The Rho GTPase Rnd1 suppresses mammary tumorigenesis and EMT by restraining Ras-MAPK signalling (original) (raw)

Accession codes

Accessions

Gene Expression Omnibus

References

  1. Collado, M. & Serrano, M. Senescence in tumours: evidence from mice and humans. Nat. Rev. Cancer 10, 51–57 (2010).
    CAS PubMed PubMed Central Google Scholar
  2. Huang, L. & Muthuswamy, S. K. Polarity protein alterations in carcinoma: a focus on emerging roles for polarity regulators. Curr. Opin. Genet. Dev. 20, 41–50 (2010).
    Article CAS PubMed PubMed Central Google Scholar
  3. Thiery, J. P., Acloque, H., Huang, R. Y. & Nieto, M. A. Epithelial–mesenchymal transitions in development and disease. Cell 139, 871–890 (2009).
    Article CAS PubMed Google Scholar
  4. Ansieau, S. et al. Induction of EMT by twist proteins as a collateral effect of tumor-promoting inactivation of premature senescence. Cancer Cell 14, 79–89 (2008).
    Article CAS PubMed Google Scholar
  5. Debnath, J. et al. The role of apoptosis in creating and maintaining luminal space within normal and oncogene-expressing mammary acini. Cell 111, 29–40 (2002).
    Article CAS PubMed Google Scholar
  6. Guo, W. et al. β4 integrin amplifies ErbB2 signaling to promote mammary tumorigenesis. Cell 126, 489–502 (2006).
    Article CAS PubMed Google Scholar
  7. Dow, L. E. et al. Loss of human Scribble cooperates with H-Ras to promote cell invasion through deregulation of MAPK signalling. Oncogene 27, 5988–6001 (2008).
    Article CAS PubMed Google Scholar
  8. Zhan, L. et al. Deregulation of scribble promotes mammary tumorigenesis and reveals a role for cell polarity in carcinoma. Cell 135, 865–878 (2008).
    Article CAS PubMed PubMed Central Google Scholar
  9. Yang, J. et al. Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell 117, 927–939 (2004).
    Article CAS PubMed Google Scholar
  10. Moody, S. E. et al. The transcriptional repressor Snail promotes mammary tumor recurrence. Cancer Cell 8, 197–209 (2005).
    Article CAS PubMed Google Scholar
  11. Jaffe, A. B. & Hall, A. Rho GTPases: biochemistry and biology. Annu. Rev. Cell Dev. Biol. 21, 247–269 (2005).
    Article CAS PubMed Google Scholar
  12. Sahai, E. & Marshall, C. J. RHO-GTPases and cancer. Nat. Rev. Cancer 2, 133–142 (2002).
    Article PubMed Google Scholar
  13. Minn, A. J. et al. Genes that mediate breast cancer metastasis to lung. Nature 436, 518–524 (2005).
    Article CAS PubMed PubMed Central Google Scholar
  14. Aguirre, A. J. et al. High-resolution characterization of the pancreatic adenocarcinoma genome. Proc. Natl Acad. Sci. USA 101, 9067–9072 (2004).
    Article CAS PubMed PubMed Central Google Scholar
  15. Rutherford, S., Hampton, G. M., Frierson, H. F. & Moskaluk, C. A. Mapping of candidate tumor suppressor genes on chromosome 12 in adenoid cystic carcinoma. Lab. Invest. 85, 1076–1085 (2005).
    Article CAS PubMed Google Scholar
  16. Yanagisawa, M. et al. A p120 catenin isoform switch affects Rho activity, induces tumor cell invasion, and predicts metastatic disease. J. Biol. Chem. 283, 18344–18354 (2008).
    Article CAS PubMed PubMed Central Google Scholar
  17. Van ’t Veer, L. J. et al. Gene expression profiling predicts clinical outcome of breast cancer. Nature 415, 530–536 (2002).
    Article PubMed Google Scholar
  18. Gyorffy, B. et al. An online survival analysis tool to rapidly assess the effect of 22,277 genes on breast cancer prognosis using microarray data of 1,809 patients. Breast Cancer Res. Treat. 123, 725–731 (2010).
    Article PubMed CAS Google Scholar
  19. Chardin, P. Function and regulation of Rnd proteins. Nat. Rev. Mol. Cell Biol. 7, 54–62 (2006).
    Article CAS PubMed Google Scholar
  20. Campisi, J. & d’Adda di Fagagna, F. Cellular senescence: when bad things happen to good cells. Nat. Rev. Mol. Cell Biol. 8, 729–740 (2007).
    Article CAS PubMed Google Scholar
  21. Luo, J. et al. A genome-wide RNAi screen identifies multiple synthetic lethal interactions with the Ras oncogene. Cell 137, 835–848 (2009).
    Article CAS PubMed PubMed Central Google Scholar
  22. Jonsson, G. et al. High-resolution genomic profiles of breast cancer cell lines assessed by tiling BAC array comparative genomic hybridization. Genes Chromosomes Cancer 46, 543–558 (2007).
    Article PubMed CAS Google Scholar
  23. Bretones, G. et al. SKP2 oncogene is a direct MYC target gene and MYC down-regulates p27(KIP1) through SKP2 in human leukemia cells. J. Biol. Chem. 286, 9815–9825 (2011).
    Article CAS PubMed PubMed Central Google Scholar
  24. Elenbaas, B. et al. Human breast cancer cells generated by oncogenic transformation of primary mammary epithelial cells. Genes Dev. 15, 50–65 (2001).
    Article CAS PubMed PubMed Central Google Scholar
