Human 8-cell embryos enable efficient induction of disease-preventive mutations without off-target effect by cytosine base editor

Yinghui Wei, Meiling Zhang, Jing Hu, Yingsi Zhou, Mingxing Xue, Jianhang Yin, Yuanhua Liu, Hu Feng, Ling Zhou, Zhifang Li, Dongshuang Wang, Zhiguo Zhang, Yin Zhou, Hongbin Liu, Ning Yao, Erwei Zuo, Jiazhi Hu, Yanzhi Du, Wen Li, Chunlong Xu, Hui Yang

PDF(9391 KB)
PDF(9391 KB)
Protein Cell ›› 2023, Vol. 14 ›› Issue (6) : 416-432. DOI: 10.1093/procel/pwac043
RESEARCH ARTICLE
RESEARCH ARTICLE

Human 8-cell embryos enable efficient induction of disease-preventive mutations without off-target effect by cytosine base editor

Author information +
History +

Abstract

Approximately 140 million people worldwide are homozygous carriers of APOE4 (ϵ4), a strong genetic risk factor for late onset familial and sporadic Alzheimer’s disease (AD), 91% of whom will develop AD at earlier age than heterozygous carriers and noncarriers. Susceptibility to AD could be reduced by targeted editing of APOE4, but a technical basis for controlling the off-target effects of base editors is necessary to develop low-risk personalized gene therapies. Here, we first screened eight cytosine base editor variants at four injection stages (from 1- to 8-cell stage), and found that FNLS-YE1 variant in 8-cell embryos achieved the comparable base conversion rate (up to 100%) with the lowest bystander effects. In particular, 80% of AD-susceptible ϵ4 allele copies were converted to the AD-neutral ϵ3 allele in human ϵ4-carrying embryos. Stringent control measures combined with targeted deep sequencing, whole genome sequencing, and RNA sequencing showed no DNA or RNA off-target events in FNLS-YE1-treated human embryos or their derived stem cells. Furthermore, base editing with FNLS-YE1 showed no effects on embryo development to the blastocyst stage. Finally, we also demonstrated FNLS-YE1 could introduce known protective variants in human embryos to potentially reduce human susceptivity to systemic lupus erythematosus and familial hypercholesterolemia. Our study therefore suggests that base editing with FNLS-YE1 can efficiently and safely introduce known preventive variants in 8-cell human embryos, a potential approach for reducing human susceptibility to AD or other genetic diseases.

Keywords

human embryo / APOE4 / disease-preventive mutations / base editor

Cite this article

Download citation ▾
Yinghui Wei, Meiling Zhang, Jing Hu, Yingsi Zhou, Mingxing Xue, Jianhang Yin, Yuanhua Liu, Hu Feng, Ling Zhou, Zhifang Li, Dongshuang Wang, Zhiguo Zhang, Yin Zhou, Hongbin Liu, Ning Yao, Erwei Zuo, Jiazhi Hu, Yanzhi Du, Wen Li, Chunlong Xu, Hui Yang. Human 8-cell embryos enable efficient induction of disease-preventive mutations without off-target effect by cytosine base editor. Protein Cell, 2023, 14(6): 416‒432 https://doi.org/10.1093/procel/pwac043

