Deletion of NF-kB p50 alters murine glioblastoma tumor-associated macrophage polarization, reduces tumor growth and prolongs survival

  • Barberi T
  • Martin A
  • Harris-Bookman S
  • et al.
N/ACitations
Citations of this article
3Readers
Mendeley users who have this article in their library.

Abstract

Glioblastoma multiforme (GBM) is a uniformly fatal brain tumor. The GBM microenvironment includes abundant tumor-associated macrophages (TAMs) that predominantly assume a pro-tumor “M2” phenotype rather than a pro-inflammatory “M1” phenotype. TAMs suppress T cell activation, produce VEGF, secrete proliferative factors, and encourage tumor invasion and metastasis. The inhibitory p50 subunit of the NF-kB transcription factor exhibits markedly increased nuclear expression in TAMs and M2- polarized macrophages [1], and p50 knockdown or deletion suppresses expression of M2-associated factors [1-3]. We hypothesize that absence of TAM p50 will convert TAMs to a pro-inflammatory M1 phenotype that will reduce tumor growth and prolong survival. The murine glioma cell line GL261-Luc was intracranially implanted into wild-type and p50(-/-) mice. Tumors grew 6-fold slower in p50(-/-) compared with wild-type mice, and p50(-/-) mice exhibited significantly increased survival. RT-qPCR analysis of CD11b+ myeloid cells isolated from the brains of tumor-bearing mice revealed decreased M2 marker expression and increased M1 marker expression in the absence of NF-kB p50. Evaluation of tumor-infiltrating T cells indicated that p50(-/-) mice possess decreased Tregcells, and that more p50(-/-) CD4 T cells induce IFNγ expression after PMA/Ionomycin stimulation than wild-type CD4 T cells. When M2-polarized p50(-/-) bone marrow-derived macrophages (BMM) are co-cultured in vitro with wild-type T cells, they do not suppress T cell proliferation to the same extent as wild-type BMM. These data suggest that NF-kB p50 is an important modulator of the suppressive TAM phenotype in GBM and that deletion of the gene encoding p50 promotes conversion to a pro-inflammatory phenotype that is less tumorpermissive. We anticipate that targeting p50 in combination with immune checkpoint inhibition might prove synergistic in facilitating anti-tumor immunity and tumor regression.

Cite

CITATION STYLE

APA

Barberi, T., Martin, A., Harris-Bookman, S., Lim, M., & Friedman, A. D. (2015). Deletion of NF-kB p50 alters murine glioblastoma tumor-associated macrophage polarization, reduces tumor growth and prolongs survival. Journal for ImmunoTherapy of Cancer, 3(S2). https://doi.org/10.1186/2051-1426-3-s2-p395

Register to see more suggestions

Mendeley helps you to discover research relevant for your work.

Already have an account?

Save time finding and organizing research with Mendeley

Sign up for free