  25. Podsypanina, K., Politi, K., Beverly, L. J. & Varmus, H. E. Oncogene cooperation in tumor maintenance and tumor recurrence in mouse mammary tumors induced by Myc and mutant Kras. Proc. Natl Acad. Sci. USA 105, 5242–5247 (2008).
    Article CAS PubMed PubMed Central Google Scholar
  26. Courtois-Cox, S. et al. A negative feedback signaling network underlies oncogene-induced senescence. Cancer Cell 10, 459–472 (2006).
    Article CAS PubMed PubMed Central Google Scholar
  27. Cichowski, K., Santiago, S., Jardim, M., Johnson, B. W. & Jacks, T. Dynamic regulation of the Ras pathway via proteolysis of the NF1 tumor suppressor. Genes Dev. 17, 449–454 (2003).
    Article CAS PubMed PubMed Central Google Scholar
  28. McCormack, J., Welsh, N. J. & Braga, V. M. Cycling around cell–cell adhesion with Rho GTPase regulators. J. Cell Sci. 126, 379–391 (2013).
    Article CAS PubMed Google Scholar
  29. Shin, S., Dimitri, C. A., Yoon, S. O., Dowdle, W. & Blenis, J. ERK2 but not ERK1 induces epithelial-to-mesenchymal transformation via DEF motif-dependent signaling events. Mol. Cell 38, 114–127 (2010).
    Article CAS PubMed PubMed Central Google Scholar
  30. Wennerberg, K. et al. Rnd proteins function as RhoA antagonists by activating p190 RhoGAP. Curr. Biol. 13, 1106–1115 (2003).
    Article CAS PubMed PubMed Central Google Scholar
  31. Oinuma, I., Ishikawa, Y., Katoh, H. & Negishi, M. The semaphorin 4D receptor plexin-B1 is a GTPase activating protein for R-Ras. Science 305, 862–865 (2004).
    Article CAS PubMed Google Scholar
  32. Tong, Y. et al. Binding of Rac1, Rnd1, and RhoD to a novel Rho GTPase interaction motif destabilizes dimerization of the plexin-B1 effector domain. J. Biol. Chem. 282, 37215–37224 (2007).
    Article CAS PubMed Google Scholar
  33. Tamagnone, L. Emerging role of semaphorins as major regulatory signals and potential therapeutic targets in cancer. Cancer Cell 22, 145–152 (2012).
    Article CAS PubMed Google Scholar
  34. Giordano, S. et al. The semaphorin 4D receptor controls invasive growth by coupling with Met. Nat. Cell Biol. 4, 720–724 (2002).
    Article CAS PubMed Google Scholar
  35. Swiercz, J. M., Kuner, R. & Offermanns, S. Plexin-B1/RhoGEF-mediated RhoA activation involves the receptor tyrosine kinase ErbB-2. J. Cell Biol. 165, 869–880 (2004).
    Article CAS PubMed PubMed Central Google Scholar
  36. Wang, Y. et al. Plexins are GTPase-activating proteins for Rap and are activated by induced dimerization. Sci. Signal. 5, ra6 (2012).
    PubMed PubMed Central Google Scholar
  37. Wang, Y., Pascoe, H. G., Brautigam, C. A., He, H. & Zhang, X. Structural basis for activation and non-canonical catalysis of the Rap GTPase activating protein domain of plexin. eLife 2, e01279 (2013).
    Article PubMed PubMed Central CAS Google Scholar
  38. Frech, M. et al. Inhibition of GTPase activating protein stimulation of Ras-p21 GTPase by the Krev-1 gene product. Science 249, 169–171 (1990).
    Article CAS PubMed Google Scholar
  39. Hata, Y. et al. Inhibition of the ras p21 GTPase-activating protein-stimulated GTPase activity of c-Ha-ras p21 by smg p21 having the same putative effector domain as ras p21s. J. Biol. Chem. 265, 7104–7107 (1990).
    CAS PubMed Google Scholar
  40. Danielson, K. G., Oborn, C. J., Durban, E. M., Butel, J. S. & Medina, D. Epithelial mouse mammary cell line exhibiting normal morphogenesis in vivo and functional differentiation in vitro. Proc. Natl Acad. Sci. USA 81, 3756–3760 (1984).
    Article CAS PubMed PubMed Central Google Scholar
  41. Barcellos-Hoff, M. H. & Ravani, S. A. Irradiated mammary gland stroma promotes the expression of tumorigenic potential by unirradiated epithelial cells. Cancer Res. 60, 1254–1260 (2000).
    CAS PubMed Google Scholar
  42. Giancotti, F. G. Mechanisms governing metastatic dormancy and reactivation. Cell 155, 750–764 (2013).
    Article CAS PubMed PubMed Central Google Scholar
  43. Yao, J. et al. Combined cDNA array comparative genomic hybridization and serial analysis of gene expression analysis of breast tumor progression. Cancer Res. 66, 4065–4078 (2006).
    Article CAS PubMed Google Scholar
  44. Sakai, W. et al. Secondary mutations as a mechanism of cisplatin resistance in BRCA2-mutated cancers. Nature 451, 1116–1120 (2008).
    Article CAS PubMed PubMed Central Google Scholar
  45. Tan, J. et al. Pharmacologic disruption of Polycomb-repressive complex 2-mediated gene repression selectively induces apoptosis in cancer cells. Genes Dev. 21, 1050–1063 (2007).
    Article CAS PubMed PubMed Central Google Scholar
  46. Ahearn, I. M., Haigis, K., Bar-Sagi, D. & Philips, M. R. Regulating the regulator: post-translational modification of RAS. Nat. Rev. Mol. Cell Biol. 13, 39–51 (2012).
    Article CAS Google Scholar
  47. Hydbring, P. et al. Phosphorylation by Cdk2 is required for Myc to repress Ras-induced senescence in cotransformation. Proc. Natl Acad. Sci. USA 107, 58–63 (2010).
    Article CAS PubMed Google Scholar
