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:

Reciprocal interactions between innate immune cells and astrocytes facilitate neuroinflammation and brain metastasis via lipocalin-2

Abstract

Brain metastasis still encompass very grim prognosis and therefore understanding the underlying mechanisms is an urgent need toward developing better therapeutic strategies. We uncover the intricate interactions between recruited innate immune cells and resident astrocytes in the brain metastatic niche that facilitate metastasis of melanoma and breast cancer. We show that granulocyte-derived lipocalin-2 (LCN2) induces inflammatory activation of astrocytes, leading to myeloid cell recruitment to the brain. LCN2 is central to inducing neuroinflammation as its genetic targeting or bone-marrow transplantation from LCN2−/− mice was sufficient to attenuate neuroinflammation and inhibit brain metastasis. Moreover, high LCN2 levels in patient blood and brain metastases in multiple cancer types were strongly associated with disease progression and poor survival. Our findings uncover a previously unknown mechanism, establishing a central role for the reciprocal interactions between granulocytes and astrocytes in promoting brain metastasis and implicate LCN2 as a prognostic marker and potential therapeutic target.

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: Systemic LCN2 signaling is upregulated in plasma and CSF in brain metastases and correlates with metastatic burden.
Fig. 2: LCN2 is systemically secreted by the microenvironment of primary tumors, upregulated in brain-tropic tumors and corelates with patient survival.
Fig. 3: LCN2 signaling is functionally important for brain metastases formation and originates mainly from granulocytes and endothelial cells.
Fig. 4: Inflammatory activation of astrocytes is partially mediated by specific LCN2 receptor signaling.
Fig. 5: LCN2 signaling facilitates recruitment of immune suppressive myeloid cells into brain metastases.
Fig. 6: LCN2 signaling from bone-marrow-derived cells plays a key functional role in facilitating brain metastasis.
Fig. 7: Microenvironment-derived LCN2 signaling is operative in human patients and correlates with decreased survival in patients with brain metastases from melanoma, breast and lung primary origin.
Fig. 8: Graphical summary.

Similar content being viewed by others

Data availability

Source data for Figs. 17 and Extended Data Figs. 17 have been provided as Source Data files.

All other data supporting the findings of this study are available from the corresponding author upon reasonable request. Source data are provided with this paper.

References

  1. Eichler, A. F. et al. The biology of brain metastases-translation to new therapies. Nat. Rev. Clin. Oncol. 8, 344–356 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Cagney, D. N. et al. Incidence and prognosis of patients with brain metastases at diagnosis of systemic malignancy: a population-based study. Neuro Oncol. 19, 1511–1521 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  3. Takei, H., Rouah, E. & Ishida, Y. Brain metastasis: clinical characteristics, pathological findings and molecular subtyping for therapeutic implications. Brain Tumor Pathol. 33, 1–12 (2016).

    Article  CAS  PubMed  Google Scholar 

  4. Doron, H., Pukrop, T. & Erez, N. A blazing landscape: neuroinflammation shapes brain metastasis. Cancer Res. 79, 423–436 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Quail, D. F. & Joyce, J. A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 19, 1423–1437 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Peinado, H. et al. Pre-metastatic niches: organ-specific homes for metastases. Nat. Rev. Cancer 17, 302–317 (2017).

    Article  CAS  PubMed  Google Scholar 

  7. Wu, A. M. L. et al. Aging and CNS myeloid cell depletion attenuate breast cancer brain metastasis. Clin. Cancer Res. https://doi.org/10.1158/1078-0432.CCR-21-1549 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Quail, D. F. & Joyce, J. A. The microenvironmental landscape of brain tumors. Cancer Cell 31, 326–341 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Schulz, M., Salamero-Boix, A., Niesel, K., Alekseeva, T. & Sevenich, L. Microenvironmental regulation of tumor progression and therapeutic response in brain metastasis. Front. Immunol. 10, 1713 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Klemm, F. et al. Interrogation of the microenvironmental landscape in brain tumors reveals disease-specific alterations of immune cells. Cell. 181, 1643–1660 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Valiente, M. et al. The evolving landscape of brain metastasis. Trends Cancer. 4, 176–196 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Chen, Q. et al. Carcinoma–astrocyte gap junctions promote brain metastasis by cGAMP transfer. Nature 533, 493–498 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Doron, H. et al. Inflammatory activation of astrocytes facilitates melanoma brain tropism via the CXCL10-CXCR3 signaling axis. Cell Rep. 28, 1785–1798 (2019).

