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

      Selective inhibition of receptor activator of NF-κB ligand (RANKL) in hematopoietic cells improves outcome after experimental myocardial infarction

      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

          Abstract

          The RANK (receptor activator of nuclear factor κB)/RANKL (RANK ligand)/OPG (osteoprotegerin) axis is activated after myocardial infarction (MI), but its pathophysiological role is not well understood. Here, we investigated how global and cell compartment-selective inhibition of RANKL affects cardiac function and remodeling after MI in mice. Global RANKL inhibition was achieved by treatment of human RANKL knock-in (huRANKL-KI) mice with the monoclonal antibody AMG161. huRANKL-KI mice express a chimeric RANKL protein wherein part of the RANKL molecule is humanized. AMG161 inhibits human and chimeric but not murine RANKL. To dissect the pathophysiological role of RANKL derived from hematopoietic and mesenchymal cells, we selectively exchanged the hematopoietic cell compartment by lethal irradiation and across-genotype bone marrow transplantation between wild-type and huRANKL-KI mice, exploiting the specificity of AMG161. After permanent coronary artery ligation, mice were injected with AMG161 or an isotype control antibody over 4 weeks post-MI. MI increased RANKL expression mainly in cardiomyocytes and scar-infiltrating cells 4 weeks after MI. Only inhibition of RANKL derived from hematopoietic cellular sources, but not global or mesenchymal RANKL inhibition, improved post-infarct survival and cardiac function. Mechanistically, hematopoietic RANKL inhibition reduced expression of the pro-inflammatory cytokine IL-1ß in the cardiac cellular infiltrate. In conclusion, inhibition of RANKL derived from hematopoietic cellular sources is beneficial to maintain post-ischemic cardiac function by reduction of pro-inflammatory cytokine production.

          Key messages

          • Experimental myocardial infarction (MI) augments cardiac RANKL expression in mice.

          • RANKL expression is increased in cardiomyocytes and scar-infiltrating cells after MI.

          • Global or mesenchymal cell RANKL inhibition has no influence on cardiac function after MI.

          • Inhibition of RANKL derived from hematopoietic cells improves heart function post-MI.

          • Hematopoietic RANKL inhibition reduces pro-inflammatory cytokines in scar-infiltrating cells.

          Electronic supplementary material

          The online version of this article (10.1007/s00109-018-1641-x) contains supplementary material, which is available to authorized users.

          Related collections

          Most cited references20

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

          Tumor necrosis factor receptor family member RANK mediates osteoclast differentiation and activation induced by osteoprotegerin ligand.

          A receptor that mediates osteoprotegerin ligand (OPGL)-induced osteoclast differentiation and activation has been identified via genomic analysis of a primary osteoclast precursor cell cDNA library and is identical to the tumor necrosis factor receptor (TNFR) family member RANK. The RANK mRNA was highly expressed by isolated bone marrow-derived osteoclast progenitors and by mature osteoclasts in vivo. Recombinant OPGL binds specifically to RANK expressed by transfected cell lines and purified osteoclast progenitors. Transgenic mice expressing a soluble RANK-Fc fusion protein have severe osteopetrosis because of a reduction in osteoclasts, similar to OPG transgenic mice. Recombinant RANK-Fc binds with high affinity to OPGL in vitro and blocks osteoclast differentiation and activation in vitro and in vivo. Furthermore, polyclonal Ab against the RANK extracellular domain promotes osteoclastogenesis in bone marrow cultures suggesting that RANK activation mediates the effects of OPGL on the osteoclast pathway. These data indicate that OPGL-induced osteoclastogenesis is directly mediated through RANK on osteoclast precursor cells.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Osteoclasts degrade endosteal components and promote mobilization of hematopoietic progenitor cells.

            Here we investigated the potential role of bone-resorbing osteoclasts in homeostasis and stress-induced mobilization of hematopoietic progenitors. Different stress situations induced activity of osteoclasts (OCLs) along the stem cell-rich endosteum region of bone, secretion of proteolytic enzymes and mobilization of progenitors. Specific stimulation of OCLs with RANKL recruited mainly immature progenitors to the circulation in a CXCR4- and MMP-9-dependent manner; however, RANKL did not induce mobilization in young female PTPepsilon-knockout mice with defective OCL bone adhesion and resorption. Inhibition of OCLs with calcitonin reduced progenitor egress in homeostasis, G-CSF mobilization and stress situations. RANKL-stimulated bone-resorbing OCLs also reduced the stem cell niche components SDF-1, stem cell factor (SCF) and osteopontin along the endosteum, which was associated with progenitor mobilization. Finally, the major bone-resorbing proteinase, cathepsin K, also cleaved SDF-1 and SCF. Our findings indicate involvement of OCLs in selective progenitor recruitment as part of homeostasis and host defense, linking bone remodeling with regulation of hematopoiesis.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Interleukin-1 receptor type I signaling critically regulates infarct healing and cardiac remodeling.

