CRISPR Gene Editing PDF
CRISPR Gene Editing PDF
CRISPR
Gene Editing
Methods and Protocols
METHODS IN MOLECULAR BIOLOGY
Series Editor
John M. Walker
School of Life and Medical Sciences
University of Hertfordshire
Hatfield, Hertfordshire, AL10 9AB, UK
Edited by
Yonglun Luo
Department of Biomedicine, Aarhus University, Aarhus, Denmark
BGI-Shenzhen, Shenzhen, China
Editor
Yonglun Luo
Department of Biomedicine
Aarhus University
Aarhus, Denmark
BGI-Shenzhen
Shenzhen, China
This Humana Press imprint is published by the registered company Springer Science+Business Media, LLC, part of
Springer Nature.
The registered company address is: 233 Spring Street, New York, NY 10013, U.S.A.
Preface
When the series editor, Dr. John M. Walker, asked me approximately 1 year ago whether I
was interested in editing a book on CRISPR technologies, I immediately took his generous
offer and started my first and challenging editorial journey. Firstly, as a junior researcher, it
has always been a great honor and encouraging experience to be capable of contributing
with knowledge and outreach to our scientific community. Secondly, the particular research
field and technology that we will be focusing on in this book is one of the most fast growing
and important breakthroughs during the past decade. And most importantly, as a researcher
working on gene editing for over 10 years, I deeply realize the importance of having a good
serial of protocols, experimental tips, and notes to increase the success rate and outcomes of
scientific projects.
When I first started the “Pig and Health” PhD project back in 2008, my objective was to
recapitulate the pathogenesis of human diseases, e.g., breast cancer and diabetes, using
genetically tailored pig models. One major milestone of my PhD study was disrupting the
BRCA1 gene in primary porcine fibroblasts by homologous recombination and subse-
quently generating a BRCA1 knockout pig by somatic cell nuclear transfer. Generating a
gene knockout (KO) animal during the pre-TALENs and pre-CRISPR era was rather
technically challenging and time-consuming. Although I spent almost 3 years and finally
got my BRCA1 KO pig, I wish my project was conducted now rather than 10 years ago.
Zinc finger nucleases (ZFNs), known as the second generation of programmable DNA
nucleases, were already available during that period. But this technology was not very
broadly adopted by the scientific community. One main reason is that the ZFN technology
is relatively difficult for the design and generation, and there is a lack of user-friendly
protocols and methods instructing the generation and functional validation of ZFNs.
When the transcription activator-like effector (TALE) protein was engineered as a
programmable DNA endonuclease (TALEN) for gene editing, as compared to ZFNs,
TALEN-based gene editing was more rapidly applied by the whole scientific community.
One important driving force of the TALEN technology is conventional web tools and
protocols developed for TALEN vector design and construction. Using one the most
popular TALEN assembly methods developed by Daniel F. Voytas’s group, although one
has to select the different modular plasmids from a large stock of premade ones, it is much
easier when compared to ZFNs to generate a couple of TALEN constructions.
Since the first proof-of-principle study of harnessing the clustered regularly interspaced
short palindromic repeats (CRISPR) and CRISPR-associated protein 9 (Cas9) for gene
editing in 2012, led by Jennifer Doudna and Emmanuelle Charpentier, the CRISPR-Cas9-
based gene editing technology rapidly took over ZFNs and TALENs and was successfully
applied for genome editing in almost all cell types and organisms. To date, many Cas9
orthologs, different CRISPR systems from bacteria and archaea have been repurposed for
gene editing. And many more genetic manipulation tools have been built on top of the
CRISPR-Cas9 system, such as CRISPR-based gene activation, gene interference, base
editing, DNA methylation, and histone acetylation. Among all the already harnessed
CRISPR-Cas systems utilized for gene editing, the CRISPR-Cas9 system is still the most
extensively developed and broadly used one.
v
vi Preface
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix
1 CRISPR-gRNA Design . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
Maria Pallarès Masmitjà, Nastassia Knödlseder, and Marc Güell
2 Tracking CRISPR’s Footprints . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
Lin Lin and Yonglun Luo
3 Rapid Quantitative Evaluation of CRISPR Genome Editing by TIDE
and TIDER . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
Eva Karina Brinkman and Bas van Steensel
4 Fast and Quantitative Identification of Ex Vivo Precise Genome
Targeting-Induced Indel Events by IDAA . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45
Saskia König, Zhang Yang, Hans Heugh Wandall, Claudio Mussolino,
and Eric Paul Bennett
5 Functional Evaluation of CRISPR Activity by the Dual-Fluorescent
Surrogate System: C-Check . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 67
Lin Lin and Yonglun Luo
vii
viii Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 359
Contributors
ix
x Contributors
DARIA SERGEEVA The Novo Nordisk Foundation Center for Biosustainability, Technical
University of Denmark, Kgs. Lyngby, Denmark
KILIAN SIMMET Institute for Molecular Animal Breeding and Biotechnology, LMU Munich,
Munich, Germany
KRISTIAN ALSBJERG SKIPPER Department of Biomedicine, Aarhus University, Aarhus,
Denmark
KIRSTEN E. SNIJDERS Wellcome Trust-MRC Stem Cell Institute, Anne McLaren Laboratory,
University of Cambridge, Cambridge, UK; Department of Surgery, University of
Cambridge, Cambridge, UK
MAGNUS STOUGAARD Department of Clinical Medicine, Aarhus University, Aarhus,
Denmark
JÜRGEN STUMM Max-Delbrück-Centrum für Molekulare Medizin, Berlin, Germany; Berlin
Institute of Health, Berlin, Germany
EMIL AAGAARD THOMSEN Department of Biomedicine, Aarhus University, Aarhus,
Denmark
LUDOVIC VALLIER Wellcome Trust-MRC Stem Cell Institute, Anne McLaren Laboratory,
University of Cambridge, Cambridge, UK; Department of Surgery, University of
Cambridge, Cambridge, UK; Wellcome Trust Sanger Institute, Hinxton, UK
BAS VAN STEENSEL Division of Gene Regulation and Oncode Institute, Netherlands Cancer
Institute, Amsterdam, The Netherlands
PETRA VOCHOZKOVA Institute for Molecular Animal Breeding and Biotechnology, LMU
Munich, Munich, Germany
HANS HEUGH WANDALL Faculty of Health Sciences, Department of Cellular and Molecular
Medicine, Copenhagen Center for Glycomics (CCG), University of Copenhagen,
Copenhagen, Denmark
YUQUAN WEI State Key Laboratory of Biotherapy and Cancer Center, West China Hospital,
Sichuan University, Chengdu, China
QINJIE WU State Key Laboratory of Biotherapy and Cancer Center, West China Hospital,
Sichuan University, Chengdu, China
ANNEGRET WÜNSCH Institute for Molecular Animal Breeding and Biotechnology, LMU
Munich, Munich, Germany
XI XIANG BGI Education Center, University of Chinese Academy of Sciences, Shenzhen,
China; BGI-Shenzhen, Shenzhen, China
SULEIXIN YANG State Key Laboratory of Biotherapy and Cancer Center, West China
Hospital, Sichuan University, Chengdu, China
ZHANG YANG Faculty of Health Sciences, Department of Cellular and Molecular Medicine,
Copenhagen Center for Glycomics (CCG), University of Copenhagen, Copenhagen,
Denmark
SANIYE YUMLU Max-Delbrück-Centrum für Molekulare Medizin, Berlin, Germany; Berlin
Institute of Health, Berlin, Germany
XIUQING ZHANG BGI-Shenzhen, Shenzhen, China
Part I
CRISPR-gRNA Design
Maria Pallarès Masmitjà, Nastassia Knödlseder, and Marc Güell
Abstract
Gene editing has great therapeutic impact, being of interest for many scientists worldwide. Clustered
regularly interspaced short palindromic repeats (CRISPR) technology has been adapted for gene editing
to serve as an efficient, rapid, and cost-effective tool. To fulfill CRISPR experiment’s goals, two components
are important: an endonuclease and a gRNA. The most commonly used endonucleases are Cpf1 and Cas9
and are described in depth in this chapter. The gRNA targets the genome site to be edited, giving great
importance to its design to obtain increased efficiency and decreased off-target events. In this chapter, we
describe different tools to design suitable gRNAs for a variety of experimental purposes.
1 Introduction
Yonglun Luo (ed.), CRISPR Gene Editing: Methods and Protocols, Methods in Molecular Biology, vol. 1961,
https://doi.org/10.1007/978-1-4939-9170-9_1, © Springer Science+Business Media, LLC, part of Springer Nature 2019
3
4 Maria Pallarès Masmitjà et al.
2 Nuclease Types
Table 1
Features of Cas9 and Cpf1
Cas9 Cpf1
Type II V
Cleavage Double-stranded DNA Double-stranded DNA
0
Protospacer-adjacent motif NGG 3 50 -TTN
(PAM)
Ends Blunt end 3 nt upstream 5 nucleotide 50 overhang 18–23 nt from
PAM PAM
CRISPR-gRNA Design 5
3 gRNA Efficiency
gRNAs will guide the CRISPR system molecules to the specific site
intended to edit in the genome. It has a scaffold that will bind the
endonuclease and a spacer of 20 nucleotides which target specific
sequence in the genome.
The efficiency of the gRNA can vary from a sequence to
another, and it is crucial for the genome editing experiment.
Some factors are known to correlate with high efficiency gRNAs
such as higher stability of the duplex between gRNA and target
DNA [11]. Scores tend to correlate with gRNA efficiency, but their
accuracy depends on the experimental system considered [12]. For
instance, Moreno-Mateos score is useful for zebrafish injection
experiments [13] (where gRNAs need to be very stable after injec-
tions including G-rich sequence) but inaccurate in cultured mam-
malian cells [14]. This score is also reliable for assays based on
delivery of gRNAs produced synthetically or by T7 in vitro tran-
scription [15]. The most common scores are based on datasets
where gRNA is continuously expressed from a U6 promoter so
they do not need to be very stable. One of the most used for
mammalian cells is the score by Doench et al. [16]; also the Wang
score is useful when designing second generation libraries of human
gRNAs in some cancer cell lines [17]. It is recommended to use
scoring systems trained with the same expression system planned to
be used. The web tool http://crispor.org provides multiple scores
such as Moreno-Mateos and Doench simultaneously to help select
efficient gRNA for different applications.
Recently, approaches based on machine learning such as
CRISTA have been developed, which determines the propensity
of a genomic site to be cleaved by a gRNA [18].
