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.

  • Brief Communication
  • Published:

Recurrent point mutations in the kinetochore gene KNSTRN in cutaneous squamous cell carcinoma

Subjects

Abstract

Here we report the discovery of recurrent mutations concentrated at an ultraviolet signature hotspot in KNSTRN, which encodes a kinetochore protein, in 19% of cutaneous squamous cell carcinomas (SCCs). Cancer-associated KNSTRN mutations, most notably those encoding p.Ser24Phe, disrupt chromatid cohesion in normal cells, occur in SCC precursors, correlate with increased aneuploidy in primary tumors and enhance tumorigenesis in vivo. These findings suggest a role for KNSTRN mutagenesis in SCC development.

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: Recurrent mutations in KNSTRN encoding p.Ser24Phe in cutaneous SCC.
Figure 2: Ser24Phe kinastrin promotes aneuploidy and enhances tumorigenesis in vivo.

Similar content being viewed by others

Accession codes

Accessions

NCBI Reference Sequence

References

  1. Lomas, A., Leonardi-Bee, J. & Bath-Hextall, F. Br. J. Dermatol. 166, 1069–1080 (2012).

    Article  CAS  Google Scholar 

  2. Durinck, S. et al. Cancer Discov. 1, 137–143 (2011).

    Article  CAS  Google Scholar 

  3. Fang, L., Seki, A. & Fang, G. Cell Cycle 8, 2819–2827 (2009).

    Article  CAS  Google Scholar 

  4. Vogelstein, B. et al. Science 339, 1546–1558 (2013).

    Article  CAS  Google Scholar 

  5. Dunsch, A.K., Linnane, E., Barr, F.A. & Gruneberg, U. J. Cell Biol. 192, 959–968 (2011).

    Article  CAS  Google Scholar 

  6. Davoli, T. et al. Cell 155, 948–962 (2013).

    Article  CAS  Google Scholar 

  7. Zack, T.I. et al. Nat. Genet. 45, 1134–1140 (2013).

    Article  CAS  Google Scholar 

  8. Biesterfeld, S., Pennings, K., Grussendorf-Conen, E.I. & Böcking, A. Br. J. Dermatol. 133, 557–560 (1995).

    Article  CAS  Google Scholar 

  9. Ziegler, A. et al. Nature 372, 773–776 (1994).

    Article  CAS  Google Scholar 

  10. Ziegler, A. et al. Proc. Natl. Acad. Sci. USA 90, 4216–4220 (1993).

    Article  CAS  Google Scholar 

  11. Kretz, M. et al. Genes Dev. 26, 338–343 (2012).

    Article  CAS  Google Scholar 

  12. Liu, X., Jian, X. & Boerwinkle, E. Hum. Mutat. 34, E2393–E2402 (2013).

    Article  CAS  Google Scholar 

  13. Li, H. & Durbin, R. Bioinformatics 25, 1754–1760 (2009).

    Article  CAS  Google Scholar 

  14. McKenna, A. et al. Genome Res. 20, 1297–1303 (2010).

    Article  CAS  Google Scholar 

  15. Koboldt, D.C. et al. Bioinformatics 25, 2283–2285 (2009).

    Article  CAS  Google Scholar 

  16. Deng, X. BMC Bioinformatics 12, 267 (2011).

    Article  Google Scholar 

  17. DePristo, M.A. et al. Nat. Genet. 43, 491–498 (2011).

    Article  CAS  Google Scholar 

  18. Ng, S.B. et al. Nature 461, 272–276 (2009).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank J.G. Rheinwald (Dana-Farber/Harvard Cancer Center) for his generous gift of SCC cell lines. We thank A.E. Oro, H.Y. Chang, M. Diehn, M.P. Scott, S.E. Artandi, G.R. Crabtree and T. Waldman as well as A. Zehnder, X. Bao, B.K. Sun, R.J. Flockhart and V. Lopez-Pajares for presubmission review and helpful comments. This work was supported by the US Veterans Affairs Office of Research and Development and by US National Institutes of Health/National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIH/NIAMS) grant AR43799 to P.A.K. C.S.L. is the recipient of career award K08 AR064732 from the NIH/NIAMS. A.S. is supported by NIH/National Institute of General Medical Science (NIGMS) grant GM074728 and American Cancer Society grant 120161-RSG. W.L.J. is the recipient of National Science Foundation (NSF) graduate research fellowship grant DGE-114747.

