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:

Bone marrow NG2+/Nestin+ mesenchymal stem cells drive DTC dormancy via TGF-β2

Abstract

In the bone marrow (BM) microenvironment, where breast cancer (BC)-disseminated tumor cells (DTCs) can remain dormant for decades, NG2+/Nestin+ mesenchymal stem cells (MSCs) promote hematopoietic stem cell quiescence. Here we reveal that periarteriolar BM-resident NG2+/Nestin+ MSCs can also instruct BC DTCs to enter dormancy. NG2+/Nestin+ MSCs produce transforming growth factor (TGF)-β2 and bone morphogenetic protein (BMP)7 and activate a quiescence pathway dependent on TGFBRIII and BMPRII, which via p38-kinase, results in p27 induction. Genetic depletion of MSCs or conditional knockout of TGF-β2 in MSCs using an NG2-CreER driver led to bone metastatic outgrowth of otherwise dormant p27+/Ki67 DTCs. Also, patients with estrogen receptor-positive BC without systemic recurrence displayed higher frequency of TGF-β2 and BMP7 detection in the BM. Our results provide direct proof that hematopoietic stem cell dormancy niches control BC DTC dormancy and suggest that aging or extrinsic factors that affect the NG2+/Nestin+ MSC niche homeostasis may result in a break from dormancy and BC bone relapse.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Depletion of NG2+/Nestin+ MSCs awakens dormant DTCs in the BM.
Fig. 2: Dormant to proliferative shift of BM E0771-GFP cancer cells upon depletion of NG2+/Nestin+ MSCs.
Fig. 3: NG2+/Nestin+ MSCs are as a source of pro-dormancy factors TGF-β2 and BMP7 in the BM.
Fig. 4: NG2+/Nestin+ MSCs activate TGF-β2 and BMP7 growth inhibitory signaling in cancer cells.
Fig. 5: Conditional KO of TGF-β2 in NG2+/Nestin+ MSCs awakens dormant DTCs in the BM.
Fig. 6: Schematic representation of the model by which HSC niches induce dormancy of BC DTCs in the BM.

Similar content being viewed by others

Data availability

The data generated during the current study that support the reported findings are available in the manuscript, including in the provided Source Data files and from the corresponding author on reasonable request. Source data are provided with this paper.

References

  1. Chaffer, C. L. & Weinberg, R. A. A perspective on cancer cell metastasis. Science 331, 1559–1564 (2011).

    Article  CAS  Google Scholar 

  2. Sherry, M. M., Greco, F. A., Johnson, D. H. & Hainsworth, J. D. Metastatic breast cancer confined to the skeletal system. An indolent disease. Am. J. Med. 81, 381–386 (1986).

    Article  CAS  Google Scholar 

  3. Engel, J. et al. The process of metastasisation for breast cancer. Eur. J. Cancer 39, 1794–1806 (2003).

    Article  CAS  Google Scholar 

  4. Husemann, Y. et al. Systemic spread is an early step in breast cancer. Cancer Cell 13, 58–68 (2008).

    Article  Google Scholar 

  5. Sanger, N. et al. Disseminated tumor cells in the bone marrow of patients with ductal carcinoma in situ. Int. J. Cancer https://doi.org/10.1002/ijc.25895 (2011).

  6. Braun, S. et al. Cytokeratin-positive cells in the bone marrow and survival of patients with stage I, II, or III breast cancer. N. Engl. J. Med. https://doi.org/10.1056/NEJM200002243420801 (2000).

  7. Chéry, L. et al. Characterization of single disseminated prostate cancer cells reveals tumor cell heterogeneity and identifies dormancy associated pathways. Oncotarget 5, 9939–9951 (2014).

    Article  Google Scholar 

  8. Borgen, E. et al. NR2F1 stratifies dormant disseminated tumor cells in breast cancer patients. Breast Cancer Res. https://doi.org/10.1186/s13058-018-1049-0(2018).

  9. Naume, B. et al. Clinical outcome with correlation to disseminated tumor cell (DTC) status after DTC-guided secondary adjuvant treatment with docetaxel in early breast cancer. J. Clin. Oncol. 32, 3848–3857 (2014).

    Article  Google Scholar 

  10. 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  Google Scholar 

  11. Carlson, P. et al. Targeting the perivascular niche sensitizes disseminated tumour cells to chemotherapy. Nat. Cell Biol. https://doi.org/10.1038/s41556-018-0267-0 (2019).