5 gRNA Design
Table 2
Selected websites for gRNA design
9 On-Target Efficiency
10 Off-Target Analysis
11 Conclusion
References
1. Mojica FJM, Dı́ez-Villaseñor C, Garcı́a- 2. Zhang F, Wen Y, Guo X (2014) CRISPR/Cas9
Martı́nez J, Almendros C (2009) Short motif for genome editing: progress, implications and
sequences determine the targets of the pro- challenges. Hum Mol Genet 23:R40–R46
karyotic CRISPR defence system. Microbiol- 3. Jinek M, Chylinski K, Fonfara I, Hauer M,
ogy 155:733–740 Doudna JA, Charpentier E (2012) A
10 Maria Pallarès Masmitjà et al.
programmable dual-RNA-guided DNA endo- scoring algorithms and integration into the
nuclease in adaptive bacterial immunity. Sci- guide RNA selection tool CRISPOR. Genome
ence 337:816–821 Biol 17:148
4. Nishimasu H, Nureki O (2017) Structures and 16. Doench JG, Fusi N, Sullender M, Hegde M,
mechanisms of CRISPR RNA-guided effector Vaimberg EW, Donovan KF et al (2016) Opti-
nucleases. Curr Opin Struct Biol 43:68–78 mized sgRNA design to maximize activity and
5. Mali P, Yang L, Esvelt KM, Aach J, Guell M, minimize off-target effects of CRISPR-Cas9.
DiCarlo JE et al (2013) RNA-guided human Nat Biotechnol 34:184–191
genome engineering via Cas9. Science 17. Wang T, Birsoy K, Hughes NW, Krupczak KM,
339:823–826 Post Y, Wei JJ et al (2015) Identification and
6. Makarova KS, Grishin NV, Shabalina SA, Wolf characterization of essential genes in the
YI, Koonin EV (2006) A putative RNA- human genome. Science 350:1096–1101
interference-based immune system in prokar- 18. Abadi S, Yan WX, Amar D, Mayrose I (2017) A
yotes: computational analysis of the predicted machine learning approach for predicting
enzymatic machinery, functional analogies with CRISPR-Cas9 cleavage efficiencies and pat-
eukaryotic RNAi, and hypothetical mechan- terns underlying its mechanism of action.
isms of action. Biol Direct 1:7 PLoS Comput Biol. Public Library of Science
7. Ran FA, Hsu PD, Lin C-Y, Gootenberg JS, 13:e1005807
Konermann S, Trevino AE et al (2013) Double 19. Kleinstiver BP, Tsai SQ, Prew MS, Nguyen NT,
nicking by RNA-guided CRISPR Cas9 for Welch MM, Lopez JM et al (2016) Genome-
enhanced genome editing specificity. Cell wide specificities of CRISPR-Cas Cpf1
154:1380–1389 nucleases in human cells. Nat Biotechnol.
8. Wen W-S, Yuan Z-M, Ma S-J, Xu J, Yuan D-T NIH Public Access 34:869
(2016) CRISPR-Cas9 systems: versatile cancer 20. Fu Y, Foden JA, Khayter C, Maeder ML,
modelling platforms and promising therapeutic Reyon D, Keith Joung J et al (2013) High
strategies. Int J Cancer 138:1328–1336 frequency off-target mutagenesis induced by
9. Kim HK, Song M, Lee J, Vipin Menon A, CRISPR-Cas nucleases in human cells. Nat
Jung S, Kang Y-M et al (2016) In vivo high- Biotechnol. NIH Public Access 31:822
throughput profiling of CRISPR–Cpf1 activity. 21. Cho SW, Kim S, Kim Y, Kweon J, Kim HS, Bae
Nat Methods 14:153–159 S et al (2014) Analysis of off-target effects of
10. Friedland AE, Baral R, Singhal P, Loveluck K, CRISPR/Cas-derived RNA-guided endonu-
Shen S, Sanchez M et al (2015) Characteriza- cleases and nickases. Genome Res. Cold Spring
tion of Staphylococcus aureus Cas9: a smaller Harbor Laboratory Press 24:132
Cas9 for all-in-one adeno-associated virus 22. Slaymaker IM, Gao L, Zetsche B, Scott DA,
delivery and paired nickase applications. Yan WX, Zhang F (2016) Rationally engi-
Genome Biol 16:257. https://doi.org/10. neered Cas9 nucleases with improved specific-
1186/s13059-015-0817-8 ity. Science 351:84–88
11. Wong N, Liu W, Wang X (2015) WU-CRISPR: 23. Zhang XH, Tee LY, Wang XG, Huang QS,
characteristics of functional guide RNAs for Yang SH (2015) Off-target effects in
the CRISPR/Cas9 system. Genome Biol CRISPR/Cas9-mediated genome engineer-
16:218 ing. Mol Ther Nucleic Acids. Cell Press 4:
12. Haeussler M, Concordet J-P (2016) Genome e264. https://doi.org/10.1038/mtna.2015.
editing with CRISPR-Cas9: can it get any bet- 37
ter? J Genet Genomics 43:239–250 24. Jeffry D, Sander JKJ (2014) CRISPR-Cas sys-
13. Moreno-Mateos MA, Vejnar CE, Beaudoin tems for genome editing, regulation and tar-
J-D, Fernandez JP, Mis EK, Khokha MK et al geting. Nat Biotechnol. NIH Public Access
(2015) CRISPRscan: designing highly efficient 32:347
sgRNAs for CRISPR-Cas9 targeting in vivo. 25. Yang L, Guell M, Byrne S, Yang JL, De Los
Nat Methods 12:982–988 Angeles A, Mali P et al (2013) Optimization of
14. Malina A, Cameron CJF, Robert F, scarless human stem cell genome editing.
Blanchette M, Dostie J, Pelletier J (2015) Nucleic Acids Res 41:9049–9061
PAM multiplicity marks genomic target sites 26. Richardson CD, Ray GJ, DeWitt MA, Curie
as inhibitory to CRISPR-Cas9 editing. Nat GL, Corn JE (2016) Enhancing homology-
Commun 6:10124 directed genome editing by catalytically active
15. Haeussler M, Schönig K, Eckert H, and inactive CRISPR-Cas9 using asymmetric
Eschstruth A, Mianné J, Renaud J-B et al donor DNA. Nat Biotechnol 34:339–344
(2016) Evaluation of off-target and on-target
CRISPR-gRNA Design 11
27. Shrock E, Güell M (2017) CRISPR in animals 30. Tsai SQ, Zheng Z, Nguyen NT, Liebers M,
and animal models. Prog Mol Biol Transl Sci. Topkar VV, Thapar V et al (2015) GUIDE-
Academic Press 152:95–114 seq enables genome-wide profiling of
28. Guell M, Yang L, Church GM (2014) Genome off-target cleavage by CRISPR-Cas nucleases.
editing assessment using CRISPR Genome Nat Biotechnol 33:187–197
Analyzer (CRISPR-GA). Bioinformatics 31. Tsai SQ, Nguyen NT, Malagon-Lopez J, Top-
30:2968–2970 kar VV, Aryee MJ, Joung JK (2017) CIRCLE-
29. Pinello L, Canver MC, Hoban MD, Orkin SH, seq: a highly sensitive in vitro screen for
Kohn DB, Bauer DE et al (2016) Analyzing genome-wide CRISPR-Cas9 nuclease
CRISPR genome-editing experiments with off-targets. Nat Methods 14:607–614
CRISPResso. Nat Biotechnol 34:695–697
Chapter 2
Abstract
The programmable clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-
associated 9 (Cas9) and CRISPR-Cas9-derived gene editing and manipulation tools have revolutionized
biomedical research over the past few years. One important category of assisting technologies in CRISPR
gene editing is methods used for detecting and quantifying indels (deletions or insertions). These indels are
caused by the repair of CRISPR-Cas9-introduced DNA double-stranded breaks (DBSs), known as
CRISPR’s DNA cleavage footprints. In addition, CRISPR-Cas9 can also leave footprints to the DNA
without introducing DSBs, known as CRISPR’s DNA-binding footprints. The indel tracking methods have
contributed greatly to the improvement of CRISPR-Cas9 activity and specificity. Here, we review and
discuss strategies developed over that past few years to track the CRISPR’s footprints, their advantages, and
limitations.
Yonglun Luo (ed.), CRISPR Gene Editing: Methods and Protocols, Methods in Molecular Biology, vol. 1961,
https://doi.org/10.1007/978-1-4939-9170-9_2, © Springer Science+Business Media, LLC, part of Springer Nature 2019
13
14 Lin Lin and Yonglun Luo
Fig. 1 Schematic illustration of DSB repair by homology-directed repair (HDR), non-homologous end joining
(NHEJ), microhomology-mediated end joining (MMEJ), and single strand annealing in mammalian cells. HDR
leads to precise and intended gene insertion, replacement, or modification. Indels (deletions or insertions) are
typically introduced by NHEJ. Variable-size small and large deletions are introduced by MMEJ and SSA,
respectively, after the repair of DSBs
gene editing tools are not very broadly adopted mostly due to the
technical difficulty and the workload of designing and generating a
working MGNs or ZFNs. The Transcription Activator-Like Effec-
tor Nucleases (TALENs) technology is the second revolution in the
gene editing history. Compared to MGNs and ZFNs, TALENs
hold a much more stringent protein-to-DNA code for targeting.
The DNA-binding domain of a TALEN is defined by the number
and order of four types of modular repeated domains. These highly
conserved 33–34 amino acids repeated domains contain two diver-
gent 12th and 13th amino acids, also known as repeat variable
diresidue (RVD): NG, HD, NI, NN/NH/NK that target T,
C, A, G nucleotide respectively. Nevertheless, it is still quite time-
consuming to generate a pair of TALEN vectors, although the
whole procedure of generating TALENs has been greatly simplified
compared to MGNs and ZFNs. The third generation of revolution
in gene editing history is the RNA-guided endonucleases, also
known as the clustered regularly interspaced short palindromic
repeats (CRISPR) and CRISPR-associated protein (Cas) gene edit-
ing technologies [12–14].
