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:

A ROR1–HER3–lncRNA signalling axis modulates the Hippo–YAP pathway to regulate bone metastasis

Abstract

Bone metastases remain a serious health concern because of limited therapeutic options. Here, we report that crosstalk between ROR1–HER3 and the Hippo–YAP pathway promotes breast cancer bone metastasis in a long noncoding RNA-dependent fashion. Mechanistically, the orphan receptor tyrosine kinase ROR1 phosphorylates HER3 at a previously unidentified site Tyr1307, following neuregulin stimulation, independently of other ErbB family members. p-HER3 Tyr1307 recruits the LLGL2–MAYA–NSUN6 RNA–protein complex to methylate Hippo/MST1 at Lys59. This methylation leads to MST1 inactivation and activation of YAP target genes in tumour cells, which elicits osteoclast differentiation and bone metastasis. Furthermore, increased ROR1, p-HER3 Tyr1307 and MAYA levels correlate with tumour metastasis and unfavourable outcomes. Our data provide insights into the mechanistic regulation and linkage of the ROR1–HER3 and Hippo–YAP pathway in a cancer-specific context, and also imply valuable therapeutic targets for bone metastasis and possible therapy-resistant tumours.

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

Figure 1: ROR1 promotes the colonization and growth of breast cancer cells within the bone.
Figure 2: ROR1-dependent phosphorylation of HER3 at Tyr1307 correlates with breast cancer clinical parameters.
Figure 3: Crosstalk between ROR1–HER3 and the Hippo–YAP pathway.
Figure 4: LncRNA MAYA is required for activation of YAP.
Figure 5: Characterization of MAYA–LLGL2–NSUN6 associations.
Figure 6: LncRNA-mediated, NSUN6-dependent methylation inhibits the kinase activity of MST1.
Figure 7: The ROR1/HER3–LLGL2/MAYA/NSUN6 signalling axis regulates YAP activity.
Figure 8: MAYA serves as a promising therapeutic target for bone metastasis.

Similar content being viewed by others

References

  1. Guise, T. A. et al. Basic mechanisms responsible for osteolytic and osteoblastic bone metastases. Clin. Cancer Res. 12, 6213s–6216s (2006).

    Article  CAS  PubMed  Google Scholar 

  2. Waning, D. L. & Guise, T. A. Molecular mechanisms of bone metastasis and associated muscle weakness. Clin. Cancer Res. 20, 3071–3077 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Kang, Y. et al. A multigenic program mediating breast cancer metastasis to bone. Cancer Cell 3, 537–549 (2003).

    Article  CAS  PubMed  Google Scholar 

  4. Pan, D. The hippo signaling pathway in development and cancer. Dev. Cell 19, 491–505 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Harvey, K. F., Zhang, X. & Thomas, D. M. The Hippo pathway and human cancer. Nat. Rev. Cancer 13, 246–257 (2013).

    Article  CAS  PubMed  Google Scholar 

  6. Meng, Z., Moroishi, T. & Guan, K. L. Mechanisms of Hippo pathway regulation. Genes Dev. 30, 1–17 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Yu, F. X. & Guan, K. L. The Hippo pathway: regulators and regulations. Genes Dev. 27, 355–371 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Hansen, C. G., Moroishi, T. & Guan, K. L. YAP and TAZ: a nexus for Hippo signaling and beyond. Trends Cell Biol. 25, 499–513 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Zhao, B., Tumaneng, K. & Guan, K. L. The Hippo pathway in organ size control, tissue regeneration and stem cell self-renewal. Nat. Cell Biol. 13, 877–883 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Thompson, B. J. & Sahai, E. MST kinases in development and disease. J. Cell Biol. 210, 871–882 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Halder, G., Dupont, S. & Piccolo, S. Transduction of mechanical and cytoskeletal cues by YAP and TAZ. Nat. Rev. Mol. Cell Biol. 13, 591–600 (2012).