    Article  CAS  PubMed  Google Scholar 

  14. Priego, N. et al. STAT3 labels a subpopulation of reactive astrocytes required for brain metastasis. Nat. Med. 24, 1024–1035 (2018).

    Article  CAS  PubMed  Google Scholar 

  15. Wasilewski, D., Priego, N., Fustero-Torre, C. & Valiente, M. Reactive astrocytes in brain metastasis. Front. Oncol. 7, 298 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Zou, Y. et al. Polyunsaturated fatty acids from astrocytes activate pparγ signaling in cancer cells to promote brain metastasis. Cancer Discov. 9, 1720–1735 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Hirata, E. et al. The brain microenvironment induces DNMT1 suppression and indolence of metastatic cancer cells. iScience. 23, 101480 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Zhang, L. et al. Microenvironment-induced PTEN loss by exosomal microRNA primes brain metastasis outgrowth. Nature 527, 100–104 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Gril, B. et al. Reactive astrocytic S1P3 signaling modulates the blood–tumor barrier in brain metastases. Nat. Commun. 9, 2705 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Schwartz, H. et al. Incipient melanoma brain metastases instigate astrogliosis and neuroinflammation. Cancer Res. https://doi.org/10.1158/0008-5472.CAN-16-0485 (2016).

    Article  PubMed  Google Scholar 

  21. Flo, T. H. et al. Lipocalin 2 mediates an innate immune response to bacterial infection by sequestrating iron. Nature 432, 917–921 (2004).

    Article  CAS  PubMed  Google Scholar 

  22. Rathore, K. I. et al. Lipocalin 2 plays an immunomodulatory role and has detrimental effects after spinal cord injury. J. Neurosci. 31, 13412–13419 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Jang, E. et al. Phenotypic polarization of activated astrocytes: the critical role of lipocalin-2 in the classical inflammatory activation of astrocytes. J Immunol. 191, 5204–5219 (2013).

    Article  CAS  PubMed  Google Scholar 

  24. Jang, E. et al. Secreted protein lipocalin-2 promotes microglial M1 polarization. FASEB J. 27, 1176–1190 (2013).

    Article  CAS  PubMed  Google Scholar 

  25. Suk, K. Lipocalin-2 as a therapeutic target for brain injury: an astrocentric perspective. Prog. Neurobiol. 144, 158–172 (2016).

    Article  CAS  PubMed  Google Scholar 

  26. Stelzer, G. et al. VarElect: the phenotype-based variation prioritizer of the GeneCards Suite. BMC Genomics 17(Suppl 2), 444 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Lee, S. et al. Lipocalin-2 Is a chemokine inducer in the central nervous system: role of chemokine ligand 10 (CXCL10) in lipocalin-2-induced cell migration. J. Biol. Chem. 286, 43855–43870 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Yan, L., Borregaard, N., Kjeldsen, L. & Moses, M. A. The high molecular weight urinary matrix metalloproteinase (MMP) activity is a complex of gelatinase B/MMP-9 and neutrophil gelatinase-associated lipocalin (NGAL): modulation of MMP-9 activity by NGAL*. J. Biol. Chem. 276, 37258–37265 (2001).

    Article  CAS  PubMed  Google Scholar 

  29. Fernández, C. A. et al. The matrix metalloproteinase-9/neutrophil gelatinase-associated lipocalin complex plays a role in breast tumor growth and is present in the urine of breast cancer patients. Clin. Cancer Res. 11, 5390–5395 (2005).

    Article  PubMed  Google Scholar 

  30. Kubben, F. J. G. M. et al. Clinical evidence for a protective role of lipocalin-2 against MMP-9 autodegradation and the impact for gastric cancer. Eur. J. Cancer 43, 1869–1876 (2007).

