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:

Radiation exposure elicits a neutrophil-driven response in healthy lung tissue that enhances metastatic colonization

An Author Correction to this article was published on 04 April 2022

This article has been updated

Abstract

Radiotherapy is one of the most effective approaches to achieve tumor control in cancer patients, although healthy tissue injury due to off-target radiation exposure can occur. In this study, we used a model of acute radiation injury to the lung, in the context of cancer metastasis, to understand the biological link between tissue damage and cancer progression. We exposed healthy mouse lung tissue to radiation before the induction of metastasis and observed a strong enhancement of cancer cell growth. We found that locally activated neutrophils were key drivers of the tumor-supportive preconditioning of the lung microenvironment, governed by enhanced regenerative Notch signaling. Importantly, these tissue perturbations endowed arriving cancer cells with an augmented stemness phenotype. By preventing neutrophil-dependent Notch activation, via blocking degranulation, we were able to significantly offset the radiation-enhanced metastases. This work highlights a pro-tumorigenic activity of neutrophils, which is likely linked to their tissue regenerative functions.

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: Radiation exposure in healthy lung tissue enhances metastasis.
Fig. 2: Radiation exposure induces the infiltration and local activation of lung neutrophils.
Fig. 3: Primed neutrophils support metastatic colonization in irradiated lungs.
Fig. 4: Radiation-primed neutrophils perturb the lung tissue environment.
Fig. 5: Notch is activated in the lung epithelium and enhances spontaneous metastasis.
Fig. 6: Radiation exposure boosts Notch signaling and stemness in metastatic cells.
Fig. 7: Radiation-primed neutrophils boost lung metastasis via their degranulation activity.
Fig. 8: Inhibition of Notch signaling attenuates the radiation-driven enhancement of metastatic growth in vivo.

Similar content being viewed by others

Data availability

The bulk RNA sequencing datasets (GSE180823) and the single-cell RNA sequencing datasets (GSE181306) are deposited in the Gene Expression Omnibus (GEO, NCBI) repository. The proteomic datasets are deposited in PRoteomics IDEntifications (PRIDE) repository (PXD027628). Source data are provided with this paper.

Change history

References

  1. Wirsdörfer, F. & Jendrossek, V. The role of lymphocytes in radiotherapy-induced adverse late effects in the lung. Front. Immunol 7, 207–220 (2016).

    Article  Google Scholar 

  2. Ombrato, L. et al. Metastatic-niche labelling reveals parenchymal cells with stem features. Nature 572, 603–608 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Vicini, F. A., Kestin, L., Huang, R. & Martinez, A. Does local recurrence affect the rate of distant metastases and survival in patients with early-stage breast carcinoma treated with breast-conserving therapy? Cancer 97, 910–919 (2003).

    Article  PubMed  Google Scholar 

  4. Kuonen, F., Secondini, C. & Rüegg, C. Molecular pathways: emerging pathways mediating growth, invasion, and metastasis of tumors progressing in an irradiated microenvironment. Clin. Cancer Res. 18, 5196–5202 (2012).

    Article  CAS  PubMed  Google Scholar 

  5. Bouchard, G. et al. Pre-irradiation of mouse mammary gland stimulates cancer cell migration and development of lung metastases. Br. J. Cancer 109, 1829–1838 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Brown, J. M. The effect of lung irradiation on the incidence of pulmonary metastases in mice. Br. J. Radiol. 46, 613–618 (1973).

    Article  CAS  PubMed  Google Scholar 

  7. Dao, T. L. & Kovaric, J. Incidence of pulmonary and skin metastases in women with breast cancer who received postoperative irradiation. Surgery 52, 203–212 (1962).

    Google Scholar 

  8. Wang, J. Neutrophils in tissue injury and repair. Cell. Tissue. Res. 371, 531–539 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Kolaczkowska, E. & Kubes, P. Neutrophil recruitment and function in health and in inflammation. Nat. Rev. Immunol. 13, 159–175 (2013).

    Article  CAS  PubMed  Google Scholar 

  10. Paris, A. J. et al. Neutrophils promote alveolar epithelial regeneration by enhancing type II pneumocyte proliferation in a model of acid-induced acute lung injury. Am. J. Physiol. 311, L1062–L1075 (2016).

