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:

NR2F2 controls malignant squamous cell carcinoma state by promoting stemness and invasion and repressing differentiation

Abstract

The nongenetic mechanisms required to sustain malignant tumor state are poorly understood. During the transition from benign tumors to malignant carcinoma, tumor cells need to repress differentiation and acquire invasive features. Using transcriptional profiling of cancer stem cells from benign tumors and malignant skin squamous cell carcinoma (SCC), we identified the nuclear receptor NR2F2 as uniquely expressed in malignant SCC. Using genetic gain of function and loss of function in vivo, we show that NR2F2 is essential for promoting the malignant tumor state by controlling tumor stemness and maintenance in mouse and human SCC. We demonstrate that NR2F2 promotes tumor cell proliferation, epithelial–mesenchymal transition and invasive features, while repressing tumor differentiation and immune cell infiltration by regulating a common transcriptional program in mouse and human SCCs. Altogether, we identify NR2F2 as a key regulator of malignant cancer stem cell functions that promotes tumor renewal and restricts differentiation to sustain a malignant tumor state.

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: NR2F2 loss of function prevents malignant transition.
Fig. 2: NR2F2 gain of function promotes a malignant tumor state.
Fig. 3: NR2F2 promotes tumor stemness in mouse and human SCC.
Fig. 4: NR2F2 is essential for the maintenance of malignant SCCs.
Fig. 5: Nr2f2 deletion promotes CSC differentiation.
Fig. 6: NR2F2 target genes regulate distinct tumor functions.
Fig. 7: Targeting NR2F2 function in human SCCs.
Fig. 8: Conservation of NR2F2 function in human SCCs.

Similar content being viewed by others

Data availability

All the raw microarray and sequencing data have been deposited in the Gene Expression Omnibus under the following accession codes: GSE164605 (reference series of the whole data), GSE175726 (human RNA-seq), GSE164597 (microarray carcinoma and papilloma CSCs), GSE164602 (microarray papilloma GOF versus Ctrl and carcinoma loss of function versus Ctrl), GSE175724 (ChIP-seq). All other relevant data are available from the corresponding author upon reasonable request. Source data are provided with this paper.

References

  1. Alam, M. & Ratner, D. Cutaneous squamous-cell carcinoma. N. Engl. J. Med. 344, 975–983 (2001).

    Article  CAS  PubMed  Google Scholar 

  2. Sanchez-Danes, A. & Blanpain, C. Deciphering the cells of origin of squamous cell carcinomas. Nat. Rev. Cancer 18, 549–561 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Owens, D. M. & Watt, F. M. Contribution of stem cells and differentiated cells to epidermal tumors. Nat. Rev. Cancer 3, 444–451 (2003).

    Article  CAS  PubMed  Google Scholar 

  4. Perez-Losada, J. & Balmain, A. Stem-cell hierarchy in skin cancer. Nat. Rev. Cancer 3, 434–443 (2003).

    Article  CAS  PubMed  Google Scholar 

  5. Abel, E. L., Angel, J. M., Kiguchi, K. & DiGiovanni, J. Multi-stage chemical carcinogenesis in mouse skin: fundamentals and applications. Nat. Protoc. 4, 1350–1362 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Kemp, C. J. Multistep skin cancer in mice as a model to study the evolution of cancer cells. Semin. Cancer Biol. 15, 460–473 (2005).

    Article  CAS  PubMed  Google Scholar 

  7. Valent, P. et al. Cancer stem cell definitions and terminology: the devil is in the details. Nat. Rev. Cancer 12, 767–775 (2012).

    Article  CAS  PubMed  Google Scholar 

  8. Beck, B. & Blanpain, C. Unravelling cancer stem cell potential. Nat. Rev. Cancer 13, 727–738 (2013).

    Article  CAS  PubMed  Google Scholar 

  9. Beck, B. et al. A vascular niche and a VEGF-Nrp1 loop regulate the initiation and stemness of skin tumors. Nature 478, 399–403 (2011).

    Article  CAS  PubMed  Google Scholar 

  10. Lapouge, G. et al. Skin squamous cell carcinoma propagating cells increase with tumor progression and invasiveness. EMBO J. 31, 4563–4575 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Malanchi, I. et al. Cutaneous cancer stem cell maintenance is dependent on β-catenin signalling. Nature 452, 650–653 (2008).