  12. Johnson, R. W. 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).

    Article  CAS  Google Scholar 

  13. 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  Google Scholar 

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

    Article  CAS  Google Scholar 

  15. Taichman, R. S. et al. GAS6 receptor status is associated with dormancy and bone metastatic tumor formation. PLoS ONE 8, e61873 (2013).

    Article  CAS  Google Scholar 

  16. Yumoto, K. et al. Axl is required for TGF-β2-induced dormancy of prostate cancer cells in the bone marrow. Sci. Rep. 6, 36520 (2016).

    Article  CAS  Google Scholar 

  17. Jung, Y. et al. Endogenous GAS6 and Mer receptor signaling regulate prostate cancer stem cells in bone marrow. Oncotarget 7, 25698–25711 (2016).

    Article  Google Scholar 

  18. Yue, X. et al. Leukemia inhibitory factor promotes EMT through STAT3- dependent miR-21 induction. Oncotarget https://doi.org/10.18632/oncotarget.6756 (2015).

  19. Agarwal, P. et al. Mesenchymal niche-specific expression of Cxcl12 controls quiescence of treatment-resistant leukemia stem cells. Cell Stem Cell 24, 769–784 (2019).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  21. Lawson, M. A. et al. Osteoclasts control reactivation of dormant myeloma cells by remodelling the endosteal niche. Nat. Commun. https://doi.org/10.1038/ncomms9983 (2015).

  22. Yu-Lee, L. Y. et al. Osteoblast-secreted factors mediate dormancy of metastatic prostate cancer in the bone via activation of the TGFbRIII–p38MAPK–pS249/ T252RB pathway. Cancer Res. 78, 2911–2924 (2018).

    Article  CAS  Google Scholar 

  23. Cackowski, F. C. & Taichman, R. S. Parallels between hematopoietic stem cell and prostate cancer disseminated tumor cell regulation. Bone. 119, 82–86 (2019).

    Article  Google Scholar 

  24. Shiozawa, Y. et al. Human prostate cancer metastases target the hematopoietic stem cell niche to establish footholds in mouse bone marrow. J. Clin. Invest. 121, 1298–1312 (2011).

    Article  CAS  Google Scholar 

  25. Wilson, A. et al. Hematopoietic stem cells reversibly switch from dormancy to self-renewal during homeostasis and repair. Cell 135, 1118–1129 (2008).

    Article  CAS  Google Scholar 

  26. Pinho, S. & Frenette, P. S. Haematopoietic stem cell activity and interactions with the niche. Nat. Rev. Mol. Cell Biol. 20, 303–320 (2019).

    Article  CAS  Google Scholar 

  27. Kunisaki, Y. et al. Arteriolar niches maintain haematopoietic stem cell quiescence. Nature 502, 637–643 (2013).

    Article  CAS  Google Scholar 

  28. Hanoun, M. et al. Acute myelogenous leukemia-induced sympathetic neuropathy promotes malignancy in an altered hematopoietic stem cell Niche. Cell Stem Cell (2014) https://doi.org/10.1016/j.stem.2014.06.020

  29. Maryanovich, M. et al. Adrenergic nerve degeneration in bone marrow drives aging of the hematopoietic stem cell niche. Nat. Med. https://doi.org/10.1038/s41591-018-0030-x (2018).

  30. Harper, K. L. et al. Mechanism of early dissemination and metastasis in Her2+ mammary cancer. Nature https://doi.org/10.1038/nature20609 (2016).

  31. Pinho, S. et al. PDGFR and CD51 mark human nestin+ sphere-forming mesenchymal stem cells capable of hematopoietic progenitor cell expansion. J. Exp. Med. 210, 1351–1367 (2013).

    Article  CAS  Google Scholar 

  32. Ranganathan, A. C., Adam, A. P., Zhang, L. & Aguirre-Ghiso, J. A. Tumor cell dormancy induced by p38SAPK and ER-stress signaling: An adaptive advantage for metastatic cells? Cancer Biol. Ther. https://doi.org/10.4161/cbt.5.7.2968 (2006).

  33. Fluegen, G. et al. Phenotypic heterogeneity of disseminated tumour cells is preset by primary tumour hypoxic microenvironments. Nat. Cell Biol. 19, 120–132 (2017).