References

[1]
Akioyamen LE, Genest J, Shan SD et al. Estimating the prevalence of heterozygous familial hypercholesterolaemia: a systematic review and meta-analysis. BMJ Open 2017;7:e016461.
CrossRef Google scholar
[2]
Alvin Huang YW, Zhou B, Nabet AM et al. Differential signaling mediated by ApoE2, ApoE3, and ApoE4 in human neurons parallels Alzheimer’s disease risk. J Neurosci 2019;39:7408–7427.
CrossRef Google scholar
[3]
Anzalone AV, Randolph PB, Davis JR et al. Search-and-replace genome editing without double-strand breaks or donor DNA. Nature 2019;576:149–157.
CrossRef Google scholar
[4]
Arnett FC, Shulman LE. Studies in familial systemic lupus erythematosus. Medicine 1976;55:313–322.
CrossRef Google scholar
[5]
Bae S, Park J, Kim JS. Cas-OFFinder: a fast and versatile algorithm that searches for potential off-target sites of Cas9 RNA-guided endonucleases. Bioinformatics 2014;30:1473–1475.
CrossRef Google scholar
[6]
Castellano JM, Kim J, Stewart FR et al. Human apoE isoforms differentially regulate brain amyloid-β peptide clearance. Sci Transl Med 2011;3:3002156.
CrossRef Google scholar
[7]
Chen AE, Egli D, Niakan K et al. Optimal timing of inner cell mass isolation increases the efficiency of human embryonic stem cell derivation and allows generation of sibling cell lines. Cell Stem Cell 2009;4:103–106.
CrossRef Google scholar
[8]
Cibulskis K, Lawrence MS, Carter SL et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat Biotechnol 2013;31:213–219.
CrossRef Google scholar
[9]
Cohen JC, Boerwinkle E, Thomas H et al. Sequence variations in PCSK9, low LDL, and protection against coronary heart disease. N Engl J Med 2006;354:1264–1272.
CrossRef Google scholar
[10]
Corder EH, Saunders AM, Strittmatter WJ et al. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science 1993;261:921–923.
CrossRef Google scholar
[11]
Dang L, Zhou X, Zhong X et al. Correction of the pathogenic mutation in TGM1 gene by adenine base editing in mutant embryos. Mol Ther 2022;30:175–183.
CrossRef Google scholar
[12]
Diogo D, Bastarache L, Liao KP et al. TYK2 protein-coding variants protect against rheumatoid arthritis and autoimmunity, with no evidence of major pleiotropic effects on non-autoimmune complex traits. PLoS One 2015;10:e0122271.
CrossRef Google scholar
[13]
Doman JL, Raguram A, Newby GA et al. Evaluation and minimization of Cas9-independent off-target DNA editing by cytosine base editors. Nat Biotechnol 2020;38:620–628.
CrossRef Google scholar
[14]
Fang H, Bergmann EA, Arora K et al. Indel variant analysis of short-read sequencing data with Scalpel. Nat Protoc 2016;11:2529–2548.
CrossRef Google scholar
[15]
Farrer LA, Cupples LA, Haines JL et al. Effects of age, sex, and ethnicity on the association between apolipoprotein E genotype and Alzheimer disease: a meta-analysis. JAMA 1997;278:1349–1356.
CrossRef Google scholar
[16]
Fu Y, Foden JA, Khayter C et al. High-frequency off-target mutagenesis induced by CRISPR-Cas nucleases in human cells. Nat Biotechnol 2013;31:822–826.
CrossRef Google scholar
[17]
Gaudelli NM, Komor AC, Rees HA et al. Programmable base editing of A•T to G•C in genomic DNA without DNA cleavage. Nature 2017;551:464–471.
CrossRef Google scholar
[18]
Gehrke JM, Cervantes O, Clement MK et al. An APOBEC3A-Cas9 base editor with minimized bystander and off-target activities. Nat Biotechnol 2018;36:977–982.
CrossRef Google scholar
[19]
Grunewald J, Zhou R, Garcia SP et al. Transcriptome-wide off-target RNA editing induced by CRISPR-guided DNA base editors. Nature 2019a;569:433–437.
CrossRef Google scholar
[20]
Grunewald J, Zhou R, Iyer S et al. CRISPR DNA base editors with reduced RNA off-target and self-editing activities. Nat Biotechnol 2019b;37:1041–1048.
