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

      Tissue-regenerative potential of the secretome of γ-irradiated peripheral blood mononuclear cells is mediated via TNFRSF1B-induced necroptosis

      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.

          Abstract

          Peripheral blood mononuclear cells (PBMCs) have been shown to produce and release a plethora of pro-angiogenetic factors in response to γ-irradiation, partially accounting for their tissue-regenerative capacity. Here, we investigated whether a certain cell subtype of PBMCs is responsible for this effect, and whether the type of cell death affects the pro-angiogenic potential of bioactive molecules released by γ-irradiated PBMCs. PBMCs and PBMC subpopulations, including CD4 + and CD8 + T cells, B cells, monocytes, and natural killer cells, were isolated and subjected to high-dose γ-irradiation. Transcriptome analysis revealed subpopulation-specific responses to γ-irradiation with distinct activation of pro-angiogenic pathways, cytokine production, and death receptor signalling. Analysis of the proteins released showed that interactions of the subsets are important for the generation of a pro-angiogenic secretome. This result was confirmed at the functional level by the finding that the secretome of γ-irradiated PBMCs displayed higher pro-angiogenic activity in an aortic ring assay. Scanning electron microscopy and image stream analysis of γ-irradiated PBMCs revealed distinct morphological changes, indicative for apoptotic and necroptotic cell death. While inhibition of apoptosis had no effect on the pro-angiogenic activity of the secretome, inhibiting necroptosis in stressed PBMCs abolished blood vessel sprouting. Mechanistically, we identified tumor necrosis factor (TNF) receptor superfamily member 1B as the main driver of necroptosis in response to γ-irradiation in PBMCs, which was most likely mediated via membrane-bound TNF-α. In conclusion, our study demonstrates that the pro-angiogenic activity of the secretome of γ-irradiated PBMCs requires interplay of different PBMC subpopulations. Furthermore, we show that TNF-dependent necroptosis is an indispensable molecular process for conferring tissue-regenerative activity and for the pro-angiogenic potential of the PBMC secretome. These findings contribute to a better understanding of secretome-based therapies in regenerative medicine.

          Related collections

          Most cited references35

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

          Paracrine mechanisms of stem cell reparative and regenerative actions in the heart.

          Stem cells play an important role in restoring cardiac function in the damaged heart. In order to mediate repair, stem cells need to replace injured tissue by differentiating into specialized cardiac cell lineages and/or manipulating the cell and molecular mechanisms governing repair. Despite early reports describing engraftment and successful regeneration of cardiac tissue in animal models of heart failure, these events appear to be infrequent and yield too few new cardiomyocytes to account for the degree of improved cardiac function observed. Instead, mounting evidence suggests that stem cell mediated repair takes place via the release of paracrine factors into the surrounding tissue that subsequently direct a number of restorative processes including myocardial protection, neovascularization, cardiac remodeling, and differentiation. The potential for diverse stem cell populations to moderate many of the same processes as well as key paracrine factors and molecular pathways involved in stem cell-mediated cardiac repair will be discussed in this review. This article is part of a special issue entitled, "Cardiovascular Stem Cells Revisited". Copyright © 2010 Elsevier Ltd. All rights reserved.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            A role of RIP3-mediated macrophage necrosis in atherosclerosis development.

            Necrotic death of macrophages has long been known to be present in atherosclerotic lesions but has not been studied. We examined the role of receptor interacting protein (RIP) 3, a mediator of necrotic cell death, in atherosclerosis and found that RIP3(-/-);Ldlr(-/-) mice were no different from RIP3(+/+);Ldlr(-/-) mice in early atherosclerosis but had significant reduction in advanced atherosclerotic lesions. Similar results were observed in Apoe(-/-) background mice. Bone marrow transplantation revealed that loss of RIP3 expression from bone-marrow-derived cells is responsible for the reduced disease progression. While no difference was found in apoptosis between RIP3(-/-);Ldlr(-/-) and RIP3(+/+);Ldlr(-/-) mice, electron microscopy revealed a significant reduction of macrophage primary necrosis in the advanced lesions of RIP3(-/-) mice. In vitro cellular studies showed that RIP3 deletion had no effect on oxidized low-density lipoprotein (LDL)-induced macrophage apoptosis, but prevented macrophage primary necrosis occurring in response to oxidized LDL under caspase inhibition or RIP3 overexpression conditions. RIP3-dependent necrosis is not postapoptotic, and the increased primary necrosis in advanced atherosclerotic lesions most likely resulted from the increase of RIP3 expression. Our data demonstrate that primary necrosis of macrophages is proatherogenic during advanced atherosclerosis development. Copyright © 2013 The Authors. Published by Elsevier Inc. All rights reserved.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Necrostatin-1 reduces histopathology and improves functional outcome after controlled cortical impact in mice.

