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:

Periostin secreted by glioblastoma stem cells recruits M2 tumour-associated macrophages and promotes malignant growth

Abstract

Tumour-associated macrophages (TAMs) are enriched in glioblastoma multiformes (GBMs) that contain glioma stem cells (GSCs) at the apex of their cellular hierarchy. The correlation between TAM density and glioma grade suggests a supportive role for TAMs in tumour progression. Here we interrogated the molecular link between GSCs and TAM recruitment in GBMs and demonstrated that GSCs secrete periostin (POSTN) to recruit TAMs. TAM density correlates with POSTN levels in human GBMs. Silencing POSTN in GSCs markedly reduced TAM density, inhibited tumour growth, and increased survival of mice bearing GSC-derived xenografts. We found that TAMs in GBMs are not brain-resident microglia, but mainly monocyte-derived macrophages from peripheral blood. Disrupting POSTN specifically attenuated the tumour-supportive M2 type of TAMs in xenografts. POSTN recruits TAMs through the integrin αvβ3 as blocking this signalling by an RGD peptide inhibited TAM recruitment. Our findings highlight the possibility of improving GBM treatment by targeting POSTN-mediated TAM recruitment.

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: POSTN is preferentially secreted by GSCs and its levels correlate with TAM density in GBMs.
Figure 2: Disrupting POSTN by shRNA markedly inhibited tumour growth and reduced TAM density in GSC-derived xenografts.
Figure 3: TAMs in human primary GBMs and xenografts are monocyte-derived macrophages from peripheral blood.
Figure 4: POSTN-recruited TAMs are maintained as the M2 subtype in GSC-derived xenografts.
Figure 5: Silencing POSTN in GSCs increased the relative fraction of M1 TAMs in GSC-derived xenografts.
Figure 6: GSC-secreted POSTN is a chemoattractant to macrophages and monocytes.
Figure 7: POSTN recruits macrophages through αvβ3 integrin signalling.
Figure 8: M2 TAMs accelerated GSC tumour growth in mouse brains.

Similar content being viewed by others

References

  1. Deorah, S., Lynch, C. F., Sibenaller, Z. A. & Ryken, T. C. Trends in brain cancer incidence and survival in the United States: Surveillance, Epidemiology, and End Results Program, 1973 to 2001. Neurosurg. Focus 20, E1 (2006).

    Article  PubMed  Google Scholar 

  2. DeAngelis, L. M. Brain tumors. N. Engl. J. Med. 344, 114–123 (2001).

    Article  CAS  PubMed  Google Scholar 

  3. Hao, C. et al. Cytokine and cytokine receptor mRNA expression in human glioblastomas: evidence of Th1, Th2 and Th3 cytokine dysregulation. Acta Neuropathol. 103, 171–178 (2002).

    Article  CAS  PubMed  Google Scholar 

  4. Abou-Ghazal, M. et al. The incidence, correlation with tumor-infiltrating inflammation, and prognosis of phosphorylated STAT3 expression in human gliomas. Clin. Cancer Res. 14, 8228–8235 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Morford, L. A., Dix, A. R., Brooks, W. H. & Roszman, T. L. Apoptotic elimination of peripheral T lymphocytes in patients with primary intracranial tumors. J. Neurosurg. 91, 935–946 (1999).

    Article  CAS  PubMed  Google Scholar 

  6. Hirano, H., Tanioka, K., Yokoyama, S., Akiyama, S. & Kuratsu, J. Angiogenic effect of thymidine phosphorylase on macrophages in glioblastoma multiforme. J. Neurosurg. 95, 89–95 (2001).

    Article  CAS  PubMed  Google Scholar 

  7. Kanamori, M., Kawaguchi, T., Berger, M. S. & Pieper, R. O. Intracranial microenvironment reveals independent opposing functions of host αVβ3 expression on glioma growth and angiogenesis. J. Biol. Chem. 281, 37256–37264 (2006).

    Article  CAS  PubMed  Google Scholar 

  8. Samaras, V. et al. Application of the ELISPOT method for comparative analysis of interleukin (IL)-6 and IL-10 secretion in peripheral blood of patients with astroglial tumors. Mol. Cell. Biochem. 304, 343–351 (2007).

    Article  CAS  PubMed  Google Scholar 

  9. Mineharu, Y. et al. Engineering the brain tumor microenvironment enhances the efficacy of dendritic cell vaccination: implications for clinical trial design. Clin. Cancer Res. 17, 4705–4718 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Pellegatta, S. et al. Neurospheres enriched in cancer stem-like cells are highly effective in eliciting a dendritic cell-mediated immune response against malignant gliomas. Cancer Res. 66, 10247–10252 (2006).