  48. Serrano, M., Lin, A. W., McCurrach, M. E., Beach, D. & Lowe, S. W. Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell 88, 593–602 (1997).
    Article CAS PubMed Google Scholar
  49. Ohtsuka, T., Shimizu, K., Yamamori, B., Kuroda, S. & Takai, Y. Activation of brain B-Raf protein kinase by Rap1B small GTP-binding protein. J. Biol. Chem. 271, 1258–1261 (1996).
    Article CAS PubMed Google Scholar
  50. Vossler, M. R. et al. cAMP activates MAP kinase and Elk-1 through a B-Raf- and Rap1-dependent pathway. Cell 89, 73–82 (1997).
    Article CAS PubMed Google Scholar
  51. York, R. D. et al. Rap1 mediates sustained MAP kinase activation induced by nerve growth factor. Nature 392, 622–626 (1998).
    Article CAS PubMed Google Scholar
  52. Mishra, S., Smolik, S. M., Forte, M. A. & Stork, P. J. Ras-independent activation of ERK signaling via the torso receptor tyrosine kinase is mediated by Rap1. Curr. Biol. 15, 366–370 (2005).
    Article CAS PubMed Google Scholar
  53. Itoh, M., Nelson, C. M., Myers, C. A. & Bissell, M. J. Rap1 integrates tissue polarity, lumen formation, and tumorigenic potential in human breast epithelial cells. Cancer Res. 67, 4759–4766 (2007).
    Article CAS PubMed PubMed Central Google Scholar
  54. Forbes, S. A. et al. COSMIC (the Catalogue of Somatic Mutations in Cancer): a resource to investigate acquired mutations in human cancer. Nucleic Acids Res. 38, D652–D657 (2010).
    Article CAS PubMed Google Scholar
  55. Eckert, L. B. et al. Involvement of Ras activation in human breast cancer cell signaling, invasion, and anoikis. Cancer Res. 64, 4585–4592 (2004).
    Article CAS PubMed Google Scholar
  56. Rody, A. et al. Poor outcome in estrogen receptor-positive breast cancers predicted by loss of plexin B1. Clin. Cancer Res. 13, 1115–1122 (2007).
    Article CAS PubMed Google Scholar
  57. Argast, G. M. et al. Plexin B1 is repressed by oncogenic B-Raf signaling and functions as a tumor suppressor in melanoma cells. Oncogene 28, 2697–2709 (2009).
    Article CAS PubMed PubMed Central Google Scholar
  58. Wong, O. G. et al. Plexin-B1 mutations in prostate cancer. Proc. Natl Acad. Sci. USA 104, 19040–19045 (2007).
    Article CAS PubMed PubMed Central Google Scholar
  59. Banerji, S. et al. Sequence analysis of mutations and translocations across breast cancer subtypes. Nature 486, 405–409 (2012).
    Article CAS PubMed PubMed Central Google Scholar
  60. Tong, Y., Hota, P. K., Hamaneh, M. B. & Buck, M. Insights into oncogenic mutations of plexin-B1 based on the solution structure of the Rho GTPase binding domain. Structure 16, 246–258 (2008).
    Article CAS PubMed PubMed Central Google Scholar
  61. Gao, J. et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci. Signal. 6, pl1 (2013).
    Article PubMed PubMed Central CAS Google Scholar
  62. Richardson, A. L. et al. X chromosomal abnormalities in basal-like human breast cancer. Cancer Cell 9, 121–132 (2006).
    Article CAS PubMed Google Scholar
  63. Chin, K. et al. Genomic and transcriptional aberrations linked to breast cancer pathophysiologies. Cancer Cell 10, 529–541 (2006).
    Article CAS PubMed Google Scholar
  64. Lu, X. et al. Predicting features of breast cancer with gene expression patterns. Breast Cancer Res. Treat. 108, 191–201 (2008).
    Article CAS PubMed Google Scholar
  65. Wang, Y. et al. Gene-expression profiles to predict distant metastasis of lymph-node negative primary breast cancer. Lancet 365, 671–679 (2005).
    Article CAS PubMed Google Scholar
  66. Ginestier, C. et al. Prognosis and gene expression profiling of 20q13-amplified breast cancers. Clin. Cancer Res. 12, 4533–4544 (2006).
    Article CAS PubMed Google Scholar
  67. Farmer, P. et al. Identification of molecular apocrine breast tumors by microarray analysis. Oncogene 24, 4660–4671 (2005).
    Article CAS PubMed Google Scholar
  68. Hess, K. R. et al. Pharmacogenomic predictor of sensitivity to preoperative chemotherapy with paclitaxel and fluorouracil. J. Clin. Oncol. 24, 4263–4244 (2006).
    Article CAS Google Scholar
  69. Neve, R. M. et al. A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell 10, 515–527 (2006).
    Article CAS PubMed PubMed Central Google Scholar
  70. 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
  71. Rhodes, D. R. et al. ONCOMINE: a cancer microarray database and integrated data-mining platform. Neoplasia 6, 1–6 (2004).
    Article CAS PubMed PubMed Central Google Scholar
  72. Bild, A. H. et al. Oncogenic pathway signatures in human cancers as a guide to targeted therapies. Nature 439, 353–357 (2006).
    Article CAS PubMed Google Scholar
  73. 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
  74. Bos, P. D. et al. Genes that mediate breast cancer metastasis to the brain. Nature 459, 1005–1009 (2009).
    Article CAS PubMed PubMed Central Google Scholar