    Article  CAS  PubMed  Google Scholar 

  31. Nuntagowat, C., Leelawat, K. & Tohtong, R. NGAL knockdown by siRNA in human cholangiocarcinoma cells suppressed invasion by reducing NGAL/MMP-9 complex formation. Clin. Exp. Metastasis 27, 295–305 (2010).

    Article  CAS  PubMed  Google Scholar 

  32. Devireddy, L. R., Gazin, C., Zhu, X. & Green, M. R. A cell-surface receptor for lipocalin 24p3 selectively mediates apoptosis and iron uptake. Cell 123, 1293–1305 (2005).

    Article  CAS  PubMed  Google Scholar 

  33. Cabedo Martinez, A. I. et al. Biochemical and structural characterization of the interaction between the siderocalin NGAL/LCN2 (neutrophil gelatinase-associated lipocalin/lipocalin 2) and the N-terminal domain of its endocytic receptor SLC22A17. J. Biol. Chem. 291, 2917–2930 (2016).

    Article  PubMed  Google Scholar 

  34. Liddelow, S. A. & Barres, B. A. Reactive astrocytes: production, function, and therapeutic potential. Immunity. 46, 957–967 (2017).

    Article  CAS  PubMed  Google Scholar 

  35. Proescholdt, M. A. et al. The management of brain metastases-systematic review of neurosurgical aspects. Cancers https://doi.org/10.3390/cancers13071616 (2021).

  36. Schag, C. C., Heinrich, R. L. & Ganz, P. A. Karnofsky performance status revisited: reliability, validity, and guidelines. J. Clin. Oncol. 2, 187–193 (1984).

    Article  CAS  PubMed  Google Scholar 

  37. Blank, A. et al. Microglia/macrophages express alternative proangiogenic factors depending on granulocyte content in human glioblastoma. J. Pathol. 253, 160–173 (2021).

    Article  CAS  PubMed  Google Scholar 

  38. Khan, S. et al. Role of neutrophils and myeloid-derived suppressor cells in glioma progression and treatment resistance. Int. J. Mol. Sci. https://doi.org/10.3390/ijms21061954 (2020).

  39. Santiago-Sanchez, G. S. et al. Biological functions and therapeutic potential of lipocalin 2 in cancer. Int. J. Mol. Sci. https://doi.org/10.3390/ijms21124365 (2020).

  40. Tong, Z. et al. Neutrophil gelatinase-associated lipocalin: a novel suppressor of invasion and angiogenesis in pancreatic cancer. Cancer Res. 68, 6100–6108 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Yang, J. et al. Lipocalin 2 promotes breast cancer progression. Proc. Natl Acad. Sci. USA. 106, 3913–3918 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Bauvois, B. & Susin, S. A. Revisiting neutrophil gelatinase-associated lipocalin (NGAL) in cancer: saint or sinner? Cancers https://doi.org/10.3390/cancers10090336 (2018).

  43. Yang, J., McNeish, B., Butterfield, C. & Moses, M. A. Lipocalin 2 is a novel regulator of angiogenesis in human breast cancer. FASEB J. 27, 45–50 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Tyagi, A. et al. Nicotine promotes breast cancer metastasis by stimulating N2 neutrophils and generating pre-metastatic niche in lung. Nat. Commun. 12, 474 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Rodvold, J. J., Mahadevan, N. R. & Zanetti, M. Lipocalin 2 in cancer: when good immunity goes bad. Cancer Lett. 316, 132–138 (2012).

    Article  CAS  PubMed  Google Scholar 

  46. Olson, B. et al. Lipocalin 2 mediates appetite suppression during pancreatic cancer cachexia. Nat. Commun. 12, 2057 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Jha, M. K. et al. Diverse functional roles of lipocalin-2 in the central nervous system. Neurosci. Biobehav. Rev. 49, 135–156 (2015).