    Google Scholar 

  11. Zemans, R. L. et al. Neutrophil transmigration triggers repair of the lung epithelium via β-catenin signaling. Proc. Natl Acad. Sci. USA 108, 15990–15995 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Hedrick, C. C. & Malanchi, I. Neutrophils in cancer: heterogeneous and multifaceted. Nat. Rev. Immunol. https://doi.org/10.1038/s41577-021-00571-6 (2021).

  13. Jones, B., Dale, R. G., Deehan, C., Hopkins, K. I. & Morgan, D. A. The role of biologically effective dose (BED) in clinical oncology. Clin Oncol (R Coll Radiol) 13, 71–81 (2001).

    CAS  Google Scholar 

  14. Emami, B. et al. Tolerance of normal tissue to therapeutic irradiation. Int. J. Radiat. Oncol. Biol. Phys. 21, 109–122 (1991).

    Article  CAS  PubMed  Google Scholar 

  15. Ballesteros, I. et al. Co-option of neutrophil fates by tissue environments. Cell https://doi.org/10.1016/j.cell.2020.10.003 (2020).

  16. Granot, Z. et al. Tumor entrained neutrophils inhibit seeding in the premetastatic lung. Cancer Cell 20, 300–314 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Takeshima, T. et al. Key role for neutrophils in radiation-induced antitumor immune responses: potentiation with G-CSF. Proc. Natl Acad. Sci. USA 113, 11300–11305 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Rayes, El,T. et al. Lung inflammation promotes metastasis through neutrophil protease-mediated degradation of Tsp-1. Proc. Natl Acad. Sci. USA 112, 16000–16005 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Park, J. et al. Cancer cells induce metastasis-supporting neutrophil extracellular DNA traps. Sci. Transl. Med. 8, 361ra138 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Albrengues, J. et al. Neutrophil extracellular traps produced during inflammation awaken dormant cancer cells in mice. Science 361, eaao4227 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Yang, L. et al. DNA of neutrophil extracellular traps promotes cancer metastasis via CCDC25. Nature 583, 133–138 (2020).

    Article  CAS  PubMed  Google Scholar 

  22. Lieschke, G. J. et al. Mice lacking granulocyte colony-stimulating factor have chronic neutropenia, granulocyte and macrophage progenitor cell deficiency, and impaired neutrophil mobilization. Blood 84, 1737–1746 (1994).

    Article  CAS  PubMed  Google Scholar 

  23. Lee, J.-H. et al. Anatomically and functionally distinct lung mesenchymal populations marked by Lgr5 and Lgr6. Cell 170, 1149–1156.e12 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. McConnell, A. M., Konda, B., Kirsch, D. G. & Stripp, B. R. Distal airway epithelial progenitor cells are radiosensitive to High-LET radiation. Sci Rep. 6, 33455–11 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Liu, J., Sato, C., Cerletti, M. & Wagers, A. Notch signaling in the regulation of stem cell self-renewal and differentiation. Curr. Top. Dev. Biol 92, 367–409 (2010).

    Article  CAS  PubMed  Google Scholar 

  26. Murtaugh, L. C., Stanger, B. Z., Kwan, K. M. & Melton, D. A. Notch signaling controls multiple steps of pancreatic differentiation. Proc. Natl Acad. Sci. USA 100, 14920–14925 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Ombrato, L. et al. Generation of neighbor-labeling cells to study intercellular interactions in vivo. Nat. Protoc. 16, 872–892 (2021).

    Article  CAS  PubMed  Google Scholar 

  28. Wang, J., Sullenger, B. A. & Rich, J. N. Notch signaling in cancer stem cells. Adv. Exp. Med. Biol 727, 174–185 (2012).

    Article  CAS  PubMed  Google Scholar 

  29. Meier-Stiegen, F. et al. Activated Notch1 target genes during embryonic cell differentiation depend on the cellular context and include lineage determinants and inhibitors. PLoS ONE 5, e11481–18 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Johnson, J. L. et al. Identification of neutrophil exocytosis inhibitors (Nexinhibs), small molecule inhibitors of neutrophil exocytosis and inflammation. J. Biol. Chem. 291, 25965–25982 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Malanchi, I. et al. Interactions between cancer stem cells and their niche govern metastatic colonization. Nature 481, 85–89 (2011).