The CRISPR-Cas gene editing technology has overcome
almost all the disadvantages of previous gene editing tools. Unlike
MGNs, ZFNs, and TALENs, the CRISPR gene editing tool is
based on RNA-to-DNA code for targeting. Thus, the sole Cas
protein, with the Streptococcus Pyogenes Cas9 protein (SpCas9)
being the most broadly used one, can be used for gene editing in
almost all cells and organisms [15]. Designing and generating a
new CRISPR targeting component is now simply achieved through
changing the guide sequences presented in a small RNA molecule,
known as guide RNA (gRNA) or small guide RNA (sgRNA). In the
SpCas9-based CRISPR gene editing system, the gRNA is a chime-
ric RNA (crRNA:tracrRNA), which interacts with SpCas9 and
guides SpCas9 to the target site through a Watson-Crick base
pairing between the guide sequences of crRNA and the target
site. As the guide or spacer sequences are the same as the target
site (protospacer), the bacterial Cas protein has evolved an
extremely smart system to distinguish self and non-self sequences,
which is the of protospacer adjacent motif (PAM)-dependent DNA
cleavage system [12]. Once all these features are met, the endonu-
clease activity of Cas9 protein is activated and a DSB is introduced
in the target sites (Fig. 2).
One essentially common feature and outcome of all these gene
editing tools is the introduction of a DSB to the target site. These
DSBs are repaired by endogenous DNA repair machineries, leading
to DNA alterations such as small deletions or insertions (called
indels) at the DSB sites. These indels are just like footprints left
by CRISPR to the DNA. Thus, quantification of the indel fre-
quency has been used to quantify the overall activity and specificity
(off-target) of these gene editing tools. Several methods have been
16 Lin Lin and Yonglun Luo
Fig. 2 Schematic illustration of CRISPR-Cas9-mediated gene targeting and the core components. The SpCas9
protein has two nuclease domains: RuvC and HNH. The first step of CRISPR gene editing is delivery of Cas9
and gRNA into cells, which can form Cas9:gRNA complex. The next step is the nucleus entry of Cas9:gRNA
complex, mediated by the nucleus localization signal (NLS) peptides fused Cas9. The last step of CRISPR gene
editing is the repair of DSBs introduced by Cas9:gRNA with DSBs repair machineries
developed over the past decade for quantifying indel frequency and
specificity (Fig. 3). To give readers a systematic overview and help
with choosing the right method for their CRISPR gene editing
experiment, this chapter reviews the advantage and disadvantage of
different methods.
Fig. 3 Schematic illustration of the procedures and methods used for CRISPR indel profiling. Mismatch-
specific enzyme assay, TIDE, IDAA, HRM, and Drop-Off methods are presented
[32, 33]. The HRM method has been used for epigenetic genotyp-
ing, high-throughput genotyping, and SNP genotyping. Com-
pared to enzymatic-based methods and TIDE, the HRM method
can provide a much more cost-effective and high-throughput
option for screening a large number of gene-edited clonal cells
(Fig. 3). For HRM, the PCR reaction and melting curve determi-
nation can be performed in the same reaction tube consecutively,
thus avoiding many practical steps such as purification of PCR
product, concentration measuring, enzymatic-reaction and gel
electrophoresis for enzymatic-based method, or preparation of
Sanger sequencing for TIDE. However, the HRM method is not
sensitive enough to measure overall indel frequency as compared to
the aforementioned methods. In addition, the HRM method can-
not quantify the frequency of different types of indels.
Fig. 4 Profiling of CRISPR indel patterns of three gRNAs by TIDE in HEK293T cells. Experiments have been
conducted in triplicates and tested with wild-type SpCas9 and the enhanced SpCas9 (eSpCas9)
Fig. 5 Profiling of CRISPR indel patterns of three gRNAs with wild-type SpCas9 or the enhanced SpCas9
(eSpCas9) by TIDE in HEK293T and Hela cells
Tracking CRISPR’s Footprints 25
11 Concluding Remarks
Acknowledgments
References
1. Chang HHY et al (2017) Non-homologous recombinational DNA repair. Cold Spring
DNA end joining and alternative pathways to Harb Perspect Biol 6(9):a016428
double-strand break repair. Nat Rev Mol Cell 8. Capecchi MR (2005) Gene targeting in mice:
Biol 18(8):495–506 functional analysis of the mammalian genome
2. McVey M, Lee SE (2008) MMEJ repair of for the twenty-first century. Nat Rev Genet 6
double-strand breaks (director’s cut): deleted (6):507–512
sequences and alternative endings. Trends 9. Epinat JC et al (2003) A novel engineered
Genet 24(11):529–538 meganuclease induces homologous recombi-
3. Ceccaldi R et al (2016) Repair pathway choices nation in yeast and mammalian cells. Nucleic
and consequences at the double-strand break. Acids Res 31(11):2952–2962
Trends Cell Biol 26(1):52–64 10. Arnould S et al (2007) Engineered I-CreI deri-
4. Lahue RS et al (1989) DNA mismatch correc- vatives cleaving sequences from the human
tion in a defined system. Science 245 XPC gene can induce highly efficient gene cor-
(4914):160–164 rection in mammalian cells. J Mol Biol 371
5. Lindahl T (1974) An N-glycosidase from (1):49–65
Escherichia coli that releases free uracil from 11. Boch J et al (2009) Breaking the code of DNA
DNA containing deaminated cytosine residues. binding specificity of TAL-type III effectors.
Proc Natl Acad Sci U S A 71(9):3649–3653 Science 326(5959):1509–1512
6. Sancar A, Rupp WD (1983) A novel repair 12. Jinek M et al (2012) A programmable dual-
enzyme: UVRABC excision nuclease of RNA-guided DNA endonuclease in adaptive
Escherichia coli cuts a DNA strand on both bacterial immunity. Science 337
sides of the damaged region. Cell 33 (6096):816–821
(1):249–260 13. Cong L et al (2013) Multiplex genome engi-
7. Mehta A, Haber JE (2014) Sources of DNA neering using CRISPR/Cas systems. Science
double-strand breaks and models of 339(6121):819–823
Tracking CRISPR’s Footprints 27
14. Mali P et al (2013) RNA-guided human 29. Dehairs J et al (2016) CRISP-ID: decoding
genome engineering via Cas9. Science 339 CRISPR mediated indels by Sanger sequenc-
(6121):823–826 ing. Sci Rep 6:28973
15. Barrangou R, Doudna JA (2016) Applications 30. Yang Z et al (2015) Fast and sensitive detection
of CRISPR technologies in research and of indels induced by precise gene targeting.
beyond. Nat Biotechnol 34(9):933–941 Nucleic Acids Res 43(9):e59
16. Yeung AT et al (2005) Enzymatic mutation 31. Ramlee MK et al (2015) High-throughput
detection technologies. BioTechniques 38 genotyping of CRISPR/Cas9-mediated
(5):749–758 mutants using fluorescent PCR-capillary gel
17. Zhu X et al (2014) An efficient genotyping electrophoresis. Sci Rep 5:15587
method for genome-modified animals and 32. D’Agostino Y et al (2016) A rapid and cheap
human cells generated with CRISPR/Cas9 sys- methodology for CRISPR/Cas9 zebrafish
tem. Sci Rep 4:6420 mutant screening. Mol Biotechnol 58
18. Liu C et al (2017) Delivery strategies of the (1):73–78
CRISPR-Cas9 gene-editing system for thera- 33. Samarut E et al (2016) A simplified method for
peutic applications. J Control Release identifying early CRISPR-induced indels in
266:17–26 zebrafish embryos using High Resolution
19. Li L et al (2018) Non-viral delivery systems for Melting analysis. BMC Genomics 17:547
CRISPR/Cas9-based genome editing: chal- 34. Findlay SD et al (2016) A digital PCR-based
lenges and opportunities. Biomaterials method for efficient and highly specific screen-
171:207–218 ing of genome edited cells. PLoS One 11(4):
20. Senis E et al (2014) CRISPR/Cas9-mediated e0153901
genome engineering: an adeno-associated viral 35. Pinheiro LB et al (2012) Evaluation of a drop-
(AAV) vector toolbox. Biotechnol J 9 let digital polymerase chain reaction format for
(11):1402–1412 DNA copy number quantification. Anal Chem
21. Schmidt F, Grimm D (2015) CRISPR genome 84(2):1003–1011
engineering and viral gene delivery: a case of 36. Kim H et al (2011) Surrogate reporters for
mutual attraction. Biotechnol J 10 enrichment of cells with nuclease-induced
(2):258–272 mutations. Nat Methods 8(11):941–943
22. Ehrke-Schulz E et al (2016) Quantification of 37. Wen Y et al (2017) A stable but reversible
designer nuclease induced mutation rates: a integrated surrogate reporter for assaying
direct comparison of different methods. Mol CRISPR/Cas9-stimulated homology-directed
Ther Methods Clin Dev 3:16047 repair. J Biol Chem 292(15):6148–6162
23. Sentmanat MF et al (2018) A survey of valida- 38. Zhou Y et al (2016) Enhanced genome editing
tion strategies for CRISPR-Cas9 editing. Sci in mammalian cells with a modified dual-
Rep 8(1):888 fluorescent surrogate system. Cell Mol Life
24. van Overbeek M et al (2016) DNA repair Sci 73(13):2543–2563
profiling reveals nonrandom outcomes at 39. Fu L et al (2016) A simple and efficient method
Cas9-mediated breaks. Mol Cell 63 to visualize and quantify the efficiency of chro-
(4):633–646 mosomal mutations from genome editing. Sci
25. Brinkman EK et al (2014) Easy quantitative Rep 6:35488
assessment of genome editing by sequence 40. Yang Y et al (2016) Highly efficient and rapid
trace decomposition. Nucleic Acids Res 42 detection of the cleavage activity of Cas9/
(22):e168 gRNA via a fluorescent reporter. Appl Biochem
26. Brinkman EK et al (2018) Easy quantification Biotechnol 180(4):655–667
of template-directed CRISPR/Cas9 editing. 41. Ramakrishna S et al (2014) Surrogate reporter-
Nucleic Acids Res 46(10):e58 based enrichment of cells containing
27. Lin L et al (2017) Fusion of SpCas9 to E. coli RNA-guided Cas9 nuclease-induced muta-
Rec A protein enhances CRISPR-Cas9 tions. Nat Commun 5:3378
mediated gene knockout in mammalian cells. 42. Hussmann D et al (2017) IGF1R depletion
J Biotechnol 247:42–49 facilitates MET-amplification as mechanism of
28. Jensen KT et al (2017) Chromatin accessibility acquired resistance to erlotinib in HCC827
and guide sequence secondary structure affect NSCLC cells. Oncotarget 8
CRISPR-Cas9 gene editing efficiency. FEBS (20):33300–33315
Lett 591(13):1892–1901
28 Lin Lin and Yonglun Luo
43. Xue LJ, Tsai CJ (2015) AGEseq: analysis of TALENs using integrase-defective lentiviral
genome editing by sequencing. Mol Plant 8 vectors. Nat Biotechnol 33(2):175–178
(9):1428–1430 58. Crosetto N et al (2013) Nucleotide-resolution
44. Pinello L et al (2016) Analyzing CRISPR DNA double-strand break mapping by next-
genome-editing experiments with CRIS- generation sequencing. Nat Methods 10
PResso. Nat Biotechnol 34(7):695–697 (4):361–365
45. Boel A et al (2016) BATCH-GE: batch analysis 59. Smith C et al (2014) Whole-genome sequenc-
of next-generation sequencing data for ing analysis reveals high specificity of CRISPR/
genome editing assessment. Sci Rep 6:30330 Cas9 and TALEN-based genome editing in
46. Qi LS et al (2013) Repurposing CRISPR as an human iPSCs. Cell Stem Cell 15(1):12–13
RNA-guided platform for sequence-specific 60. Veres A et al (2014) Low incidence of
