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:

Induction of LIFR confers a dormancy phenotype in breast cancer cells disseminated to the bone marrow

An Erratum to this article was published on 27 October 2016

This article has been updated

Abstract

Breast cancer cells frequently home to the bone marrow, where they may enter a dormant state before forming a bone metastasis. Several members of the interleukin-6 (IL-6) cytokine family are implicated in breast cancer bone colonization, but the role for the IL-6 cytokine leukaemia inhibitory factor (LIF) in this process is unknown. We tested the hypothesis that LIF provides a pro-dormancy signal to breast cancer cells in the bone. In breast cancer patients, LIF receptor (LIFR) levels are lower with bone metastases and are significantly and inversely correlated with patient outcome and hypoxia gene activity. Hypoxia also reduces the LIFR:STAT3:SOCS3 signalling pathway in breast cancer cells. Loss of the LIFR or STAT3 enables otherwise dormant breast cancer cells to downregulate dormancy-, quiescence- and cancer stem cell-associated genes, and to proliferate in and specifically colonize the bone, suggesting that LIFR:STAT3 signalling confers a dormancy phenotype in breast cancer cells disseminated to bone.

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: LIFR:STAT3 signalling is downregulated in patients with bone metastases and repressed by hypoxia.
Figure 2: Hypoxic regulation of LIFR:STAT3 signalling is independent of HIF1α or HIF2α.
Figure 3: The LIFR:STAT3 signalling pathway is intact in breast cancer cells with low metastatic potential and not intact in cells with high metastatic potential.
Figure 4: Inhibition of LIFR signalling alters dormancy-, quiescence- and cancer stem cell-associated genes.
Figure 5: Elevated Parathyroid hormone-related protein (PTHrP) signalling blocks LIFR signalling and reduces dormancy-, quiescence- and cancer stem cell-associated genes.
Figure 6: LIFR knockdown results in greater bone destruction via increased osteoclastogenesis and proliferation in vivo.
Figure 7: Loss of STAT3 signalling results in greater bone destruction in vivo.
Figure 8: Targeting STAT3 or SOCS3 mimics effects of loss of LIFR signalling on dormancy genes.

Similar content being viewed by others

Change history

  • 30 September 2016

    In the version of this Article originally published, Fig. 3b was duplicated in Fig. 3c. This has been corrected in the online versions of the Article.

References

  1. Sosa, M. S., Bragado, P. & Aguirre-Ghiso, J. A. Mechanisms of disseminated cancer cell dormancy: an awakening field. Nat. Rev. Cancer 14, 611–622 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Croucher, P. I., McDonald, M. M. & Martin, T. J. Bone metastasis: the importance of the neighbourhood. Nat. Rev. Cancer 16, 373–386 (2016).

    Article  CAS  PubMed  Google Scholar 

  3. Wang, H. et al. The osteogenic niche promotes early-stage bone colonization of disseminated breast cancer cells. Cancer Cell 27, 193–210 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Ghajar, C. M. et al. The perivascular niche regulates breast tumour dormancy. Nat. Cell Biol. 15, 807–817 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Price, T. T. et al. Dormant breast cancer micrometastases reside in specific bone marrow niches that regulate their transit to and from bone. Sci. Transl. Med. 8, 340ra373 (2016).

    Article  Google Scholar 

  6. Bragado, P., Sosa, M. S., Keely, P., Condeelis, J. & Aguirre-Ghiso, J. A. Microenvironments dictating tumor cell dormancy. Recent Results Cancer Res. 195, 25–39 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Johnson, R. W., Schipani, E. & Giaccia, A. J. HIF targets in bone remodeling and metastatic disease. Pharmacol. Ther. 150, 169–177 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Zheng, Y. et al. Targeting IL-6 and RANKL signaling inhibits prostate cancer growth in bone. Clin. Exp. Metastasis 31, 921–933 (2014).

