Cargando…

Modulation of macrophage antitumor potential by apoptotic lymphoma cells

In aggressive non-Hodgkin's lymphoma (NHL), constitutive apoptosis of a proportion of the tumor cell population can promote net tumor growth. This is associated with the accumulation of tumor-associated macrophages (TAMs) that clear apoptotic cells and exhibit pro-oncogenic transcriptional acti...

Descripción completa

Detalles Bibliográficos
Autores principales: Voss, Jorine J L P, Ford, Catriona A, Petrova, Sofia, Melville, Lynsey, Paterson, Margaret, Pound, John D, Holland, Pam, Giotti, Bruno, Freeman, Tom C, Gregory, Christopher D
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group 2017
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5442466/
https://www.ncbi.nlm.nih.gov/pubmed/28157210
http://dx.doi.org/10.1038/cdd.2016.132
Descripción
Sumario:In aggressive non-Hodgkin's lymphoma (NHL), constitutive apoptosis of a proportion of the tumor cell population can promote net tumor growth. This is associated with the accumulation of tumor-associated macrophages (TAMs) that clear apoptotic cells and exhibit pro-oncogenic transcriptional activation profiles characteristic of reparatory, anti-inflammatory and angiogenic programs. Here we consider further the activation status of these TAMs. We compare their transcriptomic profile with that of a range of other macrophage types from various tissues noting especially their expression of classically activated (IFN-γ and LPS) gene clusters – typically antitumor – in addition to their previously described protumor phenotype. To understand the impact of apoptotic cells on the macrophage activation state, we cocultured apoptotic lymphoma cells with classically activated macrophages (M((IFN-γ/LPS)), also known as M1, macrophages). Although untreated and M((IFN-γ/LPS)) macrophages were able to bind apoptotic lymphoma cells equally well, M((IFN-γ/LPS)) macrophages displayed enhanced ability to phagocytose them. We found that direct exposure of M((IFN-γ/LPS)) macrophages to apoptotic lymphoma cells caused switching towards a protumor activation state (often referred to as M2-like) with concomitant inhibition of antitumor activity that was a characteristic feature of M((IFN-γ/LPS)) macrophages. Indeed, M((IFN-γ/LPS)) macrophages exposed to apoptotic lymphoma cells displayed increased lymphoma growth-promoting activities. Antilymphoma activity by M((IFN-γ/LPS)) macrophages was mediated, in part, by galectin-3, a pleiotropic glycoprotein involved in apoptotic cell clearance that is strongly expressed by lymphoma TAMs but not lymphoma cells. Intriguingly, aggressive lymphoma growth was markedly impaired in mice deficient in galectin-3, suggesting either that host galectin-3-mediated antilymphoma activity is required to sustain net tumor growth or that additional functions of galectin-3 drive key oncogenic mechanisms in NHL. These findings have important implications for anticancer therapeutic approaches aimed at polarizing macrophages towards an antitumor state and identify galectin-3 as a potentially important novel target in aggressive NHL.