    Article  CAS  PubMed  Google Scholar 

  48. Llorens, F. et al. Cerebrospinal fluid lipocalin 2 as a novel biomarker for the differential diagnosis of vascular dementia. Nat. Commun. 11, 619 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Chi, Y. et al. Cancer cells deploy lipocalin-2 to collect limiting iron in leptomeningeal metastasis. Science 369, 276–282 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Erez, N., Truitt, M., Olson, P., Arron, S. T. & Hanahan, D. Cancer-associated fibroblasts are activated in incipient neoplasia to orchestrate tumor-promoting inflammation in an NF-κB-dependent manner. Cancer Cell 17, 135–147 (2010).

    Article  CAS  PubMed  Google Scholar 

  51. Shani, O. et al. Fibroblast-derived IL33 facilitates breast cancer metastasis by modifying the immune microenvironment and driving type 2 immunity. Cancer Res. 80, 5317–5329 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Birnboim-Perach, R., Grinberg, Y., Vaks, L., Nahary, L. & Benhar, I. Production of stabilized antibody fragments in the E. coli bacterial cytoplasm and in transiently transfected mammalian cells. Methods Mol. Biol. 1904, 455–480 (2019).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank Y. Zilberstein from the Faculty Cellular and Molecular Imaging Center for her help with intracardiac injections and mouse imaging. We thank the Rabin Medical Center Biobank and A. Levy-Barda for their support of this research. We also thank E. Vollmer, E. Ostermeier and K. Kronenberg from Regensburg University Hospital and L. Siam, M. Schaffrinski and D. Egert from Göttingen University Hospital, Germany, for their contribution in collecting and processing human samples. The study was supported by grants to N.E. from the Melanoma Research Alliance (award ID 826222), a Breakthrough Award from the US Department of Defense (BCRP award ID W81XWH2110394), Israel Science Foundation Personalized Medicine Program (IPMP no. 3495/19) and a research grant from the Tel Aviv University Cancer Biology Research Center. N.E. and T.P. were supported by a grant from the German Research Foundation (PU 355/4-1). T.P. was supported by SFB/TRR 305/1 (B03).

Author information

Authors and Affiliations

Authors

Contributions

O.A., Y.Z., H.D. and N.E. conceived of and designed this study. O.A., Y.Z., N.C., T.P. and N.E. developed the methods. O.A., Y.Z., R.B., L.M. and N.C. performed the experiments. G.G., Y.S., T.S., D.M. and L.M. performed formal analysis. A.A.K., S.H., V.Y., S.H., J.A.H., A.B., S.Y.K., L.N, I.B. and T.P. provided resources. O.A., Y.Z., N.C. and N.E. wrote the original draft. O.A., Y.Z., N.C. and N.E. revised and edited the manuscript. N.C. and N.E. administered this project. N.E. supervised the study. All authors discussed the results and provided feedback on the manuscript.

Corresponding author

Correspondence to Neta Erez.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cancer thanks Frank Winkler 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 Proteomic analysis of secreted proteins in blood of mice with melanoma or breast cancer brain metastasis.

Heatmap showing fold change values of secreted proteins measured by Proteome profiler Mouse XL Cytokine Array. Mean grey value was quantified by ImageJ. Heatmap represents Log2 (BrM/Normal) values.

Extended Data Fig. 2 LCN2 expression is induced by tumor cell-secreted factors, and is not necessary for primary tumor growth.

a. Gating strategy for FACS isolation of different cell populations from primary tumors 2.5 weeks following BT-RMS or BT-EO771 orthotopic injection. b–d. qPCR analysis of Lcn2 from BMDM, dermal fibroblasts and C166 endothelial cells treated in vitro with BT-RMS CM (BT_RMS CM) versus serum free medium (SFM). 3 biological repeats, error bars represent mean ± SD (Student’s t-test, two-sided). e. qPCR analysis of Lcn2 in primary dermal fibroblasts treated with secreted factors from RMS and sBT-RMS, error bars represent mean ± SD (SFM n = 3, RMS CM n = 3, SBT CM n = 3 biological replicates) (one-way ANOVA). f. Primary tumor growth curve of WT and Lcn2−/− mice orthotopically injected with BT-RMS melanoma cells, measured manually by caliper, (WT n = 10, Lcn2−/− n = 8 mice) (Repeated measure ANOVA). g. Primary tumor weight at time of resection of WT and Lcn2−/− mice orthotopically injected with BT-EO771 cells, (WT n = 10, Lcn2−/− n = 10 mice), dots represent individual mice, error bars represent s.e.m. (Student’s t-test, two-sided).