    Article  CAS  PubMed  Google Scholar 

  9. Bolin, C. et al. Oncostatin m promotes mammary tumor metastasis to bone and osteolytic bone degradation. Genes Cancer 3, 117–130 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Chen, D. et al. LIFR is a breast cancer metastasis suppressor upstream of the Hippo-YAP pathway and a prognostic marker. Nat. Med. 18, 1511–1517 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Iorns, E. et al. Whole genome in vivo RNAi screening identifies the leukemia inhibitory factor receptor as a novel breast tumor suppressor. Breast Cancer Res. Treat. 135, 79–91 (2012).

    Article  CAS  PubMed  Google Scholar 

  12. Sims, N. A. & Johnson, R. W. Leukemia inhibitory factor: a paracrine mediator of bone metabolism. Growth Factors 30, 76–87 (2012).

    Article  CAS  PubMed  Google Scholar 

  13. 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 

  14. 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 

  15. Starr, R. et al. A family of cytokine-inducible inhibitors of signalling. Nature 387, 917–921 (1997).

    Article  CAS  PubMed  Google Scholar 

  16. Kennecke, H. et al. Metastatic behavior of breast cancer subtypes. J. Clin. Oncol. 28, 3271–3277 (2010).

    Article  PubMed  Google Scholar 

  17. Schindl, M. et al. Overexpression of hypoxia-inducible factor 1α is associated with an unfavorable prognosis in lymph node-positive breast cancer. Clin. Cancer Res. 8, 1831–1837 (2002).

    CAS  PubMed  Google Scholar 

  18. Bos, R. et al. Levels of hypoxia-inducible factor-1α independently predict prognosis in patients with lymph node negative breast carcinoma. Cancer 97, 1573–1581 (2003).

    Article  PubMed  Google Scholar 

  19. Dales, J. P. et al. Overexpression of hypoxia-inducible factor HIF-1α predicts early relapse in breast cancer: retrospective study in a series of 745 patients. Int. J. Cancer 116, 734–739 (2005).

    Article  CAS  PubMed  Google Scholar 

  20. Generali, D. et al. Hypoxia-inducible factor-1α expression predicts a poor response to primary chemoendocrine therapy and disease-free survival in primary human breast cancer. Clin. Cancer Res. 12, 4562–4568 (2006).

    Article  CAS  PubMed  Google Scholar 

  21. Li, B. et al. Fructose-1,6-bisphosphatase opposes renal carcinoma progression. Nature 513, 251–255 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Jeong, C. H. et al. Hypoxia-inducible factor-1α inhibits self-renewal of mouse embryonic stem cells in vitro via negative regulation of the leukemia inhibitory factor-STAT3 pathway. J. Biol. Chem. 282, 13672–13679 (2007).

    Article  CAS  PubMed  Google Scholar 

  23. Narravula, S. & Colgan, S. P. Hypoxia-inducible factor 1-mediated inhibition of peroxisome proliferator-activated receptor α expression during hypoxia. J. Immunol. 166, 7543–7548 (2001).

    Article  CAS  PubMed  Google Scholar 

  24. Tausendschon, M., Dehne, N. & Brune, B. Hypoxia causes epigenetic gene regulation in macrophages by attenuating Jumonji histone demethylase activity. Cytokine 53, 256–262 (2011).

    Article  PubMed  Google Scholar 

  25. Olcina, M. M. et al. H3K9me3 facilitates hypoxia-induced p53-dependent apoptosis through repression of APAK. Oncogene 35, 793–799 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Kelly, R. D. & Cowley, S. M. The physiological roles of histone deacetylase (HDAC) 1 and 2: complex co-stars with multiple leading parts. Biochem. Soc. Trans. 41, 741–749 (2013).

    Article  CAS  PubMed  Google Scholar 

  27. Lee, H. J. & Kim, K. W. Suppression of HIF-1α by valproic acid sustains self-renewal of mouse embryonic stem cells under hypoxia in vitro. Biomol. Ther. 20, 280–285 (2012).

    Article  CAS  Google Scholar 

  28. Allan, E. H. et al. Osteoblasts display receptors for and responses to leukemia-inhibitory factor. J. Cell. Physiol. 145, 110–119 (1990).