    Article  CAS  PubMed  Google Scholar 

  11. Graf, M. R., Prins, R. M., Hawkins, W. T. & Merchant, R. E. Irradiated tumor cell vaccine for treatment of an established glioma. I. Successful treatment with combined radiotherapy and cellular vaccination. Cancer Immunol. Immunother. 51, 179–189 (2002).

    Article  PubMed  Google Scholar 

  12. Sielska, M. et al. Distinct roles of CSF family cytokines in macrophage infiltration and activation in glioma progression and injury response. J. Pathol. 230, 310–321 (2013).

    Article  CAS  PubMed  Google Scholar 

  13. Staudt, N. D. et al. Myeloid cell receptor LRP1/CD91 regulates monocyte recruitment and angiogenesis in tumors. Cancer Res. 73, 3902–3912 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Pucci, F. et al. A distinguishing gene signature shared by tumor-infiltrating Tie2-expressing monocytes, blood “resident” monocytes, and embryonic macrophages suggests common functions and developmental relationships. Blood 114, 901–914 (2009).

    Article  CAS  PubMed  Google Scholar 

  15. Komohara, Y., Ohnishi, K., Kuratsu, J. & Takeya, M. Possible involvement of the M2 anti-inflammatory macrophage phenotype in growth of human gliomas. J. Pathol. 216, 15–24 (2008).

    Article  CAS  PubMed  Google Scholar 

  16. Ling, E. A. & Wong, W. C. The origin and nature of ramified and amoeboid microglia: a historical review and current concepts. Glia 7, 9–18 (1993).

    Article  CAS  PubMed  Google Scholar 

  17. Deininger, M. H., Seid, K., Engel, S., Meyermann, R. & Schluesener, H. J. Allograft inflammatory factor-1 defines a distinct subset of infiltrating macrophages/microglial cells in rat and human gliomas. Acta Neuropathol. 100, 673–680 (2000).

    Article  CAS  PubMed  Google Scholar 

  18. Akiyama, H. & McGeer, P. L. Brain microglia constitutively express β-2 integrins. J. Neuroimmunol. 30, 81–93 (1990).

    Article  CAS  PubMed  Google Scholar 

  19. Guvenc, H. et al. Impairment of glioma stem cell survival and growth by a novel inhibitor for Survivin-Ran protein complex. Clin. Cancer Res. 19, 631–642 (2013).

    Article  CAS  PubMed  Google Scholar 

  20. Tran, C. T. et al. Differential expression of MHC class II molecules by microglia and neoplastic astroglia: relevance for the escape of astrocytoma cells from immune surveillance. Neuropathol. Appl. Neurobiol. 24, 293–301 (1998).

    Article  CAS  PubMed  Google Scholar 

  21. Ludwig, H. C. et al. Expression of nitric oxide synthase isozymes (NOS I-III) by immunohistochemistry and DNA in situ hybridization. Correlation with macrophage presence, vascular endothelial growth factor (VEGF) and oedema volumetric data in 220 glioblastomas. Anticancer Res. 20, 299–304 (2000).

    CAS  PubMed  Google Scholar 

  22. Mizutani, M. et al. The fractalkine receptor but not CCR2 is present on microglia from embryonic development throughout adulthood. J. Immunol. 188, 29–36 (2012).

    Article  CAS  PubMed  Google Scholar 

  23. Ginestier, C. et al. ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell 1, 555–567 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Singh, S. K. et al. Identification of human brain tumour initiating cells. Nature 432, 396–401 (2004).

    Article  CAS  PubMed  Google Scholar 

  25. Barker, N. et al. Crypt stem cells as the cells-of-origin of intestinal cancer. Nature 457, 608–611 (2009).

    Article  CAS  PubMed  Google Scholar 

  26. Bao, S. et al. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature 444, 756–760 (2006).

    Article  CAS  PubMed  Google Scholar 

  27. Cheng, L. et al. Glioblastoma stem cells generate vascular pericytes to support vessel function and tumor growth. Cell 153, 139–152 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Calabrese, C. et al. A perivascular niche for brain tumor stem cells. Cancer Cell 11, 69–82 (2007).

    Article  CAS  PubMed  Google Scholar 

  29. Du, R. et al. HIF1α induces the recruitment of bone marrow-derived vascular modulatory cells to regulate tumor angiogenesis and invasion. Cancer Cell 13, 206–220 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Li, Z. et al. Hypoxia-inducible factors regulate tumorigenic capacity of glioma stem cells. Cancer Cell 15, 501–513 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Lathia, J. D., Heddleston, J. M., Venere, M. & Rich, J. N. Deadly teamwork: neural cancer stem cells and the tumor microenvironment. Cell Stem Cell 8, 482–485 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Yang, I., Han, S. J., Sughrue, M. E., Tihan, T. & Parsa, A. T. Immune cell infiltrate differences in pilocytic astrocytoma and glioblastoma: evidence of distinct immunological microenvironments that reflect tumor biology. J. Neurosurg. 115, 505–511 (2011).

