Your browser doesn't support javascript.
loading
Targeting AAV vectors to the central nervous system by engineering capsid-receptor interactions that enable crossing of the blood-brain barrier.
Huang, Qin; Chen, Albert T; Chan, Ken Y; Sorensen, Hikari; Barry, Andrew J; Azari, Bahar; Zheng, Qingxia; Beddow, Thomas; Zhao, Binhui; Tobey, Isabelle G; Moncada-Reid, Cynthia; Eid, Fatma-Elzahraa; Walkey, Christopher J; Ljungberg, M Cecilia; Lagor, William R; Heaney, Jason D; Chan, Yujia A; Deverman, Benjamin E.
Afiliação
  • Huang Q; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America.
  • Chen AT; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America.
  • Chan KY; Biological and Biomedical Sciences, Harvard University, Cambridge, Massachusetts, United States of America.
  • Sorensen H; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America.
  • Barry AJ; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America.
  • Azari B; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America.
  • Zheng Q; Electrical & Computer Engineering Department, Northeastern University, Boston, Massachusetts, United States of America.
  • Beddow T; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America.
  • Zhao B; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America.
  • Tobey IG; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America.
  • Moncada-Reid C; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America.
  • Eid FE; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America.
  • Walkey CJ; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America.
  • Ljungberg MC; Department of Systems and Computer Engineering, Al-Azhar University, Cairo, Egypt.
  • Lagor WR; Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, United States of America.
  • Heaney JD; Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States of America.
  • Chan YA; Duncan Neurological Research Institute at Texas Children's Hospital, Houston, Texas, United States of America.
  • Deverman BE; Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, United States of America.
PLoS Biol ; 21(7): e3002112, 2023 07.
Article em En | MEDLINE | ID: mdl-37467291
ABSTRACT
Viruses have evolved the ability to bind and enter cells through interactions with a wide variety of cell macromolecules. We engineered peptide-modified adeno-associated virus (AAV) capsids that transduce the brain through the introduction of de novo interactions with 2 proteins expressed on the mouse blood-brain barrier (BBB), LY6A or LY6C1. The in vivo tropisms of these capsids are predictable as they are dependent on the cell- and strain-specific expression of their target protein. This approach generated hundreds of capsids with dramatically enhanced central nervous system (CNS) tropisms within a single round of screening in vitro and secondary validation in vivo thereby reducing the use of animals in comparison to conventional multi-round in vivo selections. The reproducible and quantitative data derived via this method enabled both saturation mutagenesis and machine learning (ML)-guided exploration of the capsid sequence space. Notably, during our validation process, we determined that nearly all published AAV capsids that were selected for their ability to cross the BBB in mice leverage either the LY6A or LY6C1 protein, which are not present in primates. This work demonstrates that AAV capsids can be directly targeted to specific proteins to generate potent gene delivery vectors with known mechanisms of action and predictable tropisms.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Barreira Hematoencefálica / Capsídeo Limite: Animals Idioma: En Revista: PLoS Biol Assunto da revista: BIOLOGIA Ano de publicação: 2023 Tipo de documento: Article País de afiliação: Estados Unidos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Barreira Hematoencefálica / Capsídeo Limite: Animals Idioma: En Revista: PLoS Biol Assunto da revista: BIOLOGIA Ano de publicação: 2023 Tipo de documento: Article País de afiliação: Estados Unidos