    Article  CAS  PubMed  Google Scholar 

  29. Ishimi, Y. et al. Leukemia inhibitory factor/differentiation-stimulating factor (LIF/D-factor): regulation of its production and possible roles in bone metabolism. J. Cell. Physiol. 152, 71–78 (1992).

    Article  CAS  PubMed  Google Scholar 

  30. Rathjen, P. D. et al. Developmentally programmed induction of differentiation inhibiting activity and the control of stem cell populations. Genes Dev. 4, 2308–2318 (1990).

    Article  CAS  PubMed  Google Scholar 

  31. Derigs, H. G. & Boswell, H. S. LIF mRNA expression is transcriptionally regulated in murine bone marrow stromal cells. Leukemia 7, 630–634 (1993).

    CAS  PubMed  Google Scholar 

  32. Wetzler, M. et al. Constitutive expression of leukemia inhibitory factor RNA by human bone marrow stromal cells and modulation by IL-1, TNF-α, and TGF-β. Exp. Hematol. 19, 347–351 (1991).

    CAS  PubMed  Google Scholar 

  33. Thomas, R. J. et al. Breast cancer cells interact with osteoblasts to support osteoclast formation. Endocrinology 140, 4451–4458 (1999).

    Article  CAS  PubMed  Google Scholar 

  34. Barkan, D. et al. Inhibition of metastatic outgrowth from single dormant tumor cells by targeting the cytoskeleton. Cancer Res. 68, 6241–6250 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Guise, T. A. et al. Evidence for a causal role of parathyroid hormone-related protein in the pathogenesis of human breast cancer-mediated osteolysis. J. Clin. Invest. 98, 1544–1549 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Johnson, R. W. et al. TGF-β promotion of Gli2-induced expression of parathyroid hormone-related protein, an important osteolytic factor in bone metastasis, is independent of canonical Hedgehog signaling. Cancer Res. 71, 822–831 (2011).

    Article  CAS  PubMed  Google Scholar 

  37. Kusuma, N. et al. Integrin-dependent response to laminin-511 regulates breast tumor cell invasion and metastasis. Int. J. Cancer 130, 555–566 (2012).

    Article  CAS  PubMed  Google Scholar 

  38. Kim, R. S. et al. Dormancy signatures and metastasis in estrogen receptor positive and negative breast cancer. PLoS ONE 7, e35569 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Bragado, P. et al. TGF-β2 dictates disseminated tumour cell fate in target organs through TGF-β-RIII and p38α/β signalling. Nat. Cell Biol. 15, 1351–1361 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. El Touny, L. H. et al. Combined SFK/MEK inhibition prevents metastatic outgrowth of dormant tumor cells. J. Clin. Invest. 124, 156–168 (2014).

    Article  CAS  PubMed  Google Scholar 

  41. Holmgren, L., Jackson, G. & Arbiser, J. p53 induces angiogenesis-restricted dormancy in a mouse fibrosarcoma. Oncogene 17, 819–824 (1998).

    Article  CAS  PubMed  Google Scholar 

  42. Shachaf, C. M. et al. MYC inactivation uncovers pluripotent differentiation and tumour dormancy in hepatocellular cancer. Nature 431, 1112–1117 (2004).

    Article  CAS  PubMed  Google Scholar 

  43. Almog, N. et al. Transcriptional changes induced by the tumor dormancy-associated microRNA-190. Transcription 4, 177–191 (2013).

    Article  PubMed  Google Scholar 

  44. Almog, N. et al. Transcriptional switch of dormant tumors to fast-growing angiogenic phenotype. Cancer Res. 69, 836–844 (2009).

    Article  CAS  PubMed  Google Scholar 

  45. Oki, T. et al. A novel cell-cycle-indicator, mVenus-p27K-, identifies quiescent cells and visualizes G0-G1 transition. Scientific Rep. 4, 4012 (2014).