    Article  CAS  PubMed  Google Scholar 

  33. Xing, F. et al. Hypoxia-induced Jagged2 promotes breast cancer metastasis and self-renewal of cancer stem-like cells. Oncogene 30, 4075–4086 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Auffinger, B. et al. Conversion of differentiated cancer cells into cancer stem-like cells in a glioblastoma model after primary chemotherapy. Cell Death Differ. 21, 1119–1131 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Deininger, M. H., Pater, S., Strik, H. & Meyermann, R. Macrophage/microglial cell subpopulations in glioblastoma multiforme relapses are differentially altered by radiochemotherapy. J. Neurooncol. 55, 141–147 (2001).

    Article  CAS  PubMed  Google Scholar 

  36. Bao, S. et al. Periostin potently promotes metastatic growth of colon cancer by augmenting cell survival via the Akt/PKB pathway. Cancer Cell 5, 329–339 (2004).

    Article  CAS  PubMed  Google Scholar 

  37. Baril, P. et al. Periostin promotes invasiveness and resistance of pancreatic cancer cells to hypoxia-induced cell death: role of the β4 integrin and the PI3k pathway. Oncogene 26, 2082–2094 (2007).

    Article  CAS  PubMed  Google Scholar 

  38. Malanchi, I. et al. Interactions between cancer stem cells and their niche govern metastatic colonization. Nature 481, 85–89 (2012).

    Article  CAS  Google Scholar 

  39. Michaylira, C. Z. et al. Periostin, a cell adhesion molecule, facilitates invasion in the tumor microenvironment and annotates a novel tumor-invasive signature in esophageal cancer. Cancer Res. 70, 5281–5292 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Koronyo-Hamaoui, M. et al. Attenuation of AD-like neuropathology by harnessing peripheral immune cells: local elevation of IL-10 and MMP-9. J. Neurochem. 111, 1409–1424 (2009).

    Article  CAS  PubMed  Google Scholar 

  41. Jin, X., Ishii, H., Bai, Z., Itokazu, T. & Yamashita, T. Temporal changes in cell marker expression and cellular infiltration in a controlled cortical impact model in adult male C57BL/6 mice. PLoS ONE 7, e41892 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Saederup, N. et al. Selective chemokine receptor usage by central nervous system myeloid cells in CCR2-red fluorescent protein knock-in mice. PLoS ONE 5, e13693 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Movahedi, K. et al. Different tumor microenvironments contain functionally distinct subsets of macrophages derived from Ly6C(high) monocytes. Cancer Res. 70, 5728–5739 (2010).

    Article  CAS  PubMed  Google Scholar 

  44. Qian, B. Z. & Pollard, J. W. Macrophage diversity enhances tumor progression and metastasis. Cell 141, 39–51 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Sica, A. & Mantovani, A. Macrophage plasticity and polarization: in vivo veritas. J. Clin. Invest. 122, 787–795 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Yu, J. L. & Rak, J. W. Host microenvironment in breast cancer development: inflammatory and immune cells in tumour angiogenesis and arteriogenesis. Breast Cancer Res. 5, 83–88 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Condeelis, J. & Pollard, J. W. Macrophages: obligate partners for tumor cell migration, invasion, and metastasis. Cell 124, 263–266 (2006).

    Article  CAS  PubMed  Google Scholar 

  48. Sica, A., Schioppa, T., Mantovani, A. & Allavena, P. Tumour-associated macrophages are a distinct M2 polarised population promoting tumour progression: potential targets of anti-cancer therapy. Eur. J. Cancer 42, 717–727 (2006).

    Article  CAS  PubMed  Google Scholar 

  49. Sharda, D. R. et al. Regulation of macrophage arginase expression and tumor growth by the Ron receptor tyrosine kinase. J. Immunol. 187, 2181–2192 (2011).

    Article  CAS  PubMed  Google Scholar 

  50. Arranz, A. et al. Akt1 and Akt2 protein kinases differentially contribute to macrophage polarization. Proc. Natl Acad. Sci. USA 109, 9517–9522 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Rolny, C. et al. HRG inhibits tumor growth and metastasis by inducing macrophage polarization and vessel normalization through downregulation of PlGF. Cancer Cell 19, 31–44 (2011).

