Your browser doesn't support javascript.
loading
Rapid On-Demand Extracellular Vesicle Augmentation with Versatile Oligonucleotide Tethers.
Yerneni, Saigopalakrishna S; Lathwal, Sushil; Shrestha, Pradeep; Shirwan, Haval; Matyjaszewski, Krzysztof; Weiss, Lee; Yolcu, Esma S; Campbell, Phil G; Das, Subha R.
Afiliação
  • Yerneni SS; Department of Biomedical Engineering , Carnegie Mellon University , Pittsburgh , Pennsylvania 15213 , United States.
  • Lathwal S; Department of Chemistry , Carnegie Mellon University , Pittsburgh , Pennsylvania 15213 , United States.
  • Shrestha P; Center for Nucleic Acids Science & Technology , Carnegie Mellon University , Pittsburgh , Pennsylvania 15213 , United States.
  • Shirwan H; Institute for Cellular Therapeutics and Department of Microbiology and Immunology , University of Louisville , Louisville , Kentucky 40292 , United States.
  • Matyjaszewski K; Institute for Cellular Therapeutics and Department of Microbiology and Immunology , University of Louisville , Louisville , Kentucky 40292 , United States.
  • Weiss L; Department of Chemistry , Carnegie Mellon University , Pittsburgh , Pennsylvania 15213 , United States.
  • Yolcu ES; Department of Biomedical Engineering , Carnegie Mellon University , Pittsburgh , Pennsylvania 15213 , United States.
  • Campbell PG; The Robotics Institute , Carnegie Mellon University , Pittsburgh , Pennsylvania 15213 , United States.
  • Das SR; Institute for Cellular Therapeutics and Department of Microbiology and Immunology , University of Louisville , Louisville , Kentucky 40292 , United States.
ACS Nano ; 13(9): 10555-10565, 2019 09 24.
Article em En | MEDLINE | ID: mdl-31436946
ABSTRACT
Exosomes show potential as ideal vehicles for drug delivery because of their natural role in transferring biological cargo between cells. However, current methods to engineer exosomes without negatively impacting their function remain challenging. Manipulating exosome-secreting cells is complex and time-consuming, while direct functionalization of exosome surface proteins suffers from low specificity and low efficiency. We demonstrate a rapid, versatile, and scalable method with oligonucleotide tethers to enable diverse surface functionalization on both human and murine exosomes. These exosome surface modifiers, which range from reactive functional groups and small molecules to aptamers and large proteins, can readily and efficiently enhance native exosome properties. We show that cellular uptake of exosomes can be specifically altered with a tethered AS1411 aptamer, and targeting specificity can be altered with a tethered protein. We functionalize exosomes with an immunomodulatory protein, FasL, and demonstrate their biological activity both in vitro and in vivo. FasL-functionalized exosomes, when bioprinted on a collagen matrix, allows spatial induction of apoptosis in tumor cells and, when injected in mice, suppresses proliferation of alloreactive T cells. This oligonucleotide tethering strategy is independent of the exosome source and further circumvents the need to genetically modify exosome-secreting cells.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Temas: Geral Base de dados: MEDLINE Assunto principal: Oligonucleotídeos / Vesículas Extracelulares Limite: Animals / Humans Idioma: En Revista: ACS Nano Ano de publicação: 2019 Tipo de documento: Article País de afiliação: Estados Unidos

Texto completo: 1 Coleções: 01-internacional Temas: Geral Base de dados: MEDLINE Assunto principal: Oligonucleotídeos / Vesículas Extracelulares Limite: Animals / Humans Idioma: En Revista: ACS Nano Ano de publicação: 2019 Tipo de documento: Article País de afiliação: Estados Unidos