A distinguishing gene signature shared by tumor-infiltrating Tie2-expressing monocytes, blood “resident” monocytes, and embryonic macrophages suggests common …

F Pucci, MA Venneri, D Biziato, A Nonis… - Blood, The Journal …, 2009 - ashpublications.org
F Pucci, MA Venneri, D Biziato, A Nonis, D Moi, A Sica, C Di Serio, L Naldini, M De Palma
Blood, The Journal of the American Society of Hematology, 2009ashpublications.org
We previously showed that Tie2-expressing monocytes (TEMs) have nonredundant
proangiogenic activity in tumors. Here, we compared the gene expression profile of tumor-
infiltrating TEMs with that of tumor-associated macrophages (TAMs), spleen-derived Gr1+
Cd11b+ neutrophils/myeloid-derived suppressor cells, circulating “inflammatory” and
“resident” monocytes, and tumor-derived endothelial cells (ECs) by quantitative polymerase
chain reaction–based gene arrays. TEMs sharply differed from ECs and Gr1+ Cd11b+ cells …
Abstract
We previously showed that Tie2-expressing monocytes (TEMs) have nonredundant proangiogenic activity in tumors. Here, we compared the gene expression profile of tumor-infiltrating TEMs with that of tumor-associated macrophages (TAMs), spleen-derived Gr1+Cd11b+ neutrophils/myeloid-derived suppressor cells, circulating “inflammatory” and “resident” monocytes, and tumor-derived endothelial cells (ECs) by quantitative polymerase chain reaction–based gene arrays. TEMs sharply differed from ECs and Gr1+Cd11b+ cells but were highly related to TAMs. Nevertheless, several genes were differentially expressed between TEMs and TAMs, highlighting a TEM signature consistent with enhanced proangiogenic/tissue-remodeling activity and lower proinflammatory activity. We validated these findings in models of oncogenesis and transgenic mice expressing a microRNA-regulated Tie2-GFP reporter. Remarkably, resident monocytes and TEMs on one hand, and inflammatory monocytes and TAMs on the other hand, expressed coordinated gene expression profiles, suggesting that the 2 blood monocyte subsets are committed to distinct extravascular fates in the tumor microenvironment. We further showed that a prominent proportion of embryonic/fetal macrophages, which participate in tissue morphogenesis, expressed distinguishing TEM genes. It is tempting to speculate that Tie2+ embryonic/fetal macrophages, resident blood monocytes, and tumor-infiltrating TEMs represent distinct developmental stages of a TEM lineage committed to execute physiologic proangiogenic and tissue-remodeling programs, which can be coopted by tumors.
ashpublications.org