    Article  CAS  PubMed  Google Scholar 

  52. Wang, H. et al. Activated macrophages are essential in a murine model for T cell-mediated chronic psoriasiform skin inflammation. J. Clin. Invest. 116, 2105–2114 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Gillan, L. et al. Periostin secreted by epithelial ovarian carcinoma is a ligand for α(V)β(3) and α(V)β(5) integrins and promotes cell motility. Cancer Res. 62, 5358–5364 (2002).

    CAS  PubMed  Google Scholar 

  54. Butcher, J. T., Norris, R. A., Hoffman, S., Mjaatvedt, C. H. & Markwald, R. R. Periostin promotes atrioventricular mesenchyme matrix invasion and remodeling mediated by integrin signaling through Rho/PI 3-kinase. Dev. Biol. 302, 256–266 (2007).

    Article  CAS  PubMed  Google Scholar 

  55. Grkovich, A., Johnson, C. A., Buczynski, M. W. & Dennis, E. A. Lipopolysaccharide-induced cyclooxygenase-2 expression in human U937 macrophages is phosphatidic acid phosphohydrolase-1-dependent. J. Biol. Chem. 281, 32978–32987 (2006).

    Article  CAS  PubMed  Google Scholar 

  56. Shrivastav, A. et al. Requirement of N-myristoyltransferase 1 in the development of monocytic lineage. J. Immunol. 180, 1019–1028 (2008).

    Article  CAS  PubMed  Google Scholar 

  57. Stupp, R. et al. Phase I/IIa study of cilengitide and temozolomide with concomitant radiotherapy followed by cilengitide and temozolomide maintenance therapy in patients with newly diagnosed glioblastoma. J. Clin. Oncol. 28, 2712–2718 (2010).

    Article  CAS  PubMed  Google Scholar 

  58. Mantovani, A., Sozzani, S., Locati, M., Allavena, P. & Sica, A. Macrophage polarization: tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol. 23, 549–555 (2002).

    Article  CAS  PubMed  Google Scholar 

  59. Kim, D. W. et al. High tumour islet macrophage infiltration correlates with improved patient survival but not with EGFR mutations, gene copy number or protein expression in resected non-small cell lung cancer. Br. J. Cancer 98, 1118–1124 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Popivanova, B. K. et al. Blockade of a chemokine, CCL2, reduces chronic colitis-associated carcinogenesis in mice. Cancer Res. 69, 7884–7892 (2009).

    Article  CAS  PubMed  Google Scholar 

  61. Qian, B. Z. et al. CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis. Nature 475, 222–225 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Pyonteck, S. M. et al. Deficiency of the macrophage growth factor CSF-1 disrupts pancreatic neuroendocrine tumor development. Oncogene 31, 1459–1467 (2012).

    Article  CAS  PubMed  Google Scholar 

  63. Spear, P., Barber, A., Rynda-Apple, A. & Sentman, C. L. Chimeric antigen receptor T cells shape myeloid cell function within the tumor microenvironment through IFN-γ and GM-CSF. J. Immunol. 188, 6389–6398 (2012).

    Article  CAS  PubMed  Google Scholar 

  64. Germano, G. et al. Role of macrophage targeting in the antitumor activity of trabectedin. Cancer Cell 23, 249–262 (2013).

    Article  CAS  PubMed  Google Scholar 

  65. Guryanova, O. A. et al. Nonreceptor tyrosine kinase BMX maintains self-renewal and tumorigenic potential of glioblastoma stem cells by activating STAT3. Cancer Cell 19, 498–511 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Fang, X. et al. The zinc finger transcription factor ZFX is required for maintaining the tumorigenic potential of glioblastoma stem cells. Stem Cells 32, 2033–2047 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Huang, Z. et al. Deubiquitylase HAUSP stabilizes REST and promotes maintenance of neural progenitor cells. Nat. Cell Biol. 13, 142–152 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the Brain Tumor and Neuro-Oncology Centers at Cleveland Clinic and University Hospitals Case Medical Center for providing GBM surgical specimens for this study. We are grateful to members in J.N.R.’s laboratory for their assistance and scientific discussion. We also thank C. Shemo and S. O’Bryant of the Flow Cytometry Core, J. Drazba and L. Vargo of the Imaging Core at Cleveland Clinic Lerner Research Institute, and D. Schumick of the Center for Medical Art and Photography for their assistance. This work was supported by the Cleveland Clinic Foundation and an NIH R01 grant (NS070315) to S.B.