control of gene expression. Cell 152 off-target mutations in individual CRISPR-
(5):1173–1183 Cas9 and TALEN targeted human stem cell
47. Ran FA et al (2013) Double nicking by clones detected by whole-genome sequencing.
RNA-guided CRISPR Cas9 for enhanced Cell Stem Cell 15(1):27–30
genome editing specificity. Cell 154 61. Slaymaker IM et al (2016) Rationally engi-
(6):1380–1389 neered Cas9 nucleases with improved specific-
48. Hilton IB et al (2015) Epigenome editing by a ity. Science 351(6268):84–88
CRISPR-Cas9-based acetyltransferase activates 62. Kleinstiver BP et al (2016) High-fidelity
genes from promoters and enhancers. Nat Bio- CRISPR-Cas9 nucleases with no detectable
technol 33(5):510–517 genome-wide off-target effects. Nature 529
49. Vad-Nielsen J et al (2018) Simple method for (7587):490–495
assembly of CRISPR synergistic activation 63. Kuscu C et al (2014) Genome-wide analysis
mediator gRNA expression array. J Biotechnol reveals characteristics of off-target sites bound
274:54–57 by the Cas9 endonuclease. Nat Biotechnol 32
50. Xiong K et al (2017) RNA-guided activation of (7):677–683
pluripotency genes in human fibroblasts. Cell 64. Lin L et al (2018) Genome-wide determina-
Reprogram 19(3):189–198 tion of on-target and off-target characteristics
51. Gaudelli NM et al (2017) Programmable base for RNA-guided DNA methylation by dCas9
editing of A*T to G*C in genomic DNA with- methyltransferases. Gigascience 7(3):1–19
out DNA cleavage. Nature 551 65. Kosicki M et al (2017) Dynamics of indel pro-
(7681):464–471 files induced by various CRISPR/Cas9 delivery
52. Kim D et al (2015) Digenome-seq: genome- methods. Prog Mol Biol Transl Sci 152:49–67
wide profiling of CRISPR-Cas9 off-target 66. Kosicki M et al (2018) Repair of double-strand
effects in human cells. Nat Methods 12 breaks induced by CRISPR-Cas9 leads to large
(3):237–243. 1 p following 243 deletions and complex rearrangements. Nat
53. Tsai SQ et al (2017) CIRCLE-seq: a highly Biotechnol 36:765–771
sensitive in vitro screen for genome-wide 67. Haeussler M et al (2016) Evaluation of
CRISPR-Cas9 nuclease off-targets. Nat Meth- off-target and on-target scoring algorithms
ods 14(6):607–614 and integration into the guide RNA selection
54. Cameron P et al (2017) Mapping the genomic tool CRISPOR. Genome Biol 17(1):148
landscape of CRISPR-Cas9 cleavage. Nat 68. Shou J et al (2018) Precise and predictable
Methods 14(6):600–606 CRISPR chromosomal rearrangements reveal
55. Frock RL et al (2015) Genome-wide detection principles of Cas9-mediated nucleotide inser-
of DNA double-stranded breaks induced by tion. Mol Cell 71(4):498–509.e4
engineered nucleases. Nat Biotechnol 33 69. Henrik Devitt Møller LL, Xi X, Petersen TS,
(2):179–186 Huang J, Yang L, Kjeldsen E, Jensen UB,
56. Tsai SQ et al (2015) GUIDE-seq enables Zhang X, Liu X, Xun X, Wang J, Yang H,
genome-wide profiling of off-target cleavage Church GM, Bolund L, Regenberg B, Luo Y
by CRISPR-Cas nucleases. Nat Biotechnol 33 (2018) CRISPR-C: circularization of genes
(2):187–197 and chromosome by CRISPR in human cells.
57. Wang X et al (2015) Unbiased detection of Nucleic Acids Res. https://doi.org/10.1093/
off-target cleavage by CRISPR-Cas9 and nar/gky767
Chapter 3
Abstract
Current genome editing tools enable targeted mutagenesis of selected DNA sequences in many species.
However, the efficiency and the type of introduced mutations by the genome editing method are largely
dependent on the target site. As a consequence, the outcome of the editing operation is difficult to predict.
Therefore, a quick assay to quantify the frequency of mutations is vital for a proper assessment of genome
editing actions. We developed two methods that are rapid, cost-effective, and readily applicable: (1) TIDE,
which can accurately identify and quantify insertions and deletions (indels) that arise after introduction of
double strand breaks (DSBs); (2) TIDER, which is suited for template-mediated editing events including
point mutations. Both methods only require a set of PCR reactions and standard Sanger sequencing runs.
The sequence traces are analyzed by the TIDE or TIDER algorithm (available at https://tide.nki.nl or
https://deskgen.com). The routine is easy, fast, and provides much more detailed information than current
enzyme-based assays. TIDE and TIDER accelerate testing and designing of DSB-based genome editing
strategies.
Key words CRISPR-Cas, Genome editing, Indel mutation, Mutagenesis, DNA mutational analysis/
methods, Sanger sequencing, Web tool, Algorithm
1 Introduction
Yonglun Luo (ed.), CRISPR Gene Editing: Methods and Protocols, Methods in Molecular Biology, vol. 1961,
https://doi.org/10.1007/978-1-4939-9170-9_3, © The Author(s) 2019
29
30 Eva Karina Brinkman and Bas van Steensel
2 Materials
2.1 Guide RNA TIDE and TIDER are suitable for any species in which genomic
Design editing experiments can be performed. CRISPR guide RNAs can
be designed using various online design web tools (e.g., http://
crispr.mit.edu/, https://chopchop.rc.fas.harvard.edu/, https://
www.deskgen.com/).
CRISPR Editing Evaluation by TIDE/TIDER 31
2.2 DNA Purification Usually, 1–3 days after transfection genomic DNA is isolated.
Buffers and Solutions Genomic DNA of a minimum of 1000 cells should be isolated to
get a comprehensive sampling of the complexity of the mutations
that are introduced by the repair of the CRISPR-Cas9 double
strand break. A standard genomic DNA isolation Kit (e.g., BioLine
ISOLATE II Genomic DNA Kit) can be used according to the
manufacturer’s protocol. DNA can also be isolated with the proto-
col for isolation of high-molecular-weight DNA from mammalian
cells using proteinase K and phenol/chloroform extraction [15].
2.3 PCR PCR reactions are carried out with primers surrounding the
Amplification expected break site. We advise to amplify and sequence a stretch
of Control of DNA 500–1500 bp enclosing the designed editing site. The
and Experimental projected break site should be located preferably ~200 bp down-
Sample DNA stream from the sequencing start site.
1. Genomic DNA.
2. PCR primers (see Note 1).
3. PCR master mix (example makes 50 μL):
21- μL H2O
2 μL Primer a (10 μM stock)
2 μL Primer b (10 μM stock)
μL Genomic DNA (~50 ng)
25 μL 2 pre-mix of buffer, Taq polymerase, and dNTPs
(e.g., BioLine MyTaq)
4. PCR program:
4. PCR program:
18 μL H2O
2 μL Primer a (10 μM stock)
2 μL Primer b (10 μM stock)
3 μL Annealed oligo mix
25 μL 2 pre-mix of buffer, Taq polymerase, and dNTPs (e.g., BioLine
MyTaq)
CRISPR Editing Evaluation by TIDE/TIDER 33
8. PCR program:
2.5 Sanger We strongly recommend that all PCR products (control, experi-
Sequencing mental sample(s), and for TIDER also the reference) are sequenced
in parallel. Purified PCR samples are prepared for Sanger sequenc-
ing with the following protocol or can be send for commercial
Sanger sequencing.
1. Purified PCR samples (100 ng).
2. PCR primers. Similar primers as in Subheading 2.3 can be used
(see Notes 1 and 3).
3. PCR master mix (example makes 20 μL):
15.5- μL H2O
0.5 μL Primer a or primer b (10 μM stock)
μL Purified PCR samples (100 ng)
4 μL BigDye (e.g., BigDye® terminator v3.1 of Applied Biosystems)
PCR program:
2.7 Software The TIDE and TIDER web tools are both available at https://tide.
nki.nl or https://deskgen.com.
3 Methods
3.1 Control For both methods genomic DNA is isolated from the cell pool that
and Experimental was transfected with the nuclease or guide RNA alone (control) and
Sample Generation from cells exposed to both Cas9 and guide RNA (experimental
sample). For TIDER the experimental sample is also
co-transfected with the donor template. Then a region of about
500–1500 base pairs around the target site is amplified by PCR
from DNA of the control and experimental sample (Fig. 1a, b).
Next, the PCR amplicons are subjected to conventional Sanger
sequencing. In the PCR product of the experimental sample, the
sequence trace may consist of a combination of multiple sequences
derived from unmodified DNA and DNA that has acquired a
mutation (Fig. 2a).
3.2 Reference TIDER is required for genome editing experiments in the presence
Sample Generation of a donor template. In addition to the control and experimental
(TIDER Only) sample trace (see Subheading 3.1), TIDER requires one extra San-
ger sequencing trace called “reference.” The reference is similar to
the control sequence, except that it carries the desired base pair
changes as designed in the donor template (Fig. 2e). There are two
paths to obtain the reference sequence as described below.