    Article  Google Scholar 

  46. Adam, A. P. et al. Computational identification of a p38SAPK-regulated transcription factor network required for tumor cell quiescence. Cancer Res. 69, 5664–5672 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Pernodet, N. et al. High expression of QSOX1 reduces tumorogenesis, and is associated with a better outcome for breast cancer patients. Breast Cancer Res. 14, R136 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Barkan, D. et al. Metastatic growth from dormant cells induced by a col-I-enriched fibrotic environment. Cancer Res. 70, 5706–5716 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Wang, N. et al. Mitotic quiescence, but not unique ”stemness,” marks the phenotype of bone metastasis-initiating cells in prostate cancer. FASEB J. 29, 3141–3150 (2015).

    Article  CAS  PubMed  Google Scholar 

  50. Manrique, I. et al. The inhibitor of differentiation isoform Id1b, generated by alternative splicing, maintains cell quiescence and confers self-renewal and cancer stem cell-like properties. Cancer Lett. 356, 899–909 (2015).

    Article  CAS  PubMed  Google Scholar 

  51. Chaffer, C. L. et al. Normal and neoplastic nonstem cells can spontaneously convert to a stem-like state. Proc. Natl Acad. Sci. USA 108, 7950–7955 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Liu, S. C. et al. Leukemia inhibitory factor promotes nasopharyngeal carcinoma progression and radioresistance. J. Clin. Invest. 123, 5269–5283 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Kritikou, E. A. et al. A dual, non-redundant, role for LIF as a regulator of development and STAT3-mediated cell death in mammary gland. Development 130, 3459–3468 (2003).

    Article  CAS  PubMed  Google Scholar 

  54. Biswas, S. et al. Anti-transforming growth factor ss antibody treatment rescues bone loss and prevents breast cancer metastasis to bone. PLoS ONE 6, e27090 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Gao, H. et al. The BMP inhibitor Coco reactivates breast cancer cells at lung metastatic sites. Cell 150, 764–779 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Kobayashi, A. et al. Bone morphogenetic protein 7 in dormancy and metastasis of prostate cancer stem-like cells in bone. J. Exp. Med. 208, 2641–2655 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Sosa, M. S. et al. NR2F1 controls tumour cell dormancy via SOX9- and RARβ-driven quiescence programmes. Nat. Commun. 6, 6170 (2015).

    Article  CAS  PubMed  Google Scholar 

  58. Nandy, S. B. et al. MicroRNA-125a influences breast cancer stem cells by targeting leukemia inhibitory factor receptor which regulates the hippo signaling pathway. Oncotarget 6, 17366–17378 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Tomida, M., Yamamoto-Yamaguchi, Y. & Hozumi, M. Characterization of a factor inducing differentiation of mouse myeloid leukemic cells purified from conditioned medium of mouse Ehrlich ascites tumor cells. FEBS Lett. 178, 291–296 (1984).

    Article  CAS  PubMed  Google Scholar 

  60. Li, X. et al. LIF promotes tumorigenesis and metastasis of breast cancer through the AKT-mTOR pathway. Oncotarget 5, 788–801 (2014).

    PubMed  PubMed Central  Google Scholar 

  61. Wu, L. et al. HIF-2α mediates hypoxia-induced LIF expression in human colorectal cancer cells. Oncotarget 6, 4406–4417 (2015).

    PubMed  PubMed Central  Google Scholar 

  62. Wake, M. S. & Watson, C. J. STAT3 the oncogene—still eluding therapy? FEBS J. 282, 2600–2611 (2015).

    Article  CAS  PubMed  Google Scholar 

  63. Derynck, R., Jarrett, J. A., Chen, E. Y. & Goeddel, D. V. The murine transforming growth factor-β precursor. J. Biol. Chem. 261, 4377–4379 (1986).

    CAS  PubMed  Google Scholar 

  64. Derynck, R., Akhurst, R. J. & Balmain, A. TGF-β signaling in tumor suppression and cancer progression. Nat. Genet. 29, 117–129 (2001).