Author information

Authors and Affiliations

Authors

Contributions

W.Z. and S.B. designed the experiments, analysed data and prepared the manuscript. W.Z., S.Q.K., Z.H., X.F., J.P. and L.W. performed the experiments. W.F. performed database analysis. A.E.S. and R.E.M. provided GBM surgical specimens. R.E.M. performed pathological analyses. J.N.R. and X.L. provided scientific input and helped to edit the manuscript.

Corresponding author

Correspondence to Shideng Bao.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 2 Identification of POSTN as a dominant chemoattractant to macrophages, and the correlation between POSTN expression and GBM prognosis, GSC population, or TAM density.

(a,b) Graphical analyses of migration (a) and invasion (b) assays showing that POSTN is a dominant chemoattractant in recruiting PMA-primed U937 macrophage-like cells. Migration was allowed for 24 h and invasion was allowed for 36 h. , P < 0.001; , P < 0.05; ns, P > 0.05. (n = 5 fields; mean ± s.e.m.; two-tailed unpaired t-test; experiments were repeated 3 times). (c,d) Bioinformatic analyses of the relationship between POSTN expression and overall survival (c) or progression-free survival (d) of GBM patients in TCGA dataset (The Cancer Genome Atlas) from Oncomine (http://www.oncomine.com). The threshold for high/low POSTN expression is determined by the average of POSTN expression levels in 525 GBM patients. The Kaplan–Meier survival curves demonstrate a significant invert correlation between POSTN levels and overall survival (c) (n = 525 patients; POSTN low, n = 115 patients; POSTN high, n = 400 patients; two-tailed log-rank test) or progression-free survival (d) (n = 337 patients; POSTN low, n = 164 patients; POSTN high, n = 173 patients; two-tailed log-rank test) of GBM patients. (e) Immunofluorescent staining of POSTN (green) and the GSC marker SOX2 (red) in primary GBMs. POSTN was preferentially expressed in GSCs and distributed in areas near GSCs in GBMs. In CW690 GBM with less GSCs, POSTN showed little diffusion staining and strictly co-localized with SOX2+ cells. Scale bar, 40 μm. (f) Immunofluorescent analysis of POSTN (red) and the TAM marker Iba1 (green). Three sets of representative staining from 3 tumours (B7, C4 and D8) in a tissue microarray slide (GL803a, US Biomax) were presented to show enrichment of TAMs in POSTN abundant regions in GBMs. Areas indicated with squares were enlarged and shown on right side of each picture. Scale bar, 80 μm. (g) Graphical analysis of (f) showing the enrichment of TAMs in POSTN-high areas. Average levels of POSTN and Iba1 signals were determined by ImageJ. Iba1+ TAM densities from five random POSTN-high and POSTN-low areas in each GBM were normalized to total TAMs. , P < 0.01; , P < 0.05. (n = 5 fields; mean ± s.e.m.; two tailed unpaired t-test).

Supplementary Figure 3 Silencing POSTN by shRNA reduced TAM density and extended survival of mice bearing the GSC-derived xenografts.

(a) Immunoblot analysis of POSTN in GSCs expressing non-targeting shRNA (shNT) or POSTN shRNA (shPOSTN, O55). Disrupting POSTN by the shRNA clone O55 through lentiviral infection reduced POSTN expression by >90% in GSCs. (b,c) Kaplan–Meier survival curves of mice implanted with GSCs expressing shPOSTN or shNT (control). GSCs (T3691 or T4121) were transduced with shPOSTN (O55, O56, or O57) or shNT through lentiviral infection and then transplanted intracranially into athymic/nude mice (20,000 cells per mouse). Mice bearing GSC-derived tumours expressing shPOSTN showed a significant survival extension relative to the control group. (n = 5 mice for each group; P = 0.0477 for T3691-O55 versus shNT; P = 0.0026 for T4121-O56 versus shNT; P = 0.0027 for T4121-O57 versus shNT; two-tailed log-rank test). (d) Immunofluorescent analysis of the TAM marker Iba1 (green) in GBM tumours derived from GSCs expressing shPOSTN (O55) or shNT. Frozen sections of GBM tumours derived from GSCs (T3691) expressing shPOSTN (O55) or shNT (NT) were immunostained with antibodies against Iba1 and counterstained with DAPI (blue). A dramatic reduction of TAMs was detected in the GSC-derived tumours expressing shPOSTN. Scale bar, 40 μm. (e) Graphical analysis of (d) showing a significant reduction of TAM density by 70% in the GSC-derived tumours expressing shPOSTN. POSTN intensity and TAM density were analysed with ImageJ. , P < 0.01 (n = 5 tumours; mean ± s.e.m.; two tailed unpaired t-test).

Supplementary Figure 4 TAMs in GSC-derived tumours were CD11b+ and Iba1+ macrophages that were reduced after POSTN disruption.