    Article  CAS  Google Scholar 

  34. Sosa, M. S. et al. NR2F1 controls tumour cell dormancy via SOX9- and RARβ-driven quiescence programmes. Nat. Commun. 6, 1–14 (2015).

    Article  Google Scholar 

  35. Männ, L. et al. CD11c.DTR mice develop a fatal fulminant myocarditis after local or systemic treatment with diphtheria toxin. Eur. J. Immunol. https://doi.org/10.1002/eji.201546245 (2016).

  36. Christiaansen, A. F., Boggiatto, P. M. & Varga, S. M. Limitations of Foxp3+ Treg depletion following viral infection in DEREG mice. J. Immunol. Methods https://doi.org/10.1016/j.jim.2014.03.005 (2014).

  37. Bennett, C. L. et al. Inducible ablation of mouse Langerhans cells diminishes but fails to abrogate contact hypersensitivity. J. Cell Biol. https://doi.org/10.1083/jcb.200501071 (2005).

  38. Xu, C. et al. Stem cell factor is selectively secreted by arterial endothelial cells in bone marrow. Nat. Commun. https://doi.org/10.1038/s41467-018-04726-3 (2018).

  39. Tikhonova, A. N. et al. The bone marrow microenvironment at single-cell resolution. Nature https://doi.org/10.1038/s41586-019-1104-8 (2019).

  40. Vukicevic, S., Latin, V., Chen, P., Batorsky, R. & Reddi, A. H. Localization of osteogenic protein-1 (bone morphogenetic protein-7) during human embryonic development: High affinity binding to basement membranes. Biochem. Biophys. Res. Commun. 198, 693–700 (1994).

    Article  CAS  Google Scholar 

  41. Gregory, K. E. et al. The prodomain of BMP-7 targets the BMP-7 complex to the extracellular matrix. J. Biol. Chem. 280, 27970–27980 (2005).

    Article  CAS  Google Scholar 

  42. Nakahara, F. et al. Engineering a haematopoietic stem cell niche by revitalizing mesenchymal stromal cells. Nat. Cell Biol. https://doi.org/10.1038/s41556-019-0308-3 (2019).

  43. Aguirre-Ghiso, J. A., Ossowski, L. & Rosenbaum, S. K. Green fluorescent protein tagging of extracellular signal-regulated kinase and p38 pathways reveals novel dynamics of pathway activation during primary and metastatic growth. Cancer Res. 64, 7336–7345 (2004).

    Article  CAS  Google Scholar 

  44. Regot, S., Hughey, J. J., Bajar, B. T., Carrasco, S. & Covert, M. W. High-sensitivity measurements of multiple kinase activities in live single cells. Cell 157, 1724–1734 (2014).

    Article  CAS  Google Scholar 

  45. Oki, T. et al. A novel cell-cycle-indicator, mVenus-p27K. Sci. Rep. 4, 4012 (2014).

    Article  Google Scholar 

  46. Asada, N. et al. Differential cytokine contributions of perivascular haematopoietic stem cell niches. Nat. Cell Biol. https://doi.org/10.1038/ncb3475 (2017).

  47. Zhou, B. O., Yue, R., Murphy, M. M., Peyer, J. G. & Morrison, S. J. Leptin-receptor-expressing mesenchymal stromal cells represent the main source of bone formed by adult bone marrow. Cell Stem Cell https://doi.org/10.1016/j.stem.2014.06.008 (2014).

  48. Paget, S. The distribution of secondary growths in cancer of the breast. Lancet https://doi.org/10.1016/S0140-6736(00)49915-0 (1889).

  49. Yang, Y. et al. Immunocompetent mouse allograft models for development of therapies to target breast cancer metastasis. Oncotarget 8, 30621–30643 (2017).

    Article  Google Scholar 

  50. Boudreau, N. & Bissell, M. J. Extracellular matrix signaling: Integration of form and function in normal and malignant cells. Curr. Opin. Cell Biol. 10, 640–646 (1998).

    Article  CAS  Google Scholar 

  51. Kenny, P. A. & Bissell, M. J. Tumor reversion: Correction of malignant behavior by microenvironmental cues. Int. J. Cancer 107, 688–695 (2003).

    Article  CAS  Google Scholar 

  52. Turley, R. S. et al. The type III transforming growth factor-β receptor as a novel tumor suppressor gene in prostate cancer. Cancer Res. 67, 1090–1098 (2007).

