12
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Ex vivo prime editing of patient haematopoietic stem cells rescues sickle-cell disease phenotypes after engraftment in mice

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Sickle-cell disease (SCD) is caused by an A·T-to-T·A transversion mutation in the β-globin gene ( HBB). Here we show that prime editing can correct the SCD allele ( HBB S) to wild type ( HBB A) at frequencies of 15%–41% in haematopoietic stem and progenitor cells (HSPCs) from patients with SCD. Seventeen weeks after transplantation into immunodeficient mice, prime-edited SCD HSPCs maintained HBB A levels and displayed engraftment frequencies, haematopoietic differentiation and lineage maturation similar to those of unedited HSPCs from healthy donors. An average of 42% of human erythroblasts and reticulocytes isolated 17 weeks after transplantation of prime-edited HSPCs from four SCD patient donors expressed HBB A, exceeding the levels predicted for therapeutic benefit. HSPC-derived erythrocytes carried less sickle haemoglobin, contained HBB A -derived adult haemoglobin at 28%–43% of normal levels and resisted hypoxia-induced sickling. Minimal off-target editing was detected at over 100 sites nominated experimentally via unbiased genome-wide analysis. Our findings support the feasibility of a one-time prime editing SCD treatment that corrects HBB S to HBB A, does not require any viral or non-viral DNA template and minimizes undesired consequences of DNA double-strand breaks.

          Abstract

          Prime editing can efficiently correct the sickle-cell allele to produce wild-type haemoglobin in patient haematopoietic stem cells that engraft efficiently in mice, yielding erythrocytes resistant to hypoxia-induced sickling.

          Related collections

          Most cited references64

          • Record: found
          • Abstract: found
          • Article: not found

          Search-and-replace genome editing without double-strand breaks or donor DNA

          Summary Most genetic variants that contribute to disease 1 are challenging to correct efficiently and without excess byproducts 2–5 . Here we describe prime editing, a versatile and precise genome editing method that directly writes new genetic information into a specified DNA site using a catalytically impaired Cas9 fused to an engineered reverse transcriptase, programmed with a prime editing guide RNA (pegRNA) that both specifies the target site and encodes the desired edit. We performed >175 edits in human cells including targeted insertions, deletions, and all 12 types of point mutations without requiring double-strand breaks or donor DNA templates. We applied prime editing in human cells to correct efficiently and with few byproducts the primary genetic causes of sickle cell disease (requiring a transversion in HBB) and Tay-Sachs disease (requiring a deletion in HEXA), to install a protective transversion in PRNP, and to precisely insert various tags and epitopes into target loci. Four human cell lines and primary post-mitotic mouse cortical neurons support prime editing with varying efficiencies. Prime editing shows higher or similar efficiency and fewer byproducts than homology-directed repair, complementary strengths and weaknesses compared to base editing, and much lower off-target editing than Cas9 nuclease at known Cas9 off-target sites. Prime editing substantially expands the scope and capabilities of genome editing, and in principle can correct up to 89% of known genetic variants associated with human diseases.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Repair of double-strand breaks induced by CRISPR–Cas9 leads to large deletions and complex rearrangements

            CRISPR-Cas9 is poised to become the gene editing tool of choice in clinical contexts. Thus far, exploration of Cas9-induced genetic alterations has been limited to the immediate vicinity of the target site and distal off-target sequences, leading to the conclusion that CRISPR-Cas9 was reasonably specific. Here we report significant on-target mutagenesis, such as large deletions and more complex genomic rearrangements at the targeted sites in mouse embryonic stem cells, mouse hematopoietic progenitors and a human differentiated cell line. Using long-read sequencing and long-range PCR genotyping, we show that DNA breaks introduced by single-guide RNA/Cas9 frequently resolved into deletions extending over many kilobases. Furthermore, lesions distal to the cut site and crossover events were identified. The observed genomic damage in mitotically active cells caused by CRISPR-Cas9 editing may have pathogenic consequences.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              CRISPR-Cas9 Gene Editing for Sickle Cell Disease and β-Thalassemia