The reference sequence can be easily created in a 2-step PCR
protocol based on site-directed mutagenesis [16]. Here, two addi-
tional primers are required that overlap and carry the desired muta-
tion(s) (mutated primers c, d, which are reverse complement of
each other) (Fig. 1c). These primers are used in combination with
the primers used for the amplification of the control and experi-
mental sample (control primers a, b). The control forward primer a
is combined with the reverse mutated primer c and the forward
mutated primer d with the control reverse primer b, resulting in
two PCR amplicons that incorporate the designed mutations. Then
the two amplicons are denatured and hybridized at the comple-
mentary ends in an annealing reaction. The second PCR uses the
CRISPR Editing Evaluation by TIDE/TIDER 35
TIDE
TIDER
mix, denature,
anneal, extend
primer a
primer b
PCR
designed bp changes
mutations after DSB repair
Fig. 1 Method to generate the required input samples for TIDE and TIDER. Control and test samples can be
obtained by PCR using primers spanning the CRISPR target site (primers a, b). The reference sequence (TIDER
only) can be created in a similar way as site-directed mutagenesis [16] (see Subheading 3.2 for detailed
explanation)
3.3 Web Tool The PCR products of the control, optional reference, and experi-
mental sample are processed by conventional Sanger sequencing.
The resulting sequence trace files (.ab1 or .scf format) are then
uploaded into the TIDE or TIDER web tool (both available at
http://tide.nki.nl and https://deskgen.com). In addition, a char-
acter string representing the guide RNA sequence (20 nt) is
required as input (see Notes 4 and 5). Then, the software will
perform several calculations. First, the guide RNA sequence is
36 Eva Karina Brinkman and Bas van Steensel
input output
a
wild type sgRNA
b c d
GGGATCACTCTCGGCATGGA
20 40 60 80 100
20 30 40
trace decomposition
C
% aberant sequence
alignment decomposition window 34%
% of sequence
window 31% T
A
TIDE
G
17 %
14 %
mixed pool
10
0
0
alignment window decomposition window 200 300 400 500 −10 −5 0 5 10
e
sgRNA
basepair
GGGATCACTCTCGGCATGGA deletion insertion
f g h
mixed pool donor template wild type
0 20 40 60 80 100
% designed nucletoide(s)
40
40
alignment decomposition window 35%
trace decomposition
window
% of sequence
% of sequence
30
30
TIDER
24%
20
20
16%
14%
10%
10
10
bp changes
0
0
200 300 400 500 −10 −5 0 5 HDR
basepair deletion insertion
Fig. 2 Overview of TIDE and TIDER algorithm. Due to imperfect repair (and repair by homology-directed repair
with a donor template) after cutting by a targeted nuclease, the DNA in the cell pool consists of a mixture of
indels (and designed mutations). The various introduced mutations in the pool are disentangled by TIDE or
TIDER. (a) TIDE requires as input a guide RNA sequence string and two sequences are required: (1) wild-type
control, (2) composite test sample. (b) For quality control the aberrant sequence signal is visualized in control
(black) and treated sample (green), the expected break site (vertical dotted line), region used for alignment
(pink bar), and the region used for decomposition (gray bar). A constant composite sequence signal is yielded
after the break site. (c) Trace decomposition yields the spectrum of indels with their frequencies. (d) In
presence of þ1 insertions, the base composition is estimated. (e) Input files for TIDER are identical to TIDE and
one additional sequence file with designed mutations in the used donor template. (f) Quality plot showing only
the proportion of desired mutated nucleotide(s) as designed in donor template that is/are present in the control
(black) and treated sample (green). The region for alignment (pink bar) and decomposition (gray bar) as used in
TIDER are represented. (g and h) Decomposition gives the spectrum of indels (g) and the HDR events (h) with
their frequencies
3.4 Quality Control For generation of the quality control plot the signals of all
nucleotides: A, G, T, C at each position in the sequence file are
used. In general, each position in the sequence trace is represented
CRISPR Editing Evaluation by TIDE/TIDER 37
3.5 Mutation For the detection of individual mutations with the corresponding
Detection by frequencies, the TIDE and TIDER software perform a decomposi-
Decomposition tion of the mixed sequence signal in the experimental sample. This
composite sequence trace is a linear combination of the wild type
(control) and the mutated sequences. For TIDE, the decomposi-
tion is performed on a sequence segment downstream of the break
site. As a rule of thumb, the larger the decomposition window
38 Eva Karina Brinkman and Bas van Steensel
4 Notes
20 40 60 80 100
| = expected break site
20
decomposition window
% aberant sequence R2=0.80
Cas9:
% of sequence
PAM
15
12 12 GTACGGCTCTCACTAGG|GCCTCC
11
CATGCCGAGAGTGATCC|CGGAGG
10
9.5 9.5
9
8 Cpf1:
7
PAM
5
TTTCGAGAAGTCATCTAATAAGG|CCACTGTTA
AAAGCTCTTCAGTAGATTATTCCGGTG|ACAAT
0
0
100 200 300 400 500 −10 −5 0 5 −10
basepair deletion insertion
b
expected break site
20 40 60 80 100
20
decomposition window
% aberant sequence
R2=0.94
% of sequence
15
14
13
11 11 11
10
9 9 8.5
6.5
5
0
c g
expected break site CGGACATCCTCCTTTTCCCA
20 40 60 80 100
rc gRNA:
T C
% aberant sequence
CCAATGGGAACCACGGACATCCTCCYTTTWCCAGCAGGAAGAGCAG
0
Fig. 3 Troubleshooting with TIDE and TIDER. All parameters in TIDE(R) have default settings but can be
adjusted if necessary. Different settings are often a remedy to solve error messages. (a–i) Examples of most
common error messages with the recommended setting changes. (a, b) Avoid the decomposition window to
overlap with high aberrant signal in the control. This occurs often near the ends of the sequence traces (a) or in
a stretch of repetitive sequences (b). (c, d) Alignment problems can occur when the alignment window is too
small (default is from 100 until 15 bp upstream break site) (c) or when the wrong nucleotides in the files are
aligned (d). The alignment window can be changed closer or further to 1 in the sequence trace. (e) The
presence of indels larger than the default of 10 is not included in decomposition and can result in low R2 score.
Indel size can be changed. (f) The use of other nucleases than Cas9 in TIDE(R) works when the guide RNA
string is mimicked to the 20 nt Cas9 guide RNA that cuts between nucleotide 17 and 18. (g) A mismatch in the
IUPAC nucleotide annotation that prevents the recognition of guide RNA in the control sequence can be solved
by editing the chromatogram file to the expected nucleotides. (h, i) Poor sequence quality will not give reliable
results in TIDE(R)
CRISPR Editing Evaluation by TIDE/TIDER 41
of the break site. When this window is too small or when the
break site is located upstream of nucleotide 100, the alignment
cannot be performed correctly. Then the start of the alignment
window should be set manually closer to nucleotide number
1 (Fig. 3c).
7. Incorrect alignment. When the beginning of the sequence trace
is of poor quality, the alignment function can make a mistake.
This results in a quality plot with a high aberrant sequence
signal along the whole length of the sequence trace (Fig. 3d).
The aberrant sequence signal should only increase around the
expected cut site (blue dashed line). In case of poor alignment,
the start of the alignment window needs to be adjusted until a
proper alignment is achieved (default of 100).
8. Quality plot recommendations. In the experimental sample,
around the break site a consistently elevated signal is expected,
which is due to indels introduced at the break site. The starting
position of this elevated signal may be used to verify that breaks
were induced at the expected location. The control trace
should have a low and equally distributed aberrant sequence
signal along the whole trace. The reference trace in the case of
TIDER should only have high scores at the positions of the
altered nucleotides. Fluctuations in the control and reference
signal reflect local variation in the quality of the sequence trace.
Near the end of the sequencing traces the aberrant signal is
often high, typically due to the lower quality of the trace
toward the end (Fig. 3a). When a sequence stretch of poor
local quality is present in the decomposition window the calcu-
lations of TIDE(R) are compromised. The boundaries of the
decomposition window can be manually adjusted to remove
the region that is of low quality; this will improve the estima-
tions. Another area to avoid in the decomposition window is a
stretch of repetitive sequences. These regions can be recog-
nized in the quality plot as a sudden stretch without aberrant
nucleotides (Fig. 3b). Such region might confound the decom-
position of the sequence trace.
9. Decomposition window recommendations. For TIDE, the
default decomposition window spans the entire sequence
trace from the break site until the end of the sequence minus
the size of the maximum indel. When the boundaries of the
decomposition window cannot fulfill this constraint, the soft-
ware will report that the boundaries are not acceptable. For
example, this can occur when the break site is too close to the
end of sequence trace. To address this, the decomposition
window boundaries should be set further apart or a smaller
indel size should be chosen. Alternatively, new primers have to
be designed according to Note 1. For TIDER the decomposi-
tion window is by default 20 bp upstream of the break to 80 bp
42 Eva Karina Brinkman and Bas van Steensel
15. Poor sequence quality. When the sequence has poor quality
overall, TIDE(R) will yield poor results with a low R2 value
(see Note 10) since too much noise is present in the data. The
quality plot will show an overall high aberrant sequence signal
in the control (the reference) and the experimental sample,
before and after the break site (see Note 8). It is recommended
to check the chromatograms of the samples (Fig. 3h) for poor
sequencing quality. If so, these samples cannot be analyzed
reliably by TIDE(R). Note that sometimes the peak signals in
the chromatogram appear normal, but the file can contain
wrongly unannotated or additional annotated nucleotides
(Fig. 3i). TIDE(R) gives a warning when the spacing between
the nucleotides in the chromatogram of the sequence trace is
not consistent, which is often an indication for wrongly unan-
notated or additional annotated nucleotides. In case of this
warning, the chromatograms should be carefully investigated
(use Snapgene or Chromas software).