    Article  CAS  PubMed  Google Scholar 

  12. Schober, M. & Fuchs, E. Tumor-initiating stem cells of squamous cell carcinomas and their control by TGF-β and integrin/focal adhesion kinase (FAK) signaling. Proc. Natl Acad. Sci. USA 108, 10544–10549 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Beck, B. et al. Different levels of Twist1 regulate skin tumor initiation, stemness, and progression. Cell Stem Cell 16, 67–79 (2015).

    Article  CAS  PubMed  Google Scholar 

  14. Boumahdi, S. et al. SOX2 controls tumor initiation and cancer stem-cell functions in squamous-cell carcinoma. Nature 511, 246–250 (2014).

    Article  CAS  PubMed  Google Scholar 

  15. Sastre-Perona, A. et al. De novo PITX1 expression controls bi-stable transcriptional circuits to govern self-renewal and differentiation in squamous cell carcinoma. Cell Stem Cell 24, 390–404 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Siegle, J. M. et al. SOX2 is a cancer-specific regulator of tumor initiating potential in cutaneous squamous cell carcinoma. Nat. Commun. 5, 4511 (2014).

    Article  CAS  PubMed  Google Scholar 

  17. Lin, F. J., Qin, J., Tang, K., Tsai, S. Y. & Tsai, M. J. Coup d’Etat: an orphan takes control. Endocr. Rev. 32, 404–421 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Pereira, F. A., Tsai, M. J. & Tsai, S. Y. COUP-TF orphan nuclear receptors in development and differentiation. Cell Mol. Life Sci. 57, 1388–1398 (2000).

    Article  CAS  PubMed  Google Scholar 

  19. Qin, J., Chen, X., Xie, X., Tsai, M. J. & Tsai, S. Y. COUP-TFII regulates tumor growth and metastasis by modulating tumor angiogenesis. Proc. Natl Acad. Sci. USA 107, 3687–3692 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Qin, J., Chen, X., Yu-Lee, L. Y., Tsai, M. J. & Tsai, S. Y. Nuclear receptor COUP-TFII controls pancreatic islet tumor angiogenesis by regulating vascular endothelial growth factor/vascular endothelial growth factor receptor-2 signaling. Cancer Res. 70, 8812–8821 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Xie, X., Tang, K., Yu, C. T., Tsai, S. Y. & Tsai, M. J. Regulatory potential of COUP-TFs in development: stem/progenitor cells. Semin. Cell Dev. Biol. 24, 687–693 (2013).

    Article  CAS  PubMed  Google Scholar 

  22. Hawkins, S. M. et al. Expression and functional pathway analysis of nuclear receptor NR2F2 in ovarian cancer. J. Clin. Endocrinol. Metab. 98, E1152–E1162 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Polvani, S. et al. COUP-TFII in pancreatic adenocarcinoma: clinical implication for patient survival and tumor progression. Int. J. Cancer 134, 1648–1658 (2014).

    Article  CAS  PubMed  Google Scholar 

  24. Qin, J., Tsai, S. & Tsai, M. J. COUP-TFII, a prognostic marker and therapeutic target for prostate cancer. Asian J. Androl. 15, 360–361 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Ding, W. et al. Overexpression of COUPTFII suppresses proliferation and metastasis of human gastric cancer cells. Mol. Med. Rep. 17, 2393–2401 (2018).

    CAS  PubMed  Google Scholar 

  26. Litchfield, L. M., Appana, S. N., Datta, S. & Klinge, C. M. COUP-TFII inhibits NF-κB activation in endocrine-resistant breast cancer cells. Mol. Cell Endocrinol. 382, 358–367 (2014).

    Article  CAS  PubMed  Google Scholar 

  27. Shin, S. W. et al. Clinical significance of chicken ovalbumin upstream promoter-transcription factor II expression in human colorectal cancer. Oncol. Rep. 21, 101–106 (2009).

    PubMed  Google Scholar 

  28. Wang, C. et al. High expression of COUP-TF II cooperated with negative Smad4 expression predicts poor prognosis in patients with colorectal cancer. Int. J. Clin. Exp. Pathol. 8, 7112–7121 (2015).

