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:

Single-cell transcriptomic analysis of endometriosis

Abstract

Endometriosis is a common condition in women that causes chronic pain and infertility and is associated with an elevated risk of ovarian cancer. We profiled transcriptomes of >370,000 individual cells from endometriomas (n = 8), endometriosis (n = 28), eutopic endometrium (n = 10), unaffected ovary (n = 4) and endometriosis-free peritoneum (n = 4), generating a cellular atlas of endometrial-type epithelial cells, stromal cells and microenvironmental cell populations across tissue sites. Cellular and molecular signatures of endometrial-type epithelium and stroma differed across tissue types, suggesting a role for cellular restructuring and transcriptional reprogramming in the disease. Epithelium, stroma and proximal mesothelial cells of endometriomas showed dysregulation of pro-inflammatory pathways and upregulation of complement proteins. Somatic ARID1A mutation in epithelial cells was associated with upregulation of pro-angiogenic and pro-lymphangiogenic factors and remodeling of the endothelial cell compartment, with enrichment of lymphatic endothelial cells. Finally, signatures of ciliated epithelial cells were enriched in ovarian cancers, reinforcing epidemiologic associations between these two diseases.

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: A cellular atlas of human endometriosis.
Fig. 2: The cellular landscapes of endometrioma, peritoneal endometriosis, unaffected peritoneum, eutopic endometrium and unaffected ovary.
Fig. 3: Keratin-positive components of eutopic endometrium, endometriomas and endometriosis.
Fig. 4: Gene expression signatures associated with somatic mutation of ARID1A or KRAS.
Fig. 5: Signatures of EnS and mesenchymal cells associated with endometriosis.
Fig. 6: Molecular signatures of deep and superficial peritoneal endometriosis.
Fig. 7: Deconvoluting endometriosis-associated ovarian cancers with single-cell endometriosis signatures.

Similar content being viewed by others

Data availability

The data generated during this study are available at NCBI GEO under accession number GSE213216 and can also be accessed through https://github.com/lawrenson-lab/AtlasEndometriosis.

Code availability

Scripts for the downstream analysis of the single-cell data are available on GitHub (https://github.com/lawrenson-lab/AtlasEndometriosis) and through Zenodo https://doi.org/10.5281/zenodo.6974609.

References

  1. Sainz de la Cuesta, R. et al. Histologic transformation of benign endometriosis to early epithelial ovarian cancer. Gynecol. Oncol. 60, 238–244 (1996).

    Article  CAS  PubMed  Google Scholar 

  2. Lee, A. W. et al. Evidence of a genetic link between endometriosis and ovarian cancer. Fertil. Steril. 105, 35–43.e1 (2016).

    Article  PubMed  Google Scholar 

  3. Pearce, C. L. et al. Association between endometriosis and risk of histological subtypes of ovarian cancer: a pooled analysis of case-control studies. Lancet Oncol. 13, 385–394 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Lu, Y. et al. Shared genetics underlying epidemiological association between endometriosis and ovarian cancer. Hum. Mol. Genet. 24, 5955–5964 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Tan, Y. et al. Single-cell analysis of endometriosis reveals a coordinated transcriptional programme driving immunotolerance and angiogenesis across eutopic and ectopic tissues. Nat. Cell Biol. 24, 1306–1318 (2022).

    Article  CAS  PubMed  Google Scholar 

  7. Xu, C. et al. Modeling the temporal dynamics of master regulators and CtrA proteolysis in Caulobacter crescentus cell cycle. PLoS Comput. Biol. 18, e1009847 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Wang, W. et al. Single-cell transcriptomic atlas of the human endometrium during the menstrual cycle. Nat. Med. 26, 1644–1653 (2020).

    Article  CAS  PubMed  Google Scholar 

  9. Garcia-Alonso, L. et al. Mapping the temporal and spatial dynamics of the human endometrium in vivo and in vitro. Nat. Genet. 53, 1698–1711 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Cindrova-Davies, T. et al. Menstrual flow as a non-invasive source of endometrial organoids. Commun. Biol. 4, 651 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. 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 

  12. Suda, K. et al. Clonal lineage from normal endometrium to ovarian clear cell carcinoma through ovarian endometriosis. Cancer Sci. https://doi.org/10.1111/cas.14507 (2020).

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

    Article  CAS  PubMed  Google Scholar 

  14. Sweet-Cordero, A. et al. An oncogenic KRAS2 expression signature identified by cross-species gene-expression analysis. Nat. Genet. 37, 48–55 (2005).

