Your browser doesn't support javascript.
loading
Genome editing of HBG1 and HBG2 to induce fetal hemoglobin.
Métais, Jean-Yves; Doerfler, Phillip A; Mayuranathan, Thiyagaraj; Bauer, Daniel E; Fowler, Stephanie C; Hsieh, Matthew M; Katta, Varun; Keriwala, Sagar; Lazzarotto, Cicera R; Luk, Kevin; Neel, Michael D; Perry, S Scott; Peters, Samuel T; Porter, Shaina N; Ryu, Byoung Y; Sharma, Akshay; Shea, Devlin; Tisdale, John F; Uchida, Naoya; Wolfe, Scot A; Woodard, Kaitly J; Wu, Yuxuan; Yao, Yu; Zeng, Jing; Pruett-Miller, Shondra; Tsai, Shengdar Q; Weiss, Mitchell J.
Afiliação
  • Métais JY; Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN.
  • Doerfler PA; Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN.
  • Mayuranathan T; Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN.
  • Bauer DE; Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA.
  • Fowler SC; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA.
  • Hsieh MM; Department of Pediatrics, Harvard Medical School, Boston, MA.
  • Katta V; Harvard Stem Cell Institute, Cambridge, MA.
  • Keriwala S; Broad Institute of MIT and Harvard, Cambridge, MA.
  • Lazzarotto CR; Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN.
  • Luk K; Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute, Bethesda, MD.
  • Neel MD; Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN.
  • Perry SS; Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN.
  • Peters ST; Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN.
  • Porter SN; Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA; and.
  • Ryu BY; Division of Orthopedics.
  • Sharma A; Flow Cytometry and Cell Sorting Shared Resource.
  • Shea D; Department of Cell and Molecular Biology, and.
  • Tisdale JF; Department of Cell and Molecular Biology, and.
  • Uchida N; Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN.
  • Wolfe SA; Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN.
  • Woodard KJ; Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA.
  • Wu Y; Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute, Bethesda, MD.
  • Yao Y; Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute, Bethesda, MD.
  • Zeng J; Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA; and.
  • Pruett-Miller S; Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN.
  • Tsai SQ; Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA.
  • Weiss MJ; Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN.
Blood Adv ; 3(21): 3379-3392, 2019 11 12.
Article em En | MEDLINE | ID: mdl-31698466
ABSTRACT
Induction of fetal hemoglobin (HbF) via clustered regularly interspaced short palindromic repeats/Cas9-mediated disruption of DNA regulatory elements that repress γ-globin gene (HBG1 and HBG2) expression is a promising therapeutic strategy for sickle cell disease (SCD) and ß-thalassemia, although the optimal technical approaches and limiting toxicities are not yet fully defined. We disrupted an HBG1/HBG2 gene promoter motif that is bound by the transcriptional repressor BCL11A. Electroporation of Cas9 single guide RNA ribonucleoprotein complex into normal and SCD donor CD34+ hematopoietic stem and progenitor cells resulted in high frequencies of on-target mutations and the induction of HbF to potentially therapeutic levels in erythroid progeny generated in vitro and in vivo after transplantation of hematopoietic stem and progenitor cells into nonobese diabetic/severe combined immunodeficiency/Il2rγ-/-/KitW41/W41 immunodeficient mice. On-target editing did not impair CD34+ cell regeneration or differentiation into erythroid, T, B, or myeloid cell lineages at 16 to 17 weeks after xenotransplantation. No off-target mutations were detected by targeted sequencing of candidate sites identified by circularization for in vitro reporting of cleavage effects by sequencing (CIRCLE-seq), an in vitro genome-scale method for detecting Cas9 activity. Engineered Cas9 containing 3 nuclear localization sequences edited human hematopoietic stem and progenitor cells more efficiently and consistently than conventional Cas9 with 2 nuclear localization sequences. Our studies provide novel and essential preclinical evidence supporting the safety, feasibility, and efficacy of a mechanism-based approach to induce HbF for treating hemoglobinopathies.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Hemoglobina Fetal / Gama-Globinas / Edição de Genes Limite: Animals / Humans Idioma: En Revista: Blood Adv Ano de publicação: 2019 Tipo de documento: Article País de afiliação: Tunísia

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Hemoglobina Fetal / Gama-Globinas / Edição de Genes Limite: Animals / Humans Idioma: En Revista: Blood Adv Ano de publicação: 2019 Tipo de documento: Article País de afiliação: Tunísia
...