BLESS

Last updated

BLESS, also known as breaks labeling, enrichment on streptavidin and next-generation sequencing, is a method used to detect genome-wide double-strand DNA damage. [1] In contrast to chromatin immunoprecipitation (ChIP)-based methods of identifying DNA double-strand breaks (DSBs) by labeling DNA repair proteins, BLESS utilizes biotinylated DNA linkers to directly label genomic DNA in situ which allows for high-specificity enrichment of samples on streptavidin beads and the subsequent sequencing-based DSB mapping to nucleotide resolution.

Contents

Workflow

BLESS workflow. 1) Double-stranded DNA breaks (DSBs) are labeled in situ with proximal DNA hairpin linkers containing a biotin marker. 2) Cells are fixed, lysed and treated with proteinases for extraction and subsequent shearing of genomic DNA (gDNA). 3) Labeled and unlabeled gDNA fragments are passed through streptavidin-derived beads, which capture the labeled fragments with high specificity due to the strong affinity of biotin markers to streptavidin. 4) After passage through streptavidin beads, unlabeled gDNA fragments are removed, leaving the enriched biotin-labeled gDNA fragments. 5) Distal linker is ligated to labeled gDNA fragments at the free end. 6) I-SceI endonuclease cuts linkers at the restriction site to release gDNA fragments from biotin. 7) Barcode-specific primers are used for amplification of enriched fragments by Polymerase Chain Reaction (PCR). 8) Next-Generation Sequencing of PCR products is then used for single nucleotide-resolution analysis of DSBs in the genome. BLESS workflow (update v2).png
BLESS workflow. 1) Double-stranded DNA breaks (DSBs) are labeled in situ with proximal DNA hairpin linkers containing a biotin marker. 2) Cells are fixed, lysed and treated with proteinases for extraction and subsequent shearing of genomic DNA (gDNA). 3) Labeled and unlabeled gDNA fragments are passed through streptavidin-derived beads, which capture the labeled fragments with high specificity due to the strong affinity of biotin markers to streptavidin. 4) After passage through streptavidin beads, unlabeled gDNA fragments are removed, leaving the enriched biotin-labeled gDNA fragments. 5) Distal linker is ligated to labeled gDNA fragments at the free end. 6) I-SceI endonuclease cuts linkers at the restriction site to release gDNA fragments from biotin. 7) Barcode-specific primers are used for amplification of enriched fragments by Polymerase Chain Reaction (PCR). 8) Next-Generation Sequencing of PCR products is then used for single nucleotide-resolution analysis of DSBs in the genome.

Biotinylated linker design

The biotinylated linker is designed to form a hairpin structure that specifically labels DSBs and not single-strand DNA breaks. The linker has a blunt, ligatable end with a known barcode sequence that labels the site of ligation as well as an XhoI restriction enzyme recognition site adjacent to the barcode. The hairpin loop of the linker is covalently bound to a biotin molecule, allowing for subsequent enrichment of labeled DNA with streptavidin beads. [1]

Use of biotin labels allows for specific binding without disruption of DNA due to the small size of the marker. Because biotin also has high affinity to streptavidin, further highly specific purification can be performed on streptavidin beads. [2]

Nuclei purification and in situ labeling

Following the induction of DSBs, cells are fixed with formaldehyde, lysed, and treated with proteinases to purify intact nuclei. [1] The initial fixation step stabilizes chromatin and prevents the formation of additional DSBs during sample preparation. [3] DSBs are then blunted and incubated with biotinylated linkers in the presence of T4 DNA ligase. T4 ligase does not recognize single-stranded breaks, and as such directly labels the DSB sites through covalent attachment of the biotinylated linker. [1]

DNA extraction, fragmentation, and purification

Labeled genomic DNA is extracted from nuclei and fragmented by HaeIII restriction enzyme digestion and sonication. Labeled DNA fragments are then purified using beads derived from streptavidin, a biotin-binding protein found in the bacterium Streptomyces avidinii . Because the interaction of streptavidin and biotin is strong and highly specific, purification of sample on streptavidin-coated beads allows for robust enrichment of labeled DNA fragments. [1] [2]

Distal linker DNA labeling and digestion

A second labeling step occurs after fragmentation and biotin-streptavidin affinity purification to attach primer binding sites to the free end of the captured DNA. Similar to the first labeling step, T4 DNA ligase is used to attach a distal linker to the unlabeled end of the DNA. The distal linker also has an XhoI restriction enzyme recognition site but is not covalently bound to a biotin molecule. Once the distal linker is attached, the captured DNA fragments are digested using I-SceI endonucleases that cut both the biotinylated linkers and the distal linkers to release the DNA fragments. [1]