    Article  CAS  PubMed  Google Scholar 

  12. Piccolo, S., Dupont, S. & Cordenonsi, M. The biology of YAP/TAZ: hippo signaling and beyond. Physiol. Rev. 94, 1287–1312 (2014).

    Article  CAS  PubMed  Google Scholar 

  13. Bergeron, J. J., Di Guglielmo, G. M., Dahan, S., Dominguez, M. & Posner, B. I. Spatial and temporal regulation of receptor tyrosine kinase activation and intracellular signal transduction. Annu. Rev. Biochem. 85, 573–597 (2016).

    Article  CAS  PubMed  Google Scholar 

  14. Green, J. L., Kuntz, S. G. & Sternberg, P. W. Ror receptor tyrosine kinases: orphans no more. Trends Cell Biol. 18, 536–544 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Forrester, W. C. The Ror receptor tyrosine kinase family. Cell. Mol. Life Sci. 59, 83–96 (2002).

    Article  CAS  PubMed  Google Scholar 

  16. Yoda, A., Oishi, I. & Minami, Y. Expression and function of the Ror-family receptor tyrosine kinases during development: lessons from genetic analyses of nematodes, mice, and humans. J. Recept. Signal Transduct. Res. 23, 1–15 (2003).

    Article  CAS  PubMed  Google Scholar 

  17. Baskar, S. et al. Unique cell surface expression of receptor tyrosine kinase ROR1 in human B-cell chronic lymphocytic leukemia. Clin. Cancer Res. 14, 396–404 (2008).

    Article  CAS  PubMed  Google Scholar 

  18. Daneshmanesh, A. H. et al. Orphan receptor tyrosine kinases ROR1 and ROR2 in hematological malignancies. Leuk. Lymphoma 54, 843–850 (2013).

    Article  CAS  PubMed  Google Scholar 

  19. Zhang, S. et al. ROR1 is expressed in human breast cancer and associated with enhanced tumor-cell growth. PLoS ONE 7, e31127 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Zhang, S. et al. The onco-embryonic antigen ROR1 is expressed by a variety of human cancers. Am. J. Pathol. 181, 1903–1910 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Borcherding, N., Kusner, D., Liu, G. H. & Zhang, W. ROR1, an embryonic protein with an emerging role in cancer biology. Protein Cell 5, 496–502 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Chien, H. P. et al. Expression of ROR1 has prognostic significance in triple negative breast cancer. Virchows Arch. 468, 589–595 (2016).

    Article  CAS  PubMed  Google Scholar 

  23. Gentile, A., Lazzari, L., Benvenuti, S., Trusolino, L. & Comoglio, P. M. Ror1 is a pseudokinase that is crucial for Met-driven tumorigenesis. Cancer Res. 71, 3132–3141 (2011).

    Article  CAS  PubMed  Google Scholar 

  24. Bicocca, V. T. et al. Crosstalk between ROR1 and the Pre-B cell receptor promotes survival of t(1;19) acute lymphoblastic leukemia. Cancer Cell 22, 656–667 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Masiakowski, P. & Carroll, R. D. A novel family of cell surface receptors with tyrosine kinase-like domain. J. Biol. Chem. 267, 26181–26190 (1992).

    Article  CAS  PubMed  Google Scholar 

  26. Oishi, I. et al. Spatio-temporally regulated expression of receptor tyrosine kinases, mRor1, mRor2, during mouse development: implications in development and function of the nervous system. Genes Cells 4, 41–56 (1999).

    Article  CAS  PubMed  Google Scholar 

  27. Yamaguchi, T. et al. NKX2-1/TITF1/TTF-1-induced ROR1 is required to sustain EGFR survival signaling in lung adenocarcinoma. Cancer Cell 21, 348–361 (2012).

    Article  CAS  PubMed  Google Scholar 

  28. Rashid, F., Shah, A. & Shan, G. Long non-coding RNAs in the cytoplasm. Genomics Proteomics Bioinform. 14, 73–80 (2016).

