Abstract
CRISPR/Cas9 genome editing has emerged as a promising treatment for genetic diseases like β-thalassemia. Editing γ-globin promoters to disrupt ZBTB7A/LRF or BCL11A binding sites has shown potential for reactivating fetal hemoglobin and treating sickle cell disease. However, its application to β0-thalassemia/HbE disease remains unclear. This study utilized CRISPR/Cas9 to disrupt these sites in mobilized CD34 + hematopoietic stem /progenitor cells from healthy donors and β0-thalassemia/HbE patients. The editing efficiency for the BCL11A site (75–92%) was higher than for the ZBTB7A/LRF site (57–60%). Both disruptions similarly increased fetal hemoglobin production in healthy donors (BCL11A 26.2 ± 1.4%, ZBTB7A/LRF 27.9 ± 1.5%) and β0-thalassemia/HbE cells (BCL11A 62.7 ± 0.9%, ZBTB7A/LRF 64.0 ± 1.6%). Off-target effects were absent in BCL11A-edited cells but observed at low frequencies in ZBTB7A/LRF-edited cells. Neither disruption significantly affected erythroid differentiation. These findings highlight the comparable contributions of ZBTB7A/LRF and BCL11A binding sites to γ-globin reactivation. CRISPR/Cas9 editing of either site may offer a potential therapeutic strategy for β0-thalassemia/HbE disease.
Keywords: CRISPR/Cas9 genome editing, γ-globin promoter, Fetal hemoglobin reactivation, β0-thalassemia/HbE, ZBTB7A/LRF, BCL11A
Subject terms: Diseases, Medical research, Molecular medicine
Introduction
β-Thalassemias, a common genetic disorder caused by a β-globin gene mutation that results in the reduction or loss of production of the β-globin chain, are a serious public health concern worldwide. Clinical severity varies widely, ranging from mild anemia to transfusion dependency, with complications affecting growth and overall development1,2. β0-thalassemia/HbE is a severe form of β-thalassemia, predominantly affecting Southeast Asia populations1. It results from the compound heterozygous inheritance of a β0-thalassemia mutation, which leads to the absence of β-globin production and the hemoglobin E (HbE, HBB: c.79G > A) variant. This variant causes structural hemoglobin abnormalities and reduced β-globin synthesis due to aberrant splicing3. The anemia associated with β0-thalassemia/HbE disease is driven by impaired hemoglobin production and an imbalance between α- and non-α-globin chain synthesis, leading to ineffective erythropoiesis and increased erythroid cell destruction4. At present, one of the curative treatments for transfusion-dependent thalassemia is allogeneic hematopoietic stem cell transplantation. However, the lack of HLA-matched siblings, the risk of post-transplant complications (graft rejection or graft-versus-host disease), and the high cost have restricted its availability for most patients5. Inducing fetal hemoglobin (HbF) production can alleviate the severity of β-thalassemia by ensuring an appropriate balance between α- and non-α-globin chain levels6. The γ-globin promoter contains transcriptional repressors (ZBTB7A/LRF, BCL11A) binding sites necessary for hemoglobin switching7. Hereditary persistence of fetal hemoglobin (HPFH) mutations in γ-globin promoters creates de novo binding sites for erythroid transcriptional activators or disrupts existing transcriptional repressor binding sites, leading to increased HbF production. Point mutations of the γ-globin promoter at − 113A > G, − 175T > C, and − 198T > C are benign changes that lead to HPFH, which generate binding sites for the HbF activators GATA1, TAL1, and KLF1, respectively8–10. In addition, the introduction of HPFH-associated mutations around the − 200-bp cluster (− 195C > G, − 196C > T, − 197C > T, − 201C > T, and − 202C > T/G) and − 115-bp cluster (− 114C > A, − 117G > A, and a 13-bp deletion) upstream from the transcription start site (TSS) was demonstrated to disrupt the binding sites of the two major γ-globin repressors, ZBTB7A/LRF and BCL11A, respectively10. The development of CRISPR/Cas9 technology provides the opportunity to reactivate fetal hemoglobin synthesis and develop a promising approach for the genetic repair of β-hemoglobinopathies. Previous reports have described CRISPR/Cas9-mediated β-globin gene correction via the homology-directed repair (HDR) pathway in K562 and CD34 + hematopoietic stem/progenitor cells (HSPCs) from sickle cell disease (SCD) patients11–13. CRISPR/Cas9 genome editing to create HPFH mutation in CD34 + HSPCs resulted in increased expression of γ-globin genes14,15. A previous report also showed that genetic variation of individual erythroid-lineage-specific enhancers of human BCL11A, known as DNase I hypersensitive sites (DHSs), + 55, + 58, and + 62, was associated with variation in HbF induction16–18. Preclinical studies have also demonstrated the disruption of transcriptional repressor binding sites in the γ-promoter or human BCL11A enhancer by CRISPR/Cas9, which promoted HbF reactivation in cell lines and human erythroid cells derived from patients with SCD and β-thalassemia19–21. Recently, a successful clinical trial showed that CRISPR/Cas9-mediated disruption of γ-globin promoters led to sustained increases in HbF levels and clinical improvement in SCD patients22. Several studies applying CRISPR/Cas9-based genome editing of the ZBTB7A/LRF or BCL11A binding sites in the γ-globin promoter have demonstrated a variety of indel frequencies, and increments of HbF levels in K562, HUDEP-2 and CD34 + HSPCs19–21,23. However, the preclinical profiles of indel-frequency and off-target analysis associated with CRISPR/Cas9-nuclease mediated DNA double-stranded break in β0-thalassemia/HbE, the most common β-thalassemia disease in Southeast Asian countries including Thailand, remain limited. In this present study, we explored the therapeutic potential of using CRISPR/Cas9 to disrupt the binding sites of two well-known transcriptional repressors, ZBTB7A/LRF (-197) and BCL11A (-115) in the γ-globin promoters in erythroblasts derived from CD34 + HSPCs of β0-thalassemia/HbE patients (Fig. 1A).
Fig. 1.