CrossRef Google scholar
[21]
Halliday MR, Rege SV, Ma Q et al. Accelerated pericyte degeneration and blood-brain barrier breakdown in apolipoprotein E4 carriers with Alzheimer’s disease. J Cereb Blood Flow Metab 2016;36:216–227.
CrossRef Google scholar
[22]
Harper AR, Nayee S, Topol EJ. Protective alleles and modifier variants in human health and disease. Nat Rev Genet 2015;16:689–701.
CrossRef Google scholar
[23]
Hsu PD, Scott DA, Weinstein JA et al. DNA targeting specificity of RNA-guided Cas9 nucleases. Nat Biotechnol 2013;31:827–832.
CrossRef Google scholar
[24]
Huang Y, Mahley RW. Apolipoprotein E: structure and function in lipid metabolism, neurobiology, and Alzheimer’s diseases. Neurobiol Dis 2014;72 Pt A:3–12.
CrossRef Google scholar
[25]
Huang Y, Mucke L. Alzheimer mechanisms and therapeutic strategies. Cell 2012;148:1204–1222.
CrossRef Google scholar
[26]
Iannucci J, Sen A, Grammas P. Isoform-specific effects of apolipoprotein E on markers of Inflammation and toxicity in brain glia and neuronal cells in vitro. Curr Issues Mol Biol 2021;43:215–225.
CrossRef Google scholar
[27]
Jonsson T, Atwal JK, Steinberg S et al. A mutation in APP protects against Alzheimer’s disease and age-related cognitive decline. Nature 2012;488:96–99.
CrossRef Google scholar
[28]
Komor AC, Kim YB, Packer MS et al. Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 2016;533:420–424.
CrossRef Google scholar
[29]
López-Isac E, Campillo-Davo D, Bossini-Castillo L et al.; Spanish Scleroderma Group. Influence of TYK2 in systemic sclerosis susceptibility: a new locus in the IL-12 pathway. Ann Rheum Dis 2016;75:1521–1526.
CrossRef Google scholar
[30]
Lee MT, Bonneau AR, Giraldez AJ. Zygotic genome activation during the maternal-to-zygotic transition. Annu Rev Cell Dev Biol 2014;30:581–613.
CrossRef Google scholar
[31]
Liang P, Ding C, Sun H et al. Correction of beta-thalassemia mutant by base editor in human embryos. Protein Cell 2017;8:811–822.
CrossRef Google scholar
[32]
Lin YT, Seo J, Gao F et al. APOE4 causes widespread molecular and cellular alterations associated with Alzheimer’s disease phenotypes in human iPSC-derived brain cell types. Neuron 2018;98:1141–1154.e7.
CrossRef Google scholar
[33]
Liu CC, Kanekiyo T, Xu H et al. Apolipoprotein E and Alzheimer disease: risk, mechanisms and therapy. Nat Rev Neurol 2013a;9:106–118.
CrossRef Google scholar
[34]
Liu CC, Kanekiyo T, Xu H et al. Apolipoprotein E and Alzheimer disease: risk, mechanisms and therapy. Nat Rev Neurol 2013b;9:106–118.
CrossRef Google scholar
[35]
Mahley RW, Weisgraber KH, Huang Y. Apolipoprotein E4: a causative factor and therapeutic target in neuropathology, including Alzheimer’s disease. Proc Natl Acad Sci USA 2006;103:5644–5651.
CrossRef Google scholar
[36]
Miname MH, Santos RD. Reducing cardiovascular risk in patients with familial hypercholesterolemia: risk prediction and lipid management. Prog Cardiovasc Dis 2019;62:414–422.
CrossRef Google scholar
[37]
Montagne A, Nation DA, Sagare AP et al. APOE4 leads to blood-brain barrier dysfunction predicting cognitive decline. Nature 2020;581:71–76.
CrossRef Google scholar
[38]
Nordestgaard BG, Chapman MJ, Humphries SE et al.; European Atherosclerosis Society Consensus Panel. Familial hypercholesterolaemia is underdiagnosed and undertreated in the general population: guidance for clinicians to prevent coronary heart disease: consensus statement of the European Atherosclerosis Society. Eur Heart J 2013;34:3478–390a.
[39]
Ossenkoppele R, Jansen WJ, Rabinovici GD et al.; Amyloid PET Study Group. Prevalence of amyloid PET positivity in dementia syndromes: a meta-analysis. JAMA 2015;313:1939–1949.