    Article  Google Scholar 

  29. Willingham, A. T. et al. A strategy for probing the function of noncoding RNAs finds a repressor of NFAT. Science 309, 1570–1573 (2005).

    Article  CAS  PubMed  Google Scholar 

  30. Yoon, J. H., Abdelmohsen, K. & Gorospe, M. Posttranscriptional gene regulation by long noncoding RNA. J. Mol. Biol. 425, 3723–3730 (2013).

    Article  CAS  PubMed  Google Scholar 

  31. Tichon, A. et al. A conserved abundant cytoplasmic long noncoding RNA modulates repression by Pumilio proteins in human cells. Nat. Commun. 7, 12209 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Wang, P. et al. The STAT3-binding long noncoding RNA lnc-DC controls human dendritic cell differentiation. Science 344, 310–313 (2014).

    Article  CAS  PubMed  Google Scholar 

  33. Liu, B. et al. A cytoplasmic NF-κB interacting long noncoding RNA blocks IκB phosphorylation and suppresses breast cancer metastasis. Cancer Cell 27, 370–381 (2015).

    Article  CAS  PubMed  Google Scholar 

  34. Lin, A. et al. The LINK-A lncRNA activates normoxic HIF1α signalling in triple-negative breast cancer. Nat. Cell Biol. 18, 213–224 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Park, H. W. et al. Alternative wnt signaling activates YAP/TAZ. Cell 162, 780–794 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Mohseni, M. et al. A genetic screen identifies an LKB1-MARK signalling axis controlling the Hippo–YAP pathway. Nat. Cell Biol. 16, 108–117 (2014).

    Article  CAS  PubMed  Google Scholar 

  37. Cordenonsi, M. et al. The Hippo transducer TAZ confers cancer stem cell-related traits on breast cancer cells. Cell 147, 759–772 (2011).

    Article  CAS  PubMed  Google Scholar 

  38. Zanconato, F. et al. Genome-wide association between YAP/TAZ/TEAD and AP-1 at enhancers drives oncogenic growth. Nat. Cell Biol. 17, 1218–1227 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Zhang, J. et al. YAP-dependent induction of amphiregulin identifies a non-cell-autonomous component of the Hippo pathway. Nat. Cell Biol. 11, 1444–1450 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Gomez, M., Gomez, V. & Hergovich, A. The Hippo pathway in disease and therapy: cancer and beyond. Clin. Trans. Med. 3, 22 (2014).

    Article  Google Scholar 

  41. Moroishi, T., Hansen, C. G. & Guan, K. L. The emerging roles of YAP and TAZ in cancer. Nat. Rev. Cancer 15, 73–79 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Shimo, T. et al. Pathogenic role of connective tissue growth factor (CTGF/CCN2) in osteolytic metastasis of breast cancer. J. Bone Miner. Res. 21, 1045–1059 (2006).

    Article  CAS  PubMed  Google Scholar 

  43. Engelman, J. A. et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 316, 1039–1043 (2007).

    Article  CAS  PubMed  Google Scholar 

  44. Sergina, N. V. et al. Escape from HER-family tyrosine kinase inhibitor therapy by the kinase-inactive HER3. Nature 445, 437–441 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Hellyer, N. J., Kim, M. S. & Koland, J. G. Heregulin-dependent activation of phosphoinositide 3-kinase and Akt via the ErbB2/ErbB3 co-receptor. J. Biol. Chem. 276, 42153–42161 (2001).

    Article  CAS  PubMed  Google Scholar 

  46. Gentile, A., Lazzari, L., Benvenuti, S., Trusolino, L. & Comoglio, P. M. The ROR1 pseudokinase diversifies signaling outputs in MET-addicted cancer cells. Int. J. Cancer 135, 2305–2316 (2014).

