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:

Matrix stiffness induces a tumorigenic phenotype in mammary epithelium through changes in chromatin accessibility

Abstract

In breast cancer, the increased stiffness of the extracellular matrix is a key driver of malignancy. Yet little is known about the epigenomic changes that underlie the tumorigenic impact of extracellular matrix mechanics. Here, we show in a three-dimensional culture model of breast cancer that stiff extracellular matrix induces a tumorigenic phenotype through changes in chromatin state. We found that increased stiffness yielded cells with more wrinkled nuclei and with increased lamina-associated chromatin, that cells cultured in stiff matrices displayed more accessible chromatin sites, which exhibited footprints of Sp1 binding, and that this transcription factor acts along with the histone deacetylases 3 and 8 to regulate the induction of stiffness-mediated tumorigenicity. Just as cell culture on soft environments or in them rather than on tissue-culture plastic better recapitulates the acinar morphology observed in mammary epithelium in vivo, mammary epithelial cells cultured on soft microenvironments or in them also more closely replicate the in vivo chromatin state. Our results emphasize the importance of culture conditions for epigenomic studies, and reveal that chromatin state is a critical mediator of mechanotransduction.

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: Nuclear and chromatin alterations accompany phenotypic changes induced by ECM stiffness.
Fig. 2: Chromatin accessibility changes are associated with normal and tumorigenic phenotypes.
Fig. 3: Sp1 mediates the stiffness-induced tumorigenic phenotype.
Fig. 4: HDACs 3 and 8 regulate the stiffness-induced tumorigenic phenotype.
Fig. 5: Soft hydrogel culture produces more physiologically representative chromatin accessibility profiles than standard tissue culture.

Similar content being viewed by others

Data availability

The authors declare that the main data supporting the results in this study are available within the paper and its Supplementary Information. ATAC-seq data generated in this study are available through the Gene Expression Omnibus under accession code GSE131968. Additional datasets are available from the corresponding author upon reasonable request.

Code availability

Custom analysis pipelines are available at https://github.com/kundajelab.

References

  1. 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 

  2. Provenzano, P. P., Inman, D. R., Eliceiri, K. W. & Keely, P. J. Matrix density-induced mechanoregulation of breast cell phenotype, signaling and gene expression through a FAK–ERK linkage. Oncogene 28, 4326–4343 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Choquet, D., Felsenfeld, D. P. & Sheetz, M. P. Extracellular matrix rigidity causes strengthening of integrin–cytoskeleton linkages. Cell 88, 39–48 (1997).

    Article  CAS  PubMed  Google Scholar 

  4. Paszek, M. J. et al. Tensional homeostasis and the malignant phenotype. Cancer Cell 8, 241–254 (2005).

    Article  CAS  PubMed  Google Scholar 

  5. Del Rio, A. et al. Stretching single talin rod molecules activates vinculin binding. Science 323, 638–641 (2009).

    Article  CAS  PubMed  Google Scholar 

  6. Elosegui-Artola, A. et al. Mechanical regulation of a molecular clutch defines force transmission and transduction in response to matrix rigidity. Nat. Cell Biol. 18, 540–548 (2016).

    Article  CAS  PubMed  Google Scholar 

  7. Aragona, M. et al. A mechanical checkpoint controls multicellular growth through YAP/TAZ regulation by actin-processing factors. Cell 154, 1047–1059 (2013).

    Article  CAS  PubMed  Google Scholar 

  8. Chaudhuri, O. et al. Extracellular matrix stiffness and composition jointly regulate the induction of malignant phenotypes in mammary epithelium. Nat. Mater. 13, 970–978 (2014).

    Article  CAS  PubMed  Google Scholar 

  9. Uhler, C. & Shivashankar, G. V. Regulation of genome organization and gene expression by nuclear mechanotransduction. Nat. Rev. Mol. Cell Biol. 18, 717–727 (2017).

    Article  CAS  PubMed  Google Scholar 

  10. Maniatis, T., Goodbourn, S. & Fischer, J. A. Regulation of inducible and tissue-specific gene expression. Science 236, 1237–1245 (1987).

    Article  CAS  PubMed  Google Scholar 

  11. Pique-Regi, R. et al. Accurate inference of transcription factor binding from DNA sequence and chromatin accessibility data. Genome Res. 21, 447–455 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Kouzarides, T. Chromatin modifications and their function. Cell 128, 693–705 (2007).