              Transfusion-dependent β-thalassemia (TDT) and sickle cell disease (SCD) are severe monogenic diseases with severe and potentially life-threatening manifestations. BCL11A is a transcription factor that represses γ-globin expression and fetal hemoglobin in erythroid cells. We performed electroporation of CD34+ hematopoietic stem and progenitor cells obtained from healthy donors, with CRISPR-Cas9 targeting the BCL11A erythroid-specific enhancer. Approximately 80% of the alleles at this locus were modified, with no evidence of off-target editing. After undergoing myeloablation, two patients - one with TDT and the other with SCD - received autologous CD34+ cells edited with CRISPR-Cas9 targeting the same BCL11A enhancer. More than a year later, both patients had high levels of allelic editing in bone marrow and blood, increases in fetal hemoglobin that were distributed pancellularly, transfusion independence, and (in the patient with SCD) elimination of vaso-occlusive episodes. (Funded by CRISPR Therapeutics and Vertex Pharmaceuticals; ClinicalTrials.gov numbers, NCT03655678 for CLIMB THAL-111 and NCT03745287 for CLIMB SCD-121.).
                Bookmark

                Author and article information

                Contributors
                jonathan.yen@stjude.org
                drliu@fas.harvard.edu
                Journal
                Nat Biomed Eng
                Nat Biomed Eng
                Nature Biomedical Engineering
                Nature Publishing Group UK (London )
                2157-846X
                17 April 2023
                17 April 2023
                2023
                : 7
                : 5
                : 616-628
                Affiliations
                [1 ]GRID grid.66859.34, ISNI 0000 0004 0546 1623, Merkin Institute of Transformative Technologies in Healthcare, , Broad Institute of Harvard and MIT, ; Cambridge, MA USA
                [2 ]GRID grid.38142.3c, ISNI 000000041936754X, Department of Chemistry and Chemical Biology, , Harvard University, ; Cambridge, MA USA
                [3 ]GRID grid.38142.3c, ISNI 000000041936754X, Howard Hughes Medical Institute, , Harvard University, ; Cambridge, MA USA
                [4 ]GRID grid.240871.8, ISNI 0000 0001 0224 711X, Department of Hematology, , St Jude Children’s Research Hospital, ; Memphis, TN USA
                [5 ]GRID grid.94365.3d, ISNI 0000 0001 2297 5165, Molecular and Clinical Hematology Branch, National Heart, Lung, and Blood Institute/National Institute of Diabetes and Digestive and Kidney Diseases, , National Institutes of Health, ; Bethesda, MD USA
                Author information
                http://orcid.org/0000-0003-4421-971X
                http://orcid.org/0000-0001-7869-2615
                http://orcid.org/0000-0002-3684-2950
                http://orcid.org/0000-0002-9505-0363
                http://orcid.org/0000-0003-1648-6335
                http://orcid.org/0000-0002-1402-6549
                http://orcid.org/0000-0002-9314-0971
                http://orcid.org/0000-0002-6734-4858
                http://orcid.org/0000-0002-9432-9450
                http://orcid.org/0000-0002-9943-7557
                Article
                1026
                10.1038/s41551-023-01026-0
                10195679
                37069266
                b670d8cf-60fe-49eb-9f63-4d46896556c1
                © The Author(s) 2023, corrected publication 2023

                Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

                History
                : 3 January 2023
                : 22 March 2023
                Funding
                Funded by: FundRef https://doi.org/10.13039/100000060, U.S. Department of Health & Human Services | NIH | National Institute of Allergy and Infectious Diseases (NIAID);
                Award ID: U01 AI142756
                Award Recipient :
                Funded by: FundRef https://doi.org/10.13039/100000051, U.S. Department of Health & Human Services | NIH | National Human Genome Research Institute (NHGRI);
                Award ID: RM1 HG009490
                Award Recipient :
                Funded by: FundRef https://doi.org/10.13039/100000057, U.S. Department of Health & Human Services | NIH | National Institute of General Medical Sciences (NIGMS);
                Award ID: R35 GM118062
                Award Recipient :
                Funded by: FundRef https://doi.org/10.13039/100000050, U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI);
                Award ID: R01 HL156647
                Award ID: R01 HL136135
                Award Recipient :
                Funded by: FundRef https://doi.org/10.13039/100000865, Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation);
                Funded by: FundRef https://doi.org/10.13039/100000011, Howard Hughes Medical Institute (HHMI);
                Categories
                Article
                Custom metadata
                © Springer Nature Limited 2023

                crispr-cas9 genome editing,sickle cell disease,genetic engineering

                Comments

                Comment on this article