              Necroptosis is a newly identified type of programmed necrosis initiated by the activation of tumor necrosis factor alpha (TNFalpha)/Fas. Necrostatin-1 is a specific inhibitor of necroptosis that reduces ischemic tissue damage in experimental stroke models. We previously reported decreased tissue damage and improved functional outcome after controlled cortical impact (CCI) in mice deficient in TNFalpha and Fas. Hence, we hypothesized that necrostatin-1 would reduce histopathology and improve functional outcome after CCI in mice. Compared with vehicle-/inactive analog-treated controls, mice administered necrostatin-1 before CCI had decreased propidium iodide-positive cells in the injured cortex and dentate gyrus (6 h), decreased brain tissue damage (days 14, 35), improved motor (days 1 to 7), and Morris water maze performance (days 8 to 14) after CCI. Improved spatial memory was observed even when drug was administered 15 mins after CCI. Necrostatin-1 treatment did not reduce caspase-3-positive cells in the dentate gyrus or cortex, consistent with a known caspase-independent mechanism of necrostatin-1. However, necrostatin-1 reduced brain neutrophil influx and microglial activation at 48 h, suggesting a novel anti-inflammatory effect in traumatic brain injury (TBI). The data suggest that necroptosis plays a significant role in the pathogenesis of cell death and functional outcome after TBI and that necrostatin-1 may have therapeutic potential for patients with TBI.
                Bookmark

                Author and article information

                Contributors
                +43-1-4040069770 , hendrik.ankersmit@meduniwien.ac.at
                +43-1-40400 73507 , michael.mildner@meduniwien.ac.at
                Journal
                Cell Death Dis
                Cell Death Dis
                Cell Death & Disease
                Nature Publishing Group UK (London )
                2041-4889
                30 September 2019
                30 September 2019
                October 2019
                : 10
                : 10
                : 729
                Affiliations
                [1 ]ISNI 0000 0000 9259 8492, GRID grid.22937.3d, Department of Internal Medicine III, Division of Rheumatology, , Medical University of Vienna, ; Vienna, Austria
                [2 ]ISNI 0000 0000 9259 8492, GRID grid.22937.3d, Division of Thoracic Surgery, , Medical University of Vienna, ; Vienna, Austria
                [3 ]ISNI 0000 0000 9259 8492, GRID grid.22937.3d, FFG Project 852748 “APOSEC“, , Medical University of Vienna, ; Vienna, Austria
                [4 ]ISNI 0000 0000 9259 8492, GRID grid.22937.3d, Department of Biomedical Imaging and Image-guided Therapy, , Medical University of Vienna, ; Vienna, Austria
                [5 ]Department of Radiology and Cancer Research UK Cambridge Center, Cambridge, CB2 0QQ UK
                [6 ]ISNI 0000 0000 9259 8492, GRID grid.22937.3d, Vienna Business Agency Project 2343727 “APOSEC to clinic”, , Medical University Vienna, ; Vienna, Austria
                [7 ]Synlab Analytics and Services Switzerland AG, Birsfelden, Switzerland
                [8 ]ISNI 0000 0000 9259 8492, GRID grid.22937.3d, Research Division of Biology and Pathobiology of the SkinDepartment of Dermatology, Research Division of Biology and Pathobiology of the Skin, , Medical University of Vienna, ; Vienna, Austria
                [9 ]ISNI 0000 0000 9259 8492, GRID grid.22937.3d, Division of Oral and Maxillofacial Surgery, , Medical University of Vienna, ; Vienna, Austria
                [10 ]ISNI 0000 0000 9259 8492, GRID grid.22937.3d, Research Laboratories, Core Facility Flow Cytometry, , Medical University of Vienna, ; Vienna, Austria
                Author information
                http://orcid.org/0000-0003-4388-7580
                http://orcid.org/0000-0002-5178-9967
                http://orcid.org/0000-0002-6892-925X
                Article
                1974
                10.1038/s41419-019-1974-6
                6768878
                31570701
                ed9d4714-9292-433b-a38c-7f37d3178080
                © The Author(s) 2019

                Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

                History
                : 10 June 2019
                : 5 September 2019
                : 9 September 2019
                Funding
                Funded by: FundRef https://doi.org/10.13039/501100001821, Vienna Science and Technology Fund (Wiener Wissenschafts-, Forschungs- und Technologiefonds);
                Award ID: 2343727
                Award ID: 2343727
                Award ID: 2343727
                Award ID: 2343727
                Award ID: 2343727
                Award Recipient :
                Categories
                Article
                Custom metadata
                © The Author(s) 2019

                Cell biology
                cell biology,necroptosis
                Cell biology
                cell biology, necroptosis

                Comments

                Comment on this article