Your browser doesn't support javascript.
loading
Erythrocyte-driven immunization via biomimicry of their natural antigen-presenting function.
Ukidve, Anvay; Zhao, Zongmin; Fehnel, Alexandra; Krishnan, Vinu; Pan, Daniel C; Gao, Yongsheng; Mandal, Abhirup; Muzykantov, Vladimir; Mitragotri, Samir.
Afiliação
  • Ukidve A; John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138.
  • Zhao Z; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115.
  • Fehnel A; John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138.
  • Krishnan V; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115.
  • Pan DC; John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138.
  • Gao Y; John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138.
  • Mandal A; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115.
  • Muzykantov V; John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138.
  • Mitragotri S; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115.
Proc Natl Acad Sci U S A ; 117(30): 17727-17736, 2020 07 28.
Article em En | MEDLINE | ID: mdl-32665441
ABSTRACT
Erythrocytes naturally capture certain bacterial pathogens in circulation, kill them through oxidative stress, and present them to the antigen-presenting cells (APCs) in the spleen. By leveraging this innate immune function of erythrocytes, we developed erythrocyte-driven immune targeting (EDIT), which presents nanoparticles from the surface of erythrocytes to the APCs in the spleen. Antigenic nanoparticles were adsorbed on the erythrocyte surface. By engineering the number density of adsorbed nanoparticles, (i.e., the number of nanoparticles loaded per erythrocyte), they were predominantly delivered to the spleen rather than lungs, which is conventionally the target of erythrocyte-mediated delivery systems. Presentation of erythrocyte-delivered nanoparticles to the spleen led to improved antibody response against the antigen, higher central memory T cell response, and lower regulatory T cell response, compared with controls. Enhanced immune response slowed down tumor progression in a prophylaxis model. These findings suggest that EDIT is an effective strategy to enhance systemic immunity.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Imunização / Apresentação de Antígeno / Eritrócitos / Antígenos Tipo de estudo: Prognostic_studies Limite: Animals / Female / Humans Idioma: En Revista: Proc Natl Acad Sci U S A Ano de publicação: 2020 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Imunização / Apresentação de Antígeno / Eritrócitos / Antígenos Tipo de estudo: Prognostic_studies Limite: Animals / Female / Humans Idioma: En Revista: Proc Natl Acad Sci U S A Ano de publicação: 2020 Tipo de documento: Article