Download references

Acknowledgements

We thank X. Zhang for advice, reagents, and for sharing the structure of the Plexin-GAP–Rap1 complex before its publication, R. A. Weinberg, J. S. Brugge, M. Overholtzer, A. Ridley, B. Park, L. Tamagnome, M. Resh, H. Djaballah, T. Kataoka, M. Negishi and D. Medina for reagents, K. Manova for assistance with confocal microscopy, R. Khanin and G. P. Gupta for help with multivariate analysis, M. Buck for comments on the manuscript, and members of the Giancotti laboratory for discussions. We thank the Geoffrey Beene Translational Oncology Core, the Genomic Core, and the HTG Core of MSKCC for technical assistance. This work was supported by grants from the National Institutes of Health (P01 CA094060 Project 4 to F.G.G. and P30 CA08748 to MSKCC) and the Geoffrey Beene Cancer Center at MSKCC.

Author information

Author notes

  1. Tomoyo Okada, Gaia Schiavon, Miguel A. López-Lago, Christine A. Pratilas, Adriana Heguy & Giorgio Inghirami
    Present address: Present addresses: Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York 10065, USA (T.O.); Breast Unit, Royal Marsden Hospital, London SW3 6JJ, UK (G.S.); Department of Surgery and Center for Cell Engineering, Memorial Sloan-Kettering Cancer Center, New York 10065, USA (M.A.L-L.); The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA (C.A.P.); Department of Pathology, Office for Collaborative Science, New York University, New York 10016, USA (A.H.); Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York 10065, USA (G.I.).,
  2. Tomoyo Okada, Surajit Sinha and Ilaria Esposito: These authors contributed equally to this work.

Authors and Affiliations

  1. Cell Biology Program, Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA
    Tomoyo Okada, Surajit Sinha, Ilaria Esposito, Gaia Schiavon, Miguel A. López-Lago, Wenjing Su, Jonathan M. Hernandez & Filippo G. Giancotti
  2. Molecular Pharmacology and Chemistry Program, Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA
    Christine A. Pratilas & Neal Rosen
  3. Department of Pediatrics, Memorial Hospital, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA
    Christine A. Pratilas
  4. Department of Biomedical Sciences and Human Oncology, Center of Experimental Medicine and Research, University of Torino, Torino 10126, Italy
    Cristina Abele & Giorgio Inghirami
  5. Department of Surgery, Memorial Hospital, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA
    Jonathan M. Hernandez
  6. Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima 734-8551, Japan
    Masahiro Ohara & Morihito Okada
  7. Genomics Core Facility, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA
    Agnes Viale
  8. Geoffrey Beene Translational Oncology Core Facility, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA
    Adriana Heguy
  9. Bioinformatics Core Facility, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA
    Nicholas D. Socci
  10. Department of Medical Sciences, Center of Experimental Medicine and Research, University of Torino, Torino 10126, Italy
    Anna Sapino
  11. Department of Epidemiology and Biostatistics, Memorial Hospital, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA
    Venkatraman E. Seshan
  12. Structural Biology Program, Sloan-Kettering Institute for Cancer Research, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA
    Stephen Long
  13. Department of Medicine, Memorial Hospital, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA
    Neal Rosen

