Correlation Engine 2.0
Clear Search sequence regions


  • apoptosis (3)
  • blood cells (1)
  • breast cancers (3)
  • breast tumors (2)
  • cancer (2)
  • cell growth (1)
  • cell movement (1)
  • ERK1 (1)
  • female (1)
  • humans (1)
  • IL15 (4)
  • IL15RA (9)
  • il2 receptors (1)
  • IL2RB (3)
  • IL2RG (3)
  • interleukin 15 (4)
  • JAK1 (1)
  • JAK2 (1)
  • lymphocytes (1)
  • mice (1)
  • nk cells (1)
  • PRAS40 (1)
  • prognosis (1)
  • protein human (2)
  • receptor (3)
  • receptors interleukin- 15 (2)
  • regulates (1)
  • signal (1)
  • STAT1 (1)
  • STAT2 (1)
  • STAT5 (1)
  • t cells (1)
  • Sizes of these terms reflect their relevance to your search.

    Despite its aggressive nature, triple-negative breast cancer (TNBC) often exhibits leucocyte infiltrations that correlate with favorable prognosis. In this study, we offer an explanation for this apparent conundrum by defining TNBC cell subsets that overexpress the IL15 immune receptor IL15RA. This receptor usually forms a heterotrimer with the IL2 receptors IL2RB and IL2RG, which regulates the proliferation and differentiation of cytotoxic T cells and NK cells. However, unlike IL15RA, the IL2RB and IL2RG receptors are not upregulated in basal-like TNBC breast cancer cells that express IL15RA. Mechanistic investigations indicated that IL15RA signaling activated JAK1, STAT1, STAT2, AKT, PRAS40, and ERK1/2 in the absence of IL2RB and IL2RG, whereas neither STAT5 nor JAK2 were activated. RNAi-mediated attenuation of IL15RA established its role in cell growth, apoptosis, and migration, whereas expression of the IL15 cytokine in IL15RA-expressing cells stimulated an autocrine signaling cascade that promoted cell proliferation and migration and blocked apoptosis. Notably, coexpression of IL15RA and IL15 was also sufficient to activate peripheral blood mononuclear cells upon coculture in a paracrine signaling manner. Overall, our findings offer a mechanistic explanation for the paradoxical association of some high-grade breast tumors with better survival outcomes, due to engagement of the immune stroma. ©2014 American Association for Cancer Research.

    Citation

    Pierfrancesco Marra, Sumi Mathew, Anita Grigoriadis, Yin Wu, Fernanda Kyle-Cezar, Johnathan Watkins, Mamunur Rashid, Emanuele De Rinaldis, Sonya Hessey, Patrycja Gazinska, Adrian Hayday, Andrew Tutt. IL15RA drives antagonistic mechanisms of cancer development and immune control in lymphocyte-enriched triple-negative breast cancers. Cancer research. 2014 Sep 01;74(17):4908-21

    Expand section icon Mesh Tags

    Expand section icon Substances


    PMID: 24980552

    View Full Text