    PubMed  PubMed Central  Google Scholar 

  29. Zhang, C., Han, Y., Huang, H., Qu, L. & Shou, C. High NR2F2 transcript level is associated with increased survival and its expression inhibits TGF-β-dependent epithelial–mesenchymal transition in breast cancer. Breast Cancer Res. Treat. 147, 265–281 (2014).

    Article  CAS  PubMed  Google Scholar 

  30. Qin, J. et al. COUP-TFII inhibits TGF-β-induced growth barrier to promote prostate tumorigenesis. Nature 493, 236–240 (2013).

    Article  CAS  PubMed  Google Scholar 

  31. Latil, M. et al. Cell-type-specific chromatin states differentially prime squamous cell carcinoma tumor-initiating cells for epithelial to mesenchymal transition. Cell Stem Cell 20, 191–204 (2017).

    Article  CAS  PubMed  Google Scholar 

  32. Nassar, D., Latil, M., Boeckx, B., Lambrechts, D. & Blanpain, C. Genomic landscape of carcinogen-induced and genetically induced mouse skin squamous cell carcinoma. Nat. Med. 21, 946–954 (2015).

    Article  CAS  PubMed  Google Scholar 

  33. Lapouge, G. et al. Identifying the cellular origin of squamous skin tumors. Proc. Natl Acad. Sci. USA 108, 7431–7436 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. White, A. C. et al. Defining the origins of Ras/p53-mediated squamous cell carcinoma. Proc. Natl Acad. Sci. USA 108, 7425–7430 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Caulin, C., Bauluz, C., Gandarillas, A., Cano, A. & Quintanilla, M. Changes in keratin expression during malignant progression of transformed mouse epidermal keratinocytes. Exp. Cell Res. 204, 11–21 (1993).

    Article  CAS  PubMed  Google Scholar 

  36. Marcato, P., Dean, C. A., Giacomantonio, C. A. & Lee, P. W. Aldehyde dehydrogenase: its role as a cancer stem cell marker comes down to the specific isoform. Cell Cycle 10, 1378–1384 (2011).

    Article  CAS  PubMed  Google Scholar 

  37. Marcato, P. et al. Aldehyde dehydrogenase activity of breast cancer stem cells is primarily due to isoform ALDH1A3 and its expression is predictive of metastasis. Stem Cells 29, 32–45 (2011).

    Article  CAS  PubMed  Google Scholar 

  38. Pastushenko, I. et al. Identification of the tumor transition states occurring during EMT. Nature 556, 463–468 (2018).

    Article  CAS  PubMed  Google Scholar 

  39. Vaillant, F. et al. The mammary progenitor marker CD61/β3 integrin identifies cancer stem cells in mouse models of mammary tumorigenesis. Cancer Res. 68, 7711–7717 (2008).

    Article  CAS  PubMed  Google Scholar 

  40. van Kempen, L. C. et al. Activated leukocyte cell adhesion molecule/CD166, a marker of tumor progression in primary malignant melanoma of the skin. Am. J. Pathol. 156, 769–774 (2000).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Xiao, M. et al. Cancer stem-like cell related protein CD166 degrades through E3 ubiquitin ligase ChIP in head and neck cancer. Exp. Cell Res. 353, 46–53 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Gujral, T. S. et al. A noncanonical Frizzled2 pathway regulates epithelial-mesenchymal transition and metastasis. Cell 159, 844–856 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. 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  PubMed  Google Scholar 

  44. Wang, L. et al. Small-molecule inhibitor targeting orphan nuclear receptor COUP-TFII for prostate cancer treatment. Sci. Adv. 6, eaaz8031 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Ma, I. & Allan, A. L. The role of human aldehyde dehydrogenase in normal and cancer stem cells. Stem Cell Rev. Rep. 7, 292–306 (2011).

    Article  CAS  PubMed  Google Scholar 

  46. Minn, I. et al. A red-shifted fluorescent substrate for aldehyde dehydrogenase. Nat. Commun. 5, 3662 (2014).

    Article  CAS  PubMed  Google Scholar 

  47. Pillai, S., Bikle, D. D., Mancianti, M. L., Cline, P. & Hincenbergs, M. Calcium regulation of growth and differentiation of normal human keratinocytes: modulation of differentiation competence by stages of growth and extracellular calcium. J. Cell Physiol. 143, 294–302 (1990).