    Article  CAS  PubMed  Google Scholar 

  47. Tao, J. J. et al. Antagonism of EGFR and HER3 enhances the response to inhibitors of the PI3K-Akt pathway in triple-negative breast cancer. Sci. Signal. 7, ra29 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Bae, S. Y. et al. HER3 status by immunohistochemistry is correlated with poor prognosis in hormone receptor-negative breast cancer patients. Breast Cancer Res. Treat. 139, 741–750 (2013).

    Article  CAS  PubMed  Google Scholar 

  49. Yaffe, M. B. Phosphotyrosine-binding domains in signal transduction. Nat. Rev. Mol. Cell Biol. 3, 177–186 (2002).

    Article  CAS  PubMed  Google Scholar 

  50. Johnson, R. & Halder, G. The two faces of Hippo: targeting the Hippo pathway for regenerative medicine and cancer treatment. Nat. Rev. Drug Discov. 13, 63–79 (2014).

    Article  CAS  PubMed  Google Scholar 

  51. Yu, F. X. et al. Regulation of the Hippo-YAP pathway by G-protein-coupled receptor signaling. Cell 150, 780–791 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Haag, S. et al. NSUN6 is a human RNA methyltransferase that catalyzes formation of m5C72 in specific tRNAs. RNA 21, 1532–1543 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Xing, Z. et al. lncRNA directs cooperative epigenetic regulation downstream of chemokine signals. Cell 159, 1110–1125 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Glantschnig, H., Rodan, G. A. & Reszka, A. A. Mapping of MST1 kinase sites of phosphorylation. Activation and autophosphorylation. J. Biol. Chem. 277, 42987–42996 (2002).

    Article  CAS  PubMed  Google Scholar 

  55. Sapru, M. K. Neuregulin-1 regulates expression of the Ets-2 transcription factor. Life Sci. 69, 2663–2674 (2001).

    Article  CAS  PubMed  Google Scholar 

  56. 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 

  57. Zhang, C. & Yu, D. Microenvironment determinants of brain metastasis. Cell Biosci. 1, 8 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Zhang, S. et al. SRC family kinases as novel therapeutic targets to treat breast cancer brain metastases. Cancer Res. 73, 5764–5774 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Wang, W. et al. AMPK modulates Hippo pathway activity to regulate energy homeostasis. Nat. Cell Biol. 17, 490–499 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Sorrentino, G. et al. Metabolic control of YAP and TAZ by the mevalonate pathway. Nat. Cell Biol. 16, 357–366 (2014).

    Article  CAS  PubMed  Google Scholar 

  61. Lin, S. Y. et al. Nuclear localization of EGF receptor and its potential new role as a transcription factor. Nat. Cell Biol. 3, 802–808 (2001).

    Article  CAS  PubMed  Google Scholar 

  62. Haskins, J. W., Nguyen, D. X. & Stern, D. F. Neuregulin 1-activated ERBB4 interacts with YAP to induce Hippo pathway target genes and promote cell migration. Sci. Signal. 7, ra116 (2014).

    PubMed  PubMed Central  Google Scholar 

  63. Xue, Z. et al. A G-rich motif in the lncRNA braveheart interacts with a zinc-finger transcription factor to specify the cardiovascular lineage. Mol. Cell 64, 37–50 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Castello, A. et al. Insights into RNA biology from an atlas of mammalian mRNA-binding proteins. Cell 149, 1393–1406 (2012).

    Article  CAS  PubMed  Google Scholar 

  65. Bittner, N. et al. Bone metastases and the EGFR and KRAS mutation status in lung adenocarcinoma–the results of three year retrospective analysis. Pathol. Oncol. Res. 21, 1217–1221 (2015).

    Article  CAS  PubMed  Google Scholar 

  66. Karachaliou, N. et al. ROR1 as a novel therapeutic target for EGFR-mutant non-small-cell lung cancer patients with the EGFR T790M mutation. Transl. Lung Cancer Res. 3, 122–130 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are grateful to J. Massague and X. Zhang for providing the MDA-MB-231 LM2 and BoM-1833 cell lines and to D. Yu for providing the MDA-MB-231-BRN and BT474-BRN cells. We thank D. Aten for assistance with figure presentation. This work was supported by National Institutes of Health Pathway to Independence Award (R00CA166527) and Cancer Prevention Research Institute of Texas First-time Faculty Recruitment Award (R1218) grants to L.Q.Y. and National Institutes of Health Pathway to Independence Award (R00DK094981) to C.R.L.

