Angiogenic capacity of M1-and M2-polarized macrophages is determined by the levels of TIMP-1 complexed with their secreted proMMP-9

E Zajac, B Schweighofer… - Blood, The Journal …, 2013 - ashpublications.org
E Zajac, B Schweighofer, TA Kupriyanova, A Juncker-Jensen, P Minder, JP Quigley…
Blood, The Journal of the American Society of Hematology, 2013ashpublications.org
A proangiogenic function of tissue-infiltrating monocytes/macrophages has long been
attributed to their matrix metalloproteinase-9 zymogen (proMMP-9). Herein, we evaluated
the capacity of human monocytes, mature M0 macrophages, and M1-and M2-polarized
macrophages to induce proMMP-9-mediated angiogenesis. Only M2 macrophages induced
angiogenesis at levels comparable with highly angiogenic neutrophils previously shown to
release their proMMP-9 in a unique form, free of tissue inhibitor of metalloproteinases-1 …
Abstract
A proangiogenic function of tissue-infiltrating monocytes/macrophages has long been attributed to their matrix metalloproteinase-9 zymogen (proMMP-9). Herein, we evaluated the capacity of human monocytes, mature M0 macrophages, and M1- and M2-polarized macrophages to induce proMMP-9-mediated angiogenesis. Only M2 macrophages induced angiogenesis at levels comparable with highly angiogenic neutrophils previously shown to release their proMMP-9 in a unique form, free of tissue inhibitor of metalloproteinases-1 (TIMP-1). Macrophage differentiation was accompanied by induction of low-angiogenic, TIMP-1–encumbered proMMP-9. However, polarization toward the M2, but not the M1 phenotype, caused a substantial downregulation of TIMP-1 expression, resulting in production of angiogenic, TIMP-deficient proMMP-9. Correspondingly, the angiogenic potency of M2 proMMP-9 was lost after its complexing with TIMP-1, whereas TIMP-1 silencing in M0/M1 macrophages rendered them both angiogenic. Similar to human cells, murine bone marrow–derived M2 macrophages also shut down their TIMP-1 expression and produced proMMP-9 unencumbered by TIMP-1. Providing proof that angiogenic capacity of murine M2 macrophages depended on their TIMP-free proMMP-9, Mmp9-null M2 macrophages were nonangiogenic, although their TIMP-1 was severely downregulated. Our study provides a unifying molecular mechanism for high angiogenic capacity of TIMP-free proMMP-9 that would be uniquely produced in a pathophysiological microenvironment by influxing neutrophils and/or M2 polarized macrophages.
ashpublications.org