CRISPR/Cas9-mediated double-strand break on a transcriptional repressor binding site in the γ-globin gene promoter in CD34 + HSPCs. (A) The schematic representation of the γ-globin promoter on chromosome 11 illustrates sgRNA targeted on LRF (− 197) and BCL11A (− 115) binding regions. Genome editing at these sites induces indel mutations through non-homologous end joining (NHEJ) and microhomology-mediated end joining (MMEJ) DNA repair pathways. (B) Editing efficiency, represented as the percentage of indels generated by CRISPR/Cas9-mediated genome editing using sg-LRF and sg-BCL11A in erythroblasts, as determined by deep sequencing (n = 3 per group). (C,D) Deep sequencing analysis showing genome editing profiles at the LRF and BCL11A binding sites in the γ-globin promoter in edited erythroblast-derived CD34 + HSPCs. The reference sequence is displayed at the top, with the PAM sequence highlighted in a red box and the predicted cleavage position indicated by a blue dashed line. Each row represents an observed sequence aligned to the reference sequence. Insertions are outlined in red boxes, deletions are shown as dashes, and substitutions are shown in bold text. The frequency of each sequence is displayed as a percentage of the total reads, with the corresponding read count in parentheses. Unedited alleles that perfectly match the reference sequence are marked with a red asterisk. (E,F) Measurements of on-target, indel-mutation and MMEJ frequencies after genome editing in samples from healthy donors and β0-thalassemia/HbE patients (n = 3 per group).
Results
Targeting CRISPR/Cas9 to disrupt ZBTB7A/LRF or BCL11A binding site in γ-globin promoter
To determine the specificity and efficiency of CRISPR/Cas9-mediated disruption of the ZBTB7A/LRF or BCL11A binding site in the γ-globin promoter, mobilized CD34 + HSPCs from healthy donors (n = 3) and β0-thalassemia/HbE (n = 3) were initially electroporated with specific ribonucleoprotein (RNP) containing Cas9 complexed with sgRNA targeting the ZBTB7A/LRF binding site at HBG-197 (sg-LRF) and the BCL11A binding site at HBG-115 (sg-BCL11A). These sgRNA sequences were obtained from previously published and extensively characterized studies in healthy donors and SCD HSPCs19–21. Deep sequencing analysis revealed the distribution of indel frequencies at the targeted genomic regions. The editing efficiencies of sg-LRF and sg-BCL11A were approximately 69.4 ± 7.4% and 84.9 ± 17.1% in healthy donor cells and 68.2 ± 12.2% and 88.5 ± 3.1% in β0 thalassemia/HbE cells, respectively (Fig. 1B). The results showed the indel mutation profiles, ranked by the number of reads in each allele demonstrated in genome editing profiles at the ZBTB7A/LRF binding site (Fig. 1C) and BCL11A binding site (Fig. 1D) in the γ-globin promoter. We summarized the ten most frequently identified reads, demonstrating various indel frequencies at the ZBTB7A/LRF binding site (Fig. 1E) and the BCL11A binding site (Fig. 1F). The 6-bp deletion was most frequently observed at the ZBTB7A/LRF edited binding site in both healthy donors and β0-thalassemia/HbE cells (10.8 ± 1.5% vs. 11.1 ± 0.3%, respectively) (Fig. 1E). Additionally, the 13-bp deletion was the most common indel mutation detected at the BCL11A edited binding site in healthy donors and β0-thalassemia/HbE cells (21.1 ± 0.6% vs. 21.6 ± 2.1%, respectively) (Fig. 1F).
Fetal hemoglobin reactivation by targeted disruption of ZBTB7A/LRF or BCL11A binding sites in γ-globin promoter and erythroid differentiation
Disruption of the ZBTB7A/LRF or BCL11A binding sites of the γ-globin promoter resulted in a significant increase in γ-globin transcripts. In healthy donor cells, the fold changes of γ-globin were 7.5–11.4 for sg-LRF and 6.1–11.2 for sg-BCL11A. The changes in β0-thalassemia/HbE cells were 4.0–5.3 folds for sg-LRF and 2.7–3.2 folds for sg-BCL11A. The α- and β-globin transcripts were present at similar levels upon comparing edited cells and controls (Fig. 2A, B). The gene expression of LRF and BCL11A in both healthy donors and β0-thalassemia/HbE cells (Fig. 2C, D) did not significantly change after CRISPR/Cas9-mediated disruption of the ZBTB7A/LRF and BCL11A binding sites in the γ-globin promoter.
Fig. 2.
Gene expression of globins and transcription factors after disruption of ZBTB7A/LRF or BCL11A binding sites in erythroid cells derived from CD34 + HSPCs. (A,B) Relative quantification of globin gene expression (α-, β- and γ- globin) analyzed by RT-qPCR and normalized to RPS18 (n = 3 per group). (C,D) Expression of transcriptional repressors BCL11A and LRF, normalized to GAPDH (n = 3 per group). Values are represented as means ± SD. ***p < 0.0001, **p < 0.001, *p < 0.05 compared to control.
Hemoglobin analyses by cation exchange-high performance liquid chromatography (HPLC) demonstrated that editing the γ-globin promoters in healthy donor cells at ZBTB7A/LRF and BCL11A binding sites significantly increased fetal hemoglobin levels. Specifically, the HbF induction levels were 27.9 ± 1.5% for sg-LRF and 26.17 ± 1.4% for sg-BCL11A, compared to the basal level of 3.4 ± 0.9%; all p < 0.0001). In β0-thalassemia/HbE, editing the γ-globin promoters using sg-LRF and sg-BCL11A yielded significant increases in HbF levels, reaching up to 64.0 ± 1.6% (p < 0.001) and 62.7 ± 0.9% (p < 0.0001), respectively, compared to the basal level of 27.87 ± 0.9% in controls. These increases in HbF levels led to proportional decreases in HbA levels in healthy donors or HbE levels in β0-thalassemia/HbE erythroid cells (Fig. 3A). The reverse-phase HPLC analysis revealed reactivation of the γ-globin chain. The ratio of γ-globin to α-globin was significantly increased in edited cells compared to control cells in both cell types, as shown in Fig. 3B. The percentage of γG-globin chain and γA-globin chain was increased considerably after genome editing by sg-LRF and sg-BCL11A compared to controls in healthy donors and β0-thalassemia/HbE (Fig. 3C). Remarkably, genome editing in the γ-globin gene promoters predominantly resulted in the induction of the γA-globin chain rather than the γG-globin chain in both cell types, as shown in Fig. 3C.