    Article  CAS  Google Scholar 

  53. Ajiboye, S., Sissung, T. M., Sharifi, N. & Fig, W. D. More than an accessory: Implications of type III transforming growth factor-β receptor loss in prostate cancer. BJU Int. 105, 913–916 (2010).

    Article  CAS  Google Scholar 

  54. Dong, M. et al. The type III TGF-β receptor suppresses breast cancer progression. J. Clin. Invest. 117, 206–217 (2007).

    Article  CAS  Google Scholar 

  55. Kim, I. Y. et al. Loss of expression of bone morphogenetic protein receptor type II in human prostate cancer cells. Oncogene 23, 7651–7659 (2004).

    Article  CAS  Google Scholar 

  56. Singh, A. et al. Angiocrine signals regulate quiescence and therapy resistance in bone metastasis. JCI Insight https://doi.org/10.1172/jci.insight.125679 (2019).

  57. Mignone, J. L., Kukekov, V., Chiang, A. S., Steindler, D. & Enikolopov, G. Neural stem and progenitor cells in nestin-GFP transgenic mice. J. Comp. Neurol. 469, 311–324 (2004).

    Article  CAS  Google Scholar 

  58. Ishtiaq Ahmed, A. S., Bose, G. C., Huang, L. & Azhar, M. Generation of mice carrying a knockout-first and conditional-ready allele of transforming growth factor β2 gene. Genesis 52, 817–826 (2014).

    Article  CAS  Google Scholar 

  59. Ossowski, L. & Reich, E. Changes in malignant phenotype of a human carcinoma conditioned by growth environment. Cell 33, 323–333 (1983).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the Aguirre-Ghiso laboratory for helpful discussions and the assistance of the Dean’s Flow Cytometry CoRE, Microscope CoRE and Small Animal Imaging CoRE at BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai. We are grateful to C. Prophete (Frenette’s laboratory) for mouse husbandry. We thank M. Rypdal, A.V. Pladsen and O.C. Lingjærde for preparing the BC DTC material and data. The BC DTC work in Oslo has received funding from the Norwegian Cancer Association and Norwegian Health Region South-East (H.G. Russnes and B. Naume). Grant support was received from the National Institutes of Health (NIH)/National Cancer Institute (NCI) CA109182, CA216248, CA218024, CA196521, The V-Foundation and the Samuel Waxman Cancer Research Foundation Tumor Dormancy Program to J.A.A.G.; HL069438, DK056638, DK116312 and DK112976 to P.S.F.; Portuguese Foundation for Science and Technology (SFRH/BD/100380/2014) to A.R.N.; MD/PhD program of the University of Lyon and the Ecole Normale Supérieure of Lyon to E.R.; Susan G. Komen Career Catalyst Research (CCR18547848), NCI Career Transition Award (K22CA196750), NIH (R01CA244780) and Tisch Cancer Institute NIH Cancer Center Grant (P30-CA196521) to J.J.B.C.; Instituto Serrapilheira/Serra-1708-15285 to A.B. and NIH (HL126705, CA218578, R01HL145064) and American Heart Association (17GRNT33650018) to M.A.

Author information

Authors and Affiliations

Authors

Contributions

A.R.N. designed, planned and conducted experiments, analyzed data and wrote the manuscript; E.R., D.K.S. and J.F.C. conducted experiments; J.S.d.M. and J.J.B.C. helped with optimizing the imaging experiments; J.W., J.J. and M.A. provided necessary mouse model and samples and TGF-β2 expertise input; H.G.R. and B.N. provided human data; A.B. and P.S.F. provided mouse models and BM expertise input; P.S.F. and J.A.A.G. designed experiments, analyzed data and wrote the manuscript.

Corresponding author

Correspondence to Julio A. Aguirre-Ghiso.

Ethics declarations

Competing interests

J.A.A.G. is a scientific co-founder of, scientific advisory board member and equity owner in HiberCell and receives financial compensation as a consultant for HiberCell, a Mount Sinai spin-off company focused on the research and development of therapeutics that prevent or delay the recurrence of cancer.