Author information

Authors and Affiliations

Authors

Contributions

C.S.L. designed and executed experiments, analyzed data and wrote the manuscript. A.M., A.B., C.J.A., C.B.N., W.L.J., E.J.R., A.U. and Z.S. helped execute experiments, analyzed data and contributed to the design of experimentation. A.S. helped design experiments and analyzed data. J.K. and S.Z.A. performed tumor tissue acquisition and analysis. P.A.K. designed experiments, analyzed data and wrote the manuscript.

Corresponding author

Correspondence to Paul A Khavari.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Comparison of mutation frequency in TP53, CDKN2A and HRAS between data in the COSMIC database and this study.

Supplementary Figure 2 Expression of kinastrin in human skin and SCCs.

(a) Kinastrin is expressed in a broad variety of tissues. Kinastrin expression was extracted from the NextBio Body Atlas application (PLoS One 5, e13066, 2010). The dotted line indicates median expression. (b) Kinastrin expression in progenitor human primary keratinocytes11. The average FPKM of all expressed genes (FPKM > 1.5) in this population is shown for reference. (c) Kinastrin is expressed in normal skin and SCCs. Kinastrin expression was assayed in 76 cases of cutaneous SCC and 5 normal skin samples; representative images are shown. Scale bar, 25 μm.

Supplementary Figure 3 Somatic KNSTRN p.Ser24Phe mutations in SCC.

KNSTRN p.Ser24Phe mutations were confirmed by Sanger sequencing. Sequence chromatograms from a representative SCC and normal skin (NL) sample from the same individual are shown.

Supplementary Figure 4 KNSTRN mutations in SCC compared to other cancer types.

(a) KNSTRN mutation frequency by cancer type (Cancer Discov. 2, 401–404, 2012). (b) Distribution of mutations in the kinastrin coding sequence (Cancer Discov. 2, 401–404, 2012). The labeled mutations disrupt sister chromatid cohesion in normal cells. SXLP, SXLP motif; CC, coiled-coil region.

Supplementary Figure 5 Enforced expression of kinastrin p.Ser24Phe is stable in normal cells.

Western blot analysis of early-passage primary human keratinocytes following transduction with the three kinastrin isoforms. Endogenous kinastrin expression is shown in lane 1; only isoform 3 is present. Lanes 6 and 7 show the enforced protein expression used to assess sister chromatid cohesion and DNA content. Numbers indicate molecular mass in kilodaltons.

Supplementary Figure 6 Kinastrin p.Ser24Phe disrupts chromatid cohesion in SCC-13 cells.

(a) Western blot analysis of SCC-13 cells following transduction with N-terminally tagged (Flag-HA-(poly)His) kinastrin p.Ser24Phe versus wild-type kinastrin. The lower- and higher-molecular-weight bands represent endogenous and enforced kinastrin expression, respectively. (b) Quantification of unpaired chromatids. *P = 0.013, **P = 0.008; ns, not significant.

Supplementary Figure 7 Cancer-associated KNSTRN mutations disrupt chromatid cohesion in normal cells.

(a) Western blot analysis of early-passage primary human keratinocytes following transduction with KNSTRN p.Arg11Lys, p.Pro26Ser, p.Pro28Ser and p.Ala40Glu versus wild-type kinastrin. (b) Quantification of unpaired chromatids. *P = 0.05, **P < 0.03.

Supplementary Figure 8 Kinastrin p.Ser28Phe enhances aneuploidy in normal cells.

Early-passage, pooled primary human foreskin keratinocytes were transduced with dominant-negative p53 and LacZ (CTR), wild-type kinastrin (WT) or kinastrin p.Ser28Phe (S24F). Cells were grown in paclitaxel for 68 h to induce aneuploidy. (a) Percentage of primary human keratinocytes with DNA content >4N. Data shown represents the mean ± s.d. for biological duplicates with technical triplicates. *P = 0.0014, **P = 0.0004; ns, not significant. (b) Representative plots of cell cycle analysis are shown.

Supplementary Figure 9 Kinastrin p.Ser28Phe does not affect cell growth or cell cycle kinetics.

Early-passage primary human keratinocytes were transduced with LacZ (CTR), wild-type kinastrin (WT) or kinastrin p.Ser28Phe (S24F). (a) Equal numbers of cells were seeded in duplicate, and cell growth assays were performed in technical triplicates at each of the time points shown. (b) Asynchronously proliferating cells were stained with propidium iodide and assayed by flow cytometry in technical duplicates.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–9 (PDF 2225 kb)

Supplementary Tables 1–7

Supplementary Tables 1–7 (XLS 2080 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lee, C., Bhaduri, A., Mah, A. et al. Recurrent point mutations in the kinetochore gene KNSTRN in cutaneous squamous cell carcinoma. Nat Genet 46, 1060–1062 (2014). https://doi.org/10.1038/ng.3091

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ng.3091

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