Authors

  1. Tomoyo Okada
    You can also search for this author inPubMed Google Scholar
  2. Surajit Sinha
    You can also search for this author inPubMed Google Scholar
  3. Ilaria Esposito
    You can also search for this author inPubMed Google Scholar
  4. Gaia Schiavon
    You can also search for this author inPubMed Google Scholar
  5. Miguel A. López-Lago
    You can also search for this author inPubMed Google Scholar
  6. Wenjing Su
    You can also search for this author inPubMed Google Scholar
  7. Christine A. Pratilas
    You can also search for this author inPubMed Google Scholar
  8. Cristina Abele
    You can also search for this author inPubMed Google Scholar
  9. Jonathan M. Hernandez
    You can also search for this author inPubMed Google Scholar
  10. Masahiro Ohara
    You can also search for this author inPubMed Google Scholar
  11. Morihito Okada
    You can also search for this author inPubMed Google Scholar
  12. Agnes Viale
    You can also search for this author inPubMed Google Scholar
  13. Adriana Heguy
    You can also search for this author inPubMed Google Scholar
  14. Nicholas D. Socci
    You can also search for this author inPubMed Google Scholar
  15. Anna Sapino
    You can also search for this author inPubMed Google Scholar
  16. Venkatraman E. Seshan
    You can also search for this author inPubMed Google Scholar
  17. Stephen Long
    You can also search for this author inPubMed Google Scholar
  18. Giorgio Inghirami
    You can also search for this author inPubMed Google Scholar
  19. Neal Rosen
    You can also search for this author inPubMed Google Scholar
  20. Filippo G. Giancotti
    You can also search for this author inPubMed Google Scholar

Contributions

T.O., S.S., I.E., G.S., M.A.L-L., W.S., C.A.P., J.M.H. and C.A. performed and interpreted experiments; M.Ohara, M.Okada and A.S. provided annotated breast tumour samples, A.V. designed and performed genomic analyses; G.S. and V.E.S. performed bioinformatic analyses; S.L. provided structural insight; G.I. examined the results of FISH experiments; N.R. and G.I. supervised some of the experiments; F.G.G. supervised the entire study and wrote the paper.

Corresponding author

Correspondence toFilippo G. Giancotti.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 8 Silencing of Rnd1 induces EMT in MCF-10A, HMLE, and NMuMG mammary epithelial cells and RND1 is downregulated in aggressive and metastatic breast cancers.

(a) Quantification of scattering in control and Rnd1-silenced MCF-10A cells from Fig. 1c. (be) Ectopic expression of Rnd1 rescues Rnd1-silenced cells from EMT. Q-PCR analysis of RND1 in control and Rnd1-silenced MCF-10A cells transduced with a control vector or one driving re-expression of Rnd1 (b). Phase Contrast images (top) or immunostaining (bottom) for E-cadherin (Red), Phalloidin (actin, Green), and DAPI (Scale bar, 50 μm for top panels and 15 μm for middle and bottom panels) (c). Quantification of scattering (d) and immunoblotting for E-cadherin and Rho-GDI (e). (fh) Silencing of Rnd1 induces EMT traits in NMuMG cells. Control and Rnd1-silenced NMuMG cells were subjected to Q-PCR for mouse RND1 (f), immunoblotting for E-cadherin, N-cadherin and Rho-GDI (g), and immunostaining for E-cadherin (Red), β-catenin (Green) and DAPI (top) or Zo-1 (Green) and DAPI (bottom). Scale bar, 15 μm (h). (i,j) Silencing of Rnd1 induces EMT traits in HMLE cells. Control and Rnd1-silenced HMLE cells were subjected to immunoblotting for E-cadherin, Vimentin, and Rho-GDI (i) or to immunostaining for E-cadherin (Red) and DAPI (top) or β-catenin (Green) and DAPI (bottom). Scale bar, 15 μm (j). (k,l) MCF-10A cells were transduced with a control shRNA (sh-Co.) or two shRNAs targeting RhoD (sh #1 and #2) and subjected to Q-PCR for RHOD (k) or photographed under Phase Contrast. The arrows point to apoptotic cells in the field. Scale bar, 50 μm (l). (m) Correlation between RND1 mRNA levels and the 70 Genes or the Lung Metastasis poor prognosis signature (LMS) in the MSKCC dataset (n = 117). (n) Patients from the MSKCC dataset were divided according to the Z-score for RND1 expression as indicated. (o) Kaplan–Meier analysis of the correlation between expression of RND1 and metastasis-free survival (left), lung metastasis-free survival (middle) and bone metastasis-free survival (right) in the MSKCC cohort. See Methods for statistical analysis. (p) Q-PCR for RND1 and GATA3 in a panel of breast cancer cell lines of the indicated transcriptomic subgroups. Panels a and d show averages and SD from n = 3 independent experiments. Panels b, c, e, g, and h show one representative experiment out of three performed, whereas panels, f, i, j, k, I, and p show one representative experiment out of two performed. Error bars indicate SD. P values by the Student’s _t_-test are: ∗, P = < 0.05; ∗∗, P = < 0.01; ∗∗∗, P < 0.001. For source data, see Supplementary Table 8. Uncropped images of blots are shown in Supplementary Fig. 9.