    Article  CAS  PubMed  Google Scholar 

  13. Swift, J. et al. Nuclear lamin-A scales with tissue stiffness and enhances matrix-directed differentiation. Science 341, 1240104 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Jain, N., Iyer, K. V., Kumar, A. & Shivashankar, G. V. Cell geometric constraints induce modular gene-expression patterns via redistibution of HDAC3 regulated by actomyosin contractility. Proc. Natl Acad. Sci. USA 110, 11349–11354 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Li, Y., Tang, C. B. & Kilian, K. A. Matrix mechanics influence fibroblast–myofibroblast transition by directing the localization of histone deacetylase 4. Cell. Mol. Bioeng. 10, 405–415 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Flavahan, W. A., Gaskell, E. & Bernstein, B. E. Epigenetic plasticity and the hallmarks of cancer. Science 357, eaal2380 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. McDonald, O. G., Wu, H., Timp, W., Doi, A. & Feinberg, A. P. Genome-scale epigenetic reprogramming during epithelial-to-mesenchymal transition. Nat. Struct. Mol. Biol. 18, 867–874 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Tajik, A. et al. Transcription upregulation via force-induced direct stretching of chromatin. Nat. Mater. 15, 1287–1296 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Gallo, M. et al. MLL5 orchestrates a cancer self-renewal state by repressing the histone variant H3.3 and globally reorganizing chromatin. Cancer Cell 28, 715–729 (2015).

    Article  CAS  PubMed  Google Scholar 

  20. Buenrostro, J. D., Giresi, P. G., Zaba, L. C., Chang, H. Y. & Greenleaf, W. J. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods 10, 1213–1218 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Ooi, J. Y. et al. HDAC inhibition attenuates cardiac hypertrophy by acetylation and deacetylation of target genes. Epigenetics 10, 418–430 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Rafehi, H. et al. Systems approach to the pharmacological actions of HDAC inhibitors reveals EP300 activities and convergent mechanisms of regulation in diabetes. Epigenetics 12, 991–1003 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  23. Fork, C. et al. Epigenetic control of microsomal prostaglandin E synthase-1 by HDAC-mediated recruitment of p300. J. Lipid Res. 58, 386–392 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Rafehi, H. et al. Vascular histone deacetylation by pharmacological HDAC inhibition. Genome Res. 24, 1271–1284 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Rafehi, H. & El-Osta, A. HDAC inhibition in vascular endothelial cells regulates the expression of ncRNAs. Noncoding RNA 2, 4 (2016).

    PubMed Central  Google Scholar 

  26. Chen, F. et al. Inhibition of histone deacetylase reduces transcription of NADPH oxidases and ROS production and ameliorates pulmonary arterial hypertension. Free Radic. Biol. Med 99, 167–178 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Qu, K. et al. Chromatin accessibility landscape of cutaneous T cell lymphoma and dynamic response to HDAC inhibitors. Cancer Cell 32, 27–41 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Kaczynski, J., Cook, T. & Urrutia, R. Sp1- and Kruppel-like transcription factors. Genome Biol. 4, 1–8 (2003).

    Article  Google Scholar 

  29. Han, H. et al. TRRUSTv2: an expanded reference database of human and mouse transcriptional regulatory interactions. Nucleic Acids Res. 46, D380–D386 (2018).

    Article  CAS  PubMed  Google Scholar 

  30. Hsu, M. C., Chang, H. C. & Hung, W. C. HER-2/neu represses the metastasis suppressor RECK via ERK and Sp transcription factors to promote cell invasion. J. Biol. Chem. 281, 4718–4725 (2006).

    Article  CAS  PubMed  Google Scholar 

  31. Stowers, R. S. et al. Extracellular matrix stiffening induces a malignant phenotypic transition in breast epithelial cells. Cell. Mol. Bioeng. 10, 114–123 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Zhang, Y., Liao, M. & Dufau, M. L. Phosphatidylinositol 3-kinase/protein kinase Cζ-induced phosphorylation of Sp1 and p107 repressor release have a critical role in histone deacetylase inhibitor-mediated derepression of transcription of the luteinizing hormone receptor gene. Mol. Cell Biol. 26, 6748–6761 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Doetzlhofer, A. et al. Histone deacetylase 1 can repress transcription by binding to Sp1. Mol. Cell Biol. 19, 5504–5511 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Liu, S. et al. Sp1/NFκB/HDAC/miR-29b regulatory network in KIT-driven myeloid leukemia. Cancer Cell 17, 333–347 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Petersen, O. W., Ronnov-Jessen, L., Howlett, A. R. & Bissell, M. J. Interaction with basement membrane serves to rapidly distinguish growth and differentiation pattern of normal and malignant human breast epithelial cells. Proc. Natl Acad. Sci. USA 89, 9064–9068 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Uhlen, M. et al. Tissue-based map of the human proteome. Science 347, 1260419 (2015).