PCR amplification and sequencing

The digested DNA strands are amplified using PCR with primers complementary to barcode sequences in the biotinylated linker and the distal linker. The amplified DNA is further processed by digesting with XhoI restriction enzymes to remove the I-SceI ends and purified prior to sequencing. Although use of next-generation sequencing methods is recommended for BLESS analysis, Sanger sequencing has also been shown to generate successful, albeit less robust results. [1]

Computational analysis

The BLESS sequencing reads can be analyzed using the Instant Sequencing (iSeq) software suite. [1] To detect sites of DSBs, reads are aligned to a reference genome using bowtie to determine the chromosome positions. The genome is divided into intervals and hypergeometric tests are used to identify intervals enriched with mapped reads. DSBs are identified by comparing enrichment in treated samples versus a control. A statistically significant increase in a DNA damage-induced sample suggests that the DNA at this interval is fragile and enriched in DSBs. [4]

Advantages

  1. Use of biotinylated DNA linkers designed to specifically recognize double-stranded DNA breaks allows for a less biased, more direct survey of the breakome without the need to rely on native and/or DSB-proxy proteins, such as the phosphorylated histone variant H2A.X (γH2A.X), in the cell. [5] Because of this, BLESS can be utilized in a variety of cells from different organisms.
  2. For the same reason, BLESS is also sensitive to multiple sources of double-stranded breaks, such as chemical and physical DNA disruption, replication fork stalling, as well as presence of telomere ends. [1] This makes BLESS suitable for analysis of cells at various conditions.
  3. Labeling of DSBs occurs in situ, reducing the risk of false positives form detection of DNA breaks due to mechanical shearing and chemical sample treatment.

Limitations

  1. Due to specificity of the linker design, biotinylated markers can only label double-stranded DNA breaks at blunt, not cohesive ends, leading to less efficient ligation.
  2. Compared to newer breakome survey methods, such as BLISS, BLESS requires large amounts of cellular starting material for successful analysis, resulting in tedious and time-consuming sample preparation and processing. To process 24 samples, the BLESS protocol requires 60 work-hours over the course of 15 days whereas BLISS requires 12 work-hours over 5 days. [6]
  3. Because cells require chemical fixation prior to DNA extraction, BLESS is prone to high background noise from fixation artifacts. However, stringent custom optimization has been shown to reduce this issue. [7]
  4. Due to the lack of PCR controls, BLESS is not a fully quantitative method and is prone to amplification bias, resulting in poor scalability.
  5. BLESS is only suitable for detecting double-stranded breaks at one specific time in the genome, as compared to continuous analysis.

Alternative methods

Breaks labeling in situ and sequencing (BLISS)
In BLISS, cells or tissue sections are attached to a cover glass first before DSB labeling. This allows some centrifugation steps to be omitted, thus decreasing the number of artificial DSBs introduced from sample preparation, and reducing sample loss. Importantly, it allows a much smaller amount of starting material to be used compared to BLESS. Another improvement is the use of in vitro transcription to generate and amplify RNA sequences for library preparation. BLISS uses T7 bacteriophage-mediated transcription rather than PCR, reducing errors caused by PCR amplification bias that occur with BLESS. [6]
Immobilized-BLESS (i-BLESS)
A limitation of the original BLESS method is that it is problematic in application to smaller cells such as yeast cells. While low centrifugation speeds employed during nuclei isolation are not efficient enough for small cells, increasing centrifugation speeds can shear the genomic DNA. However, in i-BLESS, cells are immobilized in agarose beads prior to DSB labeling. [8] This allows the use of higher centrifugation speeds without artificial DNA shearing. The remainder of the DSB labeling procedure follows that of the BLESS method, and labeled DNA fragments are recovered from the agarose beads prior to the streptavidin capture step. The i-BLESS method is not limited to yeast and can theoretically be applied to all cells.
DSBCapture
Similar to BLESS, DSBCapture uses biotinylated adapters to label DSBs in situ and streptavidin beads to isolate labeled DNA fragments for amplification and sequencing. [9] While labeling in BLESS relies on blunt-end ligation, DSBCapture uses more efficient cohesive-end ligation to attach biotinylated modified Illumina adapters. In addition, DSBCapture relies on fewer PCR steps compared to BLESS, reducing amplification bias. [10] This method also generates libraries with higher sequence diversity than BLESS, eliminating the need to spike in other libraries to improve diversity prior to sequencing. Furthermore, DSBCapture uses single-end sequencing in contrast to BLESS where sequencing can begin from both ends. Single-end sequencing results reflect only the sequences of DSB sites, improving data yield. [11]
GUIDE-Seq
Also known as Genome-Wide Unbiased Identification of DSBs Enabled by Sequencing, GUIDE-Seq uses the incorporation of double-stranded oligodeoxynucleotide (dsODN) sequences to label sites of DSBs in living cells. [12] It allows DSBs to be labeled over an extended time period, and the sites of DNA damage identified through GUIDE-Seq reflect accumulated DSBs. In contrast, BLESS only labels and detects transient DSBs that exist when the cells were fixed.