(a) Immunofluorescent analysis of POSTN (red) and the TAM marker CD11b (green) in GBM tumours derived from GSCs expressing POSTN shRNA (shPOSTN) or NT shRNA (shNT). Frozen sections of GBM tumours derived from GSCs with shPOSTN (O56 and O57) or shNT were immunostained with antibodies against POSTN and Iba1 and counterstained with DAPI (blue). Reductions of both POSTN and TAMs were detected in GSC-derived tumours expressing shPOSTN. Scale bar, 80 μm. (b) Graphical analysis of (a) showing a decrease of POSTN signal intensity by 80% in the GSC-derived tumours expressing POSTN shRNA. , P < 0.01 (n = 5 tumours; mean ± s.e.m; two tailed unpaired t-test). (c) Graphical analysis of (a) showing a significant reduction of TAM density by 60% in the GSC-derived tumours expressing shPOSTN (O56 or O57). POSTN intensity and TAM density were analysed with ImageJ. , P < 0.001 (n = 5 tumours; mean ± s.e.m; two tailed unpaired t-test). (d) Co-immunofluorescent staining of two TAM markers Iba1 (red) and CD11b (green) in sections of GSC-derived orthotopic xenografts. Frozen sections of GBM xenografts derived from GSCs (T387) expressing shNT or shPOSTN (O57) were immunostained with antibodies against Iba1 and CD11b and counterstained with DAPI (blue). Overlapping staining of Iba1 and CD11b was detected in the majority of TAMs in GSC-derived tumours. Scale bar, 80 μm. (e) Co-immunofluorescent staining of two TAM markers Iba1 (red) and CD11b (green) in TAMs isolated from subcutaneous xenografts. Subcutaneous tumour derived from GSCs (CW0322) were dissociated and cultured in complete Neurobasal media for 24 h, and the attached, macrophage-like cells were trypsinized and transferred to RPMI/1640 media supplemented with 10% FBS. The attached cells were immunostained with the pan macrophage marker CD11b (green) along with Iba1 (red). All CD11b+ TAMs are also Iba1+, indicating that Iba1 is a universal TAM marker. Scale bar, 80 μm.

Supplementary Figure 5 TAMs in human GBMs are mainly monocyte-derived M2 macrophages that correlate with the tumour prognosis.

(a) Immunohistochemical (IHC) staining of monocyte-derived TAM marker (CCR2) or resident microglia marker (CX3CR1) in human primary GBM tumours. Two consecutive human GBM tissue microarray slides (GL806b, US Biomax) were immunostained with antibodies against CCR2 or CX3CR1, respectively. CCR2 or CX3CR1 staining in representative tumour cases (A4, B4, and B7) were presented. The majority of TAMs in GBMs were CCR2+/CX3CR1 (monocyte-derived macrophages). Representative image of the IHC staining in normal brain (H2) were presented to show CCR2/CX3CR1+ microglia (indicated by arrays) in normal brain tissue. Scale bar, 40 μm. (bd) Immunofluorescent staining of the pan macrophage marker Iba1 (red) and M2 TAM markers CD163 (b) and Fizz1 (c), or M1 TAM marker HLA-DR (d) and in primary GBM tumours. Frozen sections of GBM tumours were immunostained with the indicated antibodies and counterstained with DAPI (blue). Scale bar, 40 μm. (eg) Graphical analyses of (b), (c), (d) showing fractions of CD163+/Fizz1+ TAMs (M2 subtype) and HLA-DR+ TAMs (M1 subtype) in primary GBM tumours (n = 5 human GBMs; mean ± s.e.m.). (h) Bioinformatic analysis of the relationship between the M2 TAM marker CD163 expression and GBM patient survival in TCGA dataset (The Cancer Genome Atlas) from Oncomine (http://www.oncomine.com). The threshold for high/low CD163 expression is determined by the average of CD163 expression levels in 525 GBM patients. The Kaplan–Meier survival curves demonstrate a significant invert correlation between CD163 levels and overall survival (n = 525 patients; CD163 low, n = 244 patients; CD163 high, n = 271 patients; two-tailed log-rank test; P = 0.0013) of GBM patients. (i) Bioinformatic analysis of the relationship between the M1 TAM marker MHCII expression and GBM patient survival in TCGA dataset (The Cancer Genome Atlas) from Oncomine (http://www.oncomine.com). The threshold for high/low MHCII expression is determined by the average of MHCII expression levels in 525 GBM patients. The Kaplan–Meier survival curves demonstrate a significant positive correlation between MHCII levels and overall survival of GBM patients. (n = 525 patients; MHCII low, n = 280 patients; MHCII high, n = 235 patients; two-tailed log-rank test; P = 0.0007).

