3
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      RIP1 kinase mediates angiogenesis by modulating macrophages in experimental neovascularization

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Significance

          Pathological angiogenesis has been implicated in diverse pathologies. Infiltrating macrophages, especially those activated to M2-like phenotype, are critically important for angiogenesis. Although the role of RIP1 kinase in the regulation of apoptosis, necroptosis, and inflammation has been well-established, its role in angiogenesis remains elusive, despite being abundantly expressed in angiogenesis-related infiltrating macrophages. This study demonstrates that RIP1 kinase inhibition attenuates angiogenesis in multiple mouse models of pathological angiogenesis in vivo and suggests a therapeutic role of RIP1 kinase inhibition in pathological angiogenesis. Mechanistically, the inhibitory effect on angiogenesis depends on RIP kinase inhibition-mediated caspase activation in infiltrating macrophages through suppression of M2-like polarization, and subsequent attenuation of pathological angiogenesis.

          Abstract

          Inflammation plays an important role in pathological angiogenesis. Receptor-interacting protein 1 (RIP1) is highly expressed in inflammatory cells and is known to play an important role in the regulation of apoptosis, necroptosis, and inflammation; however, a comprehensive description of its role in angiogenesis remains elusive. Here, we show that RIP1 is abundantly expressed in infiltrating macrophages during angiogenesis, and genetic or pharmacological inhibition of RIP1 kinase activity using kinase-inactive RIP1 K45A/K45A mice or necrostatin-1 attenuates angiogenesis in laser-induced choroidal neovascularization, Matrigel plug angiogenesis, and alkali injury-induced corneal neovascularization in mice. The inhibitory effect on angiogenesis is mediated by caspase activation through a kinase-independent function of RIP1 and RIP3. Mechanistically, infiltrating macrophages are the key target of RIP1 kinase inhibition to attenuate pathological angiogenesis. Inhibition of RIP1 kinase activity is associated with caspase activation in infiltrating macrophages and decreased expression of proangiogenic M2-like markers but not M1-like markers. Similarly, in vitro, catalytic inhibition of RIP1 down-regulates the expression of M2-like markers in interleukin-4–activated bone marrow-derived macrophages, and this effect is blocked by simultaneous caspase inhibition. Collectively, these results demonstrate a nonnecrotic function of RIP1 kinase activity and suggest that RIP1-mediated modulation of macrophage activation may be a therapeutic target of pathological angiogenesis.

          Related collections

          Most cited references57

          • Record: found
          • Abstract: found
          • Article: not found

          Anti-inflammatory M2, but not pro-inflammatory M1 macrophages promote angiogenesis in vivo.

          Macrophages show extreme heterogeneity and different subsets have been characterized by their activation route and their function. For instance, macrophage subsets are distinct by acting differently under pathophysiological conditions such as inflammation and cancer. Macrophages also contribute to angiogenesis, but the role of various specific subsets in angiogenesis has not been thoroughly investigated. Matrigel supplemented with macrophage subsets [induced by IFNγ (M1), IL-4 (M2a) or IL-10 (M2c)] was injected subcutaneously in C57BL/6 J mice and analyzed by CD31 staining after 14 days. Increased numbers of endothelial cells and tubular structures were observed in M2-enriched plugs compared to control and other subsets. Additionally, more tubular structures formed in vitro in the presence of M2 macrophages or their conditioned medium. To identify a mechanism for the pro-angiogenic effect, gene expression of angiogenic growth factors was analyzed. Induced expression of basic fibroblast growth factor (Fgf2), insulin-like growth factor-1 (Igf1), chemokine (C-C motif) ligand 2 (Ccl2) and placental growth factor (Pgf) was observed in M2 macrophages. Using a blocking antibody of PlGF to inhibit M2c induced angiogenesis resulted in mildly reduced (40 %) tube formation whereas neutralization of FGF-2 (M2a) signaling by sFGFR1-IIIc affected tube formation by nearly 75 %. These results indicate that macrophages polarized towards an M2 phenotype have a higher angiogenic potential compared to other subsets. Furthermore, we propose FGF signaling for M2a- and PlGF signaling for M2c-induced angiogenesis as possible working mechanisms, yet, further research should elucidate the exact mechanism for M2-induced angiogenesis.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            RIP3 induces apoptosis independent of pronecrotic kinase activity.