Applications

While double-stranded breaks in the DNA can be caused by various sources of disruption, they are often observed at high frequency during apoptosis and can contribute to genome instability, resulting in oncogenic mutations. [1] [13] For this reason, high-resolution, specific DSB-mapping methods like BLESS are useful for breakome surveys.

DSBs can be artificially induced using genome editing technologies such as CRISPR-Cas9 or TALEN. These technologies may lead to unintentional modifications of DNA at off-target locations on the genome. [14] Since BLESS can identify the nucleotide position of DSBs, it can be used to determine if off-target genome editing has occurred and the location of DSBs unintentionally introduced by these nuclease systems. [7]

Related Research Articles

<span class="mw-page-title-main">Complementary DNA</span> Single-stranded DNA synthesized from RNA

In genetics, complementary DNA (cDNA) is DNA synthesized from a single-stranded RNA template in a reaction catalyzed by the enzyme reverse transcriptase. cDNA is often used to express a specific protein in a cell that does not normally express that protein, or to sequence or quantify mRNA molecules using DNA based methods. cDNA that codes for a specific protein can be transferred to a recipient cell for expression, often bacterial or yeast expression systems. cDNA is also generated to analyze transcriptomic profiles in bulk tissue, single cells, or single nuclei in assays such as microarrays, qPCR, and RNA-seq.

<span class="mw-page-title-main">Polymerase chain reaction</span> Laboratory technique to multiply a DNA sample for study

The polymerase chain reaction (PCR) is a method widely used to make millions to billions of copies of a specific DNA sample rapidly, allowing scientists to amplify a very small sample of DNA sufficiently to enable detailed study. PCR was invented in 1983 by American biochemist Kary Mullis at Cetus Corporation. Mullis and biochemist Michael Smith, who had developed other essential ways of manipulating DNA, were jointly awarded the Nobel Prize in Chemistry in 1993.

In biochemistry, biotinylation is the process of covalently attaching biotin to a protein, nucleic acid or other molecule. Biotinylation is rapid, specific and is unlikely to disturb the natural function of the molecule due to the small size of biotin. Biotin binds to streptavidin and avidin with an extremely high affinity, fast on-rate, and high specificity, and these interactions are exploited in many areas of biotechnology to isolate biotinylated molecules of interest. Biotin-binding to streptavidin and avidin is resistant to extremes of heat, pH and proteolysis, making capture of biotinylated molecules possible in a wide variety of environments. Also, multiple biotin molecules can be conjugated to a protein of interest, which allows binding of multiple streptavidin, avidin or neutravidin protein molecules and increases the sensitivity of detection of the protein of interest. There is a large number of biotinylation reagents available that exploit the wide range of possible labelling methods. Due to the strong affinity between biotin and streptavidin, the purification of biotinylated proteins has been a widely used approach to identify protein-protein interactions and post-translational events such as ubiquitylation in molecular biology.

<span class="mw-page-title-main">DNA sequencing</span> Process of determining the nucleic acid sequence

DNA sequencing is the process of determining the nucleic acid sequence – the order of nucleotides in DNA. It includes any method or technology that is used to determine the order of the four bases: adenine, guanine, cytosine, and thymine. The advent of rapid DNA sequencing methods has greatly accelerated biological and medical research and discovery.

454 Life Sciences was a biotechnology company based in Branford, Connecticut that specialized in high-throughput DNA sequencing. It was acquired by Roche in 2007 and shut down by Roche in 2013 when its technology became noncompetitive, although production continued until mid-2016.

