Your browser doesn't support javascript.
loading
IL-4/IL-13 Stimulated Macrophages Enhance Breast Cancer Invasion Via Rho-GTPase Regulation of Synergistic VEGF/CCL-18 Signaling.
Little, Andrew C; Pathanjeli, Pragathi; Wu, Zhifen; Bao, Liwei; Goo, Laura E; Yates, Joel A; Oliver, C Ryan; Soellner, Matthew B; Merajver, Sofia D.
Afiliación
  • Little AC; Department of Internal Medicine, Hematology-Oncology, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States.
  • Pathanjeli P; Department of Internal Medicine, Hematology-Oncology, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States.
  • Wu Z; Department of Internal Medicine, Hematology-Oncology, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States.
  • Bao L; Department of Internal Medicine, Hematology-Oncology, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States.
  • Goo LE; Department of Internal Medicine, Hematology-Oncology, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States.
  • Yates JA; Department of Internal Medicine, Hematology-Oncology, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States.
  • Oliver CR; Department of Internal Medicine, Hematology-Oncology, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States.
  • Soellner MB; Department of Internal Medicine, Hematology-Oncology, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States.
  • Merajver SD; Department of Internal Medicine, Hematology-Oncology, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States.
Front Oncol ; 9: 456, 2019.
Article en En | MEDLINE | ID: mdl-31214501
ABSTRACT
Tumor associated macrophages (TAMs) are increasingly recognized as major contributors to the metastatic progression of breast cancer and enriched levels of TAMs often correlate with poor prognosis. Despite our current advances it remains unclear which subset of M2-like macrophages have the highest capacity to enhance the metastatic program and which mechanisms regulate this process. Effective targeting of macrophages that aid cancer progression requires knowledge of the specific mechanisms underlying their pro-metastatic actions, as to avoid the anticipated toxicities from generalized targeting of macrophages. To this end, we set out to understand the relationship between the regulation of tumor secretions by Rho-GTPases, which were previously demonstrated to affect them, macrophage differentiation, and the converse influence of macrophages on cancer cell phenotype. Our data show that IL-4/IL-13 in vitro differentiated M2a macrophages significantly increase migratory and invasive potential of breast cancer cells at a greater rate than M2b or M2c macrophages. Our previous work demonstrated that the Rho-GTPases are potent regulators of macrophage-induced migratory responses; therefore, we examined M2a-mediated responses in RhoA or RhoC knockout breast cancer cell models. We find that both RhoA and RhoC regulate migration and invasion in MDA-MB-231 and SUM-149 cells following stimulation with M2a conditioned media. Secretome analysis of M2a conditioned media reveals high levels of vascular endothelial growth factor (VEGF) and chemokine (C-C motif) ligand 18 (CCL-18). Results from our functional assays reveal that M2a TAMs synergistically utilize VEGF and CCL-18 to promote migratory and invasive responses. Lastly, we show that pretreatment with ROCK inhibitors Y-276332 or GSK42986A attenuated VEGF/CCL-18 and M2a-induced migration and invasion. These results support Rho-GTPase signaling regulates downstream responses induced by TAMs, offering a novel approach for the prevention of breast cancer metastasis by anti-RhoA/C therapies.
Palabras clave

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Tipo de estudio: Prognostic_studies Idioma: En Revista: Front Oncol Año: 2019 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Tipo de estudio: Prognostic_studies Idioma: En Revista: Front Oncol Año: 2019 Tipo del documento: Article País de afiliación: Estados Unidos
...