Highly efficient therapeutic gene editing of human hematopoietic stem cells

Re-expression of the paralogous γ-globin genes (HBG1/2) could be a universal strategy to ameliorate the severe β-globin disorders sickle cell disease (SCD) and β-thalassemia by induction of fetal hemoglobin (HbF, α2γ2)1. Previously, we and others have shown that core sequences at the BCL11A erythroid enhancer are required for repression of HbF in adult-stage erythroid cells but are dispensable in non-erythroid cells2–6. CRISPR–Cas9-mediated gene modification has demonstrated variable efficiency, specificity, and persistence in hematopoietic stem cells (HSCs). Here, we demonstrate that Cas9:sgRNA ribonucleoprotein (RNP)-mediated cleavage within a GATA1 binding site at the +58 BCL11A erythroid enhancer results in highly penetrant disruption of this motif, reduction of BCL11A expression, and induction of fetal γ-globin. We optimize conditions for selection-free on-target editing in patient-derived HSCs as a nearly complete reaction lacking detectable genotoxicity or deleterious impact on stem cell function. HSCs preferentially undergo non-homologous compared with microhomology-mediated end joining repair. Erythroid progeny of edited engrafting SCD HSCs express therapeutic levels of HbF and resist sickling, while those from patients with β-thalassemia show restored globin chain balance. Non-homologous end joining repair-based BCL11A enhancer editing approaching complete allelic disruption in HSCs is a practicable therapeutic strategy to produce durable HbF induction.Optimized conditions for ribonucleoprotein delivery of Cas9–sgRNA complexes enables precise and efficient gene editing to restore fetal hemoglobin expression in sickle cell disease patient-derived HSCs

[1]  R. Hardison,et al.  A genome-editing strategy to treat β-hemoglobinopathies that recapitulates a mutation associated with a benign genetic condition , 2016, Nature Medicine.

[2]  Dana Carroll,et al.  Selection-free genome editing of the sickle mutation in human adult hematopoietic stem/progenitor cells , 2016, Science Translational Medicine.

[3]  Matthew C. Canver,et al.  BCL11A enhancer dissection by Cas9-mediated in situ saturating mutagenesis , 2015, Nature.

[4]  A. Eddaoudi,et al.  Flow Cytometric Detection of G0 in Live Cells by Hoechst 33342 and Pyronin Y Staining. , 2018, Methods in molecular biology.

[5]  P. Gregory,et al.  Homology-driven genome editing in hematopoietic stem and progenitor cells using zinc finger nuclease mRNA and AAV6 donors , 2015, Nature Biotechnology.

[6]  Brian E. McIntosh,et al.  Nonirradiated NOD,B6.SCID Il2rγ−/−KitW41/W41 (NBSGW) Mice Support Multilineage Engraftment of Human Hematopoietic Cells , 2015, Stem cell reports.

[7]  S. Rodríguez-Perales,et al.  Therapeutic gene editing in CD34+ hematopoietic progenitors from Fanconi anemia patients , 2017, EMBO molecular medicine.

[8]  Linda Z. Shi,et al.  Microhomology-mediated End Joining and Homologous Recombination share the initial end resection step to repair DNA double-strand breaks in mammalian cells , 2013, Proceedings of the National Academy of Sciences.

[9]  Jie Li,et al.  Global transcriptome analyses of human and murine terminal erythroid differentiation. , 2014, Blood.

[10]  Martha L. Bulyk,et al.  Direct Promoter Repression by BCL11A Controls the Fetal to Adult Hemoglobin Switch , 2018, Cell.

[11]  Shih-Feng Tsai,et al.  Cloning of cDNA for the major DNA-binding protein of the erythroid lineage through expression in mammalian cells , 1989, Nature.

[12]  R. Laskey,et al.  Comparative mutagenesis of nuclear localization signals reveals the importance of neutral and acidic amino acids , 1996, Current Biology.

[13]  M. Warr,et al.  Hematopoietic stem cell quiescence promotes error-prone DNA repair and mutagenesis. , 2010, Cell stem cell.

[14]  Israel Steinfeld,et al.  Chemically modified guide RNAs enhance CRISPR-Cas genome editing in human primary cells , 2015, Nature Biotechnology.

[15]  Jussi Taipale,et al.  CRISPR–Cas9 genome editing induces a p53-mediated DNA damage response , 2018, Nature Medicine.

[16]  David A. Williams,et al.  Successful hematopoietic stem cell mobilization and apheresis collection using plerixafor alone in sickle cell patients. , 2018, Blood advances.

[17]  Matthew C. Canver,et al.  Analyzing CRISPR genome-editing experiments with CRISPResso , 2016, Nature Biotechnology.

[18]  Matthew C. Canver,et al.  CRISPResso: sequencing analysis toolbox for CRISPR genome editing , 2015, bioRxiv.

[19]  M. Trivella,et al.  Blood transfusion for preventing primary and secondary stroke in people with sickle cell disease. , 2017, The Cochrane database of systematic reviews.