SNP genotyping is the measurement of genetic variations of single nucleotide polymorphisms (SNPs) between members of a species. It is a form of genotyping, which is the measurement of more general genetic variation. SNPs are one of the most common types of genetic variation. An SNP is a single base pair mutation at a specific locus, usually consisting of two alleles. SNPs are found to be involved in the etiology of many human diseases and are becoming of particular interest in pharmacogenetics. Because SNPs are conserved during evolution, they have been proposed as markers for use in quantitative trait loci (QTL) analysis and in association studies in place of microsatellites. The use of SNPs is being extended in the HapMap project, which aims to provide the minimal set of SNPs needed to genotype the human genome. SNPs can also provide a genetic fingerprint for use in identity testing. The increase of interest in SNPs has been reflected by the furious development of a diverse range of SNP genotyping methods.

Polony sequencing is an inexpensive but highly accurate multiplex sequencing technique that can be used to “read” millions of immobilized DNA sequences in parallel. This technique was first developed by Dr. George Church's group at Harvard Medical School. Unlike other sequencing techniques, Polony sequencing technology is an open platform with freely downloadable, open source software and protocols. Also, the hardware of this technique can be easily set up with a commonly available epifluorescence microscopy and a computer-controlled flowcell/fluidics system. Polony sequencing is generally performed on paired-end tags library that each molecule of DNA template is of 135 bp in length with two 17–18 bp paired genomic tags separated and flanked by common sequences. The current read length of this technique is 26 bases per amplicon and 13 bases per tag, leaving a gap of 4–5 bases in each tag.

<span class="mw-page-title-main">Restriction site associated DNA markers</span> Type of genetic marker

Restriction site associated DNA (RAD) markers are a type of genetic marker which are useful for association mapping, QTL-mapping, population genetics, ecological genetics and evolutionary genetics. The use of RAD markers for genetic mapping is often called RAD mapping. An important aspect of RAD markers and mapping is the process of isolating RAD tags, which are the DNA sequences that immediately flank each instance of a particular restriction site of a restriction enzyme throughout the genome. Once RAD tags have been isolated, they can be used to identify and genotype DNA sequence polymorphisms mainly in form of single nucleotide polymorphisms (SNPs). Polymorphisms that are identified and genotyped by isolating and analyzing RAD tags are referred to as RAD markers. Although genotyping by sequencing presents an approach similar to the RAD-seq method, they differ in some substantial ways.

<span class="mw-page-title-main">DNA nanoball sequencing</span>

DNA nanoball sequencing is a high throughput sequencing technology that is used to determine the entire genomic sequence of an organism. The method uses rolling circle replication to amplify small fragments of genomic DNA into DNA nanoballs. Fluorescent nucleotides bind to complementary nucleotides and are then polymerized to anchor sequences bound to known sequences on the DNA template. The base order is determined via the fluorescence of the bound nucleotides This DNA sequencing method allows large numbers of DNA nanoballs to be sequenced per run at lower reagent costs compared to other next generation sequencing platforms. However, a limitation of this method is that it generates only short sequences of DNA, which presents challenges to mapping its reads to a reference genome. After purchasing Complete Genomics, the Beijing Genomics Institute (BGI) refined DNA nanoball sequencing to sequence nucleotide samples on their own platform.

Massive parallel sequencing or massively parallel sequencing is any of several high-throughput approaches to DNA sequencing using the concept of massively parallel processing; it is also called next-generation sequencing (NGS) or second-generation sequencing. Some of these technologies emerged between 1993 and 1998 and have been commercially available since 2005. These technologies use miniaturized and parallelized platforms for sequencing of 1 million to 43 billion short reads per instrument run.

Magnetic sequencing is a single-molecule sequencing method in development. A DNA hairpin, containing the sequence of interest, is bound between a magnetic bead and a glass surface. A magnetic field is applied to stretch the hairpin open into single strands, and the hairpin refolds after decreasing of the magnetic field. The hairpin length can be determined by direct imaging of the diffraction rings of the magnetic beads using a simple microscope. The DNA sequences are determined by measuring the changes in the hairpin length following successful hybridization of complementary nucleotides.

Chem-seq is a technique that is used to map genome-wide interactions between small molecules and their protein targets in the chromatin of eukaryotic cell nuclei. The method employs chemical affinity capture coupled with massively parallel DNA sequencing to identify genomic sites where small molecules interact with their target proteins or DNA. It was first described by Lars Anders et al. in the January, 2014 issue of "Nature Biotechnology".

