Enhancing prime editing in hematopoietic stem and progenitor cells by modulating nucleotide metabolism

Enhancing prime editing in hematopoietic stem and progenitor cells by modulating nucleotide metabolism

Data availability

All amplicon sequencing data are publicly accessible from the National Center for Biotechnology Information BioProject database under accession number PRJNA1090890.

References

Anzalone, A. V. et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature576, 149–157 (2019).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Chen, P. J. et al. Enhanced prime editing systems by manipulating cellular determinants of editing outcomes. Cell184, 5635–5652 (2021).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Anzalone, A. V. et al. Programmable deletion, replacement, integration and inversion of large DNA sequences with twin prime editing. Nat. Biotechnol.40, 731–740 (2022).

Article 
CAS 
PubMed 

Google Scholar 

Ayinde, D., Casartelli, N. & Schwartz, O. Restricting HIV the SAMHD1 way: through nucleotide starvation. Nat. Rev. Microbiol.10, 675–680 (2012).

Article 
CAS 
PubMed 

Google Scholar 

Ballana, E. & Esté, J. A. SAMHD1: at the crossroads of cell proliferation, immune responses, and virus restriction. Trends Microbiol.23, 680–692 (2015).

Article 
CAS 
PubMed 

Google Scholar 

Mauney, C. H. & Hollis, T. SAMHD1: recurring roles in cell cycle, viral restriction, cancer, and innate immunity. Autoimmunity51, 96–110 (2018).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Laguette, N. et al. SAMHD1 is the dendritic- and myeloid-cell-specific HIV-1 restriction factor counteracted by Vpx. Nature474, 654–657 (2011).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Hrecka, K. et al. Vpx relieves inhibition of HIV-1 infection of macrophages mediated by the SAMHD1 protein. Nature474, 658–661 (2011).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Baldauf, H. M. et al. SAMHD1 restricts HIV-1 infection in resting CD4(+) T cells. Nat. Med.18, 1682–1687 (2012).

Article 
CAS 
PubMed 

Google Scholar 

Li, D. et al. Vpx mediated degradation of SAMHD1 has only a very limited effect on lentiviral transduction rate in ex vivo cultured HSPCs. Stem Cell Res.15, 271–280 (2015).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Levesque, S. et al. Marker-free co-selection for successive rounds of prime editing in human cells. Nat. Commun.13, 5909 (2022).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Mikdar, M. et al. The equilibrative nucleoside transporter ENT1 is critical for nucleotide homeostasis and optimal erythropoiesis. Blood137, 3548–3562 (2021).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Everette, K. A. et al. Ex vivo prime editing of patient haematopoietic stem cells rescues sickle-cell disease phenotypes after engraftment in mice. Nat. Biomed. Eng.7, 616–628 (2023).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Zeng, J. et al. Gene editing without ex vivo culture evades genotoxicity in human hematopoietic stem cells. Preprint at bioRxiv https://doi.org/10.1101/2023.05.27.542323 (2023).

Fiumara, M. et al. Genotoxic effects of base and prime editing in human hematopoietic stem cells. Nat. Biotechnol. https://doi.org/10.1038/s41587-023-01915-4 (2023).

Article 
PubMed 

Google Scholar 

Ferreira da Silva, J. et al. Prime editing efficiency and fidelity are enhanced in the absence of mismatch repair. Nat. Commun.13, 760 (2022).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Chen, P. J. & Liu, D. R. Prime editing for precise and highly versatile genome manipulation. Nat. Rev. Genet.24, 161–177 (2022).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Nambiar, T. S., Baudrier, L., Billon, P. & Ciccia, A. CRISPR-based genome editing through the lens of DNA repair. Mol. Cell82, 348–388 (2022).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Skasko, M. et al. Mechanistic differences in RNA-dependent DNA polymerization and fidelity between murine leukemia virus and HIV-1 reverse transcriptases. J. Biol. Chem.280, 12190–12200 (2005).

Article 
CAS 
PubMed 

Google Scholar 

Sharma, P. L., Nurpeisov, V. & Schinazi, R. F. Retrovirus reverse transcriptases containing a modified YXDD motif. Antivir. Chem. Chemother.16, 169–182 (2005).