    Article  PubMed  Google Scholar 

  32. Martin, Y. et al. Mesenchymal cancer cell-stroma crosstalk promotes niche activation, epithelial reversion, and metastatic colonization. Cell Reports 22, 2456–2469 (2015).

    Article  Google Scholar 

  33. Rock, J. R. et al. Notch-dependent differentiation of adult airway basal stem cells. Cell. Stem Cell 8, 639–648 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Xing, Y., Li, A., Borok, Z., Li, C. & Minoo, P. NOTCH1 is required for regeneration of Clara cells during repair of airway injury. Stem Cells 30, 946–955 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Finn, J. et al. Dlk1-mediated temporal regulation of Notch signaling Is required for differentiation of alveolar type II to type I cells during repair. Cell Reports 26, 2942–2954.e5 (2019).

    Article  CAS  PubMed  Google Scholar 

  36. Vaughan, A. E. et al. Lineage-negative progenitors mobilize to regenerate lung epithelium after major injury. Nature 517, 621–625 (2015).

    Article  CAS  PubMed  Google Scholar 

  37. Giuranno, L. et al. NOTCH signaling promotes the survival of irradiated basal airway stem cells. Am. J. Physiol. 317, L414–L423 (2019).

    CAS  Google Scholar 

  38. Fan, X. et al. NOTCH pathway blockade depletes CD133-positive glioblastoma cells and inhibits growth of tumor neurospheres and xenografts. Stem Cells 28, 5–16 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Hoey, T. et al. DLL4 blockade inhibits tumor growth and reduces tumor-initiating cell frequency. Cell. Stem Cell 5, 168–177 (2009).

    Article  CAS  PubMed  Google Scholar 

  40. Zheng, Y. et al. A rare population of CD24(+)ITGB4(+)Notch(hi) cells drives tumor propagation in NSCLC and requires Notch3 for self-renewal. Cancer Cell 24, 59–74 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Mamun, M. A., Mannoor, K., Cao, J., Qadri, F. & Song, X. SOX2 in cancer stemness: tumor malignancy and therapeutic potentials. J. Mol. Cell. Biol. 12, 85–98 (2020).

    Article  PubMed  Google Scholar 

  42. Aguilar-Medina, M. et al. SOX9 stem-cell factor: clinical and functional relevance in cancer. J. Oncology 2019, 6754040 (2019).

    Article  Google Scholar 

  43. Zhang, H., Lu, J., Liu, J., Zhang, G. & Lu, A. Advances in the discovery of exosome inhibitors in cancer. J. Enzyme Inhib. Med. Chem. 35, 1322–1330 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Polverino, E., Rosales-Mayor, E., Dale, G. E., Dembowsky, K. & Torres, A. The role of neutrophil elastase inhibitors in lung diseases. Chest 152, 249–262 (2017).

    Article  PubMed  Google Scholar 

  45. Wculek, S. K. & Malanchi, I. Neutrophils support lung colonization of metastasis-initiating breast cancer cells. Nature 528, 413–417 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Lee, J.-H. et al. Lung stem cell differentiation in mice directed by endothelial cells via a BMP4-NFATc1-thrombospondin-1 axis. Cell 156, 440–455 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet J. 17, 10–12 (2011).

    Article  Google Scholar 

  48. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  49. Li, B. & Dewey, C. N. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinform. 12, 323 (2011).

    Article  CAS  Google Scholar 

  50. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Anders, S. & Huber, W. Differential expression analysis for sequence count data. Gen. Biol. 11, R106 (2010).

    Article  CAS  Google Scholar 

  52. Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Hafemeister, C. & Satija, R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Gen. Biol. 20, 296–15 (2019).

    Article  CAS  Google Scholar 

  54. DeTomaso, D. et al. Functional interpretation of single cell similarity maps. Nat. Commun. 10, 4376 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgments

We are grateful to N. Osborne from the Safety, Health and Sustainability Team at the Francis Crick Institute for his invaluable support during this work. We thank E. Nye from the Experimental Histopathology Unit at the Crick Institute for histological processing and advice and D. Barry from the Advanced Light Microscopy Facility at the Crick Institute for image analysis advice. We are grateful to P. Chakravarty from the Bioinformatics & Biostatistics Facility at the Crick Institute for bioinformatics support. We thank B. Snijders from the Proteomics Facility at the Crick Institute and R. Goldstone and A. Edwards from the Advanced Sequencing Facility at the Crick Institute for their technical support. We are also grateful for support from the Flow Cytometry Unit, the Cell Services Unit and the Biological Resources Unit at the Francis Crick Institute, and in particular we thank D. Poniskaitiene and T. Zverev specifically for their extended contributions to welfare monitoring of experimental mice and for performing oral gavages. We are grateful to S. Quezada for his support for the image-guided focused irradiations. We thank A. Wilkins, Clinician Scientist at the ICR and Honorary Consultant at the Royal Marsden, London, for providing critical input on the manuscript sections discussing the clinical relevance of the study. We thank R. Ferreira for critical reading of the manuscript. This work was supported by the Francis Crick Institute, which receives its core funding from Cancer Research UK (grant no. FC001112), the UK Medical Research Council (grant no. FC001112), and the Wellcome Trust (grant no. FC001112) and the European Research Council (grant no. ERC CoG-H2020-725492). The work at UCL was supported by the Radiation Research Unit at the Cancer Research UK City of London Centre Award (no. C7893/A28990).

Author information

Authors and Affiliations

Authors

Contributions

E.N. designed and performed most of the experiments, analyzed and interpreted the data. V.L.B. managed colony breeding and generated experimental mice, supported the animal work and provided technical support and discussion. L.O. developed the sLP-Cherry labeling tool and provided valuable discussion. A.K and N.R. performed the single-cell RNA analysis. C.A.N.S. performed the γ-H2AX immunofluorescence and SA-β-gal staining. M.V. performed the injections and harvested mice for the image-guided partial-lung irradiation. S.H. performed bioinformatic analysis. F.S.R. provided technical support. P.F. performed the neutrophil proteomics. R.C. planned and performed the image-guided, focused partial-lung radiation. L.O., A.K., N.R. and V.L.B. critically reviewed the manuscript. I.M. designed and supervised the study and interpreted the data. I.M and E.N. wrote the manuscript.

Corresponding author

Correspondence to Ilaria Malanchi.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cancer thanks Marc Vooijs and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Radiation exposure in healthy lung tissue enhances metastasis.

a, Representative H&E images of metastatic lungs from control (UT) and irradiated BALB/c mice orthotopically injected with 4T1 breast cancer cells to generate a primary tumour. The metastatic area is depicted with a dashed line (n = 4 mice per group, 2 independent experiments). Scale bar, 250 μm. b,c, FACS quantification of GFP+ tumour cells in the metastatic lung (b) and primary tumour volume (c) from control and irradiated mice (n = 4 mice per group, one experiment). d,e, Representative H&E images (d) and FACS quantification of GFP+ tumour cells (e) from metastatic lungs from control (UT) or irradiated FVB mice intravenously injected with GFP+ MMTV-PyMT primary mammary tumour cells at day 7 (n = 5 per group, one experiment). Scale bar, 100 μm. f, Table and representative immunostaining of human-specific Lamin A/C to detect human lung cancer cells growing in BALB/c Nude mice (n = 4 mice per group for each cell line tested). Mice were intravenously injected with H460 or A549 human NSCLC cells 7 days following targeted lung irradiation (or sham-irradiation for control mice) and lungs harvested 3 weeks later. The table depicts the number of mice in which h-Lamin A/C+ cancer cells were present in the lungs. No foci were detected in control mice from either experimental group. The representative images show an example of a Lamin A/C+ metastatic foci within an irradiated lung, for each cell line. The H460 metastatic lesion (left) is indicated with the arrow. Scale bar, 250 μm. g, Violin plot showing the number of metastatic foci in the non-irradiated left lung lobe from mice that received image-guided targeted radiation to the right lung (n = 5 for 8 Gy mice; n = 7 for 12 Gy mice), compared with control mice (n = 6). The violin plot displays the median, 25th and 75th percentiles as well as the density of data points. Dots represent individual mice. All data represented as mean ± s.e.m. Statistical analysis by non-parametric two-tailed Mann-Whitney test for (b), (c) and (e) and a one-way ANOVA with multiple comparisons for (g). UT, untreated. IR, irradiated. Gating strategies for FACS analysis provided in Extended Data Fig. 2. Histology quantification process outlined in Supplementary File 1.