    Article  CAS  PubMed  Google Scholar 

  15. Suryo Rahmanto, Y. et al. Inactivation of Arid1a in the endometrium is associated with endometrioid tumorigenesis through transcriptional reprogramming. Nat. Commun. 11, 2717 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Reddy, J. et al. Predicting master transcription factors from pan-cancer expression data. Sci. Adv. 7, eabf6123 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Chaves-Moreira, D. et al. The transcription factor PAX8 promotes angiogenesis in ovarian cancer through interaction with SOX17. Sci. Signal. 15, eabm2496 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Dinh, H. Q. et al. Single-cell transcriptomics identifies gene expression networks driving differentiation and tumorigenesis in the human fallopian tube. Cell Rep. 35, 108978 (2021).

    Article  CAS  PubMed  Google Scholar 

  19. Zheng, W., Aspelund, A. & Alitalo, K. Lymphangiogenic factors, mechanisms, and applications. J. Clin. Invest. 124, 878–887 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Wang, X., Park, J., Susztak, K., Zhang, N. R. & Li, M. Bulk tissue cell type deconvolution with multi-subject single-cell expression reference. Nat. Commun. 10, 380 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Tan, T. Z. et al. Analysis of gene expression signatures identifies prognostic and functionally distinct ovarian clear cell carcinoma subtypes. EBioMedicine 50, 203–210 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Tothill, R. W. et al. Novel molecular subtypes of serous and endometrioid ovarian cancer linked to clinical outcome. Clin. Cancer Res. 14, 5198–5208 (2008).

    Article  CAS  PubMed  Google Scholar 

  23. Winterhoff, B. et al. Molecular classification of high grade endometrioid and clear cell ovarian cancer using TCGA gene expression signatures. Gynecol. Oncol. 141, 95–100 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Saavalainen, L. et al. Risk of gynecologic cancer according to the type of endometriosis. Obstet. Gynecol. 131, 1095–1102 (2018).

    Article  PubMed  Google Scholar 

  25. Viganó, P., Somigliana, E., Chiodo, I., Abbiati, A. & Vercellini, P. Molecular mechanisms and biological plausibility underlying the malignant transformation of endometriosis: a critical analysis. Hum. Reprod. Update 12, 77–89 (2006).

    Article  PubMed  Google Scholar 

  26. Krausgruber, T. et al. Structural cells are key regulators of organ-specific immune responses. Nature 583, 296–302 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. 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 

  28. Praetorius, T. H. et al. Is endometriosis metastasizing? Shared somatic alterations suggest common origins across endometriotic lesions. Preprint at medRxiv https://doi.org/10.1101/2021.04.12.21255355 (2021).

  29. Moore, L. et al. The mutational landscape of normal human endometrial epithelium. Nature 580, 640–646 (2020).

    Article  CAS  PubMed  Google Scholar 

  30. Hasan, A. et al. Serum albumin and C3 complement levels in endometriosis. J. Coll. Physicians Surg. Pak. 29, 702–705 (2019).

    Article  PubMed  Google Scholar 

  31. Kabut, J., Kondera-Anasz, Z., Sikora, J. & Mielczarek-Palacz, A. Levels of complement components iC3b, C3c, C4, and SC5b-9 in peritoneal fluid and serum of infertile women with endometriosis. Fertil. Steril. 88, 1298–1303 (2007).

    Article  CAS  PubMed  Google Scholar 

  32. Rahal, D., Andrade, F. & Nisihara, R. Insights into the role of complement system in the pathophysiology of endometriosis. Immunol. Lett. 231, 43–48 (2021).

    Article  CAS  PubMed  Google Scholar 

  33. Ricklin, D., Reis, E. S., Mastellos, D. C., Gros, P. & Lambris, J. D. Complement component C3—the ‘Swiss Army Knife’ of innate immunity and host defense. Immunol. Rev. 274, 33–58 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Brosens, I., Donnez, J. & Benagiano, G. Improving the classification of endometriosis. Hum. Reprod. 8, 1792–1795 (1993).

    Article  CAS  PubMed  Google Scholar 

  35. García-Solares, J., Dolmans, M.-M., Squifflet, J.-L., Donnez, J. & Donnez, O. Invasion of human deep nodular endometriotic lesions is associated with collective cell migration and nerve development. Fertil. Steril. 110, 1318–1327 (2018).

    Article  PubMed  Google Scholar 

  36. Mita, S. et al. Dienogest inhibits nerve growth factor expression induced by tumor necrosis factor-α or interleukin-1β. Fertil. Steril. 101, 595–601 (2014).