              The proinflammatory cytokine interleukin (IL)-1 signals exclusively through the type I IL-1 receptor (IL-1RI). IL-1 expression is markedly induced in the infarcted heart; however, its role in cardiac injury and repair remains controversial. We examined the effects of disrupted IL-1 signaling on infarct healing and cardiac remodeling using IL-1RI(-/-) mice. After reperfused infarction IL-1RI-null mice exhibited decreased infiltration of the infarcted myocardium with neutrophils and macrophages and reduced chemokine and cytokine expression. In the absence of IL-1 signaling, suppressed inflammation was followed by an attenuated fibrotic response. Infarcted IL-1RI(-/-) mice had decreased myofibroblast infiltration and reduced collagen deposition in the infarcted and remodeling myocardium. IL-1RI deficiency protected against the development of adverse remodeling; however, infarct size was comparable between groups suggesting that the beneficial effects of IL-1RI gene disruption were not attributable to decreased cardiomyocyte injury. Reduced chamber dilation in IL-1RI-null animals was associated with decreased collagen deposition and attenuated matrix metalloproteinase (MMP)-2 and MMP-3 expression in the peri-infarct area, suggesting decreased fibrotic remodeling of the noninfarcted heart. IL-1beta stimulated MMP mRNA synthesis in wild-type, but not in IL-1RI-null cardiac fibroblasts. In conclusion, IL-1 signaling is essential for activation of inflammatory and fibrogenic pathways in the healing infarct, playing an important role in the pathogenesis of remodeling after infarction. Thus, interventional therapeutics targeting the IL-1 system may have great benefits in myocardial infarction.
                Bookmark

                Author and article information

                Contributors
                +43-1-250 77 4550 , Reinhold.Erben@vetmeduni.ac.at
                Journal
                J Mol Med (Berl)
                J. Mol. Med
                Journal of Molecular Medicine (Berlin, Germany)
                Springer Berlin Heidelberg (Berlin/Heidelberg )
                0946-2716
                1432-1440
                8 May 2018
                8 May 2018
                2018
                : 96
                : 6
                : 559-573
                Affiliations
                [1 ]ISNI 0000 0000 9686 6466, GRID grid.6583.8, Institute of Physiology, Pathophysiology and Biophysics, Department of Biomedical Research, , University of Veterinary Medicine Vienna, ; Veterinaerplatz 1, 1210 Vienna, Austria
                [2 ]ISNI 0000 0000 9686 6466, GRID grid.6583.8, VetCore, , University of Veterinary Medicine Vienna, ; Vienna, Austria
                [3 ]ISNI 0000 0001 2111 7257, GRID grid.4488.0, Division of Endocrinology, Diabetes, and Bone Diseases, Department of Medicine III and Center for Healthy Aging, , Technische Universität Dresden, ; Dresden, Germany
                [4 ]ISNI 0000 0001 0657 5612, GRID grid.417886.4, Amgen Inc., ; Thousand Oaks, CA USA
                [5 ]Present Address: Phylon Pharma Services, Newbury Park, CA USA
                Author information
                http://orcid.org/0000-0003-0801-6958
                Article
                1641
                10.1007/s00109-018-1641-x
                5988763
                29736604
                479a6009-6181-42ca-a76b-ae9176b35461
                © The Author(s) 2018

                Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided 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.

                History
                : 18 September 2017
                : 18 April 2018
                : 22 April 2018
                Funding
                Funded by: FundRef http://dx.doi.org/10.13039/501100002428, Austrian Science Fund;
                Award ID: FWF P 21904-B11
                Categories
                Original Article
                Custom metadata
                © Springer-Verlag GmbH Germany, part of Springer Nature 2018

                Molecular medicine
                myocardial infarction,rankl,inflammation,osteoprotegerin
                Molecular medicine
                myocardial infarction, rankl, inflammation, osteoprotegerin

                Comments

                Comment on this article