Source data

Extended Data Fig. 2 FACS gating strategy.

a-c, Example of FACS gating strategy to determine the frequency of (a) GFP+ cancer cells or (b) Ly6G+ neutrophils in the lung tissue of control/irradiated mice, or (c) Lineage/EpCAM+ epithelial cells in the irradiated metastatic niche (Cherry-labelled, left), or unlabelled distant lung (Cherry-negative, right). All samples were gated to exclude debris and doublets, followed by live cell discrimination by DAPI staining. All gates were set based on fluorescence-minus-one (FMO) controls, containing all antibodies minus the one of interest, to determine the background signal. Importantly, lung tissue displays a high level of autofluorescence, which needs to be considered when excluding dead cells (an FMO-DAPI is critical for this).

Extended Data Fig. 3 Radiation exposure induces lung perturbations including neutrophil infiltration and activation.

a,b, Representative immunofluorescent images (a) and quantification (b) of phospho-Histone H2A.X (Ser139) (green) and DAPI (blue) stained lungs from control/irradiated mice, 7 days post-irradiation (n = 3 mice per group, one experiment). Scale bar, 10 μm. 6 fields of view were quantified per mouse. c,d, Representative images of senescence-associated β-galactosidase (SA-β-gal) staining on lung cryosections (c) and quantification (d) from control/irradiated mice, 7 days post-irradiation (n = 3 mice per group, one experiment). Scale bar, 25 μm. e, Quantification of immunostaining for S100A9+ neutrophils in metastases from control and irradiated lungs at day 14 (7 days post-i.v., Fig. 1d) (n = 6 mice per group, 2 independent experiments). The number of neutrophils within the metastatic area was normalised to tumour area. f, FACS quantification of GFP+ cancer cells in control/irradiated lungs from RAG1-ko mice at day 14 (7 days post-IV). n = 9 mice per group, two independent experiments, grey dots C57BL/6J and white dots FVB background. g,h, Volcano plots showing protein expression from irradiated versus control lung (g) and bone marrow (h) neutrophils. A selection of differentially expressed proteins in the lung are depicted in red, with the same proteins shown in bone marrow samples (n = 3 mice per group). i, Table of granule proteins within primary, secondary or tertiary granules with their fold change in irradiated lung neutrophils (IR) vs untreated control (UT). Nd = not detected. Highly enriched proteins are highlighted in red. j, Mice received an intraperitoneal EdU injection (25 mg/kg) 1 h prior to lung irradiation. Lungs/bone marrow were harvested 1 h or 7 days later, and EdU incorporation assessed by FACS. k, EdU incorporation in Ly6G+ neutrophils from bone marrow (left) and lungs (right) from control (UT) and irradiated (IR) mice (n = 2 control/irradiated mice at 2 h, n = 3 control/irradiated mice at day 7). A representative of two independent experiments is shown (total for both experiments n = 5 control/irradiated at 2 h,, n = 6 control/irradiated mice at day 7). All data represented as mean ± s.e.m. Statistical analysis by two-tailed t-test with Welch’s correction for (d) and (e), two-tailed non-parametric Mann-Whitney test for (f) and (k) and one-sample t-test (for value different from 2) for (b). UT, untreated; IR, irradiated. FACS gating strategies provided in Extended Data Fig. 2.

Source data

Extended Data Fig. 4 Radiation-primed neutrophil fuel metastatic growth independently of NETosis and extravasation.

