Your browser doesn't support javascript.
loading
Long-term multilineage engraftment of autologous genome-edited hematopoietic stem cells in nonhuman primates.
Peterson, Christopher W; Wang, Jianbin; Norman, Krystin K; Norgaard, Zachary K; Humbert, Olivier; Tse, Collette K; Yan, Jenny J; Trimble, Richard G; Shivak, David A; Rebar, Edward J; Gregory, Philip D; Holmes, Michael C; Kiem, Hans-Peter.
Afiliação
  • Peterson CW; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA;
  • Wang J; Sangamo BioSciences, Richmond, CA; and.
  • Norman KK; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA;
  • Norgaard ZK; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA;
  • Humbert O; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA;
  • Tse CK; Sangamo BioSciences, Richmond, CA; and.
  • Yan JJ; Sangamo BioSciences, Richmond, CA; and.
  • Trimble RG; Sangamo BioSciences, Richmond, CA; and.
  • Shivak DA; Sangamo BioSciences, Richmond, CA; and.
  • Rebar EJ; Sangamo BioSciences, Richmond, CA; and.
  • Gregory PD; Sangamo BioSciences, Richmond, CA; and.
  • Holmes MC; Sangamo BioSciences, Richmond, CA; and.
  • Kiem HP; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Medicine and Department of Pathology, University of Washington, Seattle, WA.
Blood ; 127(20): 2416-26, 2016 05 19.
Article em En | MEDLINE | ID: mdl-26980728
ABSTRACT
Genome editing in hematopoietic stem and progenitor cells (HSPCs) is a promising novel technology for the treatment of many human diseases. Here, we evaluated whether the disruption of the C-C chemokine receptor 5 (CCR5) locus in pigtailed macaque HSPCs by zinc finger nucleases (ZFNs) was feasible. We show that macaque-specific CCR5 ZFNs efficiently induce CCR5 disruption at levels of up to 64% ex vivo, 40% in vivo early posttransplant, and 3% to 5% in long-term repopulating cells over 6 months following HSPC transplant. These genome-edited HSPCs support multilineage engraftment and generate progeny capable of trafficking to secondary tissues including the gut. Using deep sequencing technology, we show that these ZFNs are highly specific for the CCR5 locus in primary cells. Further, we have adapted our clonal tracking methodology to follow individual CCR5 mutant cells over time in vivo, reinforcing that CCR5 gene-edited HSPCs are capable of long-term engraftment. Together, these data demonstrate that genome-edited HSPCs engraft, and contribute to multilineage repopulation after autologous transplantation in a clinically relevant large animal model, an important step toward the development of stem cell-based genome-editing therapies for HIV and potentially other diseases as well.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Transplante de Medula Óssea / Transplante de Células-Tronco Hematopoéticas / Linhagem da Célula / Receptores CCR5 / Edição de Genes / Macaca nemestrina Limite: Animals Idioma: En Revista: Blood Ano de publicação: 2016 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Transplante de Medula Óssea / Transplante de Células-Tronco Hematopoéticas / Linhagem da Célula / Receptores CCR5 / Edição de Genes / Macaca nemestrina Limite: Animals Idioma: En Revista: Blood Ano de publicação: 2016 Tipo de documento: Article