    Article  CAS  PubMed  Google Scholar 

  48. Hennings, H. et al. Calcium regulation of growth and differentiation of mouse epidermal cells in culture. Cell 19, 245–254 (1980).

    Article  CAS  PubMed  Google Scholar 

  49. Angel, P., Szabowski, A. & Schorpp-Kistner, M. Function and regulation of AP-1 subunits in skin physiology and pathology. Oncogene 20, 2413–2423 (2001).

    Article  CAS  PubMed  Google Scholar 

  50. Ito, Y., Bae, S. C. & Chuang, L. S. The RUNX family: developmental regulators in cancer. Nat. Rev. Cancer 15, 81–95 (2015).

    Article  CAS  PubMed  Google Scholar 

  51. Qin, J., Tsai, S. Y. & Tsai, M. J. The critical roles of COUP-TFII in tumor progression and metastasis. Cell Biosci. 4, 58 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Zheng, J. et al. Knockdown of COUP-TFII inhibits cell proliferation and induces apoptosis through upregulating BRCA1 in renal cell carcinoma cells. Int. J. Cancer 139, 1574–1585 (2016).

    Article  CAS  PubMed  Google Scholar 

  53. Alečković, M. et al. Identification of Nidogen 1 as a lung metastasis protein through secretome analysis. Genes Dev. 31, 1439–1455 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Barker, H. E., Cox, T. R. & Erler, J. T. The rationale for targeting the LOX family in cancer. Nat. Rev. Cancer 12, 540–552 (2012).

    Article  CAS  PubMed  Google Scholar 

  55. Knowles, L. M. et al. Integrin αvβ3 and fibronectin upregulate Slug in cancer cells to promote clot invasion and metastasis. Cancer Res. 73, 6175–6184 (2013).

    Article  CAS  PubMed  Google Scholar 

  56. Levental, K. R. et al. Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell 139, 891–906 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Krebs, A. M. et al. The EMT-activator Zeb1 is a key factor for cell plasticity and promotes metastasis in pancreatic cancer. Nat. Cell Biol. 19, 518–529 (2017).

    Article  CAS  PubMed  Google Scholar 

  58. Ocana, O. H. et al. Metastatic colonization requires the repression of the epithelial–mesenchymal transition inducer Prrx1. Cancer Cell 22, 709–724 (2012).

    Article  CAS  PubMed  Google Scholar 

  59. Stemmler, M. P., Eccles, R. L., Brabletz, S. & Brabletz, T. Non-redundant functions of EMT transcription factors. Nat. Cell Biol. 21, 102–112 (2019).

    Article  CAS  PubMed  Google Scholar 

  60. de The, H. Differentiation therapy revisited. Nat. Rev. Cancer 18, 117–127 (2018).

    Article  PubMed  Google Scholar 

  61. Degos, L. et al. All-trans-retinoic acid as a differentiating agent in the treatment of acute promyelocytic leukemia. Blood 85, 2643–2653 (1995).

    Article  CAS  PubMed  Google Scholar 

  62. Storm, E. E. et al. Targeting PTPRK-RSPO3 colon tumors promotes differentiation and loss of stem-cell function. Nature 529, 97–100 (2016).

    Article  CAS  PubMed  Google Scholar 

  63. Biehs, B. et al. A cell identity switch allows residual BCC to survive Hedgehog pathway inhibition. Nature 562, 429–433 (2018).

    Article  CAS  PubMed  Google Scholar 

  64. Sanchez-Danes, A. et al. A slow-cycling LGR5 tumor population mediates basal cell carcinoma relapse after therapy. Nature 562, 434–438 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Pastushenko, I. et al. Fat1 deletion promotes hybrid EMT state, tumor stemness and metastasis. Nature 589, 448–455 (2021).