    Article  CAS  PubMed  Google Scholar 

  37. Queckbörner, S. et al. Stromal heterogeneity in the human proliferative endometrium—a single-cell RNA sequencing study. J. Pers. Med. 11, 448 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Vento-Tormo, R. et al. Single-cell reconstruction of the early maternal-fetal interface in humans. Nature 563, 347–353 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Garcia-Alonso, L. et al. Mapping the temporal and spatial dynamics of the human endometrium in vivo and in vitro. Preprint at bioRxiv https://doi.org/10.1101/2021.01.02.425073 (2021).

  40. Xi, N. M. & Li, J. J. Benchmarking computational doublet-detection methods for single-cell RNA sequencing data. Cell Syst. 12, 176–194.e6 (2021).

    Article  CAS  PubMed  Google Scholar 

  41. McGinnis, C. S., Murrow, L. M. & Gartner, Z. J. DoubletFinder: doublet detection in single-cell RNA sequencing data using artificial nearest neighbors. Cell Syst. 8, 329–337.e4 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Wolock, S. L., Lopez, R. & Klein, A. M. Scrublet: computational identification of cell doublets in single-cell transcriptomic data. Cell Syst. 8, 281–291.e9 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Gayoso, A., Shor, J., Carr, A. J., Sharma, R. & Pe’er, D. JonathanShor/DoubletDetection: HOTFIX: Correct setup.py installation. Zenodo https://doi.org/10.5281/zenodo.2678041 (2019).

  44. Finak, G. et al. MAST: a flexible statistical framework for assessing transcriptional changes and characterizing heterogeneity in single-cell RNA sequencing data. Genome Biol. 16, 278 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Becht, E. et al. Dimensionality reduction for visualizing single-cell data using UMAP. Nat. Biotechnol. 37, 38–44 (2018).

    Article  Google Scholar 

  46. Yu, G. & He, Q.-Y. ReactomePA: an R/Bioconductor package for reactome pathway analysis and visualization. Mol. Biosyst. 12, 477–479 (2016).

    Article  CAS  PubMed  Google Scholar 

  47. Khalique, S. et al. Optimised ARID1A immunohistochemistry is an accurate predictor of ARID1A mutational status in gynaecological cancers. J. Pathol. Clin. Res. 4, 154–166 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Some of the specimens were collected as part of the Biologic and Epidemiologic Markers of Endometriosis (BEME) study. We thank all the patients who donated the specimens used in this study, and the team at the Biobank and Translational Research Laboratory who supported tissue procurement and histologic analyses, plus the Cedars-Sinai Applied Genomics, Computation and Translational core. We thank C. Chow and M. Ta at the Genetic Pathology Evaluation Centre (GPEC) for technical support for the mutation analyses. This study was supported in large part by a Leon Fine Translational Science Award from Cedars-Sinai Medical Center. K.L. is supported by a Liz Tilberis Early Career Award (grant no. 599175) and a Program Project Development grant (no. 373356) from the Ovarian Cancer Research Alliance, plus a Research Scholar’s Grant from the American Society (grant no. 134005). The research described was supported in part by the National Institutes of Health (NIH)/National Center for Advancing Translational Science (NCATS) UCLA CTSI Grant no. UL1TR001881 and in part by Cedars-Sinai Cancer, a Canadian Cancer Society Research Institute Impact grant (no. 705647, to D.G.H.) and a Canadian Institutes of Health Research Foundation grant (to D.G.H.). M.S.A. receives funds through the Canadian Institutes of Health Research (Early Career Investigator Grant in Maternal, Reproductive, Child & Youth Health), a Michael Smith Foundation for Health Research Scholar Award and the Janet D. Cottrelle Foundation Scholars program (managed by the BC Cancer Foundation). The GPEC receives core support from BC’s Gynecological Cancer Research team (OVCARE), and The VGH + UBC Hospital Foundation. Y.W. is a recipient of the North Family Health Research Award (administered by the VGH + UBC Hospital Foundation). M.D.P. is a recipient of grants from the NIH/National Institute of Biomedical Imaging and Bioengineering (NIBIB; grant no. U01NIBIB12482404) and NIH/National Institute of Allergy and Infectious Diseases (NIAID) (grant no. R01AI154535). The content of the manuscript is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.

