Correction of the sickle cell disease mutation in human hematopoietic stem/progenitor cells - PubMed (original) (raw)

. 2015 Apr 23;125(17):2597-604.

doi: 10.1182/blood-2014-12-615948. Epub 2015 Mar 2.

Gregory J Cost 2, Matthew C Mendel 2, Zulema Romero 1, Michael L Kaufman 1, Alok V Joglekar 1, Michelle Ho 1, Dianne Lumaquin 1, David Gray 1, Georgia R Lill 1, Aaron R Cooper 3, Fabrizia Urbinati 1, Shantha Senadheera 1, Allen Zhu 2, Pei-Qi Liu 2, David E Paschon 2, Lei Zhang 2, Edward J Rebar 2, Andrew Wilber 4, Xiaoyan Wang 5, Philip D Gregory 2, Michael C Holmes 2, Andreas Reik 2, Roger P Hollis 1, Donald B Kohn 6

Affiliations

Correction of the sickle cell disease mutation in human hematopoietic stem/progenitor cells

Megan D Hoban et al. Blood. 2015.

Abstract

Sickle cell disease (SCD) is characterized by a single point mutation in the seventh codon of the β-globin gene. Site-specific correction of the sickle mutation in hematopoietic stem cells would allow for permanent production of normal red blood cells. Using zinc-finger nucleases (ZFNs) designed to flank the sickle mutation, we demonstrate efficient targeted cleavage at the β-globin locus with minimal off-target modification. By co-delivering a homologous donor template (either an integrase-defective lentiviral vector or a DNA oligonucleotide), high levels of gene modification were achieved in CD34(+) hematopoietic stem and progenitor cells. Modified cells maintained their ability to engraft NOD/SCID/IL2rγ(null) mice and to produce cells from multiple lineages, although with a reduction in the modification levels relative to the in vitro samples. Importantly, ZFN-driven gene correction in CD34(+) cells from the bone marrow of patients with SCD resulted in the production of wild-type hemoglobin tetramers.

© 2015 by The American Society of Hematology.

PubMed Disclaimer

Figures

Figure 1

Figure 1

Cleavage and correction at the β-globin locus in CD34+ cells using an IDLV donor. (A) A portion of exon I of β-globin showing the ZFN target site (underlined) atop the start codon (bold) and the location of the sickle mutation (bold, italic). (B) Representative gel showing targeted cleavage at the β-globin locus in CB CD34+ cells. Cells were analyzed 3 days after electroporation with in vitro transcribed mRNA encoding the ZFNs. Mock represents untreated CD34+ cells. Arrows indicate cut bands following PCR amplification and digestion with Surveyor Nuclease. (C) Schematic of site-specific gene correction at the sickle mutation. Details of the donor construct and resulting genomic DNA on cleavage by ZFNs and repair by HDR. Location of sickle mutation and _Hha_I RFLP (asterisk) is indicated. Translated regions of exons to scale, introns, and 5′ untranslated region not to scale. (D) Representative RFLP gel for targeted gene modification of β-globin. CB CD34+ cells were electroporated with in vitro transcribed ZFN mRNA and transduced with donor IDLV. Cells were harvested 4 days after treatment, PCR amplified from outside the donor region, digested with _Hha_I enzyme, and resolved on an agarose gel. Arrow shows cleaved product, indicating incorporation of the RFLP into the genome at the target site. (E) Gene modification percentages in CD34+ cells. CB CD34+ cells were electroporated with in vitro transcribed ZFN mRNA (10 μg/mL) and transduced with donor IDLV (2 × 107 TU/mL). Cells were harvested 3 days after treatment and PCR amplified from outside the donor region, and qPCR was completed with primers designed to specifically detect the incorporation of the silent base change generating the _Hha_I RFLP and normalized to primers binding in exon II of the β-globin locus in the amplicon (n = 4 for all conditions). Error bars, mean ± standard deviation.

Figure 2

Figure 2

Correction at the β-globin locus in CD34+ cells using an oligonucleotide donor. (A) Schematic of oligonucleotide-directed gene modification. The top sequence is the genomic DNA of the β-globin locus with the site of the sickle mutation in bold and italicized. The sequence of the modified locus is shown on the bottom with the inserted bases shown in italics. (B) Gene modification in mPB CD34+ cells with an oligonucleotide donor. PAGE of an _Avr_II-digested PCR amplicon of the β-globin locus. The fragment contains a native _Avr_II site, cleavage of which serves as an internal control for _Avr_II digestion (the lower band on the gel). Arrows indicate _Avr_II cleavage products. (C) Six possible sites of silent mutation in the SBS 33501 ZFN binding site. Sickle mutation italicized in bold, and all possible silent mutation sites are in bold (including those not discussed). (D) Silent mutations increase gene modification at β-globin. mPB CD34+ cells were transfected with ZFNs (30 μg/mL) and the indicated donor oligonucleotide (3 μM). Introduction of the relevant silent mutation was assayed via high-throughput sequencing. White bars indicate gene modification; gray bars indicate indels. (E) Silent mutations block ZFN recleavage. Alleles with indels were examined for evidence of homology-mediated modification. Shown are the percentages of alleles with gene modification that also have evidence of NEHJ-driven indels. (F) Optimization of ZFN concentration and donor type. NHEJ-driven indels (gray bars) and gene modification (white bars) were assayed by high-throughput DNA sequencing. Given the depth of high-throughput DNA sequencing, measurement error is expected to be very low.