Additional information

Peer review information Nature Cancer thanks the anonymous reviewers for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 Control of depletion of NG2+/Nestin+ MSCs and awakening of dormant PyMT-CFP DTCs in the BM..

a, 7-week old NG2-CreERiDTR mice (NG2-CreER- or +) were daily injected with tamoxifen for 5 days, followed by 1-day rest and 2 days with diphtheria toxin (DT). 24 h later 2 × 105 E0771-GFP cells were intra-cardiac injected and 2 weeks later mice were euthanized. b,c, FACS plots (b) and quantifications (c) confirming the depletion of NG2+/Nestin+ MSCs (CD45Ter119CD31PDGFRα+CD51+) in NG2-CreERiDTR mice upon TAM and DT treatments compared with WT mice (n = 5 WT and 5 iDTR mice, median, 2-tailed Mann–Whitney U-test, p = 0.008). d-g, Detection and characterization of MMTV-PyMT-CFP cells in BM flushes by FACS (n = 5 WT and 5 iDTR mice). e, Incidence of bone metastasis (>1000 GFP+ DTCs/106 BM cells) 2 weeks after cancer cell injections (2-tailed Fisher’s exact test). f, Number of MMTV-PyMT-CFP cancer cells per million BM cells (median, 2-tailed Mann–Whitney U-test). g, Percentage of Ki67high E0771-GFP cancer cells (median, 2-tailed Mann–Whitney U-test). *p≤0.05 (p-values indicated above). n.s. not significant.

Source data

Extended Data Fig. 2 Pro-inflammatory cytokine status, vessel permeability and BM DTC seeding in NG2+/Nestin+ MSCs-depleted mice.

a, Levels of pro-inflammatory cytokines in BM supernatant of WT and NG2-CreERiDTR mice (n = 13 WT and 11 iDTR mice, from 2 independent experiments, median and interquartile range, 2-tailed Mann–Whitney tests, *p≤0.05, n.s. not significant, n.d. not detected). b-d, NG2-CreERiDTR mice (NG2-CreER- or +) were daily i.p. injected with tamoxifen for 5 days followed by a rest day and 2 i.p. injections of DT. E0771-GFP cells were intra-cardiac injected and 24 h after 70 K Dextran-TexasRed was injected 15 minutes prior euthanasia. c, Representative images of Dextran extravasation in perfused bones. Arrows, E0771-GFP cells. d, Number of E0771-GFP cells detected after 1 week of in vitro expansion of the BM aspirates collected 24 h after injection into mice (n = 5 WT and 5 iDTR mice, median). e,f, NG2-CreERiDTR mice (NG2-CreER- or +) were daily i.p. injected with TAM for 5 days, followed by intra-cardiac injection of E0771-GFP cancer cells. Cells were allowed to disseminate and extravasate for 72 h followed by 2 i.p. injections of DT. f, Number of E0771-GFP cells/million BM cells (n = 5 WT and 5 iDTR mice, median and 2-tailed Mann–Whitney tests, p=0.015). *p≤0.05 (p-values indicated above), n.s. not significant.

Source data

Extended Data Fig. 3 Effect of NG2+/Nestin+ MSCs, TGFβ2 and BMP7 on signalling pathways and growth cancer cells.

a, 3D Matrigel assay used to track solitary cell to cluster growth. Single cells were plated on top of Matrigel in low density and the percentage of single cells, doublets and clusters was quantified 4 days after. b,c, Co-culture of human HNSCC PDX-derived T-HEp3 (b, n = 4 independent experiments, 4 wells per condition, mean and SEM, 2-tailed Mann–Whitney tests p=0.04) and mouse BC MMTV-PyMT (c, n = 2 independent experiments, 4 wells per condition, mean) cells with sorted Nestin-GFP- and Nestin-GFP+ MSCs for 4 days. d-k, E0771 cells were treated every day for 4 days with TGFβ2, BMP7 or bone marrow conditioned media (BM-CM) of TGFβ2+/+ or TGFβ2+/- mice. d and h. Percentage of cancer cells in a single cell, doublet or cluster state with the indicated treatments (d, n = 5 independent experiments, 4 wells per condition each, mean and SEM, 2-tailed Mann–Whitney tests, p=0.03 and 0.005; h, n = 4 independent experiments, 4 wells per condition each, mean and SEM, 2-tailed Mann–Whitney tests, p=0.000005, 0.0001, 0.002 (Ct vs. TGFβ2+/+: CS, doublets and clusters respectively) and 0.02 (TGFβ2+/+ vs. TGFβ2+/- clusters)). e-g and i-k. Percentage of positive cells for c-Cas-3, p-ATF2 and p27 upon the indicated treatments (n = 2 independent experiments, 4 wells per condition each, mean, minimum and maximum). l, Western blots for the indicated antigens detected in E0771 cells treated for 24 h with the TGFβ2, BMP7 and different BM-CM preparations. Molecular weight markers in kDa. *p≤0.05, p-values indicated above.

