Skip to main content

Genome-Wide Profiling of DNA Double-Strand Breaks by the BLESS and BLISS Methods

  • Protocol
  • First Online:
Genome Instability

Part of the book series: Methods in Molecular Biology ((MIMB,volume 1672))

Abstract

DNA double-strand breaks (DSBs) are major DNA lesions that are constantly formed during physiological processes such as DNA replication, transcription, and recombination, or as a result of exogenous agents such as ionizing radiation, radiomimetic drugs, and genome editing nucleases. Unrepaired DSBs threaten genomic stability by leading to the formation of potentially oncogenic rearrangements such as translocations. In past few years, several methods based on next-generation sequencing (NGS) have been developed to study the genome-wide distribution of DSBs or their conversion to translocation events. We developed Breaks Labeling, Enrichment on Streptavidin, and Sequencing (BLESS), which was the first method for direct labeling of DSBs in situ followed by their genome-wide mapping at nucleotide resolution (Crosetto et al., Nat Methods 10:361–365, 2013). Recently, we have further expanded the quantitative nature, applicability, and scalability of BLESS by developing Breaks Labeling In Situ and Sequencing (BLISS) (Yan et al., Nat Commun 8:15058, 2017). Here, we first present an overview of existing methods for genome-wide localization of DSBs, and then focus on the BLESS and BLISS methods, discussing different assay design options depending on the sample type and application.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Institutional subscriptions

References

  1. Jackson SP, Bartek J (2009) The DNA-damage response in human biology and disease. Nature 461:1071–1078

    Google Scholar 

  2. van Gent DC, Hoeijmakers JH, Kanaar R (2001) Chromosomal stability and the DNA double-strandedbreak connection. Nat Rev Genet 2:196–206

    Google Scholar 

  3. McKinnon PJ, Caldecott KW (2007) DNA strand break repair and human genetic disease. Annu Rev Genomics Hum Genet 8:37–55

    Google Scholar 

  4. Porteus MH, Carroll D (2005) Gene targeting using zinc finger nucleases. Nat Biotechnol 23:967–973

    Google Scholar 

  5. Joung JK, Sander JD (2013) TALENs: a widely applicable technology for targeted genome editing. Nat Rev Mol Cell Biol 14:49–55

    Google Scholar 

  6. Hsu PD, Lander ES, Zhang F (2014) Development and applications of CRISPR-Cas9 for genome engineering. Cell 157:1262–1278

    Google Scholar 

  7. Zetsche B, Gootenberg JS, Abudayyeh OO et al (2015) Cpf1 is a single RNA-guided endonuclease of a class 2 CRISPR-Cas system. Cell 163:759–771

    Google Scholar 

  8. Tsai SQ, Keith Joung J (2016) Defining and improving the genome-wide specificities of CRISPR-Cas9nucleases. Nat Rev Genet 17:300–312

    Google Scholar 

  9. Szilard RK, Jacques P-E, Laramée L et al (2010) Systematic identification of fragile sites via genome-wide location analysis of gamma-H2AX. Nat Struct Mol Biol 17:299–305

    Google Scholar 

  10. Iacovoni JS, Caron P, Lassadi I et al (2010) High-resolution profiling of gammaH2AX around DNA double strand breaks in the mammalian genome. EMBO J 29:1446–1457

    Google Scholar 

  11. Madabhushi R, Gao F, Pfenning AR et al (2015) Activity-induced DNA breaks govern the expression of neuronal early-response genes. Cell 161:1592–1605

    Google Scholar 

  12. Crosetto N, Mitra A, Silva MJ et al (2013) Nucleotide-resolution DNA double-strand break mapping by next-generation sequencing. Nat Methods 10:361–365

    Google Scholar 

  13. Ran FA, Cong L, Yan WX et al (2015) In vivo genome editing using Staphylococcus aureus Cas9. Nature 520:186–191

    Google Scholar 

  14. Slaymaker IM, Gao L, Zetsche B et al (2016) Rationally engineered Cas9 nucleases with improved specificity. Science 351:84–88