Supplementary Figure 6 The M2 subtype TAMs are enriched in POSTN abundant areas in GBMs and POSTN promotes M2 TAM marker expression.

(a) Immunofluorescent analysis of POSTN (red) and the M2 subtype TAM marker CD206 (green) in a GBM tissue microarray (GL806c, US Biomax). Two sets of representative staining were presented to show the enrichment of TAMs in POSTN abundant regions in GBMs. Scale bar, 80 μm. (b) Immunohistochemical staining of POSTN and the M2 subtype TAM marker CD163 in two consecutive tissue microarray slides, respectively (GL806c, US Biomax). Representative staining images were presented to show that the GBM (GL806c-F1) with higher POSTN levels contains more CD163+ cells (M2 TAMs) and the GBM (GL806c-E3) with lower POSTN levels has less CD163+ (M2 TAMs). Scale bar, 40 μm. (c) Graphical analysis of POSTN and CD163 staining in the tissue microarray slides. 63.2% of GBM cases showed POSTNHigh/CD163High staining, and 22.1% of GBM cases showed POSTNLow/CD163Low staining. Only 11.8% of GBM cases showed POSTNHigh/CD163Low staining, and 2.94% of GBM cases showed POSTNLow/CD163High staining. The majority (85.3%) of GBM cases showed that M2 TAM density correlates with POSTN protein levels (data from 68 tumours). (d) RT-qPCR analyses showing up-regulation of the M2 TAM markers induced by POSTN in the isolated mouse peritoneal macrophages. The peritoneal macrophages were stimulated with rPOSTN (0.5 μg ml−1) in the RPMI media supplemented with 0.1% BSA for 24 h. RT-qPCR analysis indicates upregulation of the M2 markers STAB1 and Lyve1, along with the downregulation of the M1 markers CCL17 and IL1b. , P < 0.01; , P < 0.001. (n = 3 biologically independent samples per group; mean ± s.e.m; two tailed unpaired t-test; experiment was repeated three times). (e) RT-qPCR analyses showing downregulation of M1 TAM markers induced by POSTN in PMA-primed macrophage-like U937 cells. PMA-primed U937 cells were treated with recombinant POSTN (0.5 μg ml−1) in RPMI media supplemented with 0.1% BSA for 24 h. RT-qPCR analysis indicated downregulation of the M1 TAM markers including CCL17, IL-12a, and iNOS. , P < 0.01; , P < 0.001; ns, P > 0.05. (n = 3 biologically independent samples per group; mean ± s.e.m; two tailed unpaired t-test; experiment was repeated three times).

Supplementary Figure 7 POSTN disruption shows negligible impact on GSC growth in vitro and differentiated glioma cells rarely express POSTN in GBMs.

(a) Cell titre assay showing proliferation of GSCs expressing shPOSTN or shNT. GSCs were attached to human embryonic stem cell matrix and then transduced with shNT (NT) or shPOSTN (O56 or O57) through lentiviral infection. Cell proliferation was monitored with the luminescent cell viability assay kit at indicated time points. No significant difference was detected in cell proliferation between GSC populations expressing shPOSTN and shNT. (n = 3 biologically independent samples per group; mean ± s.e.m.; one representative experiment shown, and the experiment was repeated 3 times). (b) Immunoblot analysis showing knock-down efficiency of shPOSTN in GSCs through lentiviral infection on Day 2. An 85 90% decrease of endogenous POSTN was detected in GSCs transduced with shPOSTN. (c) Representative images of attached GSCs expressing shNT or shPOSTN. Scale bar, 40 μm. Only mild or negligible effect of POSTN disruption on GSC morphology was detected. (d,e) Immunofluorescent staining of POSTN (green) and the neuron marker TUJ1 (red) in human GBM samples. Scale bar, 40 μm. Statistical analysis (e) showing that less than 5% of TUJ1+ cells overlapped with POSTN staining (n = 3 human GBMs; mean ± s.e.m.). (f,g) Immunofluorescent staining of POSTN (green) and the astrocyte marker GFAP (red) in human GBM samples. Scale bar, 40 μm. Statistical analysis (g) showing that approximately 5% of GFAP+ cells are positive POSTN staining. (n = 3 human GBMs; mean ± s.e.m.). (h,i) Immunofluorescent staining of POSTN (green) and the oligodendrocyte marker Galc (red) in human GBM samples. Scale bar, 40 μm. Statistical analysis (i) showing that less than 6% of Galc+ cells overlapped with POSTN staining (n = 3 human GBMs; mean ± s.e.m.).