CrossRef Google scholar
[40]
Ostrowitzki S, Deptula D, Thurfjell L et al. Mechanism of amyloid removal in patients with Alzheimer disease treated with gantenerumab. Arch Neurol 2012;69:198–207.
CrossRef Google scholar
[41]
Petrushina I, Davtyan H, Hovakimyan A et al. Comparison of efficacy of preventive and therapeutic vaccines targeting the N terminus of β-amyloid in an animal model of Alzheimer’s disease. Mol Ther 2017;25:153–164.
CrossRef Google scholar
[42]
Raber J, Huang Y, Ashford JW. ApoE genotype accounts for the vast majority of AD risk and AD pathology. Neurobiol Aging 2004;25:641–650.
CrossRef Google scholar
[43]
Ramos-Niembro F, Alarcon-Segovia D. Familial aspects of mixed connective tissue disease (MCTD). I. Occurrence of systemic lupus erythematosus in another member in two families and aggregation of MCTD in another family. J Rheumatol 1978;5:433–440.
[44]
Rebeck GW, Reiter JS, Strickland DK et al. Apolipoprotein E in sporadic Alzheimer’s disease: allelic variation and receptor interactions. Neuron 1993;11:575–580.
CrossRef Google scholar
[45]
Reiman EM, Arboleda-Velasquez JF, Quiroz YT et al.; Alzheimer’s Disease Genetics Consortium. Exceptionally low likelihood of Alzheimer’s dementia in APOE2 homozygotes from a 5,000-person neuropathological study. Nat Commun 2020;11:667.
CrossRef Google scholar
[46]
Rinne JO, Brooks DJ, Rossor MN et al. 11C-PiB PET assessment of change in fibrillar amyloid-beta load in patients with Alzheimer’s disease treated with bapineuzumab: a phase 2, double-blind, placebo-controlled, ascending-dose study. Lancet Neurol 2010;9:363–372.
CrossRef Google scholar
[47]
Saunders CT, Wong WS, Swamy S et al. Strelka: accurate somatic small-variant calling from sequenced tumor-normal sample pairs. Bioinformatics 2012;28:1811–1817.
CrossRef Google scholar
[48]
Selkoe DJ. The molecular pathology of Alzheimer’s disease. Neuron 1991;6:487–498.
CrossRef Google scholar
[49]
Seripa D, D’Onofrio G, Panza F et al. The genetics of the human APOE polymorphism. Rejuvenation Res 2011;14:491–500.
CrossRef Google scholar
[50]
Seripa D, Matera MG, Daniele A et al. The missing ApoE allele. Ann Hum Genet 2007;71:496–500.
CrossRef Google scholar
[51]
Serrano-Pozo A, Qian J, Monsell SE et al. APOEϵ2 is associated with milder clinical and pathological Alzheimer disease. Ann Neurol 2015;77:917–929.
CrossRef Google scholar
[52]
Sestak AL, Shaver TS, Moser KL et al. Familial aggregation of lupus and autoimmunity in an unusual multiplex pedigree. J Rheumatol 1999;26:1495–1499.
[53]
Sevigny J, Chiao P, Bussière T et al. The antibody aducanumab reduces Aβ plaques in Alzheimer’s disease. Nature 2016;537:50–56.
CrossRef Google scholar
[54]
Shi Y, Yamada K, Liddelow SA et al.; Alzheimer’s Disease Neuroimaging Initiative. ApoE4 markedly exacerbates tau-mediated neurodegeneration in a mouse model of tauopathy. Nature 2017;549:523–527.
CrossRef Google scholar
[55]
Smith CJ, Ashford JW, Perfetti TA. Putative survival advantages in young apolipoprotein ϵ4 carriers are associated with increased neural stress. J Alzheimers Dis 2019a;68:885–923.
CrossRef Google scholar
[56]
Smith EMD, Lythgoe H, Midgley A et al. Juvenile-onset systemic lupus erythematosus: update on clinical presentation, pathophysiology and treatment options. Clin Immunol 2019b;209:108274.
CrossRef Google scholar
[57]
Stitziel NO, Stirrups KE, Masca NG et al.; Myocardial Infarction Genetics and CARDIoGRAM Exome Consortia Investigators. Coding variation in ANGPTL4, LPL, and SVEP1 and the risk of coronary disease. N Engl J Med 2016;374:1134–1144.
CrossRef Google scholar
[58]