Supplementary Figure 9 Rnd1 is upregulated by cytostatic stimuli and inhibits cell cycle progression.

(a) MCF-10A cells were incubated with mitogens (5 ng/ml EGF, 10 μg/ml Insulin, 0.5 μg/ml Hydrocortisone, and Bovine Pituitary Extract) or TGF-β (10 ng/ml), plated under either sparse or confluent conditions, or exposed to UV (100 Joules/cm2) for the indicated times, and subjected to Q-PCR for RND1 mRNA. The graphs show average values from one experiment in which triplicate samples were assessed in parallel. The experiments on the left and right panels were repeated 2 times and the one in the middle panel once. (b,c) MCF-10A cells were infected with a retrovirus encoding HA-Rnd1 or with empty vector and subjected to immunoblotting as indicated (b) or to MTT assay at the indicated times. The graph shows the averages and SD of n = 6 technical replicates. The experiment was repeated 2 times (c). (d) MCF-10A cells transduced with either a control shRNA or 2 shRNAs targeting Rnd1 were cultured for 3 days and subjected to DNA microarray analysis. Genes concomitantly upregulated or downregulated by >1.5 fold were subjected to unsupervised hierarchical clustering with Partek. 59 were downregulated and 117 upregulated (left). Ingenuity Pathways Analysis (IPA) was conducted to assign the genes to the functional categories of Biofunctions and Canonical Pathways. The graphs show the Biofunctions and Canonical Pathways with _P_-value < 0.05 (right-tailed Fisher Exact Test). Genes in red are upregulated and those in blue downregulated. See Supplementary Table 3 for the gene list. (e) Control (Co.) and Rnd1-silenced (#1 and #2) MCF-10A cells were cultured for 5 days and subjected to Q-PCR for indicated cell cycle genes or (f) immunoblotting as indicated. (g) Control (sh-Co.) and Rnd1-silenced (sh #1 and #2) MCF-10A cells were subjected to immunoblotting with antibodies to p27, p21 and p53 at the indicated times after infection. Panels b, f, and g show representative results from one of three independent experiments, whereas panels a, c and e show results from one of two independent experiments. Biological replicates yielded similar results. For source data, see Supplementary Table 8. Uncropped images of blots are shown in Supplementary Fig. 9.

Supplementary Figure 10 Expression of Myc or loss of p53 and Rb induces neoplastic transformation of Rnd1-silenced mammary epithelial cells.

(ac) Rnd1-silenced MCF-10A cells, which are maintained in culture over a 1-month period, undergo spontaneous immortalization accompanied by overexpression of c-Myc. Phase Contrast pictures of control and Rnd1-silenced MCF-10A cells that have been cultured over a 1-month period (a). Control (sh-Contr.) and spontaneously immortalized Rnd1-silenced (sh-Rnd1 #2) MCF-10A cells were subjected to soft agar assay (b) and immunoblotting as indicated (c). Scale bars, 50 μm. (df) Expression of Myc enables neoplastic conversion of Rnd1-silenced MCF-10A cells. MCF-10A cells, which were transduced with c-Myc or left uninfected, were infected with a control shRNA or one targeting Rnd1. (d) Cells were deprived of growth factors, stimulated or not with 10% FBS, and subjected to immunoblotting as indicated (top) or Q-PCR for RND1 (bottom). Graph shows one of two representative experiments. (e) Cells were subjected to SA-βtGalactosidase staining at 12 days after infection. (f) Cells were subjected to soft agar assay. The images show representative pictures at 2 weeks. Scale bar is 50 μm. (g,h) Silencing of Rnd1 induces anchorage-independent growth of HMLE cells. (g) Cells were transduced with a control shRNA or two shRNAs targeting Rnd1 and cultured in soft agar for 2 weeks. The images show representative pictures (scale bar, 50 μm). (h) The graph shows the average number of colonies and SD from one experiment performed of n = 3 technical replicates. The experiment was repeated 2 times. (i-l) Silencing of Rnd1 induces hyperproliferation but not suppression of apoptosis in 3D Matrigel. Parental MCF-10A cells and MCF-10 cells expressing c-Myc were infected with a control shRNA or two shRNAs targeting Rnd1, cultured in 3D Matrigel for 8 days, and subjected to immunofluorescent staining with anti-Ki-67 followed by DAPI. Representative images (scale bar, 50 μm) are shown (i). The graph shows the average percentage of Ki-67-positive cells and SD from n = 3 independent experiments (j). The graph shows the average percentage of cleaved caspase 3-positive cells and SD from n = 3 independent experiments (k). The pictures show representative images of pseudoacinar structures stained with anti-cleaved caspase 3 and DAPI. The arrows point to cells that have lost their apical positioning of Golgi apparatus and invaded inot the Matrigel. Note that they possess intact nuclei. Scale bar, 15 μm (l). P values are: ∗, P = < 0.05; ∗∗, P = < 0.01; ∗∗∗, P < 0.001 by the Student’s _t_-test. Panels b-f show one representative experiment out of three and panels a and g show one representative out of two performed. Biological replicates yielded similar results. Source data is given in Supplementary Table 8. Uncropped images of blots are shown in Supplementary Fig. 9.