            Receptor-interacting protein kinase 3 (RIP3 or RIPK3) has emerged as a central player in necroptosis and a potential target to control inflammatory disease. Here, three selective small-molecule compounds are shown to inhibit RIP3 kinase-dependent necroptosis, although their therapeutic value is undermined by a surprising, concentration-dependent induction of apoptosis. These compounds interact with RIP3 to activate caspase 8 (Casp8) via RHIM-driven recruitment of RIP1 (RIPK1) to assemble a Casp8-FADD-cFLIP complex completely independent of pronecrotic kinase activities and MLKL. RIP3 kinase-dead D161N mutant induces spontaneous apoptosis independent of compound, whereas D161G, D143N, and K51A mutants, like wild-type, only trigger apoptosis when compound is present. Accordingly, RIP3-K51A mutant mice (Rip3(K51A/K51A)) are viable and fertile, in stark contrast to the perinatal lethality of Rip3(D161N/D161N) mice. RIP3 therefore holds both necroptosis and apoptosis in balance through a Ripoptosome-like platform. This work highlights a common mechanism unveiling RHIM-driven apoptosis by therapeutic or genetic perturbation of RIP3.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Sequential delivery of immunomodulatory cytokines to facilitate the M1-to-M2 transition of macrophages and enhance vascularization of bone scaffolds.

              In normal tissue repair, macrophages exhibit a pro-inflammatory phenotype (M1) at early stages and a pro-healing phenotype (M2) at later stages. We have previously shown that M1 macrophages initiate angiogenesis while M2 macrophages promote vessel maturation. Therefore, we reasoned that scaffolds that promote sequential M1 and M2 polarization of infiltrating macrophages should result in enhanced angiogenesis and healing. To this end, we first analyzed the in vitro kinetics of macrophage phenotype switch using flow cytometry, gene expression, and cytokine secretion analysis. Then, we designed scaffolds for bone regeneration based on modifications of decellularized bone for a short release of interferon-gamma (IFNg) to promote the M1 phenotype, followed by a more sustained release of interleukin-4 (IL4) to promote the M2 phenotype. To achieve this sequential release profile, IFNg was physically adsorbed onto the scaffolds, while IL4 was attached via biotin-streptavidin binding. Interestingly, despite the strong interactions between biotin and streptavidin, release studies showed that biotinylated IL4 was released over 6 days. These scaffolds promoted sequential M1 and M2 polarization of primary human macrophages as measured by gene expression of ten M1 and M2 markers and secretion of four cytokines, although the overlapping phases of IFNg and IL4 release tempered polarization to some extent. Murine subcutaneous implantation model showed increased vascularization in scaffolds releasing IFNg compared to controls. This study demonstrates that scaffolds for tissue engineering can be designed to harness the angiogenic behavior of host macrophages towards scaffold vascularization.
                Bookmark

                Author and article information

                Journal
                Proc Natl Acad Sci U S A
                Proc. Natl. Acad. Sci. U.S.A
                pnas
                pnas
                PNAS
                Proceedings of the National Academy of Sciences of the United States of America
                National Academy of Sciences
                0027-8424
                1091-6490
                19 November 2019
                4 November 2019
                4 November 2019
                : 116
                : 47
                : 23705-23713
                Affiliations
                [1] aAngiogenesis Laboratory, Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School , Boston, MA 02114;
                [2] bDepartment of Ophthalmology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo , 113-8655 Tokyo, Japan;
                [3] cDepartment of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital , 833 Kaohsiung, Taiwan;
                [4] dDepartment of Ophthalmology, Graduate School of Medical Science, Kyushu University , 812-8582 Fukuoka, Japan;
                [5] eSchepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School , Boston, MA 02114
                Author notes
                1To whom correspondence may be addressed. Email: demetrios_vavvas@ 123456meei.harvard.edu .

                Edited by Akrit Sodhi, Johns Hopkins University School of Medicine, Baltimore, MD, and accepted by Editorial Board Member Jeremy Nathans October 7, 2019 (received for review May 23, 2019)

                Author contributions: T.U. and D.G.V. designed research; T.U., K.I., S.N., J.-J.L., D.E.M., N.E.E., Y.M., E.H., K.A., T.T., and E.I.P. performed research; T.U. analyzed data; and T.U., K.I., Y.M., M.A., J.W.M., and D.G.V. wrote the paper.

                Author information
                http://orcid.org/0000-0002-7935-4244
                http://orcid.org/0000-0002-3447-4074
                http://orcid.org/0000-0002-6635-0391
                http://orcid.org/0000-0002-4544-4452
                http://orcid.org/0000-0003-2046-3996
                http://orcid.org/0000-0002-8622-6478
                Article
                201908355
                10.1073/pnas.1908355116
                6876205
                31685620
                3937202f-5f36-4531-9a32-59e9a18275eb
                Copyright © 2019 the Author(s). Published by PNAS.

                This open access article is distributed under Creative Commons Attribution-NonCommercial-NoDerivatives License 4.0 (CC BY-NC-ND).

                History
                Page count
                Pages: 9
                Categories
                PNAS Plus
                Biological Sciences
                Medical Sciences
                PNAS Plus

                ripk,necrosis,immune,neovascular,amd
                ripk, necrosis, immune, neovascular, amd

                Comments

                Comment on this article