Sturm AC, Knowles JW, Gidding SS et al.; Convened by the Familial Hypercholesterolemia Foundation. Clinical genetic testing for familial hypercholesterolemia: JACC scientific expert panel. J Am Coll Cardiol 2018;72:662–680.
CrossRef Google scholar
[59]
Tanzi RE. The genetics of Alzheimer disease. Cold Spring Harb Perspect Med 2012;2:a006296.
CrossRef Google scholar
[60]
van Dyck CH. Anti-amyloid-β monoclonal antibodies for Alzheimer’s disease: pitfalls and promise. Biol Psychiatry 2018;83:311–319.
CrossRef Google scholar
[61]
Verghese PB, Castellano JM, Garai K et al. ApoE influences amyloid-β (Aβ) clearance despite minimal apoE/Aβ association in physiological conditions. Proc Natl Acad Sci USA 2013;110:25.
CrossRef Google scholar
[62]
Walton RT, Christie KA, Whittaker MN et al. Unconstrained genome targeting with near-PAMless engineered CRISPR-Cas9 variants. Science 2020;368:290–296.
CrossRef Google scholar
[63]
Wang X, Li J, Wang Y et al. Efficient base editing in methylated regions with a human APOBEC3A-Cas9 fusion. Nat Biotechnol 2018;36:946–949.
CrossRef Google scholar
[64]
Wennberg AM, Tosakulwong N, Lesnick TG et al. Association of apolipoprotein E ϵ4 with transactive response DNA-binding protein 43. JAMA Neurol 2018;75:1347–1354.
CrossRef Google scholar
[65]
Wilm A, Aw PP, Bertrand D et al. LoFreq: a sequence-quality aware, ultra-sensitive variant caller for uncovering cell-population heterogeneity from high-throughput sequencing datasets. Nucleic Acids Res 2012;40:11189–11201.
CrossRef Google scholar
[66]
Yamada M, Johannesson B, Sagi I et al. Human oocytes reprogram adult somatic nuclei of a type 1 diabetic to diploid pluripotent stem cells. Nature 2014;510:533–536.
CrossRef Google scholar
[67]
Yang WL, Shen N, Ye DQ et al. Genome-wide association study in Asian populations identifies variants in ETS1 and WDFY4 associated with systemic lupus erythematosus. PLoS Genet 2010;6:e1000841.
CrossRef Google scholar
[68]
Yin J, Liu M, Liu Y et al. Optimizing genome editing strategy by primer-extension-mediated sequencing. Cell Discov 2019; 5:18.
CrossRef Google scholar
[69]
Zeng Y, Li J, Li G et al. Correction of the marfan syndrome pathogenic FBN1 mutation by base editing in human cells and heterozygous embryos. Mol Ther 2018;26:2631–2637.
CrossRef Google scholar
[70]
Zhang M, Zhou C, Wei Y et al. Human cleaving embryos enable robust homozygotic nucleotide substitutions by base editors. Genome Biol 2019;20:101.
CrossRef Google scholar
[71]
Zhang Y, Bo L, Zhang H et al. E26 transformation-specific-1 (ETS1) and WDFY family member 4 (WDFY4) polymorphisms in Chinese patients with rheumatoid arthritis. Int J Mol Sci 2014;15:2712–2721.
CrossRef Google scholar
[72]
Zhou C, Sun Y, Yan R et al. Off-target RNA mutation induced by DNA base editing and its elimination by mutagenesis. Nature 2019;571:275–278.
CrossRef Google scholar
[73]
Zlokovic BV. Cerebrovascular effects of apolipoprotein E: implications for Alzheimer disease. JAMA Neurol 2013;70:440–444.
CrossRef Google scholar
[74]
Zuccaro MV, Xu J, Mitchell C et al. (2020) Allele-specific chromosome removal after Cas9 cleavage in human embryos. Cell 183:1650–1664.e15 e1615.
CrossRef Google scholar
[75]
Zuo E, Sun Y, Wei W et al. Cytosine base editor generates substantial off-target single-nucleotide variants in mouse embryos. Science 2019;364:289–292.
CrossRef Google scholar
[76]
Zuo E, Sun Y, Yuan T et al. A rationally engineered cytosine base editor retains high on-target activity while reducing both DNA and RNA off-target effects. Nat Methods 2020;17:600–604.
CrossRef Google scholar

RIGHTS & PERMISSIONS

2022 The Author(s) 2022. Published by Oxford University Press on behalf of Higher Education Press.
AI Summary AI Mindmap
PDF(9391 KB)

Accesses

Citations

Detail

Sections
Recommended

/