    Article  CAS  PubMed  Google Scholar 

  65. Richard, V., Luchin, A., Brena, R. M., Plass, C. & Rosol, T. J. Quantitative evaluation of alternative promoter usage and 3′ splice variants for parathyroid hormone-related protein by real-time reverse transcription-PCR. Clin. Chem. 49, 1398–1402 (2003).

    Article  CAS  PubMed  Google Scholar 

  66. Poillet, L. et al. QSOX1 inhibits autophagic flux in breast cancer cells. PLoS ONE 9, e86641 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  67. Razorenova, O. V. et al. The apoptosis repressor with a CARD domain (ARC) gene is a direct hypoxia-inducible factor 1 target gene and promotes survival and proliferation of VHL-deficient renal cancer cells. Mol. Cell. Biol. 34, 739–751 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  68. Johnson, R. W. et al. The primary function of gp130 signaling in osteoblasts is to maintain bone formation and strength, rather than promote osteoclast formation. J. Bone Miner. Res. 29, 1492–1505 (2014).

    Article  CAS  PubMed  Google Scholar 

  69. Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature 490, 61–70 (2012).

    Article  Google Scholar 

  70. An Integrated Encyclopedia of DNA Elements in the Human Genome. The ENCODE Project Consortium. Nature 489, 57–74 (2012).

    Article  Google Scholar 

  71. 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  Google Scholar 

  72. Cerami, E. et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2, 401–404 (2012).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

The authors wish to acknowledge A. Diep, G. Nelson and the Stanford Veterinary Service Center for technical assistance, and N. Sims and J. Martin for thoughtful discussion of LIFR signalling. Cell sorting/flow cytometry analysis for this project was performed on instruments in the Stanford Shared FACS Facility. The results published here are in whole or in part based upon data generated by the TCGA Research Network: http://cancergenome.nih.gov and the ENCODE consortium (see Methods). R.W.J. received fellowship support from the Stanford Cancer Institute. This work was funded by NIH grant K99CA194198 (R.W.J.). J.A.S. was supported by VA grant 1I01BX001957 and NIH grant CA163499. A.J.G. was supported by NIH grants CA67166, CA197713 and CA198291, the Silicon Valley Foundation, The Kimmelman Fund, and the Skippy Frank Foundation.

Author information

Authors and Affiliations

Authors

Contributions

R.W.J. directed the project, developed the hypothesis, performed experiments, analysed data, and wrote the manuscript. E.C.F. designed and developed the LIFR promoter construct, performed experiments and edited the manuscript. M.M.O., M.V., T.A., Y.M., A.R.M. and J.R.J. performed experiments and edited the manuscript. J.A.S. performed experiments, conceptualized and interpreted data. J.Y.W. conceptualized and interpreted data. A.J.G. conceived the hypothesis, interpreted data and edited the manuscript.

Corresponding author

Correspondence to Amato J. Giaccia.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 LIFR, STAT3 and SOCS3 alterations frequently co-occur.

(ac) The Cancer Genome Atlas (TCGA) analysis of invasive breast carcinoma (Nature 2012 dataset) patient samples for mRNA down-regulation of (a) LIFR alone (n = 682 patients with no mRNA down-regulation, n = 17 patients with mRNA down-regulation), (b) STAT3 alone (n = 682 patients with no mRNA down-regulation, n = 22 patients with mRNA down-regulation), and (c) SOCS3 alone (n = 682 patients with no mRNA down-regulation, n = 6 patients with mRNA down-regulation). Logrank test. (d) Tendency for co-occurrence of alterations in LIFR/STAT3 (top line) and STAT3/SOCS3 (bottom line) in patient samples from invasive breast carcinoma (Nature 2012 dataset). Fisher exact test. n = 825 patients. Graphs represent the mean/group and error bars represent standard error of the mean (s.e.m.). P < 0.05 and P < 0.001.

Supplementary Figure 2 LIFR is regulated via DNA methylation in breast cancer patients likely to recur.