Fig. 3.
Reactivation of fetal hemoglobin and globin chain analysis after disruption of ZBTB7A/LRF or BCL11A binding site in erythroid cells derived from CD34 + HSPCs. (A) Cation-exchange HPLC shows the reactivation of HbF synthesis on day 14. (B) Reverse phase HPLC shows globin chain analysis and the relative ratio of γ/α-globin chain induction in edited cells. (C) The expression of globin chain (α, β, βE, γG and γA) after genome editing (n = 3 per group). Values are represented as means ± SD. ***p < 0.0001, **p < 0.001, *p < 0.05 compared to control.
We found that disruption of the ZBTB7A/LRF and BCL11A binding sites in the γ-globin promoter did not significantly affect erythroid differentiation in the healthy donors and β0-thalassemia/HbE cells (Fig. 4A, B). This was demonstrated by the comparable erythroid maturation profile observed in the edited cells and control cells. Flow cytometry dot plot and gating strategy are provided in the supplemental Figure S1.
Fig. 4.
Erythroid cell differentiation after disruption of ZBTB7A/LRF or BCL11A binding site in erythroid cells derived from CD34 + HSPCs. (A) Cell morphology of control cells and edited erythroid cells on day 10 and day 14, stained with Giemsa reagent, visualized by light microscope (scale bar = 10 μm). A total of 250–300 cells were counted per donor (n = 3 per group). The stage-specific normoblast distributions are represented in bar graphs. The error bars represent donor-to-donor variation, indicating the variability observed between individual samples. Values are represented as means ± SEM. (B) Erythroid maturation analysis by flow cytometry showing the expression of transferrin receptor (CD71) and glycophorin A (GPA) in gate R1( CD71High GPA+ ) early stage population, R2( CD71Med GPA+ ) transition stage population, R3( CD71Low GPA+ ) late stage population on day 10 and day 14 during erythroid differentiation in healthy donors and β0-thalassemia/HbE patients (n = 3 per group). Values are represented as means ± SD.
Detection of a large deletion in the γ-globin promoter and off-target analysis
As γG- and γA-globin promoters share identical base sequences, a large 4.9-kb deletion spanning the γG-globin gene and the flanking sequences was generated as a result of the γ-globin promoter editing. Gap-PCR demonstrated this deletion in sg-LRF- and sg-BCL11A-edited healthy donors and β0-thalassemia/HbE cells. Semi-quantitative Gap-PCR was used to confirm the 4.9-kb deletion of the γG-globin gene in sg-LRF and sg-BCL11A edited healthy donors and β0-thalassemia/HbE cells (Fig. 5A). The results showed that editing the LRF and BCL11A binding sites yielded two PCR amplicons (full amplicon size = 6,868-bp, amplicon size with the 4.9-kb deletion = 1,865-bp) in both cell types.
Fig. 5.
Detection of 4.9-kb deletion and off-target analysis after disruption of ZBTB7A/LRF or BCL11A binding site in erythroid cells derived from CD34 + HSPCs. (A) Agarose gel electrophoresis showing a large deletion after genome editing. (4.9-kb deletion = 1,865-bp., full amplicon = 6,868-bp). (B) Deep sequencing by NGS to determine the percentage of off-target cleavage sites (indel mutations) in editing cells compared to control cells (n = 3 per group). Values are represented as means ± SEM.
We analyzed the top five previously reported potential off-target (OT1–OT5) cleavage sites for each sgRNA based on prior GUIDE-seq and CIRCLE-seq studies19,20. The numbering of OT1–OT5 in Fig. 5B follows the ranking of off-target activity reported in these studies, with OT1 representing the site with the highest predicted off-target potential and OT5 the lowest. Amplicon deep sequencing revealed that off-targeting was not detected with OT3–OT5 of sg-LRF or with OT1–OT5 of sg-BCL11A at the detection threshold for amplicon deep sequencing of approximately 0.01% to 0.1%. However, indel mutations were detected at low frequencies with OT1-sg-LRF (2.5 ± 0.7% vs. 2.6 ± 1.3%) and OT2-sg-LRF (1.6 ± 0.3% vs. 2.0 ± 1.5%) in the healthy donor and β0-thalassemia/HbE cells, respectively (Fig. 5B).
Discussion
The CRISPR/Cas9 nuclease system constitutes a major technological genome editing advancement. Non-homologous end joining repair, facilitated by single-guide RNA incorporated into the Cas9 nuclease for site-specific cleavage to DNA double-strand break-induced indel mutations, has substantial promise for clinical treatments24–29. One potential application is in cases of compound heterozygosity for the β0-thalassemia/HbE genotype, a major public health problem in Southeast Asia, including Thailand1. Therapeutic approaches aimed at reactivating HbF production in β-thalassemia by techniques leading to the downregulation of transcription factors involved in γ-globin silencing, including BCL11A, ZBTB7A/LRF, LSD1, and KLF1, have been reported30–33. However, reducing the levels of some transcriptional repressors, such as KLF1, might also affect the maturation of erythroid cells34,35.
HPFH mutations involving the transcriptional repressor binding sites in the γ-globin promoter suggested the potential benefit of mimicking HPFH mutation to reactivate HbF expression. Leslie Weber and colleagues demonstrated that disrupting two clusters in the γ-globin promoters, the binding sites of ZBTB7A/LRF (− 195, − 196, − 197) and BCL11A (− 115), significantly increased HbF synthesis in erythroblasts derived from patients with SCD. The ZBTB7A/LRF binding site is in the − 200 region, where the − 197 sgRNA-mediated disruption exhibits the highest genome editing efficiency and HbF production20.