Figure 3

Figure 3

Transplantation of ZFN and donor-treated cells into NSG mice. (A) Gene modification rates of bulk transplanted cells treated with ZFN+IDLV and cultured in vitro as determined by qPCR for the RFLP at 7 days after electroporation. Mock cells are untreated (n = 3 independent experiments). (B) Modification at the sickle base evaluated by high-throughput sequencing for ZFN+IDLV-modified CD34+ cells. Results of sequencing of the β-globin locus showing percentage of total aligned reads containing the changed wild-type to sickle base (T), as well as insertions and deletions (indels) at the cut site. Same samples as in A. Changed base, white; indels, gray. (C) CD34+ cells were electroporated with Oligo, ZFN, or ZFN+Oligo and cultured in vitro before transplantation into NSG mice. Modification rates at the sickle base and indels are shows as in B (n = 1 experiment). (D) Engraftment in the peripheral blood of transplanted mice at 5 and 8 weeks after transplant. Human engraftment determined as a percentage of hCD45+ cells out of the total hCD45+ and mCD45+ cells by flow cytometry of cells from mice receiving either mock- or ZFN+IDLV-treated cells. Mock, open diamonds; ZFN+IDLV, closed diamonds. (n = 3 independent experiments; mock, n = 5; ZFN+IDLV, n = 12; unpaired t test). (E) Engraftment in the peripheral blood as in D of cells from mice receiving either Oligo-, ZFN-, or ZFN+Oligo-treated cells. Oligo, circles; ZFN, triangles; ZFN+Oligo, diamonds (Oligo, n = 8; ZFN, n = 7; ZFN+Oligo, n = 9); 1-way analysis of variance. n.s., not significant. *P < .05, **P < .01. Error bars, mean ± standard deviation.

Figure 4

Figure 4

Gene-modified cells persist in NSG mice. (A) Gene modification rates in the bone marrow and spleen of transplanted mice at 16 weeks in cells from mice receiving either mock- or ZFN+IDLV-treated cells. High-throughput sequencing of the β-globin locus showing percentage of total aligned reads containing the modified base at the sickle mutation. Mock, open diamonds; ZFN+IDLV, closed diamonds; unpaired t test. (B) Sequencing of the β-globin locus showing insertions and deletions (indels) at the cut site as a percentage of total aligned reads. Mock, open diamonds; ZFN+IDLV, closed diamonds (n = 3 independent experiments; mock, n = 5; ZFN+IDLV, n = 12); unpaired t test. (C) Modification at the sickle base in cells from mice receiving either Oligo-, ZFN-, or ZFN+Oligo-treated cells as described in A. (D) Sequencing results for indels of cells from mice receiving either Oligo-, ZFN-, or ZFN+Oligo-treated cells as in B. Oligo, circles; ZFN, triangles; ZFN+Oligo, diamonds (Oligo, n = 8; ZFN, n = 7; ZFN+Oligo, n = 9); 1-way analysis of variance. n.s., not significant; asterisk indicates significance: *P < .05, **P < .01. Error bars, mean ± standard deviation. Values of zero cannot be plotted on a log scale but were used to calculate the error bars.

Figure 5

Figure 5

Functional correction of sickle bone marrow CD34+ cells. Bone marrow CD34+ cells from patients with SCD were electroporated with in vitro-transcribed ZFN mRNA and transduced with donor IDLV carrying the WT base at the sickle location and grown under erythroid conditions. (A) Correction at the sickle mutation evaluated by high-throughput sequencing. Results of sequencing of the β-globin locus showing percentage of total aligned reads containing the corrected WT base (A) at the sickle mutation, as well as insertions and deletions (indels) at the cut site. Corrected base, white; indels, gray. (B) HPLC of differentiated erythroid cells at the termination of culture. Cells were pelleted and lysed, and supernatant was analyzed by HPLC. (Left) SCD mock sample. (Right) SCD ZFN+IDLV sample. Shading indicates HbA:WT adult hemoglobin peak. (C) Quantification of the percent of HbA out of the total area under the curve represented by the main peaks. HbA, WT adult hemoglobin; HbF, fetal hemoglobin; HbS, sickle hemoglobin; n.s., not significant (n = 2 independent experiments; mock, n = 3; ZFN only, n = 4; IDLV only, n = 3; ZFN+IDLV, n = 6).

Comment in

Similar articles

Cited by

References

    1. Modell B, Darlison M. Global Epidemiology of Haemoglobin Disorders and Derived Service Indicators. Bull World Health Organ. 2008;86(6):480-487. - PMC - PubMed
    1. Powars DR, Chan LS, Hiti A, Ramicone E, Johnson C. Outcome of sickle cell anemia: a 4-decade observational study of 1056 patients. Medicine (Baltimore) 2005;84(6):363–376. - PubMed
    1. Delea TE, Hagiwara M, Thomas SK, Baladi JF, Phatak PD, Coates TD. Outcomes, utilization, and costs among thalassemia and sickle cell disease patients receiving deferoxamine therapy in the United States. Am J Hematol. 2008;83(4):263–270. - PubMed
    1. Cavazzana-Calvo M, Payen E, Negre O, et al. Transfusion independence and HMGA2 activation after gene therapy of human β-thalassaemia. Nature. 2010;467(7313):318–322. - PMC - PubMed
    1. Cavazza A, Moiani A, Mavilio F. Mechanisms of retroviral integration and mutagenesis. Hum Gene Ther. 2013;24(2):119–131. - PubMed

Publication types

MeSH terms

Substances

Grants and funding

LinkOut - more resources