Author information

Authors and Affiliations

Authors

Contributions

K.L. conceptualized the study and wrote the original draft of the manuscript. M.A.S.F. performed the data analysis, wrote the methods and generated the majority of the figures. X.L., F.A. and M.H. generated the single-cell data. H.Q.D. performed preliminary data analysis and provided technical expertise. K.N.W., M.S. and M.T. provided tissue specimens and surgical annotations. I.C. provided some of the eutopic endometrium tissue. J.S., L.H. and Y.C.-K. obtained patient consent and abstracted clinical data from medical records. F.M. supervised specimen collection and provided pathology review of tissues and stains with support from H.M.M and B.L.B. A.F. and R.H. prepared tissues for Visium profiling with M.H. N.L.O., J.H., Y.W., A.E.-N., D.G.H. and M.S.A. performed somatic mutation profiling. H.S.G. assisted with interpretation of immune cell data, M.D.P. provided clinical expertise and M.T.G. provided epidemiologic expertise. All authors reviewed and edited the manuscript.

Corresponding author

Correspondence to Kate Lawrenson.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Genetics thanks Kylie Gorringe, Brett McKinnon and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

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

Extended data

Extended Data Fig. 1 Quality control metrics and cell annotation procedures.

(a) The number of genes detected per cell is not significantly different across the major classes of study, nor by fresh/frozen status. (b) Observed cell number is positively correlated with observed cell number (Pearson correlation and Analysis of Variance). (c) The number of cells passing QC filters and (d) the number of reads per cell is not significantly different across the major classes of study, nor by fresh/frozen status. Coral color denotes samples that were processed immediately - ‘Fresh’; teal denotes samples that were processed into single cells and viably cryopreserved and thawed before capture - ‘Frozen’. Number of samples in (a, b and d) – endometrioma, fresh = 6, frozen = 2; endometriosis, fresh = 12, frozen = 11; eutopic endometrium, fresh = 2, frozen = 8; no endometriosis detected, fresh = 2, frozen = 2; unaffected ovary, fresh = 1, frozen = 3. (e) Decision tree showing workflow for cell type assignment. Black boxes indicate action/processes, blue boxes indicate cell-type assignment endpoints. (f) Heatmap showing expression of cell-type specific markers, by cluster. Expression is scaled between 0-1. Possible cell type assignments are indicated by column labels with the number of cell-type specific genes for each cell type indicated in brackets. For a list of marker genes see Methods and panel (g). (g) Expression of cell-type specific markers across the 96 clusters. (h) UMAP plot with 114 clusters (using Seurat shared nearest neighbor (SNN) for cluster identification considering resolution parameter of 3). (i) Correlation values for pair-wise comparisons of the 6 unassigned clusters compared to a background distribution of correlation values for 100 pairs of clusters selected at random, red dots indicate the correlation value used for cell type assignment. (j) Pearson’s correlation between clusters, based on expression of a union set of 1,960 genes differentially expressed by one or more cluster (log2 FC > 0; p <0.05). Clusters with no cell markers were assigned the identity associated with the most correlated cell type with known identity (see Methods). (k) Correlation of gene expression across major cell types, comparing fresh and cryopreserved specimens. Correlation values shown above each plot, Pearson’s correlation. (l) Principal component analysis based on cell-type composition of endometriosis and control tissues. In the box and whisker plots shown in (a,c,d,i), boxes denote the interquartile range, bar denotes median. The limits of the whiskers represent 1.5 * IQR (interquartile range) and outlier values are indicated with individual dots. Red dots denote the correlation value for expression in the given cluster compared to the cluster used to assign identity.

Extended Data Fig. 2 Additional results, analysis of epithelial subgroups by ARID1A and KRAS mutation status.

(a) Cluster frequency for eutopic endometrial epithelial clusters with inclusion of patients taking exogenous hormones. (b) Visium analysis of an endometriosis lesion from patient BEME355. (c) ARID1A and KRAS expression across epithelial subtypes. (d) Clusters present in BEME355 and expression of key genes. (e) UMAP of endometrial-type epithelial cells by ARID1A protein expression status, (f) UMAP of cells by KRAS mutation status. UMAP structure from 3A. (g) Pathways enriched in lesions with endometrial-type epithelium exhibiting heterogeneous and homogenous positive staining for ARID1A. (h) Pathways enriched in KRAS mutant and wild-type endometrial-type epithelium. Pathway analyses were performed using the Reactome R package, with p-values calculated based on a hypergeometric model and a Bonferroni correction applied.

Supplementary information

Supplementary Information

Supplementary Note, Figs. 1–6 and references.

Reporting Summary

Peer Review File

Supplementary Table 1–25

Supplementary Tables 1–25, including associated legends.

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

Fonseca, M.A.S., Haro, M., Wright, K.N. et al. Single-cell transcriptomic analysis of endometriosis. Nat Genet 55, 255–267 (2023). https://doi.org/10.1038/s41588-022-01254-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-022-01254-1

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