Single-cell sequencing examines the nucleic acid sequence information from individual cells with optimized next-generation sequencing technologies, providing a higher resolution of cellular differences and a better understanding of the function of an individual cell in the context of its microenvironment. For example, in cancer, sequencing the DNA of individual cells can give information about mutations carried by small populations of cells. In development, sequencing the RNAs expressed by individual cells can give insight into the existence and behavior of different cell types. In microbial systems, a population of the same species can appear genetically clonal. Still, single-cell sequencing of RNA or epigenetic modifications can reveal cell-to-cell variability that may help populations rapidly adapt to survive in changing environments.

<span class="mw-page-title-main">Circulating tumor DNA</span> Tumor-derived fragmented DNA in the bloodstream

Circulating tumor DNA (ctDNA) is tumor-derived fragmented DNA in the bloodstream that is not associated with cells. ctDNA should not be confused with cell-free DNA (cfDNA), a broader term which describes DNA that is freely circulating in the bloodstream, but is not necessarily of tumor origin. Because ctDNA may reflect the entire tumor genome, it has gained traction for its potential clinical utility; "liquid biopsies" in the form of blood draws may be taken at various time points to monitor tumor progression throughout the treatment regimen.

G&T-seq is a novel form of single cell sequencing technique allowing one to simultaneously obtain both transcriptomic and genomic data from single cells, allowing for direct comparison of gene expression data to its corresponding genomic data in the same cell...

<span class="mw-page-title-main">Duplex sequencing</span>

Duplex sequencing is a library preparation and analysis method for next-generation sequencing (NGS) platforms that employs random tagging of double-stranded DNA to detect mutations with higher accuracy and lower error rates.

Off-target genome editing refers to nonspecific and unintended genetic modifications that can arise through the use of engineered nuclease technologies such as: clustered, regularly interspaced, short palindromic repeats (CRISPR)-Cas9, transcription activator-like effector nucleases (TALEN), meganucleases, and zinc finger nucleases (ZFN). These tools use different mechanisms to bind a predetermined sequence of DNA (“target”), which they cleave, creating a double-stranded chromosomal break (DSB) that summons the cell's DNA repair mechanisms and leads to site-specific modifications. If these complexes do not bind at the target, often a result of homologous sequences and/or mismatch tolerance, they will cleave off-target DSB and cause non-specific genetic modifications. Specifically, off-target effects consist of unintended point mutations, deletions, insertions inversions, and translocations.

Paleogenomics is a field of science based on the reconstruction and analysis of genomic information in extinct species. Improved methods for the extraction of ancient DNA (aDNA) from museum artifacts, ice cores, archeological or paleontological sites, and next-generation sequencing technologies have spurred this field. It is now possible to detect genetic drift, ancient population migration and interrelationships, the evolutionary history of extinct plant, animal and Homo species, and identification of phenotypic features across geographic regions. Scientists can also use paleogenomics to compare ancient ancestors against modern-day humans. The rising importance of paleogenomics is evident from the fact that the 2022 Nobel Prize in physiology or medicine was awarded to a Swedish geneticist Svante Pääbo [1955-], who worked on paleogenomics.

Ancientpathogengenomics is a scientific field related to the study of pathogen genomes recovered from ancient human, plant or animal remains. Ancient pathogens are microorganisms, now extinct, that in the past centuries caused several epidemics and deaths worldwide. Their genome, referred to as ancient DNA (aDNA), is isolated from the burial's remains of victims of the pandemics caused by these pathogens.

GUIDE-Seq is a molecular biology technique that allows for the unbiased in vitro detection of off-target genome editing events in DNA caused by CRISPR/Cas9 as well as other RNA-guided nucleases in living cells. Similar to LAM-PCR, it employs multiple PCRs to amplify regions of interest that contain a specific insert that preferentially integrates into double-stranded breaks. As gene therapy is an emerging field, GUIDE-Seq has gained traction as a cheap method to detect the off-target effects of potential therapeutics without needing whole genome sequencing.