Here, we performed single electroporation with specific sg-LRF or sg-BCL11A in each donor and compared the editing results of individual binding sites performed using the same sample. Specifically, we selected − 197 and − 115 sgRNA for binding site genome editing, with our results showing that the editing efficiency of BCL11A (− 115, approximately 90%) binding site was higher than that for ZBTB7A/LRF (− 197, approximately 70%). These editing efficiencies were the primary contributors to HbF and were consistent with previous reports19,20. The discrepancy in editing efficiency may be attributed to differences in chromatin accessibility and the structural characteristics of the two target loci. The BCL11A binding site is generally more accessible whereas the LRF binding site may be surrounded by more complex regulatory regions or chromatin architecture, which could reduce editing efficiency at this site36. Additionally, the nucleotides in both the PAM-distal and PAM-proximal regions of the sgRNA significantly correlate with on-target editing efficiency. The genomic context, GC content, and secondary structure of the sgRNA are also key factors contributing to cleavage efficiency. Many studies suggest higher editing efficiencies are achieved when the GC content of the sgRNA-targeted region is between 40 and 60%37. The GC-rich of LRF binding site (5-GGTGGTGG-3) may contribute to lower cleavage efficiency compared to the BCL11A binding site (5-TGACCA-3). The predominant indel mutations detected were 13-bp deletion at the − 115 sgRNA target site and 6-bp deletions at the − 197 sgRNA target site, which were mediated by Microhomology-Mediated End Joining (MMEJ) in both healthy donor and β0-thalassemia/HbE cells. The editing efficiency at the LRF binding site was significantly lower than at the BCL11A binding site; however, HbF induction remains comparable. This may be due to some indel mutations at the BCL11A binding site that do not completely abolish BCL11A-mediated repression, allowing residual suppression of HbF expression. Our results are consistent with previous findings33, confirming that ZBTB7A/LRF and BCL11A independently repress γ-globin gene expression.
Due to the extensive homology between the human HBG1 (γA-globin) and HBG2 (γG-globin) promoter, including the recognition site for sg-LRF and sg-BCL11A. CRISPR/Cas9-mediated genome editing at ZBTB7A/LRF or BCL11A binding site on the HBG promoter always carries potential risks to generate large deletions and gene inversions. Previous studies have reported that simultaneous genome editing at the ZBTB7A/LRF and BCL11A binding induces on-target DNA double-strand breaks, leading to approximately 10–30% occurrence of the 4.9-kb deletion containing HBG2 genes (γG-globin)19,20. Consistent with these findings, our study detected a 4.9-kb deletion by Gap-PCR in approximately 40–50% following genome editing of either −115 sgRNA or −197 sgRNA in healthy donors and β0-thalassemia/HbE cells (Fig. 5A). Notably, the 4.9-kb loss resulted in the complete loss of the HBG2 gene; however, HbF expression was still elevated, primarily due to increased expression of the HBG1 gene. This finding is supported by reverse-phase HPLC analysis, which revealed a tremendous increase in γA-globin levels compared to γG-globin in editing cells (Fig. 3C), consistent with observations from previous studies13,19,20. In both healthy donors and β0-thalassemia/HbE patients, sg-BCL11A-edited cells exhibited lower levels of γG-globin and higher levels of γA-globin compared to sg-LRF-edited cells. Despite these differences, the total γ-globin content (combined γG- and γA-globin chains) remained comparable across all edited cells. This suggests that the loss of the HBG1 gene resulted in a compensatory balance between γG- and γA-globin chains, maintaining an equivalent level of HbF induction. Notably, our study showed that an increase in the proportion of HbF relative to total hemoglobin as a result of BCL11A binding site disruption (− 115 sgRNA) was comparable to that observed previously upon disruption of the core erythroid-specific enhancer of the BCL11A (+ 58) gene itself in β0-thalassemia/HbE17. This evidence supports the concept that BCL11A is a major transcriptional repressor of the γ-globin gene in adult erythroid ontogeny32,33. Double CRISPR/Cas9-mediated genome editing, which combines BCL11A enhancer editing with editing of the γ-globin promoter at either − 115 sgRNA or − 197 sgRNA concurrently, has been shown to enhance reactivation of HbF more effectively than single genome editing in both β-thalassemia cells and healthy donors21. However, the extent of off-target effects and genetic rearrangement of double genome editing sites remains unclear compared with that of single genome editing.
Previous studies have demonstrated that genome editing at the + 58 BCL11A erythroid enhancer18,21 and HBG-11521 in β-thalassemia patient-derived cells effectively ameliorates disease phenotypes by restoring α/β-globin chain balance. Their results had been shown to increase the frequency of HbF+ cells, induce HbF expression, enhance erythroid maturation (as indicated by higher enucleation rates), and significantly reduce reactive oxygen species levels compared to untreated cells. However, the erythroid maturation in sg-LRF and sg-BCL11A edited cells in this study was slightly improved (Fig. 3B), as represented by the R3 gating; however, it was not significantly different from that in unedited β0-thalassemia/HbE cells that might be caused by various of intrinsic stress in β0-thalassemia/HbE cells.
Recently, CRISPR/Cas9 genome editing has effectively disrupted γG- and γA-globin promoters in SCD, as demonstrated in preclinical experiments and a clinical study (NCT04443907)22. The clinical trials RUBY (NCT04853576)38 and EdiThal (NCT05444894)39 involved patients with SCD and transfusion-dependent β-thalassemia major, respectively. These clinical trials suggested that the 4.9-kb intergenic deletion commonly emerges upon γ-globin promoter genome editing. In the present study, we employed Gap-PCR as a qualitative method to confirm the presence of this 4.9-kb deletion. We acknowledge that accurate quantification requires methods such as ddPCR or qPCR, which will be considered in future investigations. While these studies observed the presence of a 4.9-kb deletion in edited cells, they reported favorable therapeutic outcomes with no adverse effects attributable to the gene-editing products. These findings suggest that CRISPR/Cas-mediated genome editing targeting the γ-globin promoters with the generation of the 4.9-kb deletion is safe and effective, making it a promising approach for therapeutic applications. Moreover, the adenine or cytosine base editing strategy to create HPFH mutations in the − 200 clusters of the γ-globin promoter also produced the 4.9-kb deletion and provided a range of off-target activity of base editing in SCD-derived erythroblasts40.