Acknowledgments
References
1. Brinkman EK, Chen T, de Haas M, Holland 4. Mali P, Esvelt KM, Church GM (2013) Cas9 as
HA, Akhtar W, van Steensel B (2018) Kinetics a versatile tool for engineering biology. Nat
and fidelity of the repair of Cas9-induced dou- Methods 10:957–963
ble-strand DNA breaks. Mol Cell 70:801–813 5. Yoshimi K, Kunihiro Y, Kaneko T,
2. Cong L, Ran FA, Cox D, Lin S, Barretto R, Nagahora H, Voigt B, Mashimo T (2016)
Habib N, Hsu PD, Wu X, Jiang W, Marraffini ssODN-mediated knock-in with CRISPR-Cas
LA et al (2013) Multiplex genome engineering for large genomic regions in zygotes. Nat
using CRISPR/Cas systems. Science Commun 7:10431
339:819–823 6. Inui M, Miyado M, Igarashi M, Tamano M,
3. Jinek M, Chylinski K, Fonfara I, Hauer M, Kubo A, Yamashita S, Asahara H, Fukami M,
Doudna JA, Charpentier E (2012) A program- Takada S (2014) Rapid generation of mouse
mable dual-RNA-guided DNA endonuclease models with defined point mutations by the
in adaptive bacterial immunity. Science CRISPR/Cas9 system. Sci Rep 4:5396
337:816–821 7. Storici F, Snipe JR, Chan GK, Gordenin DA,
Resnick MA (2006) Conservative repair of a
chromosomal double-strand break by single-
44 Eva Karina Brinkman and Bas van Steensel
strand DNA through two steps of annealing. of genome editing by sequence trace decompo-
Mol Cell Biol 26:7645–7657 sition. Nucleic Acids Res 42:e168
8. Wang Z, Zhou ZJ, Liu DP, Huang JD (2008) 14. Brinkman EK, Kousholt AN, Harmsen T,
Double-stranded break can be repaired by Leemans C, Chen T, Jonkers J, van Steensel B
single-stranded oligonucleotides via the (2018) Easy quantification of template-
ATM/ATR pathway in mammalian cells. Oli- directed CRISPR/Cas9 editing. Nucleic Acids
gonucleotides 18:21–32 Res 46(10):e58
9. Ma H, Marti-Gutierrez N, Park SW, Wu J, 15. Green MR, Sambrook J (2012) Molecular
Lee Y, Suzuki K, Koski A, Ji D, Hayama T, cloning: a laboratory manual. Cold Spring
Ahmed R et al (2017) Correction of a patho- Harbor Laboratory Press, Cold Spring Harbor,
genic gene mutation in human embryos. NY
Nature 548(7668):413–419 16. Kunkel TA (1985) Rapid and efficient site-
10. Richardson CD, Ray GJ, DeWitt MA, Curie specific mutagenesis without phenotypic selec-
GL, Corn JE (2016) Enhancing homology- tion. Proc Natl Acad Sci U S A 82:488–492
directed genome editing by catalytically active 17. van Overbeek M, Capurso D, Carter MM,
and inactive CRISPR-Cas9 using asymmetric Thompson MS, Frias E, Russ C, Reece-Hoyes
donor DNA. Nat Biotechnol 34:339–344 JS, Nye C, Gradia S, Vidal B et al (2016) DNA
11. Horlbeck MA, Witkowsky LB, Guglielmi B, repair profiling reveals nonrandom outcomes at
Replogle JM, Gilbert LA, Villalta JE, Torigoe Cas9-mediated breaks. Mol Cell 63:633–646
SE, Tjian R, Weissman JS (2016) Nucleosomes 18. Dorsett Y, Zhou Y, Tubbs AT, Chen BR,
impede Cas9 access to DNA in vivo and Purman C, Lee BS, George R, Bredemeyer
in vitro. Elife 5:e12677 AL, Zhao JY, Sodergen E et al (2014)
12. Yang L, Guell M, Byrne S, Yang JL, De Los HCoDES reveals chromosomal DNA end
Angeles A, Mali P, Aach J, Kim-Kiselak C, structures with single-nucleotide resolution.
Briggs AW, Rios X et al (2013) Optimization Mol Cell 56:808–818
of scarless human stem cell genome editing. 19. Beumer KJ, Trautman JK, Mukherjee K, Car-
Nucleic Acids Res 41:9049–9061 roll D (2013) Donor DNA utilization during
13. Brinkman EK, Chen T, Amendola M, van gene targeting with zinc-finger nucleases. G3
Steensel B (2014) Easy quantitative assessment 3:657–664
Open Access This chapter is licensed under the terms of the Creative Commons Attribution 4.0 International
License (http://creativecommons.org/licenses/by/4.0/), which permits use, sharing, adaptation, distribution
and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the
source, provide a link to the Creative Commons licence and indicate if changes were made.
The images or other third party material in this chapter are included in the chapter’s Creative Commons
licence, unless indicated otherwise in a credit line to the material. If material is not included in the chapter’s
Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the
permitted use, you will need to obtain permission directly from the copyright holder.
Chapter 4
Abstract
Recent developments in gene targeting methodologies such as ZFNs, TALENs, and CRISPR/Cas9 have
revolutionized approaches for gene modifications in cells, tissues, and whole animals showing great promise
for translational applications. With regard to CRISPR/Cas9, a variety of repurposed systems have been
developed to achieve gene knock-out, base editing, targeted knock-in, gene activation/repression, epige-
netic modulation, and locus-specific labeling. A functional communality of all CRISPR/Cas9 applications is
the gRNA-dependent targeting specificity of the Cas9/gRNA complex that, for gene knock-out
(KO) purposes, has been shown to dictate the indel formation potential. Therefore, the objective of a
CRISPR/Cas9 KO set up is to identify gRNA designs that enable maximum out-of-frame insertion and/or
deletion (indel) formation and thus, gRNA design becomes a proxy for optimal functionality of CRISPR/
Cas9 KO and repurposed systems. To this end, validation of gRNA functionality depends on efficient,
accurate, and sensitive identification of indels induced by a given gRNA design. For in vitro indel profiling
the most commonly used methods are based on amplicon size discrimination or sequencing. Indel detection
by amplicon analysis (IDAA™) is an alternative sensitive, fast, and cost-efficient approach ideally suited for
profiling of indels induced by Cas9/gRNA with similar sensitivity, specificity, and resolution, down to single
base discrimination, as the preferred next-generation sequencing-based indel profiling methodologies. Here
we provide a protocol that is based on complexed Cas9/gRNA RNPs delivered to primary peripheral blood
mononuclear cells (PBMCs) isolated from healthy individuals followed by quantitative IDAA indel profiling.
Importantly, the protocol described benefits from a short “sample-to-data” turnaround time of less than 5 h.
Thus, this protocol describes a methodology that provides a suitable and effective solution to validate and
quantify the extent of ex vivo CRISPR/Cas9 targeting in primary cells.
Key words Indel detection by amplicon analysis (IDAA™), NGS, Ex vivo precise genome targeting,
PBMCs, Indel “finger print”, Primary cells, CD34+, CRISPR/Cas9, RNP, Synthetic gRNA,
ProfileIt™
1 Introduction
Yonglun Luo (ed.), CRISPR Gene Editing: Methods and Protocols, Methods in Molecular Biology, vol. 1961,
https://doi.org/10.1007/978-1-4939-9170-9_4, © Springer Science+Business Media, LLC, part of Springer Nature 2019
45
46 Saskia König et al.
A
Donor
PBMCs
2. Cas9/RNP
1.
ex vivo PBMCs
2 days
DSB
NHEJ
deleon
inseron
C * Tri-primer PCR
3 hours
* wt
* deleons
* inserons
D IDAA
TM
2 hours
deleons wt inserons
+1
-10
-9
-1
-12
Fig. 1 Schematic outline of the ex vivo IDAA indel profiling workflow. (a) Human
healthy donor PBMCs are isolated, cultured ex vivo (1) followed by CRISPR/Cas9
RNP delivery (2). (b) Designer nucleases precisely targeted to a defined genomic
locus introduce DNA DSBs that are repaired by cellular repair mechanisms and
result in indel formation at the targeted site. (c) Tri-primer amplification across
the targeted site by use of the universal fluorophore labeled FAMfor primer
enables uniform labeling of amplicons. (d) Fluorophore labeled amplicons are
analyzed by capillary electrophoretic fragment analysis, followed by quantitative
indel identification by ProfileIt™. The illustration depicts the indel “finger print,”
with WT/unmodified amplicon peak shown in yellow and indels out-of-frame or
in-frame shown in blue and white respectively
400000 wt 30000
300000 1 1
250000 20000
200000 15000
150000
10000
100000 2 2
3 5000 3
50000 5 4 5 4
0
-30 -28 -26 -24 -22 -20 -18 -16 -14 -12 -10 -8 -6 -4 -2 0 2 4 6 8 10 12 14 16 18 20 -15 -11 -8 -4 01
Indel size/bp Indel size/bp
C
wt
1
Relative fluorescence units
3
4
5
Ampliconl size/bp
Fig. 2 Comparative profiles for indel detection of a sample by NGS and IDAA™/ProfileIt™ quantitative
analysis. (a) MiSeq NGS profile of amplicons derived from a cell lysate 2 days post COSMC Cas9 nucleofection
as previously described [11]. The five most dominant indel events are labeled 1–5 with indication of
unmodified wild type peak (WT). (b) IDAA profile derived from analysis of the same cell lysate as displayed
in (a), analyzed by GeneMapper software (ThermoScientific, USA), and with labeling of the same dominant
indel events. (c) Depicts the indel “finger print” of the same IDAA sample as shown in panel A and B, but
analyzed by ProfileIt™, with quantification of the five most dominant indels shown as % of total indels
including unmodified/WT product (shown in yellow) and with all out-of-frame indels shown in blue. The
quantitative results for the profile shown are presented in Table 1
Table 1
ProfileIt quantification of indels shown in Figs. 2c and 3
Fig. 3 IDAA™/ProfileIt™ analysis of ex vivo delivered Cas9 RNP to human cells. An in vitro transcribed gRNA
(gRNA #1) and two different synthetic gRNA designs (synCCR5 gRNA #1 and #2), targeting the human CCR5
between the corresponding amino acids 80 and 120, were complexed with Cas9 and delivered to K562 cell
line, isolated donor PBMCs, or purified CD34+ cells respectively. (a) Depicts the indel “finger print” from
in vitro transcribed gRNA#1, as described in the protocol, complexed with Cas9 delivered to K562 cells (lower
panel). Out-of-frame indel formation monitored three days post nucleofection as compared to untreated K562
cells (upper panel) of 34.6% was achieved. (b) Depicts the indel “finger print” derived from RNP complexed
with synCCR5 gRNA#1 delivered to PBMCs isolated from different human healthy donors (A or B). Similar indel
profiles (“indel finger prints”) are observed three days post nucleofection in PBMCs from two different donors,
marked by black arrow heads. Relative difference in WT allele presence is attributed to differences in
nucleofection efficiency. (c) Depicts the indel “finger print” from RNP complexed with synCCR5 gRNA#2
delivered either to PBMCs or CD34+ cells isolated from the same human healthy donor A. Similar indel “finger
prints” are observed 3 days post nucleofection in PBMCs and CD34+ cells. Relative difference in WT allele
presence is attributed to differences in nucleofection efficiency. In the three panels, the quantification of the
five most dominant indels is shown as % of total indels including unmodified/WT product (shown in yellow)
and out-of-frame indel % is indicated and the respective indels shown in blue. Indel quantification for all
profiles shown can be found in Table 1
50 Saskia König et al.
human cell line and primary cells, we show that the indel profile
serves as an “finger print” that is strictly dependent on gRNA
design and on the target sequence.
2 Materials
2.1 Primary Human 1. Leukocyte reduction system (LRS) chamber containing the
PBMC Isolation and blood sample for further processing.