References

  1. 1 2 3 4 5 6 7 8 9 10 Crosetto N, Mitra A, Silva MJ, Bienko M, Dojer N, Wang Q, Karaca E, Chiarle R, Skrzypczak M, Ginalski K, Pasero P, Rowicka M, Dikic I (April 2013). "Nucleotide-resolution DNA double-strand break mapping by next-generation sequencing". Nature Methods. 10 (4): 361–5. doi:10.1038/nmeth.2408. PMC   3651036 . PMID   23503052.
  2. 1 2 "Avidin-Biotin Interaction - CA". www.thermofisher.com. Retrieved 2019-03-01.
  3. Kozubek S, Lukásová E, Amrichová J, Kozubek M, Lisková A, Slotová J (June 2000). "Influence of cell fixation on chromatin topography". Analytical Biochemistry. 282 (1): 29–38. doi:10.1006/abio.2000.4538. PMID   10860496.
  4. "BLESS: Map genome-wide DNA double-strand breaks using next generation sequencing". breakome.utmb.edu. Retrieved 2019-03-01.
  5. Sharma A, Singh K, Almasan A (2012). "Histone H2AX Phosphorylation: A Marker for DNA Damage". DNA Repair Protocols. Methods in Molecular Biology. Vol. 920. pp. 613–26. doi:10.1007/978-1-61779-998-3_40. ISBN   978-1-61779-997-6. PMID   22941631.
  6. 1 2 Yan WX, Mirzazadeh R, Garnerone S, Scott D, Schneider MW, Kallas T, Custodio J, Wernersson E, Li Y, Gao L, Federova Y, Zetsche B, Zhang F, Bienko M, Crosetto N (May 2017). "BLISS is a versatile and quantitative method for genome-wide profiling of DNA double-strand breaks". Nature Communications. 8: 15058. Bibcode:2017NatCo...815058Y. doi:10.1038/ncomms15058. PMC   5437291 . PMID   28497783.
  7. 1 2 Bouwman BA, Crosetto N (December 2018). "Endogenous DNA Double-Strand Breaks during DNA Transactions: Emerging Insights and Methods for Genome-Wide Profiling". Genes. 9 (12): 632. doi: 10.3390/genes9120632 . PMC   6316733 . PMID   30558210.
  8. Biernacka A, Zhu Y, Skrzypczak M, Forey R, Pardo B, Grzelak M, Nde J, Mitra A, Kudlicki A, Crosetto N, Pasero P, Rowicka M, Ginalski K (2018). "i-BLESS is an ultra-sensitive method for detection of DNA double-strand breaks". Communications Biology. 1 (1): 181. doi:10.1038/s42003-018-0165-9. PMC   6208412 . PMID   30393778.
  9. Lensing SV, Marsico G, Hänsel-Hertsch R, Lam EY, Tannahill D, Balasubramanian S (October 2016). "DSBCapture: in situ capture and sequencing of DNA breaks". Nature Methods. 13 (10): 855–7. doi:10.1038/nmeth.3960. PMC   5045719 . PMID   27525976.
  10. Aird D, Ross MG, Chen WS, Danielsson M, Fennell T, Russ C, Jaffe DB, Nusbaum C, Gnirke A (2011). "Analyzing and minimizing PCR amplification bias in Illumina sequencing libraries". Genome Biology. 12 (2): R18. doi: 10.1186/gb-2011-12-2-r18 . PMC   3188800 . PMID   21338519.
  11. Mitra A, Skrzypczak M, Ginalski K, Rowicka M (2015). "Strategies for achieving high sequencing accuracy for low diversity samples and avoiding sample bleeding using illumina platform". PLOS ONE. 10 (4): e0120520. Bibcode:2015PLoSO..1020520M. doi: 10.1371/journal.pone.0120520 . PMC   4393298 . PMID   25860802.
  12. Tsai SQ, Zheng Z, Nguyen NT, Liebers M, Topkar VV, Thapar V, Wyvekens N, Khayter C, Iafrate AJ, Le LP, Aryee MJ, Joung JK (February 2015). "GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases". Nature Biotechnology. 33 (2): 187–197. doi:10.1038/nbt.3117. PMC   4320685 . PMID   25513782.
  13. Aparicio T, Baer R, Gautier J (July 2014). "DNA double-strand break repair pathway choice and cancer". DNA Repair. 19: 169–75. doi:10.1016/j.dnarep.2014.03.014. PMC   4051845 . PMID   24746645.
  14. Fu Y, Foden JA, Khayter C, Maeder ML, Reyon D, Joung JK, Sander JD (September 2013). "High-frequency off-target mutagenesis induced by CRISPR-Cas nucleases in human cells". Nature Biotechnology. 31 (9): 822–6. doi:10.1038/nbt.2623. PMC   3773023 . PMID   23792628.