    Article  CAS  PubMed  Google Scholar 

  66. Sallee, J. L. & Burridge, K. Density-enhanced phosphatase 1 regulates phosphorylation of tight junction proteins and enhances barrier function of epithelial cells. J. Biol. Chem. 284, 14997–15006 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Wu, B. et al. Baicalein mediates inhibition of migration and invasiveness of skin carcinoma through Ezrin in A431 cells. BMC Cancer 11, 527 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Gautier, L., Cope, L., Bolstad, B. M. & Irizarry, R. A. affy: analysis of Affymetrix GeneChip data at the probe level. Bioinformatics 20, 307–315 (2004).

    Article  CAS  PubMed  Google Scholar 

  69. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  71. Anders, S., Pyl, P. T. & Huber, W. HTSeq: a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  CAS  PubMed  Google Scholar 

  72. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Li, H. et al. The sequence alignment/map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  74. Zhang, Y. et al. Model-based analysis of ChIP-seq (MACS). Genome Biol. 9, R137 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  75. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. McLean, C. Y. et al. GREAT improves functional interpretation of cis-regulatory regions. Nat. Biotechnol. 28, 495–501 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  79. Kuleshov, M. V. et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 44, W90–W97 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Gendoo, D. M. et al. Genefu: an R/Bioconductor package for computation of gene expression-based signatures in breast cancer. Bioinformatics 32, 1097–1099 (2016).

    Article  CAS  PubMed  Google Scholar 

  81. Hu, Y. & Smyth, G. K. ELDA: extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays. J. Immunol. Methods 347, 70–78 (2009).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We are grateful to the Erasme Hospital Biobank (Brussels, Belgium; BE_BERA1; Biobanque Hôpital Erasme-ULB; BE_NBWB1; Biothèque Wallonie Bruxelles); BBMRI−ERIC for providing a large number of human tumor samples. We thank S.Y. Tsai (Department of Molecular and Cellular Biology, Baylor College of Medicine) for kindly sharing the NR2F2f/f mice. We thank Blanpain laboratory members for their constructive comments on the manuscript. We thank the animal house facility from the ULB (Erasme campus). C.B. is an investigator of WELBIO. F.M. was supported by a National Fund for Scientific Research (FNRS) postdoctoral fellowship and by TELEVIE. G.L. was supported by an FNRS postdoctoral fellowship and by TELEVIE. This work was supported by WELBIO, the FNRS, TELEVIE, the PAI program (P7/03-CanEpi), the ERC Advanced Grant (agreement ID 885093), the ULB Fondation and the Fondation Baillet Latour. The Center for Microscopy and Molecular Imaging is supported by the Fonds Yvonne Boël, by the European Regional Development Fund and the Walloon Region (Wallonia-biomed; grant no. 411132-957270; ‘CMMI-ULB’).

Author information

Authors and Affiliations

Authors

Contributions

C.B., F.M., G.L. and C.S. designed the experiments and performed data analysis. F.M., C.S. and G.L. performed most of the experiments. B.D. started the project. B.D., S.G., I.P., J.B., M.M., A.B., Y.S. and M.Raphaël contributed to the experiments. S.R., J.A. and I.S. provided hSCC samples and performed histological analysis. Y.B. and C.S. performed TCGA data acquisition and analysis. E.N., M. Rozzi, B.D., V.M. and F.R. provided technical support. C.D. provided technical support for cell sorting. G.L. and J.V. contributed to discussion and to preparation of the manuscript. C.B. and F.M. wrote the manuscript.

Corresponding author

Correspondence to Cédric Blanpain.

Ethics declarations

Competing interests

C.B. is founder and advisor of ChromaCure SA, which develops drugs targeting NR2F2. J.V. and M.M. are employees of ChromaCure. C.B. owns shares in ChromaCure. The remaining authors declare no competing interests.

Additional information

Peer review information Nature Cancer thanks Salvador Benitah, Nick Barker 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

Extended Data Fig. 1 NR2F2 expression is associated with tumor progression in mouse squamous skin tumors.

