Your browser doesn't support javascript.
loading
Harnessing macrophage-mediated degradation of gelatin microspheres for spatiotemporal control of BMP2 release.
Annamalai, Ramkumar T; Turner, Paul A; Carson, William F; Levi, Benjamin; Kunkel, Steven; Stegemann, Jan P.
Affiliation
  • Annamalai RT; Department of Biomedical Engineering, University of Michigan, Ann Arbor, USA. Electronic address: ramta@umich.edu.
  • Turner PA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, USA.
  • Carson WF; Department of Pathology, University of Michigan, Ann Arbor, USA.
  • Levi B; Department of Surgery, University of Michigan, Ann Arbor, USA.
  • Kunkel S; Department of Pathology, University of Michigan, Ann Arbor, USA.
  • Stegemann JP; Department of Biomedical Engineering, University of Michigan, Ann Arbor, USA. Electronic address: jpsteg@umich.edu.
Biomaterials ; 161: 216-227, 2018 04.
Article in En | MEDLINE | ID: mdl-29421557
ABSTRACT
Biomaterials-based approaches to harnessing the immune and inflammatory responses to potentiate wound healing hold important promise. Bone fracture healing is characterized by an acute inflammatory phase, followed by a transition to a regenerative and repair phase. In this study, we developed genipin-crosslinked gelatin microspheres designed to be preferentially degraded by inflammatory (M1) macrophages. Highly crosslinked (>90%) microspheres allowed efficient incorporation of bioactive bone morphogenetic protein 2 (BMP2), a potent stimulator of osteogenesis in progenitor cells, via electrostatic interactions. Release of BMP2 was directly correlated with degradation of the gelatin matrix. Exposure of microspheres to polarized murine macrophages showed that degradation was significantly higher in the presence of M1 macrophages, relative to alternatively activated (M2) macrophages and unpolarized controls. Microsphere degradation in the presence of non-inflammatory cells resulted in very low degradation rates. The expression of matrix metalloproteinases (MMPs) and tissue inhibitors of MMP (TIMPs) by macrophages were consistent with the observed phenotype-dependent degradation rates. Indirect co-culture of BMP2-loaded microspheres and macrophages with isolated adipose-derived mesenchymal stem cells (MSC) showed that M1 macrophages produced the strongest osteogenic response, comparable to direct supplementation of the culture medium with BMP2. Controlled release systems that are synchronized with the inflammatory response have the potential to provide better spatiotemporal control of growth factor delivery and therefore may improve the outcomes of recalcitrant wounds.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Bone and Bones / Bone Morphogenetic Protein 2 / Gelatin / Microspheres Limits: Animals Language: En Journal: Biomaterials Year: 2018 Document type: Article

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Bone and Bones / Bone Morphogenetic Protein 2 / Gelatin / Microspheres Limits: Animals Language: En Journal: Biomaterials Year: 2018 Document type: Article