Your browser doesn't support javascript.
loading
HaloTag display enables quantitative single-particle characterisation and functionalisation of engineered extracellular vesicles.
Mitrut, Roxana E; Stranford, Devin M; DiBiase, Beth N; Chan, Jonathan M; Bailey, Matthew D; Luo, Minrui; Harper, Clare S; Meade, Thomas J; Wang, Muzhou; Leonard, Joshua N.
Afiliação
  • Mitrut RE; Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois, USA.
  • Stranford DM; Center for Synthetic Biology, Northwestern University, Evanston, Illinois, USA.
  • DiBiase BN; Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois, USA.
  • Chan JM; Center for Synthetic Biology, Northwestern University, Evanston, Illinois, USA.
  • Bailey MD; Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois, USA.
  • Luo M; Center for Synthetic Biology, Northwestern University, Evanston, Illinois, USA.
  • Harper CS; Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois, USA.
  • Meade TJ; Department of Chemistry, Northwestern University, Evanston, Illinois, USA.
  • Wang M; Department of Chemistry, Northwestern University, Evanston, Illinois, USA.
  • Leonard JN; Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois, USA.
J Extracell Vesicles ; 13(7): e12469, 2024 Jul.
Article em En | MEDLINE | ID: mdl-38965984
ABSTRACT
Extracellular vesicles (EVs) play key roles in diverse biological processes, transport biomolecules between cells and have been engineered for therapeutic applications. A useful EV bioengineering strategy is to express engineered proteins on the EV surface to confer targeting, bioactivity and other properties. Measuring how incorporation varies across a population of EVs is important for characterising such materials and understanding their function, yet it remains challenging to quantitatively characterise the absolute number of engineered proteins incorporated at single-EV resolution. To address these needs, we developed a HaloTag-based characterisation platform in which dyes or other synthetic species can be covalently and stoichiometrically attached to engineered proteins on the EV surface. To evaluate this system, we employed several orthogonal quantification methods, including flow cytometry and fluorescence microscopy, and found that HaloTag-mediated quantification is generally robust across EV analysis methods. We compared HaloTag-labelling to antibody-labelling of EVs using single vesicle flow cytometry, enabling us to measure the substantial degree to which antibody labelling can underestimate proteins present on an EV. Finally, we demonstrate the use of HaloTag to compare between protein designs for EV bioengineering. Overall, the HaloTag system is a useful EV characterisation tool which complements and expands existing methods.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Citometria de Fluxo / Vesículas Extracelulares Limite: Humans Idioma: En Revista: J Extracell Vesicles Ano de publicação: 2024 Tipo de documento: Article País de afiliação: Estados Unidos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Citometria de Fluxo / Vesículas Extracelulares Limite: Humans Idioma: En Revista: J Extracell Vesicles Ano de publicação: 2024 Tipo de documento: Article País de afiliação: Estados Unidos