Off-target Cas9 nuclease activity continues to be a significant concern in therapeutic applications41,42. Because of this, we used amplicon deep sequencing to determine the indel mutation frequency at potential off-target sites. The candidates for these potential off-target regions were selected based on previous studies that used sgRNA with sequences similar to ours through GUIDE-Seq and CIRCLE-Seq methods19,20. Our study detected a low frequency of indel mutations at off-target sites OT-1 and OT-2 of sg-LRF in both edited healthy donor and β0-thalassemia/HbE cells. These off-target events were located in the intergenic region of IFNG-IL26 on chromosome 12 (OT-1) and the intergenic region of RUN6-1047 on chromosome 3 (OT-2). These findings suggest that the observed off-target effects are likely non-clinically significant; however, further investigation is warranted to confirm their safety profile. Consistent with our results, a previous study reported that the sg-LRF targeting at positions −196 and −197 induced a low frequency of indel mutations at off-target regions containing 1–3 mismatches. Recently, chromosomal aberration analysis using CAST-seq in primary HSPCs and GUIDE-seq in 293 T cells revealed different off-target effects of genome editing targeting the LRF binding site using − 196 and − 197 sgRNAs43. The combination of these techniques offers a valuable approach for monitoring the safety of genome editing in future studies. Notably, no off-target effects were detected with sg-BCL11A at position −115 in SCD20. Furthermore, recent studies have demonstrated that genetic variations can create protospacer adjacent motif (PAM) sequences, potentially altering the off-target effects of CRISPR-Cas9 gene editing44. To ensure the safety of gene editing therapies, future studies should include comprehensive off-target analyses, such as GUIDE-seq and CAST-seq in β0-thalassemia/HbE cells to better assess the impact of genetic diversity on editing outcomes.
In conclusion, we demonstrated that CRISPR/Cas9-mediated genome editing in the ZBTB7A/LRF and BCL11A binding sites in the γ-globin promoter significantly increased HbF production in β0-thalassemia/HbE disease cells. This therapeutic strategy was achieved without affecting cell maturation. Our findings demonstrated that editing erythroblasts derived from CD34 + HSPCs resulted in a particular frequency of off-target by sgRNA targeting the LRF binding site. The obtained preclinical data suggest that this approach offers a promising strategy for the clinical treatment of β0-thalassemia/HbE patients. For future studies, utilizing cell sorting techniques could provide deeper insights into the relationship between specific indel patterns and HbF induction.
Methods
Subjects and sample collection
This study was approved by the Ethics Committee on Human Rights Related to Research Involving Human Subjects, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Thailand (COA. MURA2019/179). All samples were collected after written informed consent was obtained from the patients and/or their legal guardians. All methods were performed in accordance with the relevant guidelines and regulations. Granulocyte colony-stimulating factor-mobilized peripheral blood progenitor cells were collected by leukapheresis from healthy donors (n = 3) and β0-thalassemia/HbE patients (n = 3) at the stem cell collection unit, Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Thailand.
CD34 + HSPCs isolation and gamma genome editing
CD34 + HSPCs were separated from mononuclear cells using a CD34 + positive purification kit with magnetic microbead separation and LS-MACS columns (Miltenyi Biotech, Bergisch Gladbach, Germany), as described in the manufacturer’s protocol. CD34 + HSPCs were pre-cultured in StemSpan SFEM II (STEMCELL Technologies, Vancouver, BC, Canada) in the presence of 100 ng/ml human stem cell factor (hSCF, 300–07; Peprotech), 100 ng/ml human interleukin-6 (hIL-6, 200–06; Peprotech), 100 ng/ml human thrombopoietin (hTPO, 300–18; Peprotech), 100 ng/ml human Flt3-Ligand (hFlt-3L, 300–19; Peprotech), and 10 ng/ml human interleukin-3 (hIL-3, 200–03; Peprotech), for 48 h before being subjected to RNP transfection. The in-house wild-type 3xNLS spCas-9 recombinant protein, as previously validated and reported, was used in this process17,45. sgRNAs with 2′-O-methyl-3′-phosphorothioate modification at the 5′ and 3′ ends were obtained from Integrated DNA Technologies (Singapore). The sgRNA nucleotide sequences of sg-LRF and sg-BCL11A are shown in Table S1. RNP complexes were assembled at room temperature for 15 min using a 1:3 ratio of Cas9:sgRNA (100 pmol Cas9 and 300 pmol sgRNA). The cultured CD34 + HSPCs (2 × 105 cells) were mixed with RNPs using the P3 Primary Cell 4D-Nucleofector X Kit (Lonza, USA) and electroporated using program DZ100 in 16-well Nucleocuvette Strips. Each sample was individually subjected to single electroporation with specific sg-LRF or sg-BCL11A. Ninety-six hours later, CD34 + electroporated cells were subjected to two-phase in vitro erythroid differentiation.
In vitro erythroid differentiation of CD34 + HSPCs
The basal medium for in vitro erythroid differentiation was composed of Iscove’s Modified Dulbecco’s Medium (Gibco, Grand Island, NY, USA) supplemented with 20% fetal bovine serum (Sigma-Aldrich, St. Louis, MO, USA) and 300 μg/mL holo-transferrin (holo-TF; PromoCell, Heidelberg, Germany). Cells were cultured for 4 days in phase I, in which the basal medium was composed of 10 ng/mL hIL-3, 50 ng/mL hSCF, and 2 units/mL of human recombinant erythropoietin (EPO; CILAG GmbH, Zug, Switzerland). After 4 days, suspended cells were collected and re-seeded in phase II medium consisting of basal medium with 5 units/mL EPO. The culture was maintained under an atmosphere of 5% CO2 at 37 °C for 10 days in a phase II medium.
Indel mutation analysis
Genomic DNA from erythroblasts derived from CD34 + HSPCs was isolated using a Genomic DNA Mini Kit (Geneaid, Taipei, Taiwan). The targeted editing site of the γ-globin promoter was amplified using DreamTaq Green PCR master (Thermo Fisher Scientific, MA, USA) mix using the following cycling conditions: 95 °C for 3 min; 35 cycles of 95 °C for 30 s, 63 °C for 30 s, and 72 °C for 60 s; and 72 °C for 10 min with a specific primer located on the γ-globin promoter. The relevant primer sequences are shown in TableS2. The purified PCR products were subjected to Sanger sequencing. The sequencing data were imported into the web tool ICE to analyze the frequency of indel mutations.