a: Scheme of the strategy used to isolate and transcriptionally profile CSCs from benign papillomas and malignant carcinomas. b: Bar graph representation of the relative mRNA expression of the transcription factors that are most highly upregulated in CD34+ cells from mouse malignant carcinoma vs benign papillomas (Fold change CD34+ CSC from carcinoma / papilloma; cells isolated from independent n=4 carcinoma samples and 3 papilloma samples). Data are represented as mean ± SEM of the ratio between the gene expression level in CD34+ cells in carcinoma samples and the average of all papilloma samples. c: Immunostaining for GFP (tumor cells), NR2F2 and Ki67 (proliferating cells) in DMBA SCC. Representative images of minimum 5 independent biological replicates. d: Quantification of the percentage of proliferating tumor cells that are positive for NR2F2 expression (n=9 independent tumor samples). e: Immunostaining for K14 (Epithelial tumor cells), K10 (differentiating cells) and Itgβ4 (basal membrane), NR2F2 and CD31 (endothelial cells) in papillomas from Ctrl and NR2F2 KO mice (K14Cre Nr2f2flox). Representative images of 5 independent biological replicates. f: Immunostaining for K14 (Epithelial cells), YFP (tumor cells) and Vimentin (mesenchymal cells) in differentiated, mixed and mesenchymal genetic tumors (lgr5CreER/KRasG12D/p53flox/RosaYFP). Serial sections from the same tumor samples shown in Fig. 1c. Representative images of minimum 4 independent biological replicates per tumor type. g: NR2F2 is necessary for malignant progression in K14CreER/KRasG12D/p53flox/RosaYFP. Number of benign tumors (papillomas) and of malignant SCC per mouse in Ctrl and NR2F2 KO (n=9 Ctrl and 9 NR2F2 KO mice). Scale bar=50 µm. Data in d and g are represented as mean ± SEM. The p-values are calculated using a two-tailed Mann-Whitney test.

Source data

Extended Data Fig. 2 NR2F2 ectopic expression induces defects of epidermal differentiation.

a-c: Ectopic NR2F2 expression induces weight loss and death. a: Scheme of the transgene allowing Doxycycline inducible expression of NR2F2-3XFlag. b: Survival curve of the mice after NR2F2 induction (n=9 mice per genotype). c: Graph of the weight of NR2F2 GOF mice and control counterparts after NR2F2 induction (n=2 mice per condition). d-f: Ectopic expression of NR2F2 induces defect of epidermal differentiation. d: HE and immunostaining for K14, Flag, K10, Ki67 and ZO-1 in mouse skin in NR2F2 GOF and Control. e: HE and immunostaining for K14, NR2F2, Flag, and Loricrin in mouse tongue in NR2F2 GOF and Control. f: HE and immunostaining for K14, NR2F2, Flag, and Itgβ4 in mouse stomach in NR2F2 GOF and Control. Representative images of at least 4 independent biological replicates are shown in d-f. The p-values are calculated using the Long-Rank-Mantel-Cox test in b. Scale bar = 50 µm.

Source data

Extended Data Fig. 3 NR2F2 promotes tumor stemness in human SCC.

a: Proportion of secondary tumor formation and tumor propagating cell frequency calculated using the extreme limiting dilution analysis (ELDA) for A431 (skin SCC), SK-MES-1(lung SCC) and Kyse-70 (esophagus SCC) WT and NR2F2 KO human SCC cell lines. p-value is calculated using chi-squared test. b: Proportion of secondary tumor formation and tumor propagating cell frequency calculated by using the extreme limiting dilution analysis (ELDA) for A431, SK-MES-1 and Kyse-70 parental SCC cell lines and NR2F2 KO rescue clones carrying the NR2F2-3HA transgene. p-value is calculated using chi-squared test.

Extended Data Fig. 4 NR2F2 expression is necessary for malignant tumor maintenance in murine SCC.

a: Tumor size over time and b: HE in NR2F2 KO SCC that reaches a steady size. Higher magnification shows that the residual mass is formed by terminally differentiated cells (Area 1) or necrotic/fibrotic tissue (Area 2). c: Tumor size over time and d: immunostaining for K14, NR2F2 and CD31 show that relapsing tumors escaped tamoxifen induced Nr2f2 deletion. e: Immunostaining for K14 (epithelial tumor cells), E-Cad (adherens junctions) and K10 (differentiation marker) at 3 days, 1 week and 2 weeks after beginning of doxycycline induction in Ctrl papillomas. f: Immunostaining for K14 (epithelial tumor cells), E-Cad (adherens junctions) and K10 (differentiation marker) at 3 days, 1 week and 2 weeks after beginning of doxycycline induction in NR2F2 OE papillomas. Representative images of at least 4 independent biological replicates are shown in e and f. Scale bar = 50 µm, except overview scale bar = 1mm.