Article 
CAS 
PubMed 

Google Scholar 

Palikša, S., Alzbutas, G. & Skirgaila, R. Decreased Km to dNTPs is an essential M-MuLV reverse transcriptase adoption required to perform efficient cDNA synthesis in one-step RT-PCR assay. Protein Eng. Des. Sel.31, 79–89 (2018).

Article 
PubMed 

Google Scholar 

Ponnienselvan, K. et al. Addressing the dNTP bottleneck restricting prime editing activity. Preprint at bioRxiv https://doi.org/10.1101/2023.10.21.563443 (2023).

Li, X. et al. Highly efficient prime editing by introducing same-sense mutations in pegRNA or stabilizing its structure. Nat. Commun.13, 1669 (2022).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Su, S. S., Lahue, R. S., Au, K. G. & Modrich, P. Mispair specificity of methyl-directed DNA mismatch correction in vitro. J. Biol. Chem.263, 6829–6835 (1988).

Article 
CAS 
PubMed 

Google Scholar 

Thomas, D. C., Roberts, J. D. & Kunkel, T. A. Heteroduplex repair in extracts of human HeLa cells. J. Biol. Chem.266, 3744–3751 (1991).

Article 
CAS 
PubMed 

Google Scholar 

Lahue, R., Au, K. & Modrich, P. DNA mismatch correction in a defined system. Science245, 160–164 (1998).

Article 

Google Scholar 

Mathews, C. K. Deoxyribonucleotide metabolism, mutagenesis and cancer. Nat. Rev. Cancer15, 528–539 (2015).

Article 
CAS 
PubMed 

Google Scholar 

Traut, T. W. Physiological concentrations of purines and pyrimidines. Mol. Cell. Biochem.140, 1–22 (1994).

Article 
CAS 
PubMed 

Google Scholar 

Mjelle, R. et al. Cell cycle regulation of human DNA repair and chromatin remodeling genes. DNA Repair.30, 53–67 (2015).

Article 
CAS 
PubMed 

Google Scholar 

Longley, M. J., Pierce, A. J. & Modrich, P. D. N. A polymerase δ is required for human mismatch repair in vitro. J. Biol. Chem.272, 10917–10921 (1997).

Article 
CAS 
PubMed 

Google Scholar 

Domínguez-González, C. et al. Deoxynucleoside therapy for thymidine kinase 2–deficient myopathy. Ann. Neurol.86, 293–303 (2019).

Article 
PubMed 
PubMed Central 

Google Scholar 

Amtmann, D., Gammaitoni, A. R., Galer, B. S., Salem, R. & Jensen, M. P. The impact of TK2 deficiency syndrome and its treatment by nucleoside therapy on quality of life. Mitochondrion68, 1–9 (2023).

Article 
CAS 
PubMed 

Google Scholar 

Li, C. et al. In vivo HSC prime editing rescues sickle cell disease in a mouse model. Blood141, 2085–2099 (2023).

CAS 
PubMed 
PubMed Central 

Google Scholar 

Breda, L. et al. In vivo hematopoietic stem cell modification by mRNA delivery. Science381, 436–443 (2023).

Article 
CAS 
PubMed 

Google Scholar 

An, M. et al. Engineered virus-like particles for transient delivery of prime editor ribonucleoprotein complexes in vivo. Nat. Biotechnol. https://doi.org/10.1038/s41587-023-02078-y (2024).

Article 
PubMed 

Google Scholar 

Liu, B. et al. An efficient lentiviral CRISPRi approach to silence genes in primary human monocytes. Preprint at bioRxiv https://doi.org/10.1101/2020.12.23.424242 (2020).

Casirati, G. et al. Epitope editing enables targeted immunotherapies for acute myeloid leukemia. Nature621, 404–414 (2023).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Brinkman, E. K., Chen, T., Amendola, M. & Van Steensel, B. Easy quantitative assessment of genome editing by sequence trace decomposition. Nucleic Acids Res.42, e168 (2014).

Article 
PubMed 
PubMed Central 

Google Scholar 

Brinkman, E. K. et al. Easy quantification of template-directed CRISPR/Cas9 editing. Nucleic Acids Res.46, e58 (2018).