[20]  Gang Bao,et al.  A high-fidelity Cas9 mutant delivered as a ribonucleoprotein complex enables efficient gene editing in human haematopoietic stem and progenitor cells , 2018, Nature Medicine.

[21]  Sangsu Bae,et al.  Microhomology-based choice of Cas9 nuclease target sites , 2014, Nature Methods.

[22]  J. Joung,et al.  CIRCLE-seq: a highly sensitive in vitro screen for genome-wide CRISPR-Cas9 nuclease off-targets , 2017, Nature Methods.

[23]  A. Miccio,et al.  Plerixafor enables safe, rapid, efficient mobilization of hematopoietic stem cells in sickle cell disease patients after exchange transfusion , 2018, Haematologica.

[24]  A. Bradley,et al.  Repair of double-strand breaks induced by CRISPR–Cas9 leads to large deletions and complex rearrangements , 2018, Nature Biotechnology.

[25]  Matthew C. Canver,et al.  miRNA-embedded shRNAs for Lineage-specific BCL11A Knockdown and Hemoglobin F Induction. , 2015, Molecular therapy : the journal of the American Society of Gene Therapy.

[26]  Steven Lin,et al.  Enhanced homology-directed human genome engineering by controlled timing of CRISPR/Cas9 delivery , 2014, eLife.

[27]  G. Lettre,et al.  Fetal haemoglobin in sickle-cell disease: from genetic epidemiology to new therapeutic strategies , 2016, The Lancet.

[28]  J. D. Macklis,et al.  Strict in vivo specificity of the Bcl11a erythroid enhancer. , 2016, Blood.

[29]  M. van der Burg,et al.  Targeted Genome Editing in Human Repopulating Hematopoietic Stem Cells , 2014, Nature.

[30]  D. K. Wood,et al.  Deoxygenation Reduces Sickle Cell Blood Flow at Arterial Oxygen Tension. , 2016, Biophysical journal.

[31]  Daesik Kim,et al.  Highly efficient RNA-guided genome editing in human cells via delivery of purified Cas9 ribonucleoproteins , 2014, Genome research.

[32]  C. Beam,et al.  Metabolic and immune effects of immunotherapy with proinsulin peptide in human new-onset type 1 diabetes , 2017, Science Translational Medicine.

[33]  J. Aster,et al.  Validation and Implementation of a Custom Next-Generation Sequencing Clinical Assay for Hematologic Malignancies. , 2016, The Journal of molecular diagnostics : JMD.

[34]  Martin J Aryee,et al.  Corrigendum: CIRCLE-seq: a highly sensitive in vitro screen for genome-wide CRISPR–Cas9 nuclease off-targets , 2018, Nature Methods.

[35]  Margaret A Goodell,et al.  Highly Efficient Genome Editing of Murine and Human Hematopoietic Progenitor Cells by CRISPR/Cas9. , 2016, Cell reports.

[36]  Laura J. Norton,et al.  Natural regulatory mutations elevate the fetal globin gene via disruption of BCL11A or ZBTB7A binding , 2018, Nature Genetics.

[37]  L. Symington,et al.  Microhomology-Mediated End Joining: A Back-up Survival Mechanism or Dedicated Pathway? , 2015, Trends in biochemical sciences.

[38]  Vanessa Taupin,et al.  Human hematopoietic stem/progenitor cells modified by zinc-finger nucleases targeted to CCR5 control HIV-1 in vivo , 2010, Nature Biotechnology.

[39]  Matthew C. Canver,et al.  An Erythroid Enhancer of BCL11A Subject to Genetic Variation Determines Fetal Hemoglobin Level , 2013, Science.

[40]  Lei Zhang,et al.  Correction of the sickle cell disease mutation in human hematopoietic stem/progenitor cells. , 2015, Blood.

[41]  Gregory McAllister,et al.  p53 inhibits CRISPR–Cas9 engineering in human pluripotent stem cells , 2018, Nature Medicine.

[42]  Luca Pinello,et al.  Editing aberrant splice sites efficiently restores β-globin expression in β-thalassemia. , 2019, Blood.

[43]  Ke Zhang,et al.  Mitochondrial fusion but not fission regulates larval growth and synaptic development through steroid hormone production , 2014, eLife.

[44]  Yuri R. Bendaña,et al.  Functional footprinting of regulatory DNA , 2015, Nature Methods.

[45]  X. Yang,et al.  Long-Term Engraftment and Fetal Globin Induction upon BCL11A Gene Editing in Bone-Marrow-Derived CD34+ Hematopoietic Stem and Progenitor Cells , 2017, Molecular therapy. Methods & clinical development.

[46]  Sruthi Mantri,et al.  CRISPR/Cas9 β-globin gene targeting in human haematopoietic stem cells , 2016, Nature.

[47]  Pachai Natarajan,et al.  CRISPR-Cas9 gene repair of hematopoietic stem cells from patients with X-linked chronic granulomatous disease , 2017, Science Translational Medicine.

[48]  M. Sadelain,et al.  Safety and efficacy of plerixafor dose escalation for the mobilization of CD34+ hematopoietic progenitor cells in patients with sickle cell disease: interim results , 2018, Haematologica.