Author information

Authors and Affiliations

Authors

Contributions

C.L.L. and S.W. devised and performed most experiments. K.L., J.S. and G.E.G. helped with mouse intracardiac injections. A.L., Z.X. and Q.H. helped with biochemistry studies. D.H.H. performed mass spectrometry analysis. Clinical specimens were ascertained and processed by J.Z. and Y.Z. The histological staining and corresponding analysis were performed by K.L. P.K.P. assisted with manuscript drafting. J.Y., L.H., Z.C. and H.L. performed bioinformatics analysis. S.Z. and M.-C.H. contributed to discussion and data interpretation. L.Y. and C.R.L. initiated and supervised the project and wrote the paper with input from all authors.

Corresponding authors

Correspondence to Chunru Lin or Liuqing Yang.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 ROR1 promotes proliferation and mobility of breast cancer cells.

(a) Oncomine box plot showing ROR1 copy numbers in human ERPR-/HER2- triple negative breast cancer (TNBC) and non-TNBC subtypes (n = 73 and 495 tumors respectively, one-way ANOVA). (b) Oncomine box plots showing ROR1 expression levels in a series of human TNBC (n = 211, 178, 46, 26, 22 and 39 tumors, one-way ANOVA) and non-TNBC subtypes (n = 1,340, 320, 250, 89, 44 and 129 tumors, one-way ANOVA). (c) Oncomine box plots showing ROR1 expression levels in human Non-small Cell Lung Carcinoma (n = 33, 63, 10, 26, and 132 tumors, one-way ANOVA) and Small cell lung carcinoma (n = 9 and 6 tumors, one-way ANOVA) or squamous cell lung carcinoma cohorts (n = 154, 75, 10, 26, and 21 tumors, one-way ANOVA). (d and e) Immunoblotting (IB) of indicated proteins in parental/ROR1 KO MDA-MB-231 (d) and BoM-1833 (e) single cell clones. (f and g) Cell proliferation assay of individual (f) and pooled (g) clones of ROR1 KO MDA-MB-231 cells. (h) IB detection of indicated proteins in ROR1 KO cells expressing WT ROR1 or K506A mutant. (i) Cell invasion assay of ROR1 KO MDA-MB-231 cells with expression of indicated plasmid (left, Scale bars, 200 μm; right, quantification). (j and k) Cell migration (j) and invasion (k) assay of pooled clones of ROR1 KO MDA-MB-231 cells with overexpression of indicated plasmid (Scale bars, 200 μm). (l) TRAP staining showing the osteoclast differentiation in the presence of M-CSF only, M-CSF + RANKL, or combined M-CSF + RANKL and conditioned media (CM) from ROR1 KO BoM-1833 cells or CM from ROR1 KO BoM-1833 cells KO supplemented with PBS or recombinant CTGF (50 ng ml−1) (Scale bars, 200 μm). For a-c, the boxes show the median ± 1 quartile, with whiskers extending to the most extreme data point within 1.5 interquartile ranges from the box boundaries. For f, g and i-k, mean ± s.e.m. were derived from n = 3 independent experiments (n.s., P > 0.05, P < 0.05 and P < 0.01, P < 0.001, two-tailed paired Student’s t-test). Unprocessed original scans of all blots with size marker are shown in Supplementary Fig. 9 Statistics source data for i-l are in Supplementary Table 8.

Supplementary Figure 2 Identification of ROR1-associated proteins, characterization of ROR1 phosphorylation and examination of the role of p-HER3 (Tyr1307) in breast and lung cancer.