Culture 2. 50 mL Falcon Conical Centrifuge Tubes.
3. PBS.
4. T75 Tissue Culture Flasks.
5. Biocoll Separating Solution (Biochrom, Berlin, Germany).
6. RPMI 1640 with GlutaMAX (Thermo Fisher Scientific, Wal-
tham, USA) + 10% FCS (PAA, Pasching, Austria).
7. Human interleukin 2 improved sequence (IL-2 IS), premium
grade (Miltenyi Biotec, Bergisch Gladbach, Germany).
8. Tissue culture plates.
9. NucleoCounter NC250 (ChemoMetec, Allerod, Denmark) or
similar device.
10. Humidified incubator at 37 C and 5% CO2.
2.2 Thawing and 1. Primary human peripheral blood mononuclear cells (PBMCs;
Activation of Primary see Note 1).
Human PBMCs 2. RPMI 1640 with GlutaMAX (Thermo Fisher Scientific, Wal-
tham, USA) + 10% FCS.
3. Human T cell activation kit (Miltenyi Biotec, Bergisch Glad-
bach, Germany).
4. Human interleukin 2 improved sequence (IL-2 IS), premium
grade (Miltenyi Biotec, Bergisch Gladbach, Germany).
5. Anti-CD25 antibody, human (Miltenyi Biotec, Bergisch Glad-
bach, Germany).
6. Tissue culture plates.
7. NucleoCounter NC250 (ChemoMetec, Allerod, Denmark) or
similar device.
8. DynaMag15 (Invitrogen, Carlsbad, USA) or similar.
9. Humidified incubator at 37 C and 5% CO2.
2.4 In Vitro 1. HiScribe T7 High Yield RNA Synthesis Kit (New England
Transcription and Biolabs, Ipswich, USA).
Purification 2. DNase I 2000 units/mL (Rnase free; New England Biolabs,
Ipswich, USA).
3. MEGAclear Transcription Clean-Up Kit (Ambion, Austin,
USA).
4. DEPC-treated water.
5. 10 TBE buffer: 890 mM Tris, 890 mM boric acid, 20 mM
EDTA (see Note 3).
6. Urea.
7. Acrylamide 30% (29:1) (Biorad, Hercules, USA).
8. TEMED (Carl Roth, Karlsruhe, Germany).
9. APS (Carl Roth, Karlsruhe, Germany).
10. Low range ssRNA ladder (New England Biolabs, Ipswich,
USA).
52 Saskia König et al.
2.6 Cell Lysis and 1. CoboExtract DNA extraction solution (Cobo Technologies).
DNA Extraction
2.7 Tri-primer 1. FAMFOR, Fwd and Rev IDAA primers can be ordered in a kit
IDAA PCR format from TAG Copenhagen (http://www.TAGC.com) or
custom-synthesized on a 10-nmol scale, desalted, or reverse-
phase column purified by standard oligo service providers (see
Note 4).
2. Design IDAA Fwd and Rev primers to amplify a 200–550 bp
product spanning the nuclease cut site using Primer3 or similar
software. Add the following 50 extension to the IDAA Fwd
primer: 50 -AGCTGACCGGCAGCAAAATTG-30 (see Note 5).
3. PCR plates, 96 wells.
4. Axygen 8-strip PCR tubes (Fischer Scientific).
5. Thermocycler with programmable temperature stepping func-
tionality, 96 wells, for tri-primer fluorophore labeling of ampli-
cons a Veriti Thermocycler is recommended or Eppendorf
Mastercycler with vapo.protect.
6. Benchtop microcentrifuge.
7. Eppendorf Thermomixer R, dry block heating and cooling
shaker.
8. Agarose gel electrophoresis system.
9. Novex XCell SureLock Mini-Cell.
10. Blue-light transilluminator and orange filter goggles.
11. TEMPase Hot Start DNA Polymerase with 10 ammonium
buffer and MgCl2 (Ampliqon).
Ex vivo CRISPR/Cas9 Induced Indel Quantification by IDAA 53
2.9 ProfileIT Indel 1. ProfileIt™ software will work on any modern PC or mobile
Profiling and device supporting one of the following web browsers: Google
Quantification Chrome 32+, Firefox 45+, Safari 10+, Microsoft Edge 12+.
2. License to Viking ProfileIt™ indel profiling software analysis
program (https://viking.sdu.dk/pages/software/profileit/).
3 Methods
Amount Final
Component (μL) concentration
Ammonium PCR buffer (Ampliqon), 10 1.25 1
(Important Note 39)
dNTP, 100 mM (25 mM each) 0.125 1 mM
IDAA Fwd primer, 2.5 μM 0.125 0.025 μM
IDAA Rev primer, 25 μM 0.125 0.25 μM
FAMFOR primer, 25 μM (see Note 38) 0.125 0.25 μM
DNA polymerase (Ampliqon) (see Note 0.12 0.5 U
39, important)
MgCl2, 25 mM 0.7 2.5 mM
DMSO (see Note 39, important) 0.625 5%
CoboExtract cell lysatea from Subheading 3.6, 1 20–20,000
step 2 (see Note 37) cells
ddH2O 8.33
Total 12.5
a
Alternatively, 1–100 ng purified genomic DNA may be used
4 Notes
Fig. 4 Illustration of the 30 -template independent Taq DNA polymerase activity. The 30 template independent
activity of several DNA polymerases is well documented [20–22]. Indel profiles shown are derived from
ST6GALNAC1 locus amplified from human genomic DNA [7]. From top to bottom the panels show how
optimization of PCR buffer, from standard KCl buffer, ammonium buffer, standard KCl buffer plus 5% DMSO,
ammonium buffer plus 5% DMSO, and in the latter case followed by ProfileIt™ correction can improve the 30
template independent activity from split “stutter” peak formation to a single peak. The fully 30 elongated
fragment/peak is indicated with an arrow head. Importantly, careful inspection of WT peak profile and optimal
30 extension of amplicons by use of optimal 5’- “end base” design [24] is required for reliable IDAA
quantification
64 Saskia König et al.
Acknowledgments
References
1. Chandrasegaran S, Carroll D (2016) Origins of various CRISPR/Cas9 delivery methods. Prog
programmable nucleases for genome engineer- Mol Biol Transl Sci. Elsevier Inc 152:49–67
ing. J Mol Biol 428:963–989 10. Bennett EP, Jacobi AM, Garrett RR, Behlke
2. Sfeir A, Symington LS (2015) MA (2018) Detection of insertion/deletion
Microhomology-mediated end joining: a (indel) events after genome targeting: pro’s
back-up survival mechanism or dedicated path- and con’s of the available methods. In: Appa-
way Trends Biochem Sci 40:701–714 sani K (ed) GENOME EDITING and ENGI-
3. Sehn JK (2014) Insertions and deletions NEERING: from Talens, ZFNs and CRISPRs
(Indels). In: Clinical genomics. Elsevier, to molecular surgery. Cambridge University
Amsterdam, pp 129–150 Press, Cambridge, pp 181–194
4. Deriano L, Roth DB (2013) Modernizing the 11. Yang Z, Steentoft C, Hauge C, Hansen L,
nonhomologous end-joining repertoire: alter- Thomsen AL, Niola F, Vester-Christensen
native and classical NHEJ share the stage. MB, Frodin M, Clausen H, Wandall HH et al
Annu Rev Genet 47:433–455 (2015) Fast and sensitive detection of indels
5. van Overbeek M, Capurso D, Carter MM, induced by precise gene targeting. Nucleic
Frias E, Russ C, Reece-Hoyes JS, Nye C, Acids Res 43:e59. 1–8
Vidal B, Zheng J, Hoffman GR et al (2016) 12. Sandmann S, De Graaf AO, Karimi M, Van Der
DNA repair profiling reveals nonrandom out- Reijden BA, Hellström-Lindberg E, Jansen JH,
comes at. Mol Cell 63:633–646 Dugas M (2017) Evaluating variant calling
6. Paquet D, Kwart D, Chen A, Sproul A, tools for non-matched next-generation
Jacob S, Teo S, Olsen KM, Gregg A, sequencing data. Sci Rep 7:1–12
Noggle S, Tessier-Lavigne M (2016) Efficient 13. Loman NJ, Misra RV, Dallman TJ,
introduction of specific homozygous and het- Constantinidou C, Gharbia SE, Wain J, Pallen
erozygous mutations using CRISPR/Cas9. MJ (2012) Performance comparison of bench-
Nature 533:125–129 top high-throughput sequencing platforms.
7. Narimatsu Y, Joshi H, Zhang Y, Gomes C, Nat Biotechnol 30:434–439
Yen H, Lorenzetti F, Furukawa S, 14. Brinkman EK, Chen T, Amendola M, van
Schjoldager K, Hansen L, Clausen H, Bennett Steensel B (2014) Easy quantitative assessment
EP et al (2018) A validated gRNA library for of genome editing by sequence trace decompo-
CRISPR/Cas9 targeting of the human glyco- sition. Nucleic Acids Res 42:e168–e168
syltransferase genome. Glycobiology 28 15. Hsiau T, Maures T, Waite K, Yang J, Kelso R,
(5):295–305 Holden K, Stoner R (2018) Inference of
8. Lonowski LA, Narimatsu Y, Riaz A, Delay CE, CRISPR edits from sanger trace data. bioRxiv
Yang Z, Niola F, Duda K, Ober EA, 1101:10. https://doi.org/10.1101/251082
Clausen H, Wandall HH et al (2017) Genome 16. Jamuar S, Lam A-TN, Kircher M, D’Gama A,
editing using FACS enrichment of nuclease- Wang J, Barry BJ, Zhang X, Hill RS, Partlow
expressing cells and indel detection by ampli- JN, Topcu M et al (2014) Somatic mutations in
con analysis. Nat Protoc 12:581–603 cerebral cortical malformations. N Engl J Med
9. Kosicki M, Rajan SS, Lorenzetti FC, Wandall 371:733–743
HH, Narimatsu Y, Metzakopian E, Bennett EP 17. Mohamed S, Penaranda G, Gonzalez D,
(2017) Dynamics of indel profiles induced by Camus C, Khiri H, Boulmé R, Sayada C,
66 Saskia König et al.
Philibert P, Olive D, Halfon P (2014) Compar- 30 end of a DNA fragment. DNA Cell Biol
ison of ultra-deep versus Sanger sequencing 12:763–770
detection of minority mutations on the 22. Smith JR, Carpten JD, Brownstein MJ,
HIV-1 drug resistance interpretations after Ghosh S, Magnuson VL, Gilbert DA, Trent
virological failure. AIDS 28:1315–1324 JM, Collins FS (1995) Approach to genotyping
18. Yang Z, Wang S, Halim A, Schulz MA, errors caused by nontemplated nucleotide
Frodin M, Rahman SH, Vester-Christensen addition by Taq DNA polymerase. Genome
MB, Behrens C, Kristensen C, Vakhrushev SY Res 5:312–317
et al (2015) Engineered CHO cells for produc- 23. Sentmanat MF, Peters ST, Florian CP, Con-
tion of diverse, homogeneous glycoproteins. nelly JP, Pruett-Miller SM (2018) A survey of
Nat Biotechnol 33:2014–2017 validation strategies for CRISPR-Cas9 editing.