(a) Western blot for LIFR in MCF7 whole cell lysates following lentiviral knockdown. MCF7 non-silencing control (MCF7NSC) and MCF7shRNA #3 cells were used in all remaining experiments. Vinculin = loading control. (b) PGK1 mRNA levels for MCF7NSC cells cultured in normoxia (Nx) or hypoxia (Hx; 0.5% oxygen) for 24 h. 3 technical replicates from a single experiment representative of 2 independent experiments. (c) LIFR mRNA levels in SUM159 human breast cancer cells following 24 h culture in normoxia (Nx) or hypoxia (Hx; 0.5% oxygen). 3 technical replicates from a single experiment. (d) Correlation of LIFR mRNA levels (y-axis) or (e) STAT3 mRNA levels (y-axis) and LIFR DNA methylation (x-axis) in patients with invasive breast carcinoma (TCGA Provisional dataset, n = 1,104 patients). Pearson Correlation and Spearman Correlation. (f) LIFR mRNA levels (n = 850 patients disease free; n = 68 patients recurred/progressed) and (g) LIFR DNA methylation (n = 575 patients disease free; n = 51 patients recurred/progressed) in patients with invasive breast carcinoma (TCGA Provisional dataset). Student’s unpaired t-test. Source data for Suppl. 2b available in Supplementary Table 1. Graphs represent the mean/group and error bars represent standard error of the mean (SEM). P < 0.05, P < 0.01, and P < 0.0001.

Supplementary Figure 3 LIF inhibits proliferation in breast cancer cells with low metastatic potential.

(ac) XTT assay to determine proliferation of MCF7, MDA-MB-231b, and 4T1BM2 cancer cells in response to recombinant LIF (50 ng ml−1) or TGF-β (5 ng ml−1) over 3 days. Two-Way ANOVA with Tukey’s multiple comparisons test. n = 3 biological replicates/day. (d,e) SOCS3 mRNA levels in MCF7 and MDA-MB-231b cells following 1 or 6 h treatment with recombinant LIF (0–100 ng ml−1). One-Way ANOVA with Sidak’s multiple comparisons test. n = 3 biological replicates, each being an average from 3 independent experiments. Source data for Suppl. 3ac available in Supplementary Table 1. Graphs represent the mean/group and error bars represent standard error of the mean (SEM). P < 0.05, P < 0.01, and P < 0.0001.

Supplementary Figure 4 LIF stimulates STAT3:SOCS3 induction in breast cancer cells with low metastatic potential.

(a,c,e) Western blot for LIFR, pSTAT3 (Y705), total Stat3, and β-actin or vinculin (loading control) after 15 or 30 min treatment with PBS (vehicle control), recombinant OSM (50 ng ml−1) or recombinant LIF (50 ng ml−1) in (a) PyMT and (c) D2.0R cells with low metastatic potential and (e) D2A1 cells with high metastatic potential. Blots represent 3 independent biological replicates/group. (b,d,f) SOCS3 mRNA levels after 1 or 6 h treatment with PBS, recombinant OSM (50 ng ml−1) or recombinant LIF (50 ng ml−1). h: Mann-Whitney test. j,l: Student’s unpaired t-test.n = 3 biological replicates, each being an average from 3 independent experiments. (g,i) Western blot for LIFR, pSTAT3 (Y705), total Stat3, and vinculin (loading control) after 15 or 30 min treatment with PBS (vehicle control), recombinant OSM (50 ng ml−1) or recombinant LIF (50 ng ml−1) in (g) 4T1 parental cells and (i) MDA-MB-231 parental cells. Blots represent 3 independent biological replicates/group. (h,j) SOCS3 mRNA levels after 1 or 6 h treatment with PBS, recombinant OSM (50 ng ml−1) or recombinant LIF (50 ng ml−1) in (h) 4T1 parental cells and (j) MDA-MB-231 parental cells. One-Way ANOVA. n = 3 biological replicates, each being an average from 3 independent experiments. Unprocessed blots in Supplementary Fig. 9. Graphs represent the mean/group and error bars represent standard error of the mean (SEM). P < 0.05, P < 0.01, P < 0.001, and P < 0.0001.