(a) A list of top ROR1-associated proteins identified by protein pull-down and MS in MDA-MB-231 cells. (b) IB detection of p-HER3 (Tyr1307) in MDA-MB-231 cells treated with or without NRG1 using antibodies pre-incubated with indicated blocking peptides. Antibodies generated from two independent rabbits were tested. (c and d) Immunoprecipitation (IP) and IB detection of indicated proteins in MDA-MB-231 cells transfected with indicated plasmids (c) or treated with indicated small compound inhibitors (d) followed by NRG1 treatment. (e) IP and IB detection of indicated proteins in 32D cells transfected with indicated expression vectors followed by NRG1 stimulation. (f) IB detection of indicated proteins in parental/HER3 KO MDA-MB-231 single cell clones. (g and h) Cell proliferation assay of individual (g) and pooled (h) clones of HER3 KO MDA-MB-231 cells. (i) Cell invasion assay of individual clones of HER3 KO MDA-MB-231 cells with overexpression of indicated plasmid (left, Scale bars, 200 μm; right, quantification). (j and k) Cell migration (j) and invasion (k) assay of pooled clones of HER3 KO MDA-MB-231 cells (left, Scale bars, 200 μm; right, quantification). (l) Immunohistochemical (IHC) staining of p-HER3 (Tyr1307) in normal lung tissues, lung adenocarcinomas, non-metastatic (TnN0M0) and metastatic (TnN > 0M ≥ 0) lung adenocarcinomas (left, Scale bars, 40 μm; right, n = 75, 75, 43 and 32 lung tissues/adenocarcinomas respectively, median, one-way ANOVA) (m) Kaplan-Meier survival analysis of p-HER3 (Tyr1307) low and high lung adenocarcinoma patients (n = 20 and 55 patients respectively, log rank test). For gk, mean ± s.e.m. were derived from n = 3 independent experiments (P < 0.05, P < 0.01 and P < 0.001, two-tailed paired Student’s t-test). Unprocessed original scans of all blots with size marker are shown in Supplementary Fig. 9 Statistics source data for i-l are in Supplementary Table 8.

Supplementary Figure 3 Identification and characterization of p-LLGL2 (Tyr499) and MST1 (Lys59me2).

(a) Summary of top LLGL2-associated proteins identified by LLGL2 pull-down followed by MS in MDA-MB-231 cells. (b and e) Annotated MS/MS spectrum assigned to the LLGL2 peptide VGSFDP[p]YSDDPR, at 717.782 Da. (b) and the STK4/MST1 peptide ETGQIVAI[dime] KQVPVESDLQEIIK, at 1233.200 Da. (e). Data acquired from analysis of the tryptic digest by high-sensitivity LC-MS/MS on an Orbitrap Elite high-resolution mass spectrometer. (c and f) IB detection of p-LLGL2 (Tyr499) (c) and MST1 (Lys59me2) (f) in MDA-MB-231 cells treated with or without NRG1, using antibodies pre-incubated with indicated blocking peptides. Antibodies generated from two independent rabbits were tested. (d) IB detection of indicated proteins in cells transfected with indicated expression vectors followed by NRG1 stimulation. Scanning densitometric analysis was performed for p-LLGL2 (Tyr499) blot. (g) IB detection of indicated proteins in MDA-MB-231 cells treated with LPA or NRG1. Unprocessed original scans of all blots with size marker are shown in Supplementary Fig. 9.

Supplementary Figure 4 Identification of MAYA-associated proteins, characterization of MAYA transcript and its correlation with lung cancer.