Supplementary Figure 8 Disrupting POSTN or its signalling showed very mild effect on tumour vascularization.

(a) Immunofluorescent analysis of blood vessels marked by Glut1 (red) and TAMs labelled by Iba1 (green) in GBM xenografts derived from GSCs expressing shPOSTN or shNT. Frozen sections of GBM tumours derived from GSCs (T387) expressing shPOSTN (O56 and O57) or shNT were immunostained with antibodies against Glut1 and Iba1 and counterstained with DAPI (blue). A dramatic reduction of TAMs in the GSC-derived tumours expressing shPOSTN was confirmed, while the blood vessels were not affected by POSTN disruption. Scale bar, 100 μm. (b) Graphical analysis of (a) showing a significant reduction of TAM density by 70% in the GSC-derived tumours expressing shPOSTN. , P < 0.001 (n = 5 tumours; mean ± s.e.m; two tailed unpaired t-test). (c) Graphical analysis of (a) showing no significant difference in vessel intensity between control tumours (shNT) and the tumours expressing shPOSTN. ns, P > 0.05. Vessel density and TAM density were analysed with ImageJ. (n = 5 tumours; mean ± s.e.m.; two tailed unpaired t-test). (d) Immunofluorescent staining of the vessel marker Glut1 (red) in GBM xenografts treated with RGD inhibitory or control peptide. Mice bearing GBM xenografts were treated with the RGD inhibitory or control peptide at 25 mg kg−1 for 5 days through intraperitoneal delivery. Frozen tumour sections were immunostained with antibodies against Glut1 (red) and counterstained with DAPI (blue). Scale bar, 100 μm. (e) Graphical analysis of (d) showing no significant difference of vessel density between the GBM xenografts treated with the RGD inhibitory peptide and control peptide. ns, P > 0.05 (n = 5 tumours; mean ± s.e.m.; two tailed unpaired t-test).

Supplementary Figure 9 POSTN disruption did not affect GSC population and cell proliferation but induced apoptosis in GSC-derived xenografts.

(a) Immunofluorescent staining of SOX2 in GBM xenografts derived from GSCs expressing shPOSTN or shNT to determine the effect of POSTN knockdown on GSC populations. Frozen sections of GBM xenografts derived from GSCs (T387) expressing shNT or shPOSTN were immunostained with the antibody against SOX2 (red) and counterstained with DAPI (blue). Scale bar, 40 μm. (b) Graphical analysis of (a) showing no significant difference in SOX2+ populations between tumours expressing shPOSTN and shNT (control). Scale bar, 40 μm. P > 0.05 (n = 5 tumours; mean ± s.e.m.; two tailed unpaired t-test). (c) Immunohistochemical (IHC) staining of Ki67 (brown) in GBM xenografts derived from GSCs expressing shPOSTN (O56 or O57) or shNT to determine the effect of POSTN down-regulation on cell proliferation. Scale bar, 40 μm. (d) Graphical analysis of (c) showing no significant difference in Ki67 staining between the tumours expressing shPOSTN and shNT. Scale bar, 40 μm. P > 0.05 (n = 5 tumours; mean ± s.e.m.; two tailed unpaired t-test). (e) TUNEL assay detecting cell apoptosis in GSC-derived tumours expressing shPOSTN or shNT. More apoptotic cell deaths were detected in the tumours with POSTN down-regulation (shPOSTN, O56 or O57) than the control tumours (shNT). Scale bar, 20 μm. (f) Graphical analysis of (e) showing a significant increase (250–300%) in TUNEL positive cells in tumours derived from GSCs expressing shPOSTN relative to the control tumours (shNT). , P < 0.001 (n = 5 tumours; mean ± s.e.m.; two tailed unpaired t-test). (g) qPCR analysis of multiple M1 and M2 macrophage markers in CD11b+ populations from normal mouse brains or GBM xenografts. M2 macrophage markers STAB1 and Lyve1 were markedly up-regulated in CD11b+ population isolated from GBM xenografts, whereas M1 marker IL1b was significantly down-regulated. , P < 0.001; ns, P > 0.05. (mean ± s.e.m.; n = 3 biologically independent samples per group, one representative experiment shown, and the experiment was repeated 3 times).

Supplementary Figure 10

Scans of uncropped blots.

Supplementary Table 1 A list of the secreted proteins differentially expressed by GSCs relative to matched non-stem tumour cells (NSTCs) from primary GBMs.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhou, W., Ke, S., Huang, Z. et al. Periostin secreted by glioblastoma stem cells recruits M2 tumour-associated macrophages and promotes malignant growth. Nat Cell Biol 17, 170–182 (2015). https://doi.org/10.1038/ncb3090

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3090

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