Article 
PubMed 
PubMed Central 

Google Scholar 

Xu, L., Liu, Y. & Han, R. BEAT: a Python program to quantify base editing from Sanger sequencing. Cris. J.2, 223–229 (2019).

Article 
CAS 

Google Scholar 

Wu, Y. et al. Highly efficient therapeutic gene editing of human hematopoietic stem cells. Nat. Med.25, 776–783 (2019).

Article 
CAS 
PubMed 
PubMed Central 

Google Scholar 

Bloh, K. et al. Deconvolution of complex DNA repair (DECODR): establishing a novel deconvolution algorithm for comprehensive analysis of CRISPR-edited Sanger sequencing data. Cris. J.4, 120–131 (2021).

Article 
CAS 

Google Scholar 

Conant, D. et al. Inference of CRISPR edits from Sanger trace data. Cris. J.5, 123–130 (2022).

Article 
CAS 

Google Scholar 

Clement, K. et al. CRISPResso2 provides accurate and rapid genome editing sequence analysis. Nat. Biotechnol.37, 215–226 (2019).

Article 

Google Scholar 

Download references

Acknowledgements

This work was supported by the Doris Duke Foundation, the St. Jude Children’s Research Hospital Collaborative Research Consortium, the Harvard Stem Cell Institute and the National Institutes of Health (R01HL150669 and R01HL170629). S.L. holds a Banting Postdoctoral Fellowship from the Canadian Institutes of Health Research. A.C. was partially supported by a fellowship from the American Italian Cancer Foundation. CD34+ cells were provided by the Fred Hutch Cooperative Center of Excellence in Hematology (U54 DK106829). We thank D. Klatt and C. Brendel for their support in producing in-house Vpx VLPs, G. Casirati for his support in mRNA IVT, P. Wang and N. Kanarek for performing metabolic studies, W. Mannherz and S. Agarwal for insightful discussions, J. Zeng and N.R. Neri for technical support, and the GPP of the Broad Institute of MIT and Harvard for providing VSV-G envelope and SIV Vpx vectors.

Author information

Authors and Affiliations

Division of Hematology/Oncology, Boston Children’s Hospital, Boston, MA, USA

Sébastien Levesque, Andrea Cosentino, Archana Verma, Pietro Genovese & Daniel E. Bauer

Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA

Sébastien Levesque, Andrea Cosentino, Archana Verma, Pietro Genovese & Daniel E. Bauer

Harvard Stem Cell Institute, Cambridge, MA, USA

Sébastien Levesque, Archana Verma, Pietro Genovese & Daniel E. Bauer

Broad Institute, Cambridge, MA, USA

Sébastien Levesque, Archana Verma, Pietro Genovese & Daniel E. Bauer

Department of Pediatrics, Harvard Medical School, Boston, MA, USA

Sébastien Levesque, Andrea Cosentino, Archana Verma, Pietro Genovese & Daniel E. Bauer

Milano-Bicocca University, Milan, Italy

Andrea Cosentino

Contributions

S.L. and D.E.B. conceived the study; S.L. and D.E.B. devised the methods; S.L., A.C., A.V., P.G. and D.E.B. carried out the investigation; S.L. wrote the original draft; S.L. and D.E.B. wrote, reviewed and edited the manuscript; P.G. and D.E.B. acquired the funding; and P.G. and D.E.B. provided supervision.

Corresponding author

Correspondence to
Daniel E. Bauer.

Ethics declarations

Competing interests

S.L. and D.E.B. have filed a provisional patent application related to this work. All other authors have no competing interests.

Peer review

Peer review information

Nature Biotechnology thanks the anonymous reviewers for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Vpx VLP and dN supplementation enhance prime editing in HSPCs with the first-generation PE2 prime editor.