(a) Summary of the top MAYA-associated proteins identified by RNA pull-down followed by MS in MDA-MB-231 cells. (b) Determination of MAYA transcript in MDA-MB-231 cells by 5′-and 3′-RACE. R1: repeat 1, R2: repeat 2. (c) Top, northern blot detection of MAYA in pre-made human tissue RNA blots; bottom, RNA gel electrophoresis. (d) Top, northern blot detection of MAYA in MDA-MB-231 cells with indicated treatment; bottom, RNA gel electrophoresis. (e) Examination of MAYA expression in multiple tumor types and their normal counterparts by RNAscope assay. (f) MAYA RNAscope staining intensities in normal lung tissues, lung adenocarcinomas, non-metastatic (TnN0M0) and metastatic (TnN > 0M ≥ 0) lung adenocarcinomas (n = 75, 75, 50 and 25 lung tissues/adenocarcinomas respectively, median, one-way ANOVA) (g) Kaplan-Meier survival analysis of MAYA low and high lung cancer patients (n = 21 and 54 patients respectively, log rank test). (h and i) RT-qPCR detection of MAYA and CTGF expression level in MCF-7 cells transfected with the indicated siRNAs that grown under spare or confluent culture conditions (h) or followed by lysophosphatidic acid (LPA) treatment (1 μM, 2 h) (i). For h and i, mean ± s.e.m. were derived from n = 3 independent experiments (n.s., P > 0.05 andP < 0.01, two-tailed paired Student’s t-test). Unprocessed original scans of all blots/gels with size marker are shown in Supplementary Fig. 9.

Supplementary Figure 5 Characterization of MAYA-LLGL2 and MAYA-NSUN6 interactions.

(a) Electrophoresis of in vitro transcribed Xef1α (left) or MAYA (right) sense (sen.) and anti-sense (as.) transcripts. (b) Streptavidin RNA pull-down assay, followed by IB detection of indicated proteins, using MAYA sense (sen.), anti-sense (as.) and Xef1α sense RNA. The presence of RNA transcripts was detected by streptavidin-HRP using dot-blot assay. (c) Electrophoresis of in vitro transcribed full-length MAYA or truncated transcripts. (d) RNA REMSA assay was performed using recombinant GST-tagged LLGL2 (aa. 101-200) or FLAG-tagged NSUN6 in the presence of 32P-labeled or cold MAYA RNA probes (nt. 251-290 and nt. 851-890), respectively. (e) Cell fractionation followed by RT-qPCR detecting relative MAYA expression in MDA-MB-231 cells with indicated stimuli (mean ± s.e.m. were derived from n = 3 independent experiments, n.s., P > 0.05, two-tailed paired Student’s t-test). Unprocessed original scans of all blots/gels with size marker are shown in Supplementary Fig. 9. An unprocessed original scans of EMSA image for d is also shown in Supplementary Fig. 9.

Supplementary Figure 6 Examination of the role of LLGL2 and NSUN6 in breast cancer cells.

(a and b) IB detection of the indicated proteins in LLGL2 KO (a) and NSUN6 KO (b) MDA-MB-231 cells. (c and d) Cell proliferation assay of individual (c) and pooled (d) clones of LLGL2 KO (left panels) and NSUN6 KO (right panels) MDA-MB-231 cells. (e and f) Cell invasion assay of individual clones of LLGL2 KO (e) and NSUN6 KO (f) MDA-MB-231 cells with overexpression of indicated plasmid (left, Scale bars, 200 μm; right, quantification). (g and h) Cell migration (g) and invasion (h) assay of pooled clones of LLGL2 KO and NSUN6 KO MDA-MB-231 cells (left, Scale bars, 200 μm; right, quantification). For ch, mean ± s.e.m. were derived from n = 3 independent experiments (n.s., P > 0.05, P < 0.05, P < 0.01 and P < 0.001, two-tailed paired Student’s t-test). Unprocessed original scans of all blots with size marker are shown in Supplementary Fig. 9 Source data for eh are in Supplementary Table 8.

Supplementary Figure 7 MAYA is required for NRG1-triggered ROR1-HER3-MST1-YAP signaling axis.