Supplementary Figure 11 Loss of Rnd1 promotes hyperproliferation and EMT through activation of the Raf-ERK cascade.

(a) Control (sh-Co.) and Rnd1-silenced (sh #1 and #2) MCF-10A cells were subjected to GST-pull down assay to measure activated Cdc42, Rac1, and Rho (A, B, and C). (b) MCF-10A cells expressing either a control shRNA (sh-Co.) or one targeting Rnd1 (sh #1) were starved for 24 hours, stimulated with growth factors for the indicated times, and subjected to GST-RBD pull down assay to measure GTP-Ras. (c) HMLE (top) and HEK 293T (middle) cells expressing a control shRNA (sh-Co.) or two sh-RNAs targeting Rnd1 (sh #1 and #2) were starved for 24 h, and HUVECs (bottom) untransfected or transfected with a control si-RNA or one targeting Rnd1 (si-Rnd1) were subjected to pulldown assay to measure GTP-Ras. (d) MCF-10A cells carrying either a control shRNA or one targeting Rnd1 (sh #1) were cultured for 24 h with PD98059 (MEK inhibitor) or vehicle, and subjected to immunofluorescent staining as indicated. Scale bar, 15 μm. (e) MCF-10A cells expressing either a control shRNA (sh-Co.) or one targeting Rnd1 (sh #2) were cultured for 3 days and treated with Rho kinase inhibitor (Y27632), myosin light chain kinase inhibitor (Blebbistatin) or DMSO for 24 h. Scale bar, 15 μm. (f) MCF-10A cells expressing either a control shRNA or 2 shRNAs targeting Rnd1 (sh #1 and #2) were cultured for 2 days and treated with PD98059 (MEKi), Wortmannin (PI-3Ki), or vehicle for 24 h. Total lysates were subjected to immunoblotting with the indicated antibodies. (g) Gene set enrichment analysis plots showing that low RND1 mRNA levels do not correlate with the expression of a Src or a β-catenin signature in the 3 breast cancer datasets. (h) The levels of mRNAs encoding Myc and Rnd1 exhibit an inverse correlation in 3 breast cancer DNA microarray datasets. Spearman correlation = − 0.46 for EMC192, − 0.36 for EMC286 and − 0.43 for MSKCC99; P < 0.001. See Method for analysis. Data in a, b, c and e are from one of two independent experiments, whereas d and f are from one of three independent experiments. Biological replicates yielded similar results. Uncropped images of blots are shown in Supplementary Fig. 9.

Supplementary Figure 12 Plexin B1 does not modulate MET or HER2 signaling in breast epithelial cells.

(a) Total lysates from the indicated normal mammary epithelial cells and breast cancer cells were subjected to immunoblotting to assess the expression of Plexin B1, Plexin A1, and Semaphorin 4D. (b) Phase Contrast images of control and Plexin B1-silenced MCF10A cells. Scale bar, 50 μm. (c) HMLE cells were starved in growth factor-deprived medium supplemented with 0.2% serum for 15 h and incubated with HGF with or without Semaphorin 4D for the indicated times. Total lysates were subjected to immunoblotting with the indicated antibodies. (d) ZR751 cells were starved as in c and incubated with HRG with or without Semaphorin 4D for the indicated times. Total lysates were subjected to immunoblotting with the indicated antibodies. Representative data from two independent experiments are shown. Uncropped images of blots are shown in Supplementary Fig. 9.

Supplementary Figure 13 Rnd1-PlexinB1-Rasa1 signaling axis is altered in breast cancer.

(a) Analysis of mRNA levels of RND1, PLXNB1 and major RasGAPs in the PAM50 basal-like breast tumor dataset from TCGA using cbioportal. (b) Immunoblotting of MAPK signaling pathway components in p120RasGAP-silenced MCF10A cells. Blots are representative images from two independent experiments. Uncropped images of blots are shown in Supplementary Fig. 9.

Supplementary Figure 14 Silencing of Rnd1 accelerates tumorigenesis, whereas ectopic expression of Rnd1 exerts the opposite effect.

