Your browser doesn't support javascript.
loading
Engineered lentivirus-derived nanoparticles (LVNPs) for delivery of CRISPR/Cas ribonucleoprotein complexes supporting base editing, prime editing and in vivo gene modification.
Haldrup, Jakob; Andersen, Sofie; Labial, Alexander Rafael LaVilla; Wolff, Jonas Holst; Frandsen, Frederik Plum; Skov, Thomas Wisbech; Rovsing, Anne Bruun; Nielsen, Ian; Jakobsen, Thomas Stax; Askou, Anne Louise; Thomsen, Martin K; Corydon, Thomas J; Thomsen, Emil Aagaard; Mikkelsen, Jacob Giehm.
Afiliación
  • Haldrup J; Department of Biomedicine, Aarhus University, Aarhus C, Denmark.
  • Andersen S; Department of Biomedicine, Aarhus University, Aarhus C, Denmark.
  • Labial ARL; Department of Biomedicine, Aarhus University, Aarhus C, Denmark.
  • Wolff JH; Department of Biomedicine, Aarhus University, Aarhus C, Denmark.
  • Frandsen FP; Department of Biomedicine, Aarhus University, Aarhus C, Denmark.
  • Skov TW; Department of Biomedicine, Aarhus University, Aarhus C, Denmark.
  • Rovsing AB; Department of Biomedicine, Aarhus University, Aarhus C, Denmark.
  • Nielsen I; Department of Biomedicine, Aarhus University, Aarhus C, Denmark.
  • Jakobsen TS; Department of Biomedicine, Aarhus University, Aarhus C, Denmark.
  • Askou AL; Department of Ophthalmology, Aarhus University Hospital, Aarhus N, Denmark.
  • Thomsen MK; Department of Biomedicine, Aarhus University, Aarhus C, Denmark.
  • Corydon TJ; Department of Ophthalmology, Aarhus University Hospital, Aarhus N, Denmark.
  • Thomsen EA; Department of Biomedicine, Aarhus University, Aarhus C, Denmark.
  • Mikkelsen JG; Department of Biomedicine, Aarhus University, Aarhus C, Denmark.
Nucleic Acids Res ; 51(18): 10059-10074, 2023 Oct 13.
Article en En | MEDLINE | ID: mdl-37678882
ABSTRACT
Implementation of therapeutic in vivo gene editing using CRISPR/Cas relies on potent delivery of gene editing tools. Administration of ribonucleoprotein (RNP) complexes consisting of Cas protein and single guide RNA (sgRNA) offers short-lived editing activity and safety advantages over conventional viral and non-viral gene and RNA delivery approaches. By engineering lentivirus-derived nanoparticles (LVNPs) to facilitate RNP delivery, we demonstrate effective administration of SpCas9 as well as SpCas9-derived base and prime editors (BE/PE) leading to gene editing in recipient cells. Unique Gag/GagPol protein fusion strategies facilitate RNP packaging in LVNPs, and refinement of LVNP stoichiometry supports optimized LVNP yield and incorporation of therapeutic payload. We demonstrate near instantaneous target DNA cleavage and complete RNP turnover within 4 days. As a result, LVNPs provide high on-target DNA cleavage and lower levels of off-target cleavage activity compared to standard RNP nucleofection in cultured cells. LVNPs accommodate BE/sgRNA and PE/epegRNA RNPs leading to base editing with reduced bystander editing and prime editing without detectable indel formation. Notably, in the mouse eye, we provide the first proof-of-concept for LVNP-directed in vivo gene disruption. Our findings establish LVNPs as promising vehicles for delivery of RNPs facilitating donor-free base and prime editing without formation of double-stranded DNA breaks.

Texto completo: 1 Bases de datos: MEDLINE Idioma: En Revista: Nucleic Acids Res Año: 2023 Tipo del documento: Article País de afiliación: Dinamarca

Texto completo: 1 Bases de datos: MEDLINE Idioma: En Revista: Nucleic Acids Res Año: 2023 Tipo del documento: Article País de afiliación: Dinamarca