1
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Direct osteogenesis and immunomodulation dual function via sustained release of naringin from the polymer scaffold

      Read this article at

      ScienceOpenPublisher
      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

          In this work, the NG-MBG/PLLA scaffolds not only have a good ability to promote osteogenic differentiation of mBMSCs but also provide suitable bone immune microenvironment capabilities for bone regeneration during the inflammatory phase.

          Abstract

          Many traditional Chinese medicine monomers, such as naringin (NG), can regulate the local immune microenvironment to benefit osteogenesis. However, the rapid release of NG from scaffolds severely influences the osteogenesis-promoting effect. Herein, NG was loaded into mesoporous bioglass (MBG) to achieve sustained release through physical adsorption and the barrier role of mesoporous channels, then MBG loaded with NG was added to poly( l-lactic acid) (PLLA) to fabricate composite scaffolds by selective laser sintering (SLS) technology. The results showed that the NG-MBG/PLLA scaffolds could continuously and slowly release NG for 14 days compared with NG/PLLA scaffolds, and the cumulative release amount for the NG-MBG/PLLA scaffolds was 44.26%. In addition, the NG-MBG/PLLA scaffolds can promote the proliferation and osteogenesis differentiation of mouse bone marrow mesenchymal stem cells (mBMSCs). Meanwhile, the composite scaffolds decreased the reactive oxygen species (ROS) level of RAW264.7 under the stimulation of lipopolysaccharide (LPS) and significantly suppressed interleukin-6 (IL-6) and enhanced arginase-1 (Arg-1) protein expressions. Moreover, calcium nodule and alkaline phosphatase production of mBMSCs in a macrophage-conditioned medium for the NG-MBG/PLLA group also evidently increased compared with the PLLA and MBG/PLLA groups. These NG sustained-release composite scaffolds with osteo-immunomodulation function have great application prospects in the clinic.

          Related collections

          Most cited references44

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

          Anti-inflammatory effects of flavonoids

          Inflammation plays a key role in diseases such as diabetes, asthma, cardiovascular diseases and cancer. Diet can influence different stages of inflammation and can have an important impact on several inflammatory diseases. Increasing scientific evidence has shown that polyphenolic compounds, such as flavonoids, which are found in fruits, vegetables, legumes, or cocoa, can have anti-inflammatory properties. Recent studies have demonstrated that flavonoids can inhibit regulatory enzymes or transcription factors important for controlling mediators involved in inflammation. Flavonoids are also known as potent antioxidants with the potential to attenuate tissue damage or fibrosis. Consequently, numerous studies in vitro and in animal models have found that flavonoids have the potential to inhibit the onset and development of inflammatory diseases. In the present review, we focused in flavonoids, the most abundant polyphenols in the diet, to give an overview of the most recent scientific knowledge about their impact on different inflammatory diseases.
            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
              • Record: found
              • Abstract: found
              • Article: not found

              A Biomimetic Hierarchical Nanointerface Orchestrates Macrophage Polarization and Mesenchymal Stem Cell Recruitment To Promote Endogenous Bone Regeneration

              The host immune response to bone biomaterials is vital in determining scaffold fates and bone regeneration outcomes. The nanometer-scale interface of biomaterials, which independently controls physical inputs to cells, regulates osteogenic differentiation of stem cells and local immune response. Herein, we fabricated biomimetic hierarchical intrafibrillarly mineralized collagen (HIMC) with a bone-like staggered nanointerface and investigated its immunomodulatory properties and mesenchymal stem cell (MSC) recruitment during endogenous bone regeneration. The acquired HIMC potently induced neo-bone formation by promoting CD68+CD163+ M2 macrophage polarization and CD146+STRO-1+ host MSC recruitment in critical-sized bone defects. Mechanistically, HIMC facilitated M2 macrophage polarization and interleukin (IL)-4 secretion to promote MSC osteogenic differentiation. An anti-IL4 neutralizing antibody significantly reduced M2 macrophage-mediated osteogenic differentiation of MSCs. Moreover, HIMC-loaded-IL-4 implantation into critical-sized mandible defects dramatically enhanced bone regeneration and CD68+CD163+ M2 macrophage polarization. The depletion of monocyte/macrophages by clodronate liposomes significantly impaired bone regeneration by HIMC, but did not affect MSC recruitment. Thus, in emulating natural design, the hierarchical nanointerface possesses the capacity to recruit host MSCs and promote endogenous bone regeneration by immunomodulation of macrophage polarization through IL-4.
                Bookmark

                Author and article information

                Contributors
                Journal
                JMCBDV
                Journal of Materials Chemistry B
                J. Mater. Chem. B
                Royal Society of Chemistry (RSC)
                2050-750X
                2050-7518
                November 22 2023
                2023
                : 11
                : 45
                : 10896-10907
                Affiliations
                [1 ]Jiangxi University of Chinese Medicine, Nanchang 330004, China
                [2 ]Department of ophthalmology, The Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang 330006, China
                [3 ]School of Energy and Mechanical Engineering, Jiangxi University of Science and Technology, Nanchang 330013, China
                [4 ]Institute of Additive Manufacturing, Jiangxi University of Science and Technology, Nanchang 330013, China
                [5 ]State Key Laboratory of Precision Manufacturing for Extreme Service Performance, College of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
                [6 ]College of Mechanical Engineering, Xinjiang University, Urumqi 830017, China
                [7 ]Department of Orthopedics and Trauma, The Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang 330006, China
                [8 ]National Engineering Research Center for Manufacturing Technology of Traditional Chinese Medicine Solid Preparations, Jiangxi University of Chinese Medicine, Nanchang 330004, China
                Article
                10.1039/D3TB01555F
                6d3399f5-9995-4e9e-aff9-629f3996c627
                © 2023

                http://rsc.li/journals-terms-of-use

                History

                Comments

                Comment on this article