(a-g) Comma-D cells infected with a control shRNA (sh-Co.) or two shRNA targeting Rnd1 (sh #4 and #5) were subjected to Q-PCR to verify depletion of Rnd1. Data are averages and SD from n = 3 independent experiments (a). Cells were photographed under Phase Contrast. Scale bar, 50 μm (b). Cells were subjected to double immunofluorescent staining for β-catenin (Green) and E-cadherin (Red) followed by DAPI. Scale bar, 15 μm (c). Cells were subjected to MTT assay. Data are from one experiment shown as averages of five technical repeats (n = 2 biological replicates) (d). Cells were subjected to immunoblotting as indicated (e, f). Cells were cultured in soft agar for 2 weeks. The graph indicates the average number of colonies from five technical repeats (n = 2 biological replicates) (left) and the pictures representative images (right) (g). (h) 4T1 and ErbB2 cells infected with control vector or one encoding HA-Rnd1 were subjected to immunoblotting. (i,j) 4T1 cells infected with retroviral vector encoding Rnd1 or control vector were inoculated intravenously. Lung metastasis was measured by bioluminescent imaging. The panels shows representative images at the indicated times (i). The graph shows individual data points for normalized photon flux (lung, dotted area in i) at different time points after tail-vein injection from n = 5 mice in each cohort (j). (k) Quantification of bioluminescence from lungs from the spontaneous metastasis experiment of Fig. 7f. n = 3 mice and individual data points are shown. Panels b and c show one representative experiment out of three independent experiments. Error bars are SD in a, d and g and SEM in j and k. P values are: ∗, P = < 0.05; ∗∗, P = < 0.01; ∗∗∗, P < 0.001 by the Student’s _t_-test. Biological replicates yielded similar results. For source data, see Supplementary Table 8. Uncropped images of blots are shown in Supplementary Fig. 9.

Supplementary Figure 15 Genetic and epigenetic inactivation of RND1 in human breast cancer cell lines.

(a) Box plots depicting the distribution of RND1 copy number loss defined as the ratio of signal observed for the _RND1_-specific and centromeric chr 12 probes in wild type (blue dots) and deleted (red dots) tumor cell subpopulations. Black bars indicate the mean values for each subpopulation. Samples are indicated with Arabic numbers, N stands for normal cells. (b) The indicated breast cancer cells were treated with SAHA alone (24 h), 5-AZA alone (72 h) or a combination of SAHA and 5-AZA (72 h) and subjected to Q-PCR for RND1. Data are from one experiment and are shown as averages of three technical replicates (n = 2 biological replicates). (c) Subcellular localization of wild type Rnd1. Rnd1 is localized at the plasma membrane and on the Golgi. HUVECs were transfected with a plasmid encoding HA-tagged Rnd1 and a Farnesylated CAAX motif fused to GFP and subjected to immunostaining with anti-HA (Red), anti-Giantin (Cyan) and DAPI. (d) Co-localization of Rnd1 with H-Ras and K-Ras. HUVECs transfected with plasmids encoding HA-tagged Rnd1 and GFP-H-Ras (Top) or GFP-K-Ras4B (Bottom) were subjected to immunostaining with anti-HA (Red), anti-Giantin (Cyan) and DAPI (Blue). Images in (c) and (d) are representative of three independent experiments. Scale bar, 15 μm. (e) EpiTYPER was carried out to analyse the methylation state of CpGs islands in the RND1 gene promoter in selected breast tumour cell lines. Each circle indicates a CpG dinucleotide. WGA: negative control. IVD: positive control. (f) HCC1428 cells infected with a control shRNA (Co.) or 3 shRNAs targeting EZH2 (#1, #2, #3) were subjected to immunoblotting with the indicated antibodies. A representative blot from two independent experiments is shown. (g) The indicated cell lines were infected with a shRNA targeting EZH2 (#1) or not, treated with SAHA (6 h) or vehicle control, and then subjected to Q-PCR for RND1 (left). HCC1428 cells expressing a control shRNA or 2 shRNAs targeting EZH2 (#2 and #3) were treated with SAHA (24 h) or vehicle control and subjected to Q-PCR for RND1 (right). One of two independent experiments is shown. For source data, see Supplementary Table 8. Uncropped images of blots are shown in Supplementary Fig. 9.

Supplementary Table 1 Metastasis-Free Survival analysis for all RhoGTPases

Full size table

Supplementary Table 2 Univariate and Multivariate analysis for survival

Full size table

Supplementary Table 3 list of genes up- or down-regulated in RND1 knocked down cells

Full size table

Supplementary Table 4 Cases positive for RND1 deletion by FISH related to Fig. 8a and Supplementary Fig. 8a

Full size table

Supplementary Table 5 Solid sequencing results for each mutation of _RND1_related to Fig. 8

Full size table

Supplementary Table 6 Antibody list used in this study

Full size table

Supplementary Table 7 Q-PCR Primer list

Full size table

Supplementary information

Rights and permissions

About this article

Cite this article

Okada, T., Sinha, S., Esposito, I. et al. The Rho GTPase Rnd1 suppresses mammary tumorigenesis and EMT by restraining Ras-MAPK signalling.Nat Cell Biol 17, 81–94 (2015). https://doi.org/10.1038/ncb3082

Download citation