Supplementary Figure 5 LIFR knockdown does not alter proliferation but confers an invasive phenotype in vitro.

(a) Western blot for pSTAT3 (Y705), total Stat3, and β-actin (loading control) in MCF7NSC and MCF7shLIFR (#3 from Supplementary Fig. 1g) following 15 min treatment with PBS (vehicle control), recombinant OSM (50 ng ml−1) or recombinant LIF (50 ng ml−1). (b) SOCS3 mRNA levels in MCF7NSC and MCF7shLIFR cells following 1 h treatment with PBS, recombinant OSM (50 ng ml−1) or recombinant LIF (50 ng ml−1). 3 technical replicates from a single experiment representative of 2 independent experiments. (c) Cell counting assay to assess changes in proliferation in MCF7NSC and MCF7shLIFR cells over 3 days. 2 biological replicates/point. (d) Average tumor volume for MCF7NSC (n = 4 mice) or MCF7shLIFR (n = 3 mice) tumors inoculated into the mammary fat pad (500K cells/fat pad). Student’s unpaired t-test. (e) Images (left) and quantification (right) of scratch wound assay in MCF7NSC and MCF7shLIFR cells in vitro. Scale bar = 200 μm. Student’s unpaired t-test. 2 biological replicates, one each from 2 independent experiments. (f) 3D culture of MCF7NSC and MCF7shLIFR cells in type I rat collagen. Images taken at day 5 (scale bar = 200 μm) and day 9 (scale bar = 50 μm) after tumor cell seeding. Images representative of 3 independent experiments. Source data for Suppl. 5b-e available in Supplementary Table 1 and unprocessed blots in Supplementary Fig. 9. Graphs represent the mean/group and error bars represent standard error of the mean (SEM). P < 0.05, P < 0.001 and P < 0.0001.##P < 0.01 versus MCF7NSC + LIF treatment.

Supplementary Figure 6 Quiescence and cancer stem cell-associated genes implicated in tumor cell dormancy.

(a) Table of genes identified as validated dormancy genes in ER + ve breast cancer cells and quiescence-related genes. (b) LIFR mRNA levels in SUM159 cells with LIFR lentiviral knockdown. 3 technical replicates from a single experiment. Source data for Suppl. 6b available in Supplementary Table 1. Graphs represent the mean/group and error bars represent standard error of the mean (SEM). P < 0.001.

Supplementary Figure 7 The effects of LIFR signaling on dormancy are mediated via STAT3 versus PI3K:mTOR or MAPK signaling.

(a) TCGA analysis of invasive breast carcinoma (Nature 2012 dataset) patient samples for mRNA down-regulation of LIFR, PIK3CA, and mTOR genes (n = 543 patients with no down-regulation, n = 32 patients with mRNA down-regulation). Logrank test. (b) TCGA analysis of invasive breast carcinoma (Nature 2012 dataset) patient samples for mRNA down-regulation of LIFR, MAPK3, and MAPK1 genes (n = 666 patients with no down-regulation, n = 52 patients with mRNA down-regulation). Logrank test. (c) Western blot for phospho-Akt, Akt, and loading control Actin protein levels in MCF7 cells treated with vehicle (water), 10 nM, 100 nM, or 1,000 nM BEZ235 (PI3K inhibitor) for 24 h. (d) mRNA levels for dormancy and quiescence-associated genes in MCF7 cells after 24 h treatment with BEZ235. Multiple t-tests with Holm-Sidak post-test. n = 3 biological replicates, each being an average from 3 independent experiments. (e) Western blot for phospho-Erk1/2, Erk1/2, and loading control Actin protein levels in MCF7 cells treated with vehicle (DMSO), 0.1 μM, 1 μM, or 10 μM AZD6244 (ERK1/2 inhibitor) for 24 h. (f) mRNA levels for dormancy and quiescence-associated genes in MCF7 cells after 24 h treatment with AZD6244. Multiple t-tests with Holm-Sidak post-test. n = 3 biological replicates, each being an average from 3 independent experiments. (g,h) mRNA levels for cancer stem cell-associated genes in MCF7 cells after 24 h treatment with (g) BEZ235 or (h) AZD6244. Multiple t-tests with Holm-Sidak post-test. n = 3 biological replicates, each being an average from 3 independent experiments. Unprocessed blots in Supplementary Fig. 9. Graphs represent the mean/group and error bars represent standard error of the mean (SEM). P < 0.05, P < 0.01, and P < 0.001.

