Your browser doesn't support javascript.
loading
Transient inhibition of 53BP1 increases the frequency of targeted integration in human hematopoietic stem and progenitor cells.
Baik, Ron; Cromer, M Kyle; Glenn, Steve E; Vakulskas, Christopher A; Chmielewski, Kay O; Dudek, Amanda M; Feist, William N; Klermund, Julia; Shipp, Suzette; Cathomen, Toni; Dever, Daniel P; Porteus, Matthew H.
Afiliação
  • Baik R; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Lorry I. Lokey Stem Cell Research Building, 265 Campus Drive, Stanford, CA, USA.
  • Cromer MK; Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA.
  • Glenn SE; Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
  • Vakulskas CA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Lorry I. Lokey Stem Cell Research Building, 265 Campus Drive, Stanford, CA, USA.
  • Chmielewski KO; Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA.
  • Dudek AM; Integrated DNA Technologies, Inc., Coralville, IA, USA.
  • Feist WN; Integrated DNA Technologies, Inc., Coralville, IA, USA.
  • Klermund J; Institute for Transfusion Medicine and Gene Therapy, Medical Center - University of Freiburg, 79106, Freiburg, Germany.
  • Shipp S; Center for Chronic Immunodeficiency, University of Freiburg, 79106, Freiburg, Germany.
  • Cathomen T; Ph.D. Program, Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany.
  • Dever DP; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Lorry I. Lokey Stem Cell Research Building, 265 Campus Drive, Stanford, CA, USA.
  • Porteus MH; Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA.
Nat Commun ; 15(1): 111, 2024 01 02.
Article em En | MEDLINE | ID: mdl-38169468
ABSTRACT
Genome editing by homology directed repair (HDR) is leveraged to precisely modify the genome of therapeutically relevant hematopoietic stem and progenitor cells (HSPCs). Here, we present a new approach to increasing the frequency of HDR in human HSPCs by the delivery of an inhibitor of 53BP1 (named "i53") as a recombinant peptide. We show that the use of i53 peptide effectively increases the frequency of HDR-mediated genome editing at a variety of therapeutically relevant loci in HSPCs as well as other primary human cell types. We show that incorporating the use of i53 recombinant protein allows high frequencies of HDR while lowering the amounts of AAV6 needed by 8-fold. HDR edited HSPCs were capable of long-term and bi-lineage hematopoietic reconstitution in NSG mice, suggesting that i53 recombinant protein might be safely integrated into the standard CRISPR/AAV6-mediated genome editing protocol to gain greater numbers of edited cells for transplantation of clinically meaningful cell populations.
Assuntos

Texto completo: 1 Coleções: 01-internacional Temas: Geral Base de dados: MEDLINE Assunto principal: Transplante de Células-Tronco Hematopoéticas / Edição de Genes Tipo de estudo: Guideline Limite: Animals / Humans Idioma: En Revista: Nat Commun Assunto da revista: BIOLOGIA / CIENCIA Ano de publicação: 2024 Tipo de documento: Article País de afiliação: Estados Unidos

Texto completo: 1 Coleções: 01-internacional Temas: Geral Base de dados: MEDLINE Assunto principal: Transplante de Células-Tronco Hematopoéticas / Edição de Genes Tipo de estudo: Guideline Limite: Animals / Humans Idioma: En Revista: Nat Commun Assunto da revista: BIOLOGIA / CIENCIA Ano de publicação: 2024 Tipo de documento: Article País de afiliação: Estados Unidos