Abstract

Macrophages regulate host responses to implants through their dynamic adhesion, release, and activation. Herein, we employ bisphosphonate (BP)-coated gold nanoparticle template (BNP) to direct the swift and convertible formation of Mg2+-functional Mg2+-BP nanoparticle (NP) on the BP-AuNP surface via reversible Mg2+-BP coordination, thus producing (Mg2+-BP)-Au dimer (MgBNP). Ethylenediaminetetraacetic acid-based Mg2+ chelation facilitates the dissolution of Mg2+-BP NP, thus enabling the reversion of the MgBNP to the BNP. This convertible nanoassembly incorporating cell-adhesive Mg2+ moieties directs reversible attachment and detachment of macrophages by BP and EDTA, without physical scraping or trypsin that could damage cells. The swift formation of RGD ligand- and Mg2+-bifunctional RGD-Mg2+-BP NP that yields (RGD-Mg2+-BP)-Au dimer (RGDBNP) further stimulates the adhesion and pro-regenerative M2-type polarization of macrophages, both in vitro and in vivo, including rho-associated protein kinase. This swift and non-toxic dimer formation can include diverse bio-functional moieties to regulate host responses to implants.

Highlights

  • Macrophages regulate host responses to implants through their dynamic adhesion, release, and activation

  • We hypothesized that the BP ligand on the surface of the AuNP would direct the binding of Mg2+, which facilitates the localized formation of the Mg2+-BP NP driven by the Mg2+BP coordination on the surface of the BP-AuNP

  • The detachment of macrophages by physical scraping was highly efficient but resulted in the low viability (62%) of the detached macrophages (Supplementary Fig. 13b–d). These findings indicate that the non-toxic, efficient, and reversible attachment and detachment of macrophages induced by this convertible formation of the Mg2+-BP NPs without the use of physical scraping or proteolytic enzyme, offer unique advantages for applications in culturing and studying macrophages

Read more

Summary

Introduction

Macrophages regulate host responses to implants through their dynamic adhesion, release, and activation. Ethylenediaminetetraacetic acid-based Mg2+ chelation facilitates the dissolution of Mg2+-BP NP, enabling the reversion of the MgBNP to the BNP This convertible nanoassembly incorporating cell-adhesive Mg2+ moieties directs reversible attachment and detachment of macrophages by BP and EDTA, without physical scraping or trypsin that could damage cells. The swift formation of RGD ligand- and Mg2+-bifunctional RGD-Mg2+-BP NP that yields (RGD-Mg2+-BP)-Au dimer (RGDBNP) further stimulates the adhesion and pro-regenerative M2-type polarization of macrophages, both in vitro and in vivo, including rho-associated protein kinase. This swift and non-toxic dimer formation can include diverse bio-functional moieties to regulate host responses to implants. It is desirable to develop materials that can manipulate the attachment and detachment of macrophages as well as their phenotypic polarization by dynamically presenting bio-functional moieties

Methods
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.