a, Experimental setup. Irradiated mice were given daily injections of an anti-Ly6G neutrophil depletion antibody or IgG control, beginning the day before an IV injection of 4T1-GFP+ cancer cells. Mice were collected 72 h post-i.v. b,c, Frequency of Ly6G+ neutrophils (b) and GFP+ cancer cells (c) among live cells by FACS (n = 7 mice per group, 2 independent experiments). d, Representative immunofluorescent images of myeloperoxidase (MPO, green), citrullinated histone-H3 (Cit-H3, red) and DAPI (blue) stained lungs at 2 h, 24 h and 7 days post-irradiation (n = 3 mice per group, each timepoint). The positive control represents lung tissue from a mouse infected intratracheally with C.albicans and harvested 24 h later. Scale bar, 100 μm. e, Schematic of 3D co-cultures. GFP+MMTV-PyMT cancer cells were seeded in AlvetexTM Scaffold 96-well plates with MACS-sorted Ly6G+ neutrophils from control or irradiated mice, harvested 7 days after irradiation. f, GFP signal quantification. Cancer cell growth on the scaffold is shown as the fold change compared to cancer cells alone (n = 3 mice, each with at least 3 technical replicates, see methods). g, Schematic of GFP+MMTV-PyMT cancer cell proliferation in 2D co-culture with MACS-sorted Ly6G+ lung neutrophils from control (UT) or irradiated mice, harvested 7 days after irradiation (n = 6 mice per group). Cells were treated with EdU (20 μM) and incorporation was assessed by FACS 6 h later. h, Quantification of EdU+ cells among GFP+ cancer cells. PyMT cancer cells co-cultured with neutrophils isolated from control (UT) or irradiated (IR) lungs (n = 6 mice per group) were compared to PyMT cells cultured alone (n = 2 replicates). All data represented as mean ± s.e.m. Statistical analysis by one-way ANOVA for (b) and (c), two-way ANOVA for (f) and an unpaired two-tailed t-test for (h). UT, untreated; IR, irradiated; NT, neutrophils. Gating strategies for FACS analysis provided in Extended Data Fig. 2.

Source data

Extended Data Fig. 5 Recombinant G-CSF treatment permits controlled neutrophil recruitment.

a, Experimental setup for rG-CSF time course. Mice were given a subcutaneous injection of rGCSF every second day for a total of 4 doses, beginning the day before irradiations. On day 0, 2, 4, 6, 7 and 8, a blood sample was taken from each mouse to quantify Ly6G+ neutrophils. Mice were harvested at day 8. b, FACS analysis of Ly6G+CD11b+ neutrophils among CD45+ cells in the blood (n = 6 mice per group). The day of rGCSF injections is depicted in blue and indicated by an arrow. The day of irradiation (day 0), the day of cancer cell injection (day 7) and the day of lung seeding (day 8) are indicated. Each dot represents an individual mouse, treated with either rGCSF (blue dots) or PBS (black dots). IV, intravenous; rG-CSF, recombinant G-CSF; ko, knock-out.

Source data

Extended Data Fig. 6 Primed neutrophils influence the lung epithelial cell response to radiation-induced injury.

a, Principle Component Analysis (PCA) of Lin-EpCAMmesenchymal cell signatures following RNA-seq analysis of control, irradiated and neutrophil-depleted irradiated lungs. Each dot represents an individual mouse, ovals enclose samples from each group to highlight their similarity in the PCA plot (n = 4 mice per group). b,c, 3D co-culture of GFP+MMTV-PyMT+ cancer cells on AlvetexTM Scaffolds with EpCAM+ lung epithelial cells isolated from control (UT), irradiated (IR) and neutrophil-depleted irradiated (IR + α-Ly6G) mice, 7 days after irradiation with (b) showing GFP signal quantification at day 4, normalised to cancer cell growth alone and (c) displaying representative images of GFP intensity at day 4 (n = 9 mice total per group, 3 independent experiments). Each dot in (b) represents the average of n = 3 mice for an independent experiment, with at least 3 technical replicates quantified per mouse in each experiment. Scale bar, 400 μm. d,e, 3D culture of GFP+EpCAM+ epithelial cells to assess survival on the scaffold, with (d) showing representative images of GFP intensity on the scaffold at day 4 (n = 3 mice per group, at least 3 technical replicates per mouse) and (e) showing GFP signal quantification (n = 3 mice per group, at least 3 technical replicates per mouse). Each dot represents an individual mouse. GFP+EpCAM+ cells were sorted from the lungs of control, irradiated and neutrophil-depleted irradiated actin-GFP mice 7 days after irradiation, and seeded in AlvetexTM Scaffolds. f, Heatmap of Lin-EpCAMmesenchymal cells from control, irradiated and neutrophil-depleted irradiated lungs (hierarchically clustered samples in columns and genes in rows) (n = 4 mice per group). All data represented as mean ± s.e.m. Statistical analysis by one-way ANOVA for (b). UT, untreated; IR, irradiated; Ep, epithelial.