Analysis of 4.9-kb deletion of the γ-globin promoter
The detection of the region encompassing the 4.9-kb deletion at the γ-globin gene promoter was analyzed using gap-PCR. Genomic DNA was amplified using Hercules II Fusion DNA Polymerase (Agilent Technologies, CA, USA) using the following cycling conditions: 95 °C for 2 min; 25 cycles of 95 °C for 10 s, 60 °C for 20 s, and 72 °C for 3.5 min; and finally, 72 °C for 5 min, with the specific primers shown in Table S3. The full-amplicon and large-deletion PCR products were 6.7-kb and 1.8-kb in size, as determined by agarose gel electrophoresis.
RNA isolation and reverse-transcription quantitative PCR (RT-qPCR)
In accordance with the manufacturer’s instructions, total RNA was extracted from erythroid cultures (approximately 2 × 106 cells) using TRIzol Reagent (Thermo Fisher Scientific, MA, USA). cDNA was synthesized by a reverse-transcription reaction using the RevertAid First Strand cDNA synthesis kit (Thermo Fisher Scientific), following the manufacturer’s protocol. RT-qPCR was performed in duplicate with the specific primers (Table S4) using FastStart Essential DNA Green Master Mix (Roche Diagnostics, CA, USA) and analyzed using a LightCycler 96 System (Roche Molecular Systems). All globin gene expression levels were normalized to ribosomal protein S18 (RPS18), whereas the expression levels of transcription factors were normalized to GAPDH. The 2−∆∆Ct method46 was used to analyze the relative fold change.
Erythroid differentiation analysis
Erythroid cell differentiation was monitored via the analysis of cell surface markers using flow cytometry on a FACSVerse flow cytometer (BD Biosciences, San Jose, CA, USA), in which cells were immunostained with allophycocyanin-conjugated anti-transferrin receptor (CD71-APC) (BD Biosciences) and fluorescein isothiocyanate-conjugated anti-glycophorin A (GPA-FITC) (BioLegend, CA, USA) antibodies. In addition, cell maturation was monitored using Giemsa-stained cytospin preparations. Cell morphology was observed under a light microscope.
Quantification of fetal hemoglobin
Hemolysates were prepared from at least 1 × 106 cultured cells on day 14 in VAR-β-THAL Elution buffer 1 (Bio-Rad, CA, USA) and used for high-performance liquid chromatography (HPLC) for hemoglobin type analysis using a Bio-Rad VARIANT II Hemoglobin Testing System with the β-Thalassemia Short Program (Bio-Rad), following the manufacturer’s recommendations.
Globin chain analysis by reverse-phase high-performance liquid chromatography
Erythroid cells were lysed in deionized water and subjected to two freeze–thaw cycles. A clear cell lysate was separated by centrifugation at 14,000×g for 10 min at 4 °C, and the supernatant was transferred into an HPLC micro vial. Analyses were performed on a Waters HPLC Alliance e2695 (Waters Corporation, MA, USA) separation module and detector. The stationary phase was collected on an Aeris 3.6-µm WIDEPORE-C4 200 Å column behind a SecurityGuard UHPLC Wide-pore C18; 4.6 mm guard column (Phenomenex, CA, USA). The globin chain separation was performed as previously described47. Empower 3 chromatography software was used for data acquisition and analysis.
Analysis of on- and off-target of sgRNA by amplicon deep sequencing
The off-target sg-LRF and sg-BCL11A cleavage sites were predicted by GUIDE-Seq and Circle-Seq, as previously described19,20. The top 5 off-target sequences were selected for examination in genome-edited erythroblasts from healthy donors and β0-thalassemia/HbE patients. Briefly, genomic DNA of genome-edited erythroblasts derived from CD34 + HSPCs was then subjected to amplification of the selected potential on- and off-target sgRNA cleavage sites using locus-specific primers (Table S5). Two-step PCR amplification with the proofreading enzyme Herculase II Fusion DNA Polymerase (Agilent Technologies) was performed for library preparation. First-PCR reactions used locus-specific primers overhanging the Illumina Nextera handle sequence. PCR thermal cycling was performed as follows: 95 °C for 2 min; 15 cycles of 95 °C for 10 s, 60–63 °C for 20 s, and 72 °C for 20 s; and final extension 72 °C for 5 min. Purified first-PCR amplicons were used as a template, and a unique Illumina Nextera index (Index i5 vs. i7 barcodes) was used for the second PCR reaction. PCR thermal cycling was performed as follows: 95 °C for 2 min; 15 cycles of 95 °C for 10 s, 60 °C for 20 s, and 72 °C for 20 s; and final extension 72 °C for 5 min. The PCR products were purified with AMPure XP beads (Beckman Coulter) and quantified using Qubit™ dsDNA Quantification Assay Kit (Thermo Fisher Scientific). Library quantification, normalization, and pooling were performed as described in the 16S metagenomic sequencing library guide (https://support.illumina.com/downloads/16s_metagenomic_sequencing_library_preparation.html). The Illumina MiSeq platform performed 2 × 250 paired-end sequencing, generating approximately 100,000 paired-end reads per amplicon. Sequencing data were analyzed using CRISPResso2 software version 2.2.14 with the following specific parameters48. A minimum alignment identity of 75% was applied to include high-confidence alignments. The quantification window size was set to ± 2-bp around the predicted cleavage site to identify insertion and deletion events. Reads with a mean PHRED quality score below 30 were excluded from the analysis to maintain data quality.
Statistical analysis
All statistical analyses were performed using unpaired Student’s t-test by Prism 8 version 8.4.3 (GraphPad Software, San Diego, CA, USA). Results are presented as means ± SD, and differences were considered significant when the p-value was less than 0.05.
Supplementary Information
Acknowledgements
We sincerely thank the Mahidol University Frontier Research Facility (MU-FRF) for supporting MiSeq sequencing.
Author contributions
C.W.: Investigation, Validation, Formal analysis, Writing – original draft, Writing – review & editing. P.I.: Investigation. C.S.: Resources. N.J., A. Tub., W.C.: Supervision, Writing – review & editing. P.K., J.P.: Data curation, Formal analysis. D.S., U.A., S.H.: Resources, Supervision. D.S.: Conceptualization, Methodology, Writing – review & editing. A. Tang: Conceptualization, Methodology, Investigation, Validation, Writing – original draft, Writing – review & editing, Supervision. All authors have read and approved the final version of the manuscript.