Source data

Extended Data Fig. 5 Downstream changes induced by NR2F2 loss of function in mouse SCC.

a: Gene ontology analysis of the genes that are upregulated in NR2F2 KO carcinomas, showing categories that are significantly enriched (dotted line p=0,05). The p-value is calculated according to the Benjamini-Hochberg method for multiple hypothesis testing. b: Bar graph representation of selected genes that are upregulated in NR2F2 KO tumors, grouped by their respective function (Fold change KO/Control; n=2 Ctrl and NR2F2 KO carcinomas, independent biological replicates). c: Immunostaining for K14 (tumor cells) and K8 (progression marker) in NR2F2 KO and Ctrl SCC. d: Quantification of K8 expressing tumor cells in NR2F2 KO and Ctrl SCC (n=22 Ctrl and 13 NR2F2 KO tumors). Scale bar = 50 μm. Data in d are represented as mean ± s.e.m. The p-value is calculated using a two-tailed Mann-Whitney U-test.

Source data

Extended Data Fig. 6 Effects of NR2F2 inhibition in a human skin SCC model.

a: Relative expression level of NR2F2 in A431, SK-BR3 and MDA-MB-468 cells (n=2 biological replicates). Data are normalized to MDA-MB-468. Data are represented as mean ±SEM. b: Viability of A431, SK-BR3 and MDA-MB-468 cells 72h after CIA1 treatment in vitro (n=3 biological replicates). Data are represented as mean ± SD. c: Proportion of secondary tumor formation and tumor propagating cell frequency calculated by using the extreme limiting dilution analysis (ELDA) of human skin SCC cells (A431) following CIA1 treatment. p-value is calculated using a chi-squared test. d: Histology of CIA1 treated xenograft tumors. Immunostaining (left) for PHH3 (mitotic cells) and K14 (Tumor cells) and quantification (right) of PHH3+ cells/mm2 (n=4 Ctrl and 4 CIA1 treated tumors). Scale bar = 50 µm. Data are represented as mean ± s.e.m. The p-value is calculated using the two-tailed Mann-Whitney U-test.

Source data

Extended Data Fig. 7 NR2F2 loss of function characterization in skin human SCC cell lines.

a: WB for NR2F2 and β-actin in A431 control and NR2F2 shRNA KD #1 and #2 human cell lines. b: NR2F2 knockdown alters the growth of spheroids of A431 skin SCC cell lines. Pictures were taken 4 and 11 days after seeding. Representative images of 3 independent experiments. c, d: Wound-healing assay of NR2F2 shRNA KD #1 (c) and #2 (d) following doxycycline induction. Scale bar=500 µm. Representative images of 3 independent experiments. e: qRT-PCR of upregulated and downregulated genes following NR2F2 KD and CIA1 exposure in A431 cell line (n=3 technical replicates). Data are represented as mean ± s.e.m. f: WB for NR2F2, HA and β-actin in A431 NR2F2 GOF human cell line (A431 WT and 3HA-NR2F2 GOF). g: Overlap of the A431 NR2F2 ChIP-seq peaks with the downregulated and upregulated genes in the NR2F2 KD#2 cell line (p-values are calculated using a Two-sided hypergeometric test). h: Overlap of the A431 NR2F2 ChIP-seq peaks with the downregulated and upregulated genes in the NR2F2 KO mouse SCC. p-values are calculated using a Two-sided hypergeometric test. WBs are representative of two independent experiments with similar results in f, and three independent experiments with similar results in a.

Source data

Supplementary information

Reporting Summary

Supplementary Table 1

Sequences of the oligonucleotides used in the manuscript.

Supplementary Data 1

Example of FACS gating strategy as listed in the Reporting Summary.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 7

Unprocessed western blots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mauri, F., Schepkens, C., Lapouge, G. et al. NR2F2 controls malignant squamous cell carcinoma state by promoting stemness and invasion and repressing differentiation. Nat Cancer 2, 1152–1169 (2021). https://doi.org/10.1038/s43018-021-00287-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-021-00287-5

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