(a) IP and IB detection of indicated proteins in ROR1 KO MDA-MB-231cells transfected with indicated expression vectors followed by NRG1 stimulation. (b and c) RT-qPCR detection of indicated genes in ROR1 KO cells transfected with indicated expression vectors followed by NRG1 stimulation. (d) MAYA expression level in MDA-MB-231 cells transfected with indicated individual LNAs targeting MAYA. (e) RT-qPCR detection of MAYA expression in MDA-MB-231 cells transfected with LNAs against MAYA followed by overexpression of indicated plasmids and NRG1 treatment. (f) IB detection of indicated proteins in A549 cells transfected with LNAs against MAYA followed by overexpression of indicated plasmids and NRG1 treatment. (gi) RT-qPCR detection of indicated gene expression in MDA-MB-231 cells transfected with LNAs against MAYA followed by overexpression of indicated plasmids and NRG1 treatment (100 ng ml−1 for 1 h). (j) Osteoclast differentiation in the presence of M-CSF only, M-CSF + RANKL, or combined M-CSF + RANKL and CM from scramble (Scr) or MAYA LNA-transfected BoM-1833 cells rescued with indicated plasmids (Scale bars, 200 μm) For be and gi, mean ± s.e.m. were derived from n = 3 independent experiments (n.s., P > 0.05, P < 0.05, P < 0.01 and P < 0.001, two-tailed paired Student’s t-test). Unprocessed original scans of all blots with size marker are shown in Supplementary Fig. 9. Statistics source data for j are in Supplementary Table 8.

Supplementary Figure 8 Targeting MAYA inhibits the migration of lung cancer cells in vitro and tumor bone metastasis in vivo.

(a) RT-qPCR detection of MAYA expression level in a panel of breast cancer cell lines. (b) RT-qPCR detection of MAYA expression level in BoM-1833 cells harboring indicated shRNAs. (c) Cell proliferation assay of BoM-1833 cells harboring indicated shRNAs. (d and e) Cell migration (d) and invasion (e) assays using BoM-1833 cells harboring indicated shRNAs (left, Scale bars, 200 μm; right, quantification). (f) RT-qPCR detection of MAYA expression level in bone tumors of nude mice inoculated with BoM-1833 cells harboring indicated shRNAs. (g) RT-qPCR detection of MAYA expression level in A549-Luc cells harboring indicated shRNAs. (h) Cell proliferation assay of A549-Luc cells harboring indicated shRNAs. (i and j) Cell migration (i) and invasion (j) assays using A549-Luc cells harboring indicated shRNAs (left, Scale bars, 200 μm; right, quantification). (k and l) Representative BLI images (k) and bone colonization (l) of nude mice with intra-cardiac injection of A549-Luc cells harboring indicated shRNAs (n = 5 mice per group). (m) Graphic illustration of the functional role of NRG1-HER3 triggered, MAYA-mediated Hippo signaling suppression and YAP targets activation for promoting bone metastasis. For aj and l, mean ± s.e.m. were derived from n = 3 independent experiments (P < 0.05, P < 0.01 and P < 0.001, two-tailed paired Student’s t-test). Statistics source data for d,e,i,j and kl are in Supplementary Table 8.

Supplementary information

Supplementary Information

Supplementary Information (PDF 13724 kb)

Supplementary Table 1

Supplementary Information (XLSX 130 kb)

Supplementary Table 2

Supplementary Information (XLSX 93 kb)

Supplementary Table 3

Supplementary Information (XLSX 42 kb)

Supplementary Table 4

Supplementary Information (XLSX 14 kb)

Supplementary Table 5

Supplementary Information (XLSX 33 kb)

Supplementary Table 6

Supplementary Information (XLSX 10 kb)

Supplementary Table 7

Supplementary Information (XLSX 23 kb)

Supplementary Table 8

Supplementary Information (XLSX 74979 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, C., Wang, S., Xing, Z. et al. A ROR1–HER3–lncRNA signalling axis modulates the Hippo–YAP pathway to regulate bone metastasis. Nat Cell Biol 19, 106–119 (2017). https://doi.org/10.1038/ncb3464

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3464

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