Funding
This research is supported by Faculty of Medicine Ramathibodi Hospital, Mahidol University, Thailand (Grant ID: RF_64041) and Mahidol University (Grant ID: MRC-IM 02/2565; Basic Research Fund: the fiscal year 2022 Grant ID: BRF1-016/2565).
Data availability
The amplicon deep sequencing datasets generated in this study for both on-target and off-target sgRNA sequences have been deposited in the NCBI Sequence Read Archive (SRA) under accession number PRJNA1211571 (https://www.ncbi.nlm.nih.gov/sra/PRJNA1211571). Additional data or materials supporting this study’s findings are available from the corresponding author upon request.
Declarations
Competing interests
The authors declare no competing interests.
Ethics declarations
This study was performed after obtaining institutional ethical approval (COA.MURA2019/179) from the Ethics Committee on Human Rights Related to Research Involving Human Subjects, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Thailand.
Footnotes
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary Information
The online version contains supplementary material available at 10.1038/s41598-025-10791-8.
References
- 1.Fucharoen, S. & Weatherall, D. J. The hemoglobin E thalassemias. Cold Spring Harb. Perspect. Med.2, a011734. 10.1101/cshperspect.a011734 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Sripichai, O. et al. A scoring system for the classification of beta-thalassemia/Hb E disease severity. Am. J. Hematol.83, 482–484. 10.1002/ajh.21130 (2008). [DOI] [PubMed] [Google Scholar]
- 3.Orkin, S. H. et al. Linkage of beta-thalassaemia mutations and beta-globin gene polymorphisms with DNA polymorphisms in human beta-globin gene cluster. Nature10.1038/296627a0 (1982). [DOI] [PubMed] [Google Scholar]
- 4.Schrier, S. L. Pathophysiology of thalassemia. Curr. Opin. Hematol.9, 123–126. 10.1097/00062752-200203000-00007 (2002). [DOI] [PubMed] [Google Scholar]
- 5.Origa, R. β-thalassemia. Genet. Med.19, 609–619. 10.1038/gim.2016.173 (2017). [DOI] [PubMed] [Google Scholar]
- 6.Razak, S. A. A. et al. Genetic modifiers of fetal haemoglobin (HbF) and phenotypic severity in β-thalassemia patients. Curr. Mol. Med.18, 295–305. 10.2174/1566524018666181004121604 (2018). [DOI] [PubMed] [Google Scholar]
- 7.Hariharan, P. & Nadkarni, A. Insight of fetal to adult hemoglobin switch: genetic modulators and therapeutic targets. Blood Rev.49, 100823. 10.1016/j.blre.2021.100823 (2021). [DOI] [PubMed] [Google Scholar]
- 8.Wienert, B. et al. Editing the genome to introduce a beneficial naturally occurring mutation associated with increased fetal globin. Nat. Commun.6, 7085. 10.1038/ncomms8085 (2015). [DOI] [PubMed] [Google Scholar]
- 9.Martyn, G. E. et al. A natural regulatory mutation in the proximal promoter elevates fetal globin expression by creating a de novo GATA1 site. Blood133, 852–856. 10.1182/blood-2018-07-863951 (2019). [DOI] [PubMed] [Google Scholar]
- 10.Wienert, B. et al. KLF1 drives the expression of fetal hemoglobin in British HPFH. Blood130, 803–807. 10.1182/blood-2017-02-767400 (2017). [DOI] [PubMed] [Google Scholar]
- 11.Hoban, M. D. et al. CRISPR/Cas9-mediated correction of the sickle mutation in human CD34+ cells. Mol. Ther.24, 1561–1569. 10.1038/mt.2016.148 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Cottle, R. N., Lee, C. M., Archer, D. & Bao, G. Controlled delivery of β-globin-targeting TALENs and CRISPR/Cas9 into mammalian cells for genome editing using microinjection. Sci. Rep.5, 16031. 10.1038/srep16031 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Park, S. H. et al. Highly efficient editing of the β-globin gene in patient-derived hematopoietic stem and progenitor cells to treat sickle cell disease. Nucleic Acids Res.47, 7955–7972. 10.1093/nar/gkz475 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Martyn, G. E. et al. Natural regulatory mutations elevate the fetal globin gene via disruption of BCL11A or ZBTB7A binding. Nat. Genet.50, 498–503. 10.1038/s41588-018-0085-0 (2018). [DOI] [PubMed] [Google Scholar]
- 15.Ye, L. et al. Genome editing using CRISPR-Cas9 to create the HPFH genotype in HSPCs: An approach for treating sickle cell disease and β-thalassemia. Proc. Natl. Acad. Sci. U. S. A.113, 10661–10665. 10.1073/pnas.1612075113 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Canver, M. C. et al. BCL11A enhancer dissection by Cas9-mediated in situ saturating mutagenesis. Nature527, 192–197. 10.1038/nature15521 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Wu, Y. et al. Highly efficient therapeutic gene editing of human hematopoietic stem cells. Nat. Med.25, 776–783. 10.1038/s41591-019-0401-y (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Psatha, N. et al. Disruption of the BCL11A erythroid enhancer reactivates fetal hemoglobin in erythroid cells of patients with β-thalassemia major. Mol. Ther. Methods Clin. Dev.10, 313–326. 10.1016/j.omtm.2018.08.003 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Métais, J. Y. et al. Genome editing of HBG1 and HBG2 to induce fetal hemoglobin. Blood Adv.3, 3379–3392. 10.1182/bloodadvances.2019000820 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Weber, L. et al. Editing a γ-globin repressor binding site restores fetal hemoglobin synthesis and corrects the sickle cell disease phenotype. Sci Adv.6, eaay9392. 10.1126/sciadv.aay9392 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Psatha, N. et al. Enhanced HbF reactivation by multiplex mutagenesis of thalassemic CD34+ cells in vitro and in vivo. Blood138, 1540–1553. 10.1182/blood.2020010020 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Sharma, A. et al. CRISPR-Cas9 editing of the HBG1 and HBG2 promoters to treat sickle cell disease. N. Engl. J. Med.389, 820–832. 10.1056/NEJMoa2215643 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Han, Y. et al. CRISPR/Cas9-based multiplex genome editing of BCL11A and HBG efficiently induces fetal hemoglobin expression. Eur. J. Pharmacol.918, 174788. 10.1016/j.ejphar.2022.174788 (2022). [DOI] [PubMed] [Google Scholar]