Extended Data Fig. 3 Brain metastatic burden and survival in mice injected with breast cancer cells.

a. Experimental scheme analyzed in (b,c). b. Survival curve analysis of WT and Lcn2-/- mice injected intracardially with BT-EO771 cells, (WT n = 10, Lcn2-/- n = 9 mice) (Kaplan–Meier curve, log-rank test). c. Quantification of brain metastatic burden for mice in (a), quantified as % CD45- mCherry+ tumor cells/live cells, (ctrl n = 9, WT n = 9, Lcn2-/- n = 7 mice) (one-way ANOVA). d. Gating strategy for isolation by FACS of different cells populations from WT and Lcn2-/- BrM mice injected intracardially with BT-RMS or BT-EO771 cells 18 days after injection.

Extended Data Fig. 4 Astrocytes activate pro-inflammatory signaling in an LCN2-dependent manner, and SLC22A17 is required for astrocyte response to LCN2.

a. Expression of SLC22A17 in bulk RNA-seq of different cell populations isolated from samples of human gliomas (CD45- n = 23, MG n = 21, MDM n = 17, Neutrophils n = 16, T cells n = 22 patients) (one-way ANOVA), (Brain TIME dataset). b. LCN2 is sufficient to induce pro-inflammatory signaling in primary astrocytes. qPCR analysis of inflammatory gene signature in primary astrocytes incubated with 10ug/ml rLCN2. Error bars represent SD, dots represent two biological repeats with technical replicates (two-way ANOVA). c. qPCR analysis of inflammatory gene signature in primary astrocytes transfected with siRNA targeting Slc22a17 or with control siRNA (siSlc22a17 or siScramble). Astrocytes were then treated with 10ug/ml rLCN2 or SFM for 24 h. Error bars represent SD, 4 biological repeats in duplicates. d. LCN2 measured in CM of BT-RMS cells or granulocytes activated by BT-RMS CM, error bars represent SD, dots represent 3 biological repeats (two-way ANOVA). e, f. Activation of astrocytes was quantified by number of GFAP + cells/ field. Representative images are shown from n = 6,8,8 mice per group, error bars represent mean ± SD. 6 fields X 1 sections per mouse were analyzed (one-way ANOVA). g, h. Expression level of inflammatory gene signature measured by qPCR in RNA of FACS sorted astrocytes in vivo from WT or Lcn2-/- mice with BrM following BT-RMS or BT-EO771 injection. Dots represent individual mice, line indicates median, plot shows mean to max (melanoma: Ctrl n = 4, WT n = 7,10, Lcn2-/- n = 5,9 mice), (breast: Ctrl n = 6, WT n = 5, Lcn2-/- n = 5,7 mice) (two-way ANOVA). i. Expression level of inflammatory gene signature measured by qPCR of FACS sorted astrocytes in vivo from WT or Lcn2-/- normal mice, (WT n = 3, Lcn2-/- n = 2 mice). j, k. Expression level of immunosuppressive gene signature measured by qPCR of sorted MG/MDM in vivo from WT or Lcn2-/- mice, 3 weeks following BT-RMS or BT-EO771 intracardiac injection, line indicates median, plot shows mean to max (Melanoma WT n = 7, Lcn2-/- n = 5, Breast WT n = 5, Lcn2-/- n = 6 mice) (two-way ANOVA).