19. Cornu TI, Mussolino C, Cathomen T (2017) Sci Rep 8(1):888
Refining strategies to translate genome editing 24. Magnuson VL, Ally DS, Nylund SJ, Karanja-
to the clinic. Nat Med 23:415–423 wala ZE, Rayman JB, Knapp JI, Lowe AL,
20. Clark JM (1988) Novel non-templated nucle- Ghosh S, Collins FS (1996) Substrate nucleo-
otide addition reactions catalyzed by procaryo- tide-determined non-templated addition of
tic and eucaryotic DNA polymerases. Nucleic adenine by Taq DNA polymerase: implications
Acids Res 16:9677–9686 for PCR-based genotyping and cloning. Bio-
21. Hu G (1993) DNA polymerase-catalyzed addi- techniques 21(4):700–9
tion of nontemplated extra nucleotides to the
Chapter 5
Abstract
Rapid evaluation of the CRISPR gRNA activity is an essential step of employing the technology in editing
genes. Through machine learning strategy, the rule sets for in silico designing gRNAs with high activity has
greatly improved. However, there are still discrepancies between different prediction rule sets, and between
the predicted and actual gRNA activities. Thus, experimentally validating gRNA activity is still the gold
standard in defining the best gRNAs for gene editing experiments. One such approach for experimentally
selecting gRNAs with high activity is fluorescent surrogate reporter vectors. We had previously developed a
dual-fluorescent surrogate system, called C-Check, which based on single-strand annealing repair of the
DNA double-strand breaks introduced by CRISPR-Cas9 to generate a functional EGFP. The system offers
a tool for rapid functional evaluation of CRISPR gRNA activity, as well as for enrichment of gene edited
cells. In this chapter, we will give a step-by-step instruction on the design, generation, and application of the
C-Check system for quantifying gRNA activities.
Key words CRISPR, Cas9, Reporter vector, Surrogate vector, C-Check, Gene editing
1 Introduction
Yonglun Luo (ed.), CRISPR Gene Editing: Methods and Protocols, Methods in Molecular Biology, vol. 1961,
https://doi.org/10.1007/978-1-4939-9170-9_5, © Springer Science+Business Media, LLC, part of Springer Nature 2019
67
68 Lin Lin and Yonglun Luo
Fig. 1 Schematic illustration of the C-Check vector. The C-Check vector contains two expression cassettes.
First, a consecutive AsRED expression cassette is used as a reference control for normalization. Second, a
truncated EGFP expression cassette, of which the EGFP genes is splitted into two parts with 500 nt in
homology. A BasI-based golden-gate cloning site is used for insertion of any surrogate DNA for C-Check-
based CRISPR activity assay. Upon introduction of double-stranded DNA breaks (DSB) at the surrogate DNA by,
e.g., CRISPR, DSB repaired by single-strand annealing with the 500 nt homologous sequences will generate a
functional EGFP expression cassette
CRISPR Activity Quantification by C-Check 69
2 Materials
3 Methods
3.1 The Principle The C-Check vectors can be generated through (1) assembly of
of Choosing synthesized oligonucleotides (synthetic approach) or (2) PCR
the Optimal C-Check amplification of the targeted region (PCR approach). The principal
Design Approach of choosing synthetic or PCR approach depends on the number of
gRNAs to be evaluated and how the gRNA target sites are
distributed in the genome. Please follow these principles to choose
the optimal approach:
1. Number of gRNAs (1–3), use the synthetic approach.
2. Number of gRNA (over 3), if all gRNAs target the same
genomic locus within a very close region (less than 500 bp),
use PCR approach.
3. Number of gRNA (over 3), if all gRNAs are located in a broad
genomic region (over 500 bp), use a combination of synthetic
and PCR approach.
4. Design one C-Check vector for each gene/locus.
Table 1
Guide for designing C-Check oligonucleotides by synthetics
3.3 C-Check Vector Most steps of the PCR approach are the same as the synthetic
Generation—PCR approach. Here we highlight those steps unique for the PCR
Approach approach.
72 Lin Lin and Yonglun Luo
Table 2
Primer designing scheme for PCR-based generation of C-Check vectors
6. Use a sterile 100 μL pipette tip, pick one white clone, firstly dip
into LB1, gently swirl for 1 s.
7. Then place the tip in LS1 of the lysate tube.
8. Repeat pick the rest of colonies as steps 6 and 7.
9. Cap the lysate set and LB set tubes.
10. Place the LB set tubes at 37 C incubator.
11. Place the lysate set tubes in a thermal cycler, lyse the cells at
98 C for 10 min.
12. Save the lysate at 4 C, and use 1 μL lysate as template for PCR
screening.
13. If using the synthetic approach for generating C-Check vector,
PCR screening is carried out using the C-Check screening
primer (Forward) and the antisense C-Check oligo (see Note 8).
14. If using the PCR approach for generating C-Check vector,
PCR screening can be carried out using the same condition as
amplifying the fragment as in Subheading 3.3, step 3.
15. Analyze PCR with a 1.0% agarose gel. For synthetic approach,
the length of PCR product should be 600 bp + the length of
C-Check antisense oligo. For PCR approach, the length of
PCR product will be the same as the cloning fragment.
16. Set up LB culture of the PCR positive clones, 5 mL
LB + 50 μg/mL spectinomycin.
17. Purify C-Check plasmid using a miniprep kit.
18. Validate the C-Check plasmids with restriction enzymes
(RE) digestion: BamHI and KpnI. Positive clones will result
in two bands: one of 6.6 kb and the other of 600 bp+C-Check
oligo/fragment.
19. Set up LB culture of one RE positive clone for midi-prep.
20. Purify plasmid using a commercial midi-prep kit.
21. Save plasmid at 4 C until used.
3.5 Transfection 1. Seed 50,000 HEK293T cells per well to a 24-well plate, pre-
of HEK293T Cells pare triplicate for each transfection and enough wells of cells
with C-Check (see Note 9).
and CRISPR 2. Transfect HEK293T cells with X-tremeGENE 9 or other trans-
fection reagents. For each transfection, use a molar ratio of
Cas9:gRNA:C-Check ¼ 1:1:1.
3. Inspect cells using a fluorescent microscope 48 h post transfec-
tion (Fig. 2).
Fig. 3 FACS analysis of C-Check-based measurement of CRISPR activity. Gatings for transfected cells
(AsRED+) and for cells that underwent SSA-mediated repair (EGFP+AsRED+) are presented
4 Notes
Acknowledgments
References
1. Mali P et al (2013) RNA-guided human 3. Henrik Devitt Møller LL, Xi X, Petersen TS,
genome engineering via Cas9. Science 339 Huang J, Yang L, Kjeldsen E, Jensen UB,
(6121):823–826 Zhang X, Liu X, Xun X, Wang J, Yang H,
2. Boch J et al (2009) Breaking the code of DNA Church GM, Bolund L, Regenberg B, Luo Y
binding specificity of TAL-type III effectors. (2018) CRISPR-C: circularization of genes
Science 326(5959):1509–1512 and chromosome by CRISPR in human cells.
CRISPR Activity Quantification by C-Check 77
Nucleic Acids Res. https://doi.org/10.1093/ 8. Yang Y et al (2016) Highly efficient and rapid
nar/gky767 detection of the cleavage activity of Cas9/
4. Leenay RT, Beisel CL (2017) Deciphering, gRNA via a fluorescent reporter. Appl Biochem
communicating, and engineering the CRISPR Biotechnol 180(4):655–667
PAM. J Mol Biol 429(2):177–191 9. Jensen KT et al (2017) Chromatin accessibility
5. Brinkman EK et al (2014) Easy quantitative and guide sequence secondary structure affect
assessment of genome editing by sequence CRISPR-Cas9 gene editing efficiency. FEBS
trace decomposition. Nucleic Acids Res 42 Lett 591(13):1892–1901
(22):e168 10. Liu B et al (2018) STAT3 associates with vacu-
6. Yang Z et al (2015) Fast and sensitive detection olar H(+)-ATPase and regulates cytosolic and
of indels induced by precise gene targeting. lysosomal pH. Cell Res 28(10):996–1012
Nucleic Acids Res 43(9):e59 11. Lin L et al (2017) Fusion of SpCas9 to E. coli
7. Zhou Y et al (2016) Enhanced genome editing Rec A protein enhances CRISPR-Cas9
in mammalian cells with a modified dual- mediated gene knockout in mammalian cells.
fluorescent surrogate system. Cell Mol Life J Biotechnol 247:42–49
Sci 73(13):2543–2563
Part II
Abstract
Since its first harnessing in gene editing in 2012 and successful application in mammalian gene editing in
2013, the CRISPR-Cas9 system exerted magnificent power in all gene-editing-related applications, indi-
cating a sharp thrive of this novel technology. However, there are still some critical drawbacks of the
CRISPR-Cas9 system that hampered its broad application in gene editing. Efficient delivery of the Cas9
protein and its partner small guide RNA (sgRNA) to the target cells or tissue is one of the technical
bottlenecks. CRISPR-Cas9 delivery via DNA plasmids still plays the big role in gene editing methods. With
regard to the disadvantages of CRISPR-Cas9 plasmids, the most acute barrier lies in its large size (>10 kb)
and the subsequent low transfection efficiency by conventional transfection method. In this chapter, what
we present is an easy method by fabricating CRISPR-Cas9 plasmids into nanoparticle system and efficiently
delivered into target cells to achieve gene editing.
Key words CRISPR-Cas9, Transfection, Artificial virus, Branched polyethylene imine, Heptafluor-
obutyric anhydride
1 Introduction
Yonglun Luo (ed.), CRISPR Gene Editing: Methods and Protocols, Methods in Molecular Biology, vol. 1961,
https://doi.org/10.1007/978-1-4939-9170-9_6, © Springer Science+Business Media, LLC, part of Springer Nature 2019
81
82 Suleixin Yang et al.