Source data

Extended Data Fig. 4 Effect of TGFβ2 and BMP7 on signalling pathways (using ELK, p38 and p27 sensors) and MSCs in E0771 cancer cell growth.

a-d, T-HEp3 cells with ERK, p38 and p27 activity biosensors were reversed transfected with control siRNA or siRNAs for TGFBRIII and BMPRII followed by 24-hour treatments with TGFβ2 and BMP7. a, TGFBRIII and BMPRII mRNA levels 48 h after transfection with the indicated siRNAs (n = 3 independent experiments, mean, minimum and maximum, 2-tailed Mann–Whitney tests, p=0.01 (TGFbR3) and 0.002 (BMPR2)). bd, Quantification of the T-HEp3-biosensors activity (n = 3 independent experiments each, mean, minimum and maximum, 2-tailed Mann–Whitney tests, p=0.05). e, Percentage of E0771 cells in a single cell (p = 0.002), doublet or cluster (p = 0.005) state after co-culture with Control (passaged) or revitalized (r) MSCs (n = 4 independent experiments, 4 wells per condition per experiment, mean and SEM, 2-tailed Mann–Whitney tests). f, qPCR of TGFβ1, TGFβ2 and BMP7 from Control and rMSCs (n = 2). *p≤0.05, p-values indicated above.

Source data

Extended Data Fig. 5 NG2-CreERTGFβ2 mouse model controls.

a, TGFβ1 mRNA levels in NG2+/Nestin+ MSCs (sorted using CD45Ter119CD31PDGFRα+CD51+ markers) in NG2-CreERTGFβ2 mice upon TAM treatments compared with WT mice (n = 4 independent experiments). b, TGFβ1, TGFβ2 and BMP7 mRNA levels in MSCs (CD45CD31Ter119PDGFRα+CD51+), osteo-progenitor (CD45Ter119CD31ALCAM+, OPCs) and endothelial (CD45Ter119CD31+vEcad+, ECs) cells from NG2-CreERTGFβ2 mice upon TAM treatments compared with WT mice (n = 2 independent experiments). *p≤0.05, p-values indicated above.

Source data

Supplementary information

Reporting Summary

Supplementary Fig. 1

FACS strategy and gates used to sort BM cells. a, FACS strategy and gates used to sort CD45CD31Nestin-GFP or +/bright BM cells. b, FACS strategy and gates used to sort MSCs (CD45CD31Ter119PDGFRa+CD51+, MSCs), osteo-progenitor cells (CD45Ter119CD31ALCAM+, OPCs) and endothelial cells (CD45Ter119CD31+vEcad+, ECs) from WT mice.

Supplementary Video 1

Movie of a typical sonogram used to conduct imaging-guided injection of cells into the left ventricle of the heart. This methodology ensures intracardiac injection efficiency, minimizing variability of injections on BM DTC burden analysis.

Source data

Source Data Fig. 1

Raw numerical data behind graphs from Fig. 1.

Source Data Fig. 2

Raw numerical data behind graphs from Fig. 2.

Source Data Fig. 3

Raw numerical data behind graphs from Fig. 3.

Source Data Fig. 4

Raw numerical data behind graphs from Fig. 4.

Source Data Fig. 5

Raw numerical data behind graphs from Fig. 5.

Source Data Extended Data Fig. 1

Raw numerical data behind graphs from Extended Data Fig. 1.

Source Data Extended Data Fig. 2

Raw numerical data behind graphs from Extended Data Fig. 2.

Source Data Extended Data Fig. 3

Raw numerical data behind graphs from Extended Data Fig. 3.

Source Data Extended Data Fig. 3

Unprocessed western blots from Extended Data Fig. 3.

Source Data Extended Data Fig. 4

Raw numerical data behind graphs from Extended Data Fig. 4.

Source Data Extended Data Fig. 5

Raw numerical data behind graphs from Extended Data Fig. 5.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nobre, A.R., Risson, E., Singh, D.K. et al. Bone marrow NG2+/Nestin+ mesenchymal stem cells drive DTC dormancy via TGF-β2. Nat Cancer 2, 327–339 (2021). https://doi.org/10.1038/s43018-021-00179-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-021-00179-8

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