Supplementary Figure 8 Loss of LIFR in MCF7 cells does not alter lung colonization.

(a) Negative controls for Ki67 staining in MCF7NSC and MCF7shLIFR tumor-bearing tibiae (8 mice/group). Scale bar = 100 μm. (b) Representative PIMO staining at the tumor-bone interface for 2 mice/group (of a total 3 mice/group where this was observed) in MCF7NSC and MCF7shLIFR tumor-bearing bones. Scale bar = 100 μm. (c) Tumor burden/total area histomorphometric analysis of lung sections from MCF7NSC and MCF7shLIFR tumor-inoculated mice. 3/10 MCF7NSC and 1/10 MCF7shLIFR tumor-inoculated mice had tumor detectable by H&E. Representative H&E images on right. Student’s unpaired t-test.n = 10 mice/group. Scale bar = 100 μm. (d) Human β-2microglobulin mRNA levels in homogenized lungs from MCF7NSC and MCF7shLIFR inoculated mice. 6/10 MCF7NSC and 5/10 MCF7shLIFR tumor-inoculated mice had detectable human mRNA in the lungs. HMBS = housekeeping gene. Student’s unpaired t-test.n = 10 mice/group. (e) Analysis of Minn et al. dataset for LIFR mRNA levels in breast cancer patients with a poor prognosis based on Van ’t Veer signature (n = 32 patients with no lung metastasis, n = 13 patients with lung metastasis). Student’s unpaired t-test with Welch’s Correction. (f) Structure of the Stat3 inhibitor ML116 (Stat3i). (g) 3D model of Stat3i docking in predicted DNA binding site of a Stat3 monomer. (h) SOCS3 mRNA levels in MCF7 cells following 1 h treatment with LIF (50 ng ml−1) ± 1 h Stat3i pre-treatment (1–50 μM). 3 technical replicates from a single experiment. (i) mRNA levels of LIFR-independent dormancy/quiescence-associated genes in MCF7 cells following 24 h treatment with 5 μM or 50 μM of Stat3 inhibitor ML116 (Stat3i). Multiple t-tests with Holm-Sidak post-test. n = 3 biological replicates, one each from 3 independent experiments. (j) SOCS3 mRNA levels in MCF7 cells transfected with siRNA against SOCS3. Student’s unpaired t-test.n = 3 biological replicates, each being an average from 3 independent experiments. (k,l) mRNA levels for (k) dormancy and quiescence and (l) cancer stem cell-associated genes in SUM159 cells transfected with siRNA against SOCS3 for 48 h. Student’s unpaired t-test.n = 3 biological replicates, each being an average from 3 independent experiments. Source data for Suppl. 8c,d,h, available in Supplementary Table 1. Graphs represent the mean/group and error bars represent standard error of the mean (SEM). P < 0.05, P < 0.01, P < 0.001, and P < 0.0001.

Supplementary Figure 9 Unprocessed Western Blots.

Unprocessed Western blots developed on a Chemidoc XRS are provided for figures as indicated.

Supplementary information

Supplementary Information

Supplementary Information (PDF 4757 kb)

Supplementary Table 1

Supplementary Information (XLSX 165 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Johnson, R., Finger, E., Olcina, M. et al. Induction of LIFR confers a dormancy phenotype in breast cancer cells disseminated to the bone marrow. Nat Cell Biol 18, 1078–1089 (2016). https://doi.org/10.1038/ncb3408

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3408

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