- 24.Laurent, M. et al. CRISPR-based gene therapies: from preclinical to clinical treatments. Cells13, 800. 10.3390/cells13100800 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Li, T. et al. CRISPR/Cas9 therapeutics: progress and prospects. Signal Transduct. Target Ther.8, 36. 10.1038/s41392-023-01309-7 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Dimitrievska, M. et al. Revolutionising healing: Gene editing’s breakthrough against sickle cell disease. Blood Rev.65, 101185. 10.1016/j.blre.2024.101185 (2024). [DOI] [PubMed] [Google Scholar]
- 27.Hardouin, G. et al. Sickle cell disease: From genetics to curative approaches. Annu. Rev. Genom. Hum. Genet.24, 255–275. 10.1146/annurev-genom-120122-081037 (2023). [DOI] [PubMed] [Google Scholar]
- 28.Ma, L. et al. CRISPR/Cas9-based gene-editing technology for sickle cell disease. Gene874, 147480. 10.1016/j.gene.2023.147480 (2023). [DOI] [PubMed] [Google Scholar]
- 29.Zarghamian, P., Klermund, J. & Cathomen, T. Clinical genome editing to treat sickle cell disease-A brief update. Front. Med.9, 1065377. 10.3389/fmed.2022.1065377 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Xu, J. et al. Corepressor-dependent silencing of fetal hemoglobin expression by BCL11A. Proc. Natl. Acad. Sci. U. S. A.110, 6518–6523. 10.1073/pnas.1303976110 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Kerenyi, M. A. et al. Histone demethylase Lsd1 represses hematopoietic stem and progenitor cell signatures during blood cell maturation. Elife2, e00633. 10.7554/eLife.00633 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Dai, Y. et al. Therapeutic fetal-globin inducers reduce transcriptional repression in hemoglobinopathy erythroid progenitors through distinct mechanisms. Blood Cells Mol. Dis.56, 62–69. 10.1016/j.bcmd.2015.10.004 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Masuda, T. et al. Transcription factors LRF and BCL11A independently repress expression of fetal hemoglobin. Science351, 285–289. 10.1126/science.aad3312 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Gnanapragasam, M. N. et al. EKLF/KLF1-regulated cell cycle exit is essential for erythroblast enucleation. Blood12, 1631–1641. 10.1182/blood-2016-03-706671 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Khamphikham, P. et al. Genetic variation of Krüppel-like factor 1 (KLF1) and fetal hemoglobin (HbF) levels in β0-thalassemia/HbE disease. Int. J. Hematol.3, 297–310. 10.1007/s12185-017-2357-3 (2018). [DOI] [PubMed] [Google Scholar]
- 36.Jung, W. J. et al. Factors affecting the cleavage efficiency of the CRISPR-Cas9 system. Anim. Cells Syst.28, 75–83. 10.1080/19768354.2024.2322054 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Liu, X. et al. Sequence features associated with the cleavage efficiency of CRISPR/Cas9 system. Sci. Rep.6, 19675. 10.1038/srep19675 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Hanna, R. et al. S264: Edit-301 shows promising preliminary safety and efficacy results in the phase I/II clinical trial (Ruby) of patients with severe sickle cell disease using highly specific and efficient asCas12a enzyme. HemaSphere10.1097/01.HS9.0000967968.05170.e0 (2023). [Google Scholar]
- 39.Frangoul, H. et al. AsCas12a gene editing of HBG1/2 promoters with Edit-301 results in early and sustained normalization of hemoglobin and increased fetal hemoglobin in patients with severe sickle cell disease and transfusion-dependent beta-thalassemia. Transplant. Cell Ther.30, S238–S239. 10.1016/j.jtct.2023.12.312 (2024). [Google Scholar]
- 40.Antoniou, P. et al. Base-editing-mediated dissection of a γ-globin cis-regulatory element for the therapeutic reactivation of fetal hemoglobin expression. Nat. Commun.13, 6618. 10.1038/s41467-022-34493-1 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Guo, C. et al. Off-target effects in CRISPR/Cas9 gene editing. Front. Bioeng. Biotechnol.11, 1143157. 10.3389/fbioe.2023.1143157 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Khoshandam, M. et al. Clinical applications of the CRISPR/Cas9 genome-editing system: Delivery options and challenges in precision medicine. Genes Dis.11, 268–282. 10.1016/j.gendis.2023.02.027 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Frati, G. et al. Safety and efficacy studies of CRISPR-Cas9 treatment of sickle cell disease highlights disease-specific responses. Mol. Ther.32, 4337–4352. 10.1016/j.ymthe.2024.07.015 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Cancellieri, S. et al. Human genetic diversity alters off-target outcomes of therapeutic gene editing. Nat. Genet.55, 34–43. 10.1038/s41588-022-01257-y (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Singpant, P. et al. Recombinant Cas9 protein production in an endotoxin-free system and evaluation with editing the BCL11A gene in human cells. Protein Expr. Purif.210, 106313. 10.1016/j.pep.2023.106313 (2023). [DOI] [PubMed] [Google Scholar]
- 46.Livak, K. J. & Schmittgen, T. D. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-delta delta C(T)) METHOD. Methods25, 402–408. 10.1006/meth.2001.1262 (2001). [DOI] [PubMed] [Google Scholar]
- 47.Wongborisuth, C. et al. Down-regulation of the transcriptional repressor ZNF802 (JAZF1) reactivates fetal hemoglobin in β0-thalassemia/HbE. Sci. Rep.12, 4952. 10.1038/s41598-022-08920-8 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Clement, K. et al. CRISPResso2 provides accurate and rapid genome editing sequence analysis. Nat. Biotechnol.37, 224–226. 10.1038/s41587-019-0032-3 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The amplicon deep sequencing datasets generated in this study for both on-target and off-target sgRNA sequences have been deposited in the NCBI Sequence Read Archive (SRA) under accession number PRJNA1211571 (https://www.ncbi.nlm.nih.gov/sra/PRJNA1211571). Additional data or materials supporting this study’s findings are available from the corresponding author upon request.