Source data

Extended Data Fig. 7 Radiation-induced Notch signalling in the lung environment is boosted by the presence of neutrophils.

a, Representative images and quantification of immunofluorescent staining for the Notch Intracellular Domain (NCID) in irradiated EpCAM+ lung epithelial cells that were MACS-sorted and seeded on coverslips. Cells were harvested at day 7 from irradiated mice that were treated daily with an anti-Ly6G neutrophil depletion antibody or an IgG control antibody. Cells were quantified using CellProfiler, each dot represents the intensity of nuclear NCID staining in an individual cell (n = 130 IgG cells, n = 205 α-Ly6G cells, see methods). n = 3 mice per group, 3 technical replicates quantified per mouse. Scale bar, 5 μm. b, Quantitative RT-PCR validation of differentially-expressed genes identified by RNA-sequencing in sorted Lin-EpCAM+ lung epithelial cells. Lungs were harvested from an independent cohort of control, irradiated and neutrophil-depleted irradiated mice (n = 3 mice per group). c, Quantitative RT-PCR expression of Notch genes in sorted CD31+ lung endothelial cells from control, irradiated and neutrophil-depleted irradiated mice (n = 4 mice per group). Gapdh was used as a housekeeper gene for normalisation in (b) and (c). All data represented as mean ± s.e.m. Statistical analysis by unpaired two-tailed t-test for (a) and one-way ANOVA for (b) and (c). UT, untreated; IR, irradiated; Ep, epithelial.

Source data

Extended Data Fig.8 Radiation-exposure boosts Notch-signalling and stemness in metastatic cells.

a, Quantitative RT-PCR analysis of Notch1 and target gene Hes1 in lineage-traced lung alveolar type 2 cells from SPC-Cre-ERT2 control mice and SPC-Cre-ERT2/Rosa26NICD-IRES-GFP Notch-activated mice. Mice were administered tamoxifen by oral gavage (40 mg/kg) over three consecutive days. Lungs were harvested 14 days after the last tamoxifen dose and GFP+ cells were sorted by flow cytometry (n = 3 control mice, n = 4 Notch mice). b, Primary tumour weight from PyMT/Control (n = 6) and PyMT/Notch mice (n = 8) harvested two weeks post-tamoxifen induction. c, Quantification and representative images of Hes1 immunostaining in lung metastases from PyMT/Control (n = 5) and PyMT/Notch mice (n = 7) mice (metastatic lungs harvested over n = 5 independent tamoxifen administrations, immunostaining quantification in methods). The enlarged inset shows nuclear localisation. Scale bar, 100 μm (main image), 10 μm (enlarged inset). d, Combined Uniform Manifold Approximation and Projection (UMAP) plot of cells from the mCherry+ niche and mCherry- distant lung from irradiated mice (n = 10 mice, pooled). The expression level of EpCAM (distinguishing epithelial cells), Pdgfrα and Pdgfrβ (fibroblasts) and Pecam1(CD31) (endothelial cells) is indicated in blue. e, Representative immunostaining for RBPJ (top panel) and Hes1 (lower panel) in metastatic lungs from control, irradiated, and neutrophil-depleted irradiated mice harvested at day 14 (n = 7 mice per group, 2 independent experiments). Quantification shown in Fig. 6d,e. The enlarged inset shows nuclear localisation within tumour cells. Hes1+ cells are indicated by arrows. Scale bar, 100 μm (main image), 10 μm (enlarged inset). f, Representative immunostaining and quantification of RBPJ staining intensity in metastatic lungs from irradiated FVB mice pre-treated with rGCSF or PBS prior to injection of MMTV-PyMT cancer cells (Fig. 3e, n = 5 mice per group). Staining intensity within the metastatic area was measured using ImageJ (see methods). Scale bar, 100 μm. All data represented as mean ± s.e.m. Statistical analysis by non-parametric two-tailed Mann-Whitney test for (a), a two-tailed unpaired t-test for (b), (c) and (f). UT, untreated; IR, irradiated. The violin plot displays the median, 25th and 75th percentiles as well as the density of data points. Dots represent individual mice. Gating strategies for FACS sorting for (d) provided in Extended Data Fig. 2.

Source data

Supplementary information

Supplementary Information

Supplementary File 1 (Metastasis quantification strategy) and Tables 1 and 2.

Reporting Summary

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nolan, E., Bridgeman, V.L., Ombrato, L. et al. Radiation exposure elicits a neutrophil-driven response in healthy lung tissue that enhances metastatic colonization. Nat Cancer 3, 173–187 (2022). https://doi.org/10.1038/s43018-022-00336-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-022-00336-7

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