    Google Scholar 

  15. Lensing SV, Marsico G, Hänsel-Hertsch R et al (2016) DSBCapture: in situ capture and sequencing of DNA breaks. Nat Methods 13:855–857

    Google Scholar 

  16. Canela A, Sridharan S, Sciascia N et al (2016) DNA breaks and end resection measured genome-wide by end sequencing. Mol Cell 63:898–911

    Google Scholar 

  17. Yan WX, Mirzazadeh R, Garnerone S et al (2017) BLISS is a versatile and quantitative method for genome-wide profiling of DNA double-strand breaks. Nat Commun 8:15058

    Google Scholar 

  18. Kim D, Bae S, Park J et al (2015) Digenome-seq: genome-wide profiling of CRISPR-Cas9 off-targeteffects in human cells. Nat Methods 12:237–243. 1 p following 243

    Google Scholar 

  19. Kim D, Kim J, Hur JK et al (2016) Genome-wide analysis reveals specificities of Cpf1 endonucleases in human cells. Nat Biotechnol 34:863–868

    Google Scholar 

  20. Tsai SQ, Zheng Z, Nguyen NT et al (2015) GUIDE-seq enables genome-wide profiling of off-targetcleavage by CRISPR-Cas nucleases. Nat Biotechnol 33:187–197

    Google Scholar 

  21. Kleinstiver BP, Tsai SQ, Prew MS et al (2016) Genome-wide specificities of CRISPR-Cas Cpf1 nucleases in human cells. Nat Biotechnol 34:869–874

    Google Scholar 

  22. Wang X, Wang Y, Wu X et al (2015) Unbiased detection of off-target cleavage by CRISPR-Cas9 and TALENs using integrase-defective lentiviral vectors. Nat Biotechnol 33:175–178

    Google Scholar 

  23. Frock RL, Hu J, Meyers RM et al (2015) Genome-wide detection of DNA double-stranded breaks induced by engineered nucleases. Nat Biotechnol 33:179–186

    Google Scholar 

  24. Wei P-C, Chang AN, Kao J et al (2016) Long neural genes harbor recurrent DNA break clusters in neural stem/progenitor cells. Cell 164:644–655

    Google Scholar 

  25. Kivioja T, Vähärautio A, Karlsson K et al (2012) Counting absolute numbers of molecules using unique molecular identifiers. Nat Methods 9:72–74

    Google Scholar 

  26. Baslan T, Kendall J, Rodgers L et al (2012) Genome-wide copy number analysis of single cells. Nat Protoc 7:1024–1041

    Google Scholar 

  27. Naumova N, Imakaev M, Fudenberg G et al (2013) Organization of the mitotic chromosome. Science 342:948–953

    Google Scholar 

  28. Dunnett CW (1955) A multiple comparison procedure for comparing several treatments with a control. J Am Stat Assoc 50:1096–1121

    Google Scholar 

  29. Zhang Y, Liu T, Meyer CA et al (2008) Model-based analysis of ChIP-Seq (MACS). Genome Biol 9:R137

    Google Scholar 

  30. Heinz S, Benner C, Spann N et al (2010) Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol Cell 38:576–589

    Google Scholar 

  31. Zang C, Schones DE, Zeng C et al (2009) A clustering approach for identification of enriched domains from histone modification ChIP-Seq data. Bioinformatics 25:1952–1958

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Magda Bienko or Nicola Crosetto .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2018 Springer Science+Business Media LLC

About this protocol

Cite this protocol

Mirzazadeh, R., Kallas, T., Bienko, M., Crosetto, N. (2018). Genome-Wide Profiling of DNA Double-Strand Breaks by the BLESS and BLISS Methods. In: Muzi-Falconi, M., Brown, G. (eds) Genome Instability. Methods in Molecular Biology, vol 1672. Humana Press, New York, NY. https://doi.org/10.1007/978-1-4939-7306-4_14

Download citation

  • DOI: https://doi.org/10.1007/978-1-4939-7306-4_14

  • Published:

  • Publisher Name: Humana Press, New York, NY

  • Print ISBN: 978-1-4939-7305-7

  • Online ISBN: 978-1-4939-7306-4

  • eBook Packages: Springer Protocols

Publish with us

Policies and ethics