Your browser doesn't support javascript.
loading
Biallelic correction of sickle cell disease-derived induced pluripotent stem cells (iPSCs) confirmed at the protein level through serum-free iPS-sac/erythroid differentiation.
Haro-Mora, Juan J; Uchida, Naoya; Demirci, Selami; Wang, Qi; Zou, Jizhong; Tisdale, John F.
Afiliação
  • Haro-Mora JJ; Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institutes (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, Maryland.
  • Uchida N; Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institutes (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, Maryland.
  • Demirci S; Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institutes (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, Maryland.
  • Wang Q; iPS Cell Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland.
  • Zou J; iPS Cell Core Facility, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland.
  • Tisdale JF; Cellular and Molecular Therapeutics Branch, National Heart Lung and Blood Institutes (NHLBI)/National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, Maryland.
Stem Cells Transl Med ; 9(5): 590-602, 2020 05.
Article em En | MEDLINE | ID: mdl-32034898
ABSTRACT
New technologies of induced pluripotent stem cells (iPSCs) and genome editing have emerged, allowing for the development of autologous transfusion therapies. We previously demonstrated definitive ß-globin production from human embryonic stem cell (hESC)-derived erythroid cell generation via hemangioblast-like ES-sacs. In this study, we demonstrated normal ß-globin protein production from biallelic corrected sickle cell disease (SCD) iPSCs. We optimized our ES/iPS-sac method for feeder cell-free hESC maintenance followed by serum-free ES-sac generation, which is preferred for electroporation-based genome editing. Surprisingly, the optimized protocol improved yields of ES-sacs (25.9-fold), hematopoietic-like spherical cells (14.8-fold), and erythroid cells (5.8-fold), compared with our standard ES-sac generation. We performed viral vector-free gene correction in SCD iPSCs, resulting in one clone with monoallelic and one clone with biallelic correction, and using this serum-free iPS-sac culture, corrected iPSC-generated erythroid cells with normal ß-globin, confirmed at DNA and protein levels. Our serum-free ES/iPS-sac protocol with gene correction will be useful to develop regenerative transfusion therapies for SCD.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Células Eritroides / Células-Tronco Pluripotentes Induzidas / Edição de Genes / Anemia Falciforme Idioma: En Ano de publicação: 2020 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Células Eritroides / Células-Tronco Pluripotentes Induzidas / Edição de Genes / Anemia Falciforme Idioma: En Ano de publicação: 2020 Tipo de documento: Article