(a) PE quantification as determined by BEAT analysis from Sanger sequencing. 5 ×105 HSPCs were electroporated with PE3 RNAs (PE2 mRNA + epegRNA + nicking sgRNA) targeting ATP1A1 and treated with 5X Vpx VLP (GPP) or vehicle control. Genomic DNA was harvested 3 days post-nucleofection. n = 1 experiment (donor 1) replicated with HSPCs from donor 2 with similar results (See Extended Data Fig. 1b). (b) PE and indels quantification as determined by BEAT and TIDE analysis from Sanger sequencing. HSPCs were thawed and cultured in the presence or absence of 100 µM dNs for 24 hours. Following dN treatment, 5 ×105 HSPCs were electroporated with PE3 RNAs targeting ATP1A1 and cultured in the presence or absence of 100 µM each dN and the indicated concentration of Vpx VLPs (GPP) or vehicle control for 72 hours. Genomic DNA was harvested 3 days post-nucleofection. n = 1 experiment. (c) PE and indels quantification as determined by BEAT and TIDE analysis from Sanger sequencing. HSPCs were thawed and cultured in the presence or absence of the indicated concentration of each dN for 24 hours (before electroporation). Following dN treatment, 1.25 ×105 HSPCs were electroporated with PE3 RNAs targeting ATP1A1 and cultured in the presence or absence of the indicated concentration of each dN for 72 hours (after electroporation). Genomic DNA was harvested 3 days post-nucleofection. n = 1 experiment. Donor 2, circle. Donor 3, diamond.

Extended Data Fig. 2 Modulation of nucleotide metabolism has minimal impact on prime editing in cancer cell lines.

(a) PE and indels quantification as determined by BEAT and TIDE analysis from Sanger sequencing. K562 cells were electroporated with PE3max vectors targeting ATP1A1 and the indicated Vpx or SAMHD1 vector and cultured with the indicated concentration of each dN for 72 hours. An empty pUC19 vector was used as a negative control to normalize the concentration of DNA in all nucleofections. Genomic DNA was harvested 3 days post-nucleofection. n = 2 independent biological replicates performed at different times. (b) Same as in (a) with Jurkat cells.

Extended Data Fig. 3 Staggered PE3 nicks generate tandem duplications at ATP1A1 in HSPCs.

(a) CRISPResso2 amplicon sequencing allele plots after PE3 at ATP1A1. 2.5 ×105 HSPCs were electroporated with PE3max RNAs targeting ATP1A1 (Q118R_v2, +3 TTG to CCT) and cultured in the presence or absence of 5X Vpx VLPs (GPP) and 50 µM each dN. Genomic DNA was harvested 3 days post-nucleofection. Representative allele plots are from one of three independent biological replicates performed with CD34+ HSPCs from three different donors with equivalent results. Insertions are illustrated in red squares, and they represent tandem duplications of the genomic sequence found between the two nicking sites.

Extended Data Fig. 4 Modulation of nucleotide metabolism modestly improves PE at HBB.

(a) PE and indels quantification as determined by CRISPResso2 analysis from amplicon sequencing. 2.5 ×105 HSPCs were electroporated with PE3/PE3bmax RNAs targeting HBB and cultured in the presence or absence of 5X Vpx VLPs (GPP) and 50 µM each dNs. Genomic DNA was harvested 3 days post-nucleofection. Data are plotted as mean ± SEM from n = 3 independent biological replicates performed with CD34+ HSPCs from one donor homozygous for the rs713040 allele (donor 2). Each biological replicate is illustrated with the same shade of grey. (b) Same as in (a) but HSPCs were electroporated with PE2max or PE4max (PE2max mRNA + hMLH1dn mRNA) RNAs targeting HBB and cultured in the presence or absence of 50 µM each dNs. Where indicated, an equimolar ratio of Vpx mRNA was co-delivered during electroporation. The mRNA molarity was normalized between all conditions with EGFP mRNA. Data are plotted as mean ± SEM from n = 3 independent biological replicates performed with CD34+ HSPCs from three different donors. Donor 2, circle. Donor 6, empty diamond. Donor 7, triangle (down). (c) Representative CRISPResso2 amplicon sequencing allele plots from the experiments shown in (a) and (b). Representative allele plots are from one of three independent biological replicates performed with CD34+ HSPCs from three different donors with equivalent results. Reads with ≥ 0.2% frequency are shown, and the frequency of indels is indicated for each sample.

Extended Data Fig. 5 Modulation of nucleotide metabolism positively interacts with MMR-evading epegRNAs in quiescent HSPCs.

