57BL/6 mice. Scale bars, 100 µm. (d) Histology scoring for the presence of lymphocytic infiltrate in multiple tissues from aged (>4 month) Krt17+/+, Krt17−/−, Aire+/+ and Aire−/− mice. *P values are from non-parametric ANOVA with Dunn’s post-testing. n.s., no statistical significance. (e) Summary of histology scoring (each pie represents an individual mouse) with each tissue represented by a different color. A filled wedge indicates >10% of the tissue exhibiting inflammation.

Supplementary Figure 4 Characterization of Aire and nuclear K17 punctae in tumor epithelia.

(a) A431 keratinocytes transfected with mCherry-Aire and immunostained in green with markers for lysosomes (LAMP-1), stress granules (eIF3η) or PML bodies (PML). Scale bars, 5 µm. (b) A431 keratinocytes stably expressing shRNA targeting Krt17 (shKrt17) or shControl plasmids transiently transfected with mCherry-Aire and immunostained for HSP70 (green). Scale bars, 20 µm. (c) Single-plane confocal images of HeLa cells stably expressing GFP-K17 and either mock treated or treated with LMB. Scale bars, 5 µm. (d) Single-plane confocal images through the nucleus of HeLa cells either mock treated or treated with LMB and immunostained for K17 (green) and Hoechst (blue) using mouse (left panels) or rabbit (right panels) anti-K17 antibodies. The z plane is on top. Arrows denote intranuclear punctae immunopositive for K17. Scale bars, 20 µm. (e,f) Single-plane confocal images of immortalized keratinocytes from wild-type (e) or Krt17−/− (f) mouse epidermis immunostained for K17 and K5. Scale bars, 20 µm. (g) Immunoblots for subcellular fractions of DMSO- or TPA-treated A431 cells transfected with mCherry-Aire compared to unfused mCherry control. Sol, soluble (GAPDH positive); NE, nuclear envelope enriched (nesprin 3 positive); Chr, chromatin enriched (histone H3 positive). (h) Immunoblots of subcellular fractions of DMSO- or TPA-treated A431 keratinocytes probed for various type I keratins, as indicated on the left. Sol, soluble (GAPDH positive); NE, nuclear envelope enriched (nesprin 3 positive); Chr, chromatin enriched (histone H3 positive). All images in ag are representative of three biological replicates. Blots in h are representative of five biological replicates.

Supplementary Figure 5 ChIP and EMSA assays.

(a) Immunoblot for K17 after immunoprecipitation of K17, relative to preimmune sera (PIS), from the nuclear extract used for the ChIP analysis presented in Figure 4. Input represents the sample removed before immunoprecipitation. The image represents one of seven biological replicates. (b) DNA amplicons from the K17 ChIP assay (Fig. 4) observed after equal numbers of qPCR cycles by DNA electrophoresis. The images represent one of three biological replicates. (c,d) 5′ upstream sequences of the C XCL10 (c) and CCL19 (d) transcripts. The locations of the forward and reverse primer sites used for the ChIP assays are depicted with colored arrows. NF-κB consensus sequences are denoted with bold text. The ATG translation start site is italicized. (e) EMSA analysis of radiolabeled Oct1 consensus oligonucleotides using nuclear extracts from A431 keratinocytes treated with TPA, relative to DMSO control. Supershift analysis was conducted with antibodies against K17, relative to preimmune sera (PIS) control. Cold competition was conducted using 50-fold excess of non-labeled oligonucleotide. The image represents one of three biological replicates. (f) Reciprocal coimmunoprecipitation demonstrating p65 enrichment with K17 immunoprecipitation relative to preimmune sera (PIS) (left panel; n = 7 biological replicates) and K17 enrichment with p65 immunoprecipitation relative to IgG control (right panel; n = 3 biological replicates).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–5 and Supplementary Tables 1–7. (PDF 2001 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hobbs, R., DePianto, D., Jacob, J. et al. Keratin-dependent regulation of Aire and gene expression in skin tumor keratinocytes. Nat Genet 47, 933–938 (2015). https://doi.org/10.1038/ng.3355

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3355

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer