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:

The mutational landscape of normal human endometrial epithelium

Abstract

All normal somatic cells are thought to acquire mutations, but understanding of the rates, patterns, causes and consequences of somatic mutations in normal cells is limited. The uterine endometrium adopts multiple physiological states over a lifetime and is lined by a gland-forming epithelium1,2. Here, using whole-genome sequencing, we show that normal human endometrial glands are clonal cell populations with total mutation burdens that increase at about 29 base substitutions per year and that are many-fold lower than those of endometrial cancers. Normal endometrial glands frequently carry ‘driver’ mutations in cancer genes, the burden of which increases with age and decreases with parity. Cell clones with drivers often originate during the first decades of life and subsequently progressively colonize the epithelial lining of the endometrium. Our results show that mutational landscapes differ markedly between normal tissues—perhaps shaped by differences in their structure and physiology—and indicate that the procession of neoplastic change that leads to endometrial cancer is initiated early in life.

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: Clonality of normal endometrial glands.
Fig. 2: Mutation burden correlates with age in normal endometrial glands.
Fig. 3: Histology images and reconstructed phylogenetic trees for two individuals in whom every normal endometrial gland contained at least one driver mutation.
Fig. 4: Phylogenetic trees of endometrial glands for donors aged 19 to 40 years.
Fig. 5: Timing of driver mutations in normal endometrial glands.

Similar content being viewed by others

Data availability

Whole-genome sequencing data are deposited in the European Genome–Phenome Archive (EGA) with accession number EGAS00001002471.

Code availability

Code for statistical analyses on total substitution and driver mutation burdens is included in the Supplementary Information, and is deposited on GitHub at https://github.com/LuizaMoore/Endometrium. All other code is available on request.

References

  1. Gargett, C. E., Schwab, K. E. & Deane, J. A. Endometrial stem/progenitor cells: the first 10 years. Hum. Reprod. Update 22, 137–163 (2016).

    CAS  PubMed  Google Scholar 

  2. Kaitu’u-Lino, T. J., Ye, L. & Gargett, C. E. Reepithelialization of the uterine surface arises from endometrial glands: evidence from a functional mouse model of breakdown and repair. Endocrinology 151, 3386–3395 (2010).

    Article  PubMed  Google Scholar 

  3. Alexandrov, L. B. et al. Signatures of mutational processes in human cancer. Nature 500, 415–421 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Alexandrov, L. B. et al. The repertoire of mutational signatures in human cancer. Nature 578, 94–101 (2020).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  5. Stratton, M. R., Campbell, P. J. & Futreal, P. A. The cancer genome. Nature 458, 719–724 (2009).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  6. Blokzijl, F. et al. Tissue-specific mutation accumulation in human adult stem cells during life. Nature 538, 260–264 (2016).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  7. Lee-Six, H. et al. Population dynamics of normal human blood inferred from somatic mutations. Nature 561, 473–478 (2018).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  8. Franco, I. et al. Somatic mutagenesis in satellite cells associates with human skeletal muscle aging. Nat. Commun. 9, 800 (2018).

    Article  PubMed  PubMed Central  ADS  Google Scholar 

  9. Martincorena, I. et al. High burden and pervasive positive selection of somatic mutations in normal human skin. Science 348, 880–886 (2015).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  10. Martincorena, I. et al. Somatic mutant clones colonize the human esophagus with age. Science 362, 911–917 (2018).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  11. Yokoyama, A. et al. Age-related remodelling of oesophageal epithelia by mutated cancer drivers. Nature 565, 312–317 (2019).

    Article  CAS  PubMed  ADS  Google Scholar 

  12. Zhu, M. et al. Somatic mutations increase hepatic clonal fitness and regeneration in chronic liver disease. Cell 177, 608–621 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Suda, K. et al. Clonal expansion and diversification of cancer-associated mutations in endometriosis and normal endometrium. Cell Rep. 24, 1777–1789 (2018).

    Article  CAS  PubMed  Google Scholar 

  14. Lee-Six, H. et al. The landscape of somatic mutation in normal colorectal epithelial cells. Nature 574, 532–537 (2019).

    Article  CAS  PubMed  ADS  Google Scholar 

  15. Brunner, S. F. et al. Somatic mutations and clonal dynamics in healthy and cirrhotic human liver. Nature 574, 538–542 (2019).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  16. Schmitt, M. W. et al. Detection of ultra-rare mutations by next-generation sequencing. Proc. Natl Acad. Sci. USA 109, 14508–14513 (2012).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  17. Hoang, M. L. et al. Genome-wide quantification of rare somatic mutations in normal human tissues using massively parallel sequencing. Proc. Natl Acad. Sci. USA 113, 9846–9851 (2016).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  18. Lodato, M. A. et al. Aging and neurodegeneration are associated with increased mutations in single human neurons. Science 359, 555–559 (2018).

    Article  CAS  PubMed  ADS  Google Scholar 

  19. Jaiswal, S. et al. Age-related clonal hematopoiesis associated with adverse outcomes. N. Engl. J. Med. 371, 2488–2498 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Genovese, G. et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N. Engl. J. Med. 371, 2477–2487 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Xie, M. et al. Age-related mutations associated with clonal hematopoietic expansion and malignancies. Nat. Med. 20, 1472–1478 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Tempest, N., Maclean, A. & Hapangama, D. K. Endometrial stem cell markers: current concepts and unresolved questions. Int. J. Mol. Sci. 19, E3240 (2018).

    Article  Google Scholar 

  23. CRUK. Uterine cancer risk. https://www.cancerresearchuk.org/health-professional/cancer-statistics/statistics-by-cancer-type/uterine-cancer/risk-factors#heading-Zero (accessed 28 March 2020).

  24. Morice, P., Leary, A., Creutzberg, C., Abu-Rustum, N. & Darai, E. Endometrial cancer. Lancet 387, 1094–1108 (2016).

    Article  PubMed  Google Scholar 

  25. Le Gallo, M. & Bell, D. W. The emerging genomic landscape of endometrial cancer. Clin. Chem. 60, 98–110 (2014).

    Article  PubMed  Google Scholar 

  26. Onstad, M. A., Schmandt, R. E. & Lu, K. H. Addressing the role of obesity in endometrial cancer risk, prevention, and treatment. J. Clin. Oncol. 34, 4225–4230 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Setiawan, V. W. et al. Type I and II endometrial cancers: have they different risk factors? J. Clin. Oncol. 31, 2607–2618 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  28. Martincorena, I. et al. Universal patterns of selection in cancer and somatic tissues. Cell 171, 1029–1041 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. The Cancer Genome Atlas Research Network. Integrated genomic characterization of endometrial carcinoma. Nature 497, 67–73 (2013).

    Article  PubMed  PubMed Central  ADS  Google Scholar 

  30. Anglesio, M. S. et al. Cancer-associated mutations in endometriosis without cancer. N. Engl. J. Med. 376, 1835–1848 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Lac, V. et al. Iatrogenic endometriosis harbors somatic cancer-driver mutations. Hum. Reprod. 34, 69–78 (2019).

    Article  CAS  PubMed  Google Scholar 

  32. Lac, V. et al. Oncogenic mutations in histologically normal endometrium: the new normal? J. Pathol. 249, 173–181 (2019).

    Article  CAS  PubMed  Google Scholar 

  33. Lopez-Garcia, C., Klein, A. M., Simons, B. D. & Winton, D. J. Intestinal stem cell replacement follows a pattern of neutral drift. Science 330, 822–825 (2010).

    Article  CAS  PubMed  ADS  Google Scholar 

  34. Barker, N. et al. Lgr5+ve stem cells drive self-renewal in the stomach and build long-lived gastric units in vitro. Cell Stem Cell 6, 25–36 (2010).

    Article  CAS  PubMed  Google Scholar 

  35. Rouhani, F. J. et al. Mutational history of a human cell lineage from somatic to induced pluripotent stem cells. PLoS Genet. 12, e1005932 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Nicholson, A. M. et al. Fixation and spread of somatic mutations in adult human colonic epithelium. Cell Stem Cell 22, 909–918 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Zhang, Y. et al. A pan-cancer compendium of genes deregulated by somatic genomic rearrangement across more than 1,400 cases. Cell Rep. 24, 515–527 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Roerink, S. F. et al. Intra-tumour diversification in colorectal cancer at the single-cell level. Nature 556, 457–462 (2018).

    Article  CAS  PubMed  ADS  Google Scholar 

  39. Wu, Q. J. et al. Parity and endometrial cancer risk: a meta-analysis of epidemiological studies. Sci. Rep. 5, 14243 (2015).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  40. Greaves, M. In utero origins of childhood leukaemia. Early Hum. Dev. 81, 123–129 (2005).

    Article  PubMed  Google Scholar 

  41. Greaves, M. Pre-natal origins of childhood leukemia. Rev. Clin. Exp. Hematol. 7, 233–245 (2003).

    CAS  PubMed  Google Scholar 

  42. Mitchell, T. J. et al. Timing the landmark events in the evolution of clear cell renal cell cancer: TRACERx renal. Cell 173, 611–623 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Anderson, N. D. et al. Rearrangement bursts generate canonical gene fusions in bone and soft tissue tumors. Science 361, eaam8419 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Maura, F. et al. Genomic landscape and chronological reconstruction of driver events in multiple myeloma. Nat. Commun. 10, 3835 (2019).

    Article  PubMed  PubMed Central  ADS  Google Scholar 

  45. Gerstung, M. et al. The evolutionary history of 2,658 cancers. Nature 578, 122–128 (2020).

    Article  CAS  PubMed  PubMed Central  ADS  Google Scholar 

  46. Li, H. & Durbin, R. Fast and accurate long-read alignment with Burrows–Wheeler transform. Bioinformatics 26, 589–595 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Nik-Zainal, S. et al. Mutational processes molding the genomes of 21 breast cancers. Cell 149, 979–993 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Raine, K. M. et al. cgpPindel: identifying somatically acquired insertion and deletion events from paired end sequencing. Curr. Protoc. Bioinformatics 52, 15.7.1–15.7.2 (2015).

    Article  PubMed  Google Scholar 

  49. Ye, K., Schulz, M. H., Long, Q., Apweiler, R. & Ning, Z. Pindel: a pattern growth approach to detect break points of large deletions and medium sized insertions from paired-end short reads. Bioinformatics 25, 2865–2871 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Van Loo, P. et al. Allele-specific copy number analysis of tumors. Proc. Natl Acad. Sci. USA 107, 16910–16915 (2010).

    Article  PubMed  PubMed Central  ADS  Google Scholar 

  51. Raine, K. M. et al. ascatNgs: identifying somatically acquired copy-number alterations from whole-genome sequencing data. Curr. Protoc. Bioinformatics 56, 15.19.11–15.19.17 (2016).

    Article  Google Scholar 

  52. Buels, R. et al. JBrowse: a dynamic web platform for genome visualization and analysis. Genome Biol. 17, 66 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Yates, L. R. et al. Subclonal diversification of primary breast cancer revealed by multiregion sequencing. Nat. Med. 21, 751–759 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Hoang, D. T. et al. MPBoot: fast phylogenetic maximum parsimony tree inference and bootstrap approximation. BMC Evol. Biol. 18, 11 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the staff of WTSI Sample Logistics, Genotyping, Pulldown, Sequencing and Informatics facilities for their contribution; L. O’Neil, C. Latimer and P. Scott for technical support; F. Nadeu, N. Roberts and J. Wang for their advice on mutational signature extraction; A. R. J. Lawson, F. Abascal and S. Grossmann for their assistance with data analysis; and the Cambridge Biorepository for Translational Medicine for the provision of samples from deceased transplant organ donors. This work was supported by the Wellcome Trust. L.M. is a recipient of a CRUK Clinical PhD fellowship (C20/A20917) and Pathological Society of Great Britain and Ireland Trainee Small Grant (grant reference no. 1175). S.F.B. was supported by the Swiss National Science Foundation (P2SKP3-171753 and P400PB-180790). M.A.S. is supported by a Rubicon fellowship from NWO (019.153LW.038).

Author information

Authors and Affiliations

Authors

Contributions

M.R.S. and L.M. designed the study and wrote the manuscript with contributions from all authors. K.S.-P., C.A.I.-D., J.J.B., K.T.M., M.J.-L. and L.M. obtained samples. P.E. and L.M. devised the protocol for laser-capture microscopy, DNA extraction and sequencing of endometrial glands. L.M. prepared sections, reviewed histology, and microdissected and lysed endometrial glands. Y.H. assisted with tissue processing and section preparation. L.M. performed data curation and analysis with help from D.L., T.H.H.C., M.A.S., S.C.D., K.J.D., T.B., R.R., T.J.M., J.N., P.S.T., S.F.B. and H.L.-S. T.H.H.C. reconstructed phylogenetic trees. S.C.D. performed formal clonality assessment with dpclust. M.A.S. devised filters for substitutions and structural variants. D.L., F.M. and S.M. assisted with signature analyses. I.M. assisted with statistical and dN/dS analyses. P.J.C. oversaw statistical analyses and performed analysis of structural variants. M.R.S. supervised the study.

Corresponding author

Correspondence to Michael R. Stratton.

Ethics declarations

Competing interests

M.R.S. is on the Scientific Advisory Board for GRAIL.

Additional information

Peer review information Nature thanks Michael Lawrence and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 Clonality of endometrial glands and driver mutations.

a, The majority of the sampled normal endometrial glands (n = 257 individual endometrial glands) were clonal with a median VAF for all identified indels of 0.3 or above. b, The presence of a driver mutation did not have a significant effect on the observed monoclonality of the glands (two-sided Mann–Whitney U-test, P = 0.1). Here, we compared the median VAF of endometrial glands with drivers (median = 0.33, range 0.17–0.5, n = 145) to that of glands without drivers (median = 0.31, range 0.16–0.5, n = 112). Box plots were constructed with the upper and lower edge of the box defining the 25th (Q1) and 75th (Q3) percentile, respectively, outliers (plotted as circles) are defined as values beyond the whiskers (upper, Q1 − 1.5 × interquartile range (IQR) and lower Q3 + 1.5 × IQR). c, All glands from the 19-year-old donor (donor PD37506) (n = 10 individual endometrial glands) were clonal with a median VAF ≥ 0.3, but there were no detectable driver mutations.

Extended Data Fig. 2 Assessment of clonal composition of individual endometrial glands using the mutation clustering method dpclust.

ac, Each column contains a summary of the clonality analysis for individual donors, showing the fraction of samples in which 1, 2 or 3 or more mutation clusters were found (a), the fraction of mutations assigned per cluster for each sample (b) and at the total number of SNVs per sample (c).

Extended Data Fig. 3 Phylogenetic trees of endometrial glands for donors aged 42 to 81 years.

Phylogenetic trees for twelve donors aged 42 to 81 years were also reconstructed using SBSs with branch length proportional to the number of variants; the stacked bar plots represent the attributed SBS mutational signatures that contributed to each branch. Signature extraction was not performed on branches with fewer than 100 substitutions. The ordering of signatures within each branch is for visualization purposes only. Every single studied gland from donors PD39952 (44 year old) and PD40659 (81 year old) had at least one driver mutation.

Extended Data Fig. 4 An example of copy-number neutral loss of heterozygosity in a normal endometrial gland.

a, A biallelic truncating mutation is seen in ZFHX3 (p.R715*), with every read carrying the variant. b, Associated copy-number neutral loss of heterozygosity is observed on chromosome 16.

Extended Data Fig. 5 Oncoplot of all driver mutations and their distribution across individual endometrial-gland samples and donors.

Each cell represents an individual endometrial gland sample and is colour-coded to represent the total number of detected driver mutations (0–3). PIK3CA was the most frequently mutated gene, with at least 1 mutation detected in 54% (15 out of 28) of women. In some glands, these mutations in PIK3CA co-occurred with mutations in ZFHX3, ARHGAP35, FGFR2, FOXA2 and other genes that are also selected for in endometrial cancer.

Extended Data Fig. 6 Timing of all driver mutations.

To time the driver mutations, we used the reconstructed SNV-based phylogenetic trees for 25 out the 28 donors. To estimate the time interval in which specific mutations occurred, we calculated a patient-specific mutation rate by taking the ratio of the mean mutation burden per endometrial gland of the patient and the age of the patient. The mutation number at the start and end of a branch in the phylogenetic tree was then converted to a lower and upper age by dividing these numbers by the estimated mutation rate. This approach relies on the assumption of a constant mutation rate for endometrial glands throughout the life of a patient. The same approach was used for dating indels. We dated the driver mutations that occurred in the trunks and branches.

Extended Data Fig. 7 Timing of driver mutations using patient-based and cohort-based estimates of mutation rates.

To estimate the time interval in which specific mutations occurred, we applied two approaches: (a) ‘patient-based’, in which we calculated a patient-specific mutation rate by taking the ratio of the mean mutation burden per endometrial gland of the patient and the age of the patient; (b) ‘cohort-based’, in which the mutation rate for each patient was derived from the linear mixed-effect model for total mutation rate that included data from the entire cohort. The mutation number at the start and end of a branch in the phylogenetic tree was then converted to a lower and upper age by dividing these numbers by the estimated mutation rate. Both approaches rely on the assumption of a constant mutation rate for endometrial glands throughout the life of the patient. The dotted line represents time from the upper bound of the estimate to sampling.

Extended Data Fig. 8 Comparison between normal endometrial epithelium and endometrial cancer.

a, b, Normal endometrial glands show a lower total mutation burden (substitutions (a) and indels (b)) than do endometrial cancers (data from PCAWG2). c, Genes under significant positive selection (dN/dS > 1) in normal endometrial epithelium (n = 257 individual endometrial glands) and endometrial cancer (n = 218 biologically independent samples). q values were calculated for the whole exome, and under a restricted hypothesis test of 369 known endometrial cancer genes28. d, Venn diagram showing the overlap between the genes under positive selection in normal endometrium and endometrial cancer from the results described in c. e, Identified driver mutations and their distribution in normal endometrial glands and the two major types of endometrial cancer (endometroid and serous carcinoma).

Supplementary information

Supplementary Information

This file contains a guide to Supplementary Results 1-15.

Reporting Summary

Supplementary Data

This file contains Supplementary Results 1-15 – see Supplementary Information document for full details.

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

Moore, L., Leongamornlert, D., Coorens, T.H.H. et al. The mutational landscape of normal human endometrial epithelium. Nature 580, 640–646 (2020). https://doi.org/10.1038/s41586-020-2214-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-020-2214-z

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: 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