(a) Schematic representation of the RNAs used to co-deliver Vpx with PE3max components. The timeline for prime editing in quiescent CD34+ HSPCs is illustrated on the right. (b) PE and indels quantification as determined by BEAT and TIDE analysis from Sanger sequencing. 5 ×105 quiescent HSPCs were electroporated directly after thawing with Vpx mRNA and PE3max RNAs targeting HBB or B2M and cultured in the presence or absence of 50 µM each dN. Genomic DNA was harvested 3 days post-nucleofection. Data are plotted as mean ± SEM from n = 3 independent biological replicates performed with CD34+ HSPCs from three different donors. (c) Same as in (b) for prime editing at ATP1A1 with MMR-evading substitutions. Donor 2, circle. Donor 6, empty diamond. Donor 7, triangle (down).

Extended Data Fig. 6 Omitting the nicking sgRNA abrogates indels and tandem duplications at ATP1A1 in HSPCs.

(a) CRISPResso2 amplicon sequencing allele plots from Fig. 2a. 2.5 ×105 HSPCs were electroporated with PE2max RNAs targeting ATP1A1 (Q118R_v3, +3 TTGG to CCTA) and cultured in the presence or absence of 25X in-house Vpx VLPs and 50 µM each dN. Genomic DNA was harvested 3 days post-nucleofection. Representative allele plots are from one of three independent biological replicates performed with CD34+ HSPCs from three different donors with equivalent results. Reads with ≥ 0.2% frequency are shown, and the frequency of indels is indicated for each sample.

Extended Data Fig. 7 Comparison of quantification accuracy between Sanger and amplicon sequencing at ATP1A1, B2M, and HBB (related to Fig. 2 and Extended Data Fig. 4).

(a) PE and indel quantification as determined by BEAT and TIDE analysis from Sanger sequencing. (See Fig. 2a for comparison with amplicon sequencing.) The MMR evasion strategy based on the installation of additional silent mutations and the epegRNAs used to target the ATP1A1 locus are illustrated. Point mutations are illustrated in orange. As described in Figs. 2, 2.5 ×105 HSPCs were electroporated with PE2max RNAs targeting ATP1A1 and cultured in the presence or absence of 50 µM each dN and 25X in-house Vpx VLPs. Genomic DNA was harvested 3 days post-nucleofection. Data are from two out of three independent biological replicates performed with both sequencing methods. (b) Same as in (a), but HSPCs were electroporated with Vpx mRNA and PE2max RNAs targeting B2M (See Fig. 2b for comparison with amplicon sequencing). Data are plotted as mean ± SEM from n = 3 independent biological replicates performed with CD34+ HSPCs from three different donors. (c) PE and indel quantification as determined by BEAT and TIDE analysis from Sanger sequencing (See Extended Data Fig. 4a for comparison with amplicon sequencing). As described in Extended Data Fig. 4, 2.5 x 105 HSPCs were electroporated with PE3/PE3bmax RNAs targeting HBB and cultured in the presence or absence of 5X Vpx VLPs (GPP) and 50 µM each dNs. Genomic DNA was harvested 3 days post-nucleofection. Data are plotted as mean ± SEM from n = 3 independent biological replicates performed with CD34+ HSPCs from one donor homozygous for the rs713040 allele (donor 2). Each biological replicate is illustrated with the same shade of grey. (d) Correlation of Sanger and amplicon sequencing quantifications at ATP1A1 (PE2max), B2M (PE2max), and HBB (PE3max and PE3bmax) for the 96 samples shown in (a-c).

Supplementary information

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Cite this article

Levesque, S., Cosentino, A., Verma, A. et al. Enhancing prime editing in hematopoietic stem and progenitor cells by modulating nucleotide metabolism.
Nat Biotechnol (2024). https://doi.org/10.1038/s41587-024-02266-4

Download citation

Received: 16 October 2023

Accepted: 26 April 2024

Published: 28 May 2024

DOI: https://doi.org/10.1038/s41587-024-02266-4

>>> Read full article>>>
Copyright for syndicated content belongs to the linked Source : Nature.com – https://www.nature.com/articles/s41587-024-02266-4

Exit mobile version