    Article  PubMed  CAS  Google Scholar 

  37. Heo, S. J. et al. Biophysical regulation of chromatin architecture instills a mechanical memory in mesenchymal stem cells. Sci. Rep. 5, 16895 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Jain, N., Iyer, K. V., Kumar, A. & Shivashankar, G. V. Cell geometric constraints induce modular gene-expression patterns via redistribution of HDAC3 regulated by actomyosin contractility. Proc. Natl Acad. Sci. USA 110, 11349–11354 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Demmerle, J., Koch, A. J. & Holaska, J. M. The nuclear envelope protein emerin binds directly to histone deacetylase 3 (HDAC3) and activates HDAC3 activity. J. Biol. Chem. 287, 22080–22088 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Le, H. Q. et al. Mechanical regulation of transcription controls Polycomb-mediated gene silencing during lineage commitment. Nat. Cell Biol. 18, 864–875 (2016).

    Article  CAS  PubMed  Google Scholar 

  41. Xu, R. et al. Sustained activation of STAT5 is essential for chromatin remodeling and maintenance of mammary-specific function. J. Cell Biol. 184, 57–66 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Le Beyec, J. et al. Cell shape regulates global histone acetylation in human mammary epithelial cells. Exp. Cell Res. 313, 3066–3075 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Fraga, M. F. et al. Loss of acetylation at Lys16 and trimethylation at Lys20 of histone H4 is a common hallmark of human cancer. Nat. Genet. 37, 391–400 (2005).

    Article  CAS  PubMed  Google Scholar 

  44. Bibikova, M. et al. Human embryonic stem cells have a unique epigenetic signature. Genome Res. 16, 1075–1083 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Wisdom, K. & Chaudhuri, O. 3D cell culture in interpenetrating networks of alginate and rBM matrix. Methods Mol. Biol. 1612, 29–37 (2017).

    Article  CAS  PubMed  Google Scholar 

  46. Thevenaz, P., Ruttimann, U. E. & Unser, M. A pyramid approach to subpixel registration based on intensity. IEEE Trans. Image Process. 7, 27–41 (1998).

    Article  CAS  PubMed  Google Scholar 

  47. Thielicke, W. & Stamhuis, E. J. Towards user-friendly, affordable and accurate digital particle image velocimetry in MATLAB. J. Open Res. Softw. 2, e30 (2014).

    Article  Google Scholar 

  48. Debnath, J., Muthuswamy, S. K. & Brugge, J. S. Morphogenesis and oncogenesis of MCF-10A mammary epithelial acini grown in three-dimensional basement membrane cultures. Methods 30, 256–268 (2003).

    Article  CAS  PubMed  Google Scholar 

  49. Buenrostro, J. D., Wu, B., Chang, H. Y. & Greenleaf, W. J. ATAC-seq: a method for assaying chromatin accessibility genome-wide. Curr. Protoc. Mol. Biol. 109, 21–29 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  50. 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 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Machanick, P. & Bailey, T. L. MEME-ChIP: motif analysis of large DNA datasets. Bioinformatics 27, 1696–1697 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. 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 

  55. Szklarczyk, D. et al. STRING v10: protein–protein interaction networks, integrated over the tree of life. Nucleic Acids Res. 43, D447–D452 (2015).

    Article  CAS  PubMed  Google Scholar 

  56. ACTB: Tissue-breast (Human Protein Atlas, 2019); https://www.proteinatlas.org/ENSG00000075624-ACTB/tissue/breast#

Download references

Acknowledgements

The authors acknowledge helpful discussions with the Chaudhuri laboratory, C. Hsueh for assistance with nuclear curvature analysis, M. Black for assistance with chromatin thickness analysis, the Bollyky laboratory for use of the Licor imager, the Stanford Genome Sequencing Service Center for sequencing (NIH S10OD020141), and the Stanford Cell Science Imaging Facility for assistance with confocal image analysis and TEM preparation and imaging (NIH 1S10RR026780-01). This work was supported by an NIH F32 fellowship to R.S.S. (F32CA210431), and an American Cancer Society grant (RSG-16-028-01) and NIH grant (R37-CA214136) to O.C.

Author information

Authors and Affiliations

Authors

Contributions

R.S.S. and O.C. conceived the study. R.S.S., A.S., A.K. and O.C. wrote the manuscript. R.S.S., J.J.G., J.C., A.R., M.N.T., M.P.S. and O.C. designed the experiments. R.S.S., J.J.G., J.C., A.R. and S.N. conducted experiments. R.S.S., A.S., J.I., J.C., A.K. and O.C. analysed the data.

Corresponding author

Correspondence to Ovijit Chaudhuri.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Supplementary information

Supplementary Information

Supplementary figures and tables.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Stowers, R.S., Shcherbina, A., Israeli, J. et al. Matrix stiffness induces a tumorigenic phenotype in mammary epithelium through changes in chromatin accessibility. Nat Biomed Eng 3, 1009–1019 (2019). https://doi.org/10.1038/s41551-019-0420-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41551-019-0420-5

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