7
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Optimisation of lithium-substituted bioactive glasses to tailor cell response for hard tissue repair

      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

          Bioactive glasses (BG) are used clinically because they can both bond to hard tissue and release therapeutic ions that can stimulate nearby cells. Lithium has been shown to regulate the Wnt/β-catenin cell signalling pathway, which plays important roles in the formation and repair of bone and teeth. Lithium-releasing BG, therefore, have the potential to locally regulate hard tissue formation; however, their design must be tailored to induce an appropriate biological response. Here, we optimised the release of lithium from lithium-substituted BG by varying BG composition, particle size and concentration to minimise toxicity and maximise upregulation of the Wnt target gene Axin2 in in vitro cell cultures. Our results show that we can tailor lithium release from BG over a wide therapeutic and non-toxic range. Increasing the concentration of BG in cell culture medium can induce toxicity, likely due to modulations in pH. Nevertheless, at sub-toxic concentrations, lithium released from BG can upregulate the Wnt pathway in 17IA4 cells, similarly to treatment with LiCl. Taken together, these data demonstrate that ion release from lithium-substituted BG can be tailored to maximise biological response. These data may be important in the design of BG that can regulate the Wnt/β-catenin pathway to promote hard tissue repair or regeneration.

          Related collections

          Most cited references29

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

          Wnt/beta-catenin signaling in mesenchymal progenitors controls osteoblast and chondrocyte differentiation during vertebrate skeletogenesis.

          Chondrocytes and osteoblasts are two primary cell types in the skeletal system that are differentiated from common mesenchymal progenitors. It is believed that osteoblast differentiation is controlled by distinct mechanisms in intramembranous and endochondral ossification. We have found that ectopic canonical Wnt signaling leads to enhanced ossification and suppression of chondrocyte formation. Conversely, genetic inactivation of beta-catenin, an essential component transducing the canonical Wnt signaling, causes ectopic formation of chondrocytes at the expense of osteoblast differentiation during both intramembranous and endochondral ossification. Moreover, inactivation of beta-catenin in mesenchymal progenitor cells in vitro causes chondrocyte differentiation under conditions allowing only osteoblasts to form. Our results demonstrate that beta-catenin is essential in determining whether mesenchymal progenitors will become osteoblasts or chondrocytes regardless of regional locations or ossification mechanisms. Controlling Wnt/beta-catenin signaling is a common molecular mechanism underlying chondrocyte and osteoblast differentiation and specification of intramembranous and endochondral ossification.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Can bioactivity be tested in vitro with SBF solution?

            A large part of the scientific community has accepted the paradigm that a simulated body solution (SBF) can be used to test the bioactivity of a material. This is exemplified by the rapidly increasing number of publications using this test. The aim of this document is to demonstrate that (i) there is presently not enough scientific data to support this assumption, and (ii) even though the assumption was valid, the way the test is generally conducted leaves room for improvement. Theoretical arguments and facts supporting these statements are provided, together with possible improvements of the proposed bioactivity test.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Lrp5-independent activation of Wnt signaling by lithium chloride increases bone formation and bone mass in mice.

              One of the well characterized cell biologic actions of lithium is the inhibition of glycogen synthase kinase-3beta and the consequent activation of canonical Wnt signaling. Because deficient Wnt signaling has been implicated in disorders of reduced bone mass, we tested whether lithium could improve bone mass in mice. We gavage-fed lithium chloride to 8-week-old mice from three different strains (Lrp5(-/-), SAMP6, and C57BL/6) and assessed the effect on bone metabolism after 4 weeks of therapy. Lrp5(-/-) mice lack the Wnt coreceptor low-density lipoprotein receptor-related protein 5 and have markedly reduced bone mass. Lithium, which is predicted to act downstream of this receptor, restored bone metabolism and bone mass to near wild-type levels in these mice. SAMP6 mice have accelerated osteoporosis due to inadequate osteoblast renewal. Lithium significantly improved bone mass in these mice and in wild-type C57BL/6 mice. We found that lithium activated canonical Wnt signaling in cultured calvarial osteoblasts from Lrp5(-/-) mice ex vivo and that lithium-treated mice had increased expression of Wnt-responsive genes in their bone marrow cells in vivo. These data lead us to conclude that lithium enhances bone formation and improves bone mass in mice and that it may do so via activation of the canonical Wnt pathway. Lithium has been used safely and effectively for over half a century in the treatment of bipolar illness. Prospective studies in patients receiving lithium should determine whether it also improves bone mass in humans.
                Bookmark

                Author and article information

                Contributors
                +44 (0) 20 7188 7388 , eileen.gentleman@kcl.ac.uk
                Journal
                J Mater Sci
                J Mater Sci
                Journal of Materials Science
                Springer US (New York )
                0022-2461
                1573-4803
                9 February 2017
                9 February 2017
                2017
                : 52
                : 15
                : 8832-8844
                Affiliations
                [1 ]ISNI 0000 0001 2322 6764, GRID grid.13097.3c, Craniofacial Development and Stem Cell Biology, , King’s College London, ; 27th Floor, Guy’s Hospital, London, SE1 9RT UK
                [2 ]ISNI 0000 0001 1939 2794, GRID grid.9613.d, Otto Schott Institute of Materials Research, , Friedrich Schiller University Jena, ; 07743 Jena, Germany
                Author information
                http://orcid.org/0000-0003-0447-5137
                Article
                838
                10.1007/s10853-017-0838-7
                5644509
                29056759
                b02824a5-5bea-4e81-9402-218eb9a1c3cb
                © The Author(s) 2017

                Open AccessThis 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
                : 29 September 2016
                : 24 January 2017
                Funding
                Funded by: FundRef http://dx.doi.org/10.13039/100004440, Wellcome Trust;
                Funded by: FundRef http://dx.doi.org/10.13039/501100000833, Rosetrees Trust;
                Funded by: FundRef http://dx.doi.org/10.13039/501100000275, Leverhulme Trust;
                Funded by: FundRef http://dx.doi.org/10.13039/501100000265, Medical Research Council;
                Categories
                In Honour of Larry Hench
                Custom metadata
                © Springer Science+Business Media New York 2017

                Comments

                Comment on this article