Extended Data Fig. 5 LCN2 induces recruitment of immune suppressive granulocytes to brain in the in the context of BrM.

a. Quantification of migrated Ly6G+ granulocytes or Ly6C+ Ly6G monocytes toward normal astrocytes or astrocytes treated 24 h with 10ug/ml rLCN2. Dots represent individual wells, 4 biological repeats in duplicates, error bars represent mean ± SD (Student’s t-test, two-sided). b. Immune profiling of bone marrow cell populations by flow cytometry of 8-week-old C57BL/6 WT or Lcn2−/− mice, (WT n = 5, Lcn2−/− n = 5 mice). c. Immune profiling of CD11b+ myeloid cells by flow cytometry of normal WT or Lcn2−/− mice (male WT n = 5, Lcn2−/− n = 3, female WT n = 6, Lcn2−/− n = 3 mice). d,e. Expression level of immunosuppressive gene signature measured by qPCR of FACS sorted granulocytes in vivo from WT or Lcn2−/− mice, 3 weeks following BT-RMS intracardiac injection, line indicates median, plot shows mean to max (Melanoma WT n = 11, Lcn2−/− n = 9, Breast WT n = 5, Lcn2−/− n = 6 mice) (two-way ANOVA).

Extended Data Fig. 6 Bone marrow-derived granulocyte induce increasing systemic LCN2 levels in plasma and inflammatory activation of astrocytes.

a. qPCR analysis of Lcn2 expression in different cell populations sorted from bone marrow of normal C57BL/6 males: CD45-, CD45+CD11b+Ly6CinterLy6G+ granulocytes, CD45+CD11b+Ly6C+Ly6G- monocytes, CD45+CD11b-CD3+ B220- T cells, CD45+CD11b-CD3-B220+ B cells. Dots represent individual mice (n = 4 mice per group), line indicates median, whisker shows mean to max (one-way ANOVA). b. ‘LCN2 BM contribution’ calculated by expression of LCN2 in different cell populations from a, multiplied by their abundance in BM from Extended Data Fig. 5b. Results are presented as percent of total. c. LCN2 ELISA in blood, one and two weeks following BMT. Dots represent individual mice, error bars represent s.e.m., (WT n = 9, Lcn2-/- n = 9 mice) (one-way ANOVA). d. LCN2 levels in blood of mice at endpoint measured by ELISA, (WT n = 7, Lcn2-/- n = 7 mice), dots represent individual mice, error bars represent s.e.m. (Student’s t-test, two-sided). e. Expression level of inflammatory gene signature measured by qPCR in RNA of FACS sorted astrocytes in vivo from WT mice that underwent BMT as described in (Fig. 4a). Dots represent individual mice, line indicates median, plot shows mean to max (Ctrl n = 4, WT n = 6, Lcn2-/- n = 6 mice) (two-way ANOVA).

Extended Data Fig. 7 LCN2 is highly expressed in granulocytes in human BrM and can help direct patient’s care in combination with clinically used prognostic factors.

a. Immunofluorescence staining in frozen sections of resected human brain metastases from melanoma, breast and lung primary origin. Co-localization of LCN2 with CD66b (granulocytes). Representative images of separate staining are shown from n = 2 patients samples stained per cancer type. b. Five-year survival curve analysis of patients with low versus high LCN2 levels in patients with lung cancer BrM. The cutoff between high and low levels was defined as the median LCN2 level (Kaplan–Meier curve, log-rank test). c. Five-year survival curve analysis of patients with KPS score over or under 70, in patients with lung cancer BrM. d. 5-year survival curve analysis of patients with KPS score < 70, stratified to low versus high LCN2 levels in patients from Fig. 7h.

Extended Data Table. 1 Data of patients with brain metastasis from melanoma, breast and lung cancer
Extended Data Table. 2 Data of patients with untreated stage IV melanoma
Extended Data Table. 3 Data of patients with brain metastasis from melanoma and lung cancer

Supplementary information

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. 1

Unprocessed cytokine array membrane.

Source Data Extended Data Fig. 1

Statistical Source Data.

Source Data Extended Data Fig. 2

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.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Adler, O., Zait, Y., Cohen, N. et al. Reciprocal interactions between innate immune cells and astrocytes facilitate neuroinflammation and brain metastasis via lipocalin-2. Nat Cancer 4, 401–418 (2023). https://doi.org/10.1038/s43018-023-00519-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-023-00519-w

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