877
Views
0
CrossRef citations to date
0
Altmetric
Research Article

Genetic confounds of transgenerational epigenetic inheritance in mice

ORCID Icon &
Article: 2318519 | Received 15 Oct 2023, Accepted 07 Feb 2024, Published online: 18 Feb 2024

References

  • van Otterdijk SD, Michels KB. Transgenerational epigenetic inheritance in mammals: how good is the evidence? FASEB J. 2016;30(7):2457–10. doi: 10.1096/fj.201500083
  • Takahashi Y, Morales Valencia M, Yu Y, et al. Transgenerational inheritance of acquired epigenetic signatures at CpG islands in mice. Cell. 2023;186(4):715–731.e19. doi: 10.1016/j.cell.2022.12.047
  • Zeng J, Li G. Tfmapper: a tool for searching putative factors regulating gene expression using ChIP-seq data. Int J Biol Sci. 2018;14(12):1724–1731. doi: 10.7150/ijbs.28850
  • Noris P, Perrotta S, Seri M, et al. Mutations in ANKRD26 are responsible for a frequent form of inherited thrombocytopenia: analysis of 78 patients from 21 families. Blood. 2011;117(24):6673–80. doi: 10.1182/blood-2011-02-336537
  • Pippucci T, Savoia A, Perrotta S, et al. Mutations in the 5' UTR of ANKRD26, the ankirin repeat domain 26 gene, cause an autosomal-dominant form of inherited thrombocytopenia, THC2. Am J Hum Genet. 2011;88(1):115–120. doi: 10.1016/j.ajhg.2010.12.006
  • Skelly DA, Czechanski A, Byers C, et al. Mapping the effects of genetic variation on chromatin state and gene expression reveals loci that control ground state pluripotency. Cell Stem Cell. 2020;27(3):459–469.e8. doi: 10.1016/j.stem.2020.07.005
  • Onos KD, Uyar A, Keezer KJ, et al. Enhancing face validity of mouse models of Alzheimer’s disease with natural genetic variation. PLoS Genet. 2019;15(5):e1008155. doi: 10.1371/journal.pgen.1008155
  • Westra HJ, Franke L. From genome to function by studying eQtls. Biochim Biophys Acta. 2014;1842(10):1896–1902. doi: 10.1016/j.bbadis.2014.04.024
  • Zhou W, Hinoue T, Barnes B, et al. DNA methylation dynamics and dysregulation delineated by high-throughput profiling in the mouse. Cell Genom. 2022;2(7):100144. doi: 10.1016/j.xgen.2022.100144
  • 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(9):822–6. doi: 10.1038/nbt.2623
  • Chung CH, Allen AG, Sullivan NT, et al. Computational analysis concerning the impact of DNA accessibility on CRISPR-Cas9 cleavage efficiency. Mol Ther. 2020;28(1):19–28. doi: 10.1016/j.ymthe.2019.10.008
  • Papathanasiou S, Markoulaki S, Blaine LJ, et al. Whole chromosome loss and genomic instability in mouse embryos after CRISPR-Cas9 genome editing. Nat Commun. 2021;12(1):5855. doi: 10.1038/s41467-021-26097-y
  • Tsuchida CA, Brandes N, Bueno R, et al. Mitigation of chromosome loss in clinical CRISPR-Cas9-engineered T cells. Cell. 2023;186(21):4567–4582. doi: 10.1016/j.cell.2023.08.041
  • Cullot G, Boutin J, Toutain J, et al. CRISPR-Cas9 genome editing induces megabase-scale chromosomal truncations. Nat Commun. 2019;10(1):1136. doi: 10.1038/s41467-019-09006-2
  • Przewrocka J, Rowan A, Rosenthal R, et al. Unintended on-target chromosomal instability following CRISPR/Cas9 single gene targeting. Ann Oncol. 2020;31(9):1270–1273. doi: 10.1016/j.annonc.2020.04.480
  • Lazar NH, Celik S, Chen L, et al. High-resolution genome-wide mapping of chromosome-arm-scale truncations induced by CRISPR-Cas9 editing. bioRxiv. 2023. 2023.04.15.537038
  • Kuijk E, Jager M, van der Roest B, et al. The mutational impact of culturing human pluripotent and adult stem cells. Nat Commun. 2020;11(1):2493. doi: 10.1038/s41467-020-16323-4
  • Laurent LC, Ulitsky I, Slavin I, et al. Dynamic changes in the copy number of pluripotency and cell proliferation genes in human ESCs and iPscs during reprogramming and time in culture. Cell Stem Cell. 2011;8(1):106–18. doi: 10.1016/j.stem.2010.12.003
  • Kwon EM, Connelly JP, Hansen NF, et al. iPscs and fibroblast subclones from the same fibroblast population contain comparable levels of sequence variations. Proc Natl Acad Sci USA. 2017;114(8):1964–1969. doi: 10.1073/pnas.1616035114
  • Ben-David U, Mayshar Y, Benvenisty N. Large-scale analysis reveals acquisition of lineage-specific chromosomal aberrations in human adult stem cells. Cell Stem Cell. 2011;9(2):97–102. doi: 10.1016/j.stem.2011.06.013
  • Taapken SM, Nisler BS, Newton MA, et al. Karyotypic abnormalities in human induced pluripotent stem cells and embryonic stem cells. Nat Biotechnol. 2011;29(4):313–314. doi: 10.1038/nbt.1835
  • Ben-David U, Siranosian B, Ha G, et al. Genetic and transcriptional evolution alters cancer cell line drug response. Nature. 2018;560(7718):325–330. doi: 10.1038/s41586-018-0409-3
  • Quevedo R, Smirnov P, Tkachuk D, et al. Assessment of Genetic Drift in Large Pharmacogenomic Studies. Cell Systems. 2020;11(4):393–401.e2. doi: 10.1016/j.cels.2020.08.012
  • Albalat R, Canestro C. Evolution by gene loss. Nat Rev Genet. 2016;17(7):379–91. doi: 10.1038/nrg.2016.39
  • Abyzov A, Urban AE, Snyder M, et al. Cnvnator: an approach to discover, genotype, and characterize typical and atypical CNVs from family and population genome sequencing. Genome Res. 2011;21(6):974–84. doi: 10.1101/gr.114876.110
  • Vojta A, Dobrinić P, Tadić V, et al. Repurposing the CRISPR-Cas9 system for targeted DNA methylation. Nucleic Acids Res. 2016;44(12):5615–28. doi: 10.1093/nar/gkw159
  • Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15(12):550. doi: 10.1186/s13059-014-0550-8
  • Langmead B, Salzberg SL. Fast gapped-read alignment with bowtie 2. Nat Methods. 2012;9(4):357–9. doi: 10.1038/nmeth.1923
  • Li H, Handsaker B, Wysoker A, et al. The sequence Alignment/Map format and SAMtools. Bioinformatics. 2009;25(16):2078–9. doi: 10.1093/bioinformatics/btp352
  • McKenna A, Hanna M, Banks E, et al. The genome analysis toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 2010;20(9):1297–303. doi: 10.1101/gr.107524.110
  • Chen K, Wallis JW, McLellan MD, et al. BreakDancer: an algorithm for high-resolution mapping of genomic structural variation. Nat Methods. 2009;6(9):677–81. doi: 10.1038/nmeth.1363
  • 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(10):1473–5. doi: 10.1093/bioinformatics/btu048
  • Quinlan AR, Hall IM. Bedtools: a flexible suite of utilities for comparing genomic features. Bioinformatics. 2010;26(6):841–2. doi: 10.1093/bioinformatics/btq033