文档库 最新最全的文档下载
当前位置:文档库 › 2014-Feng ZHANG-Book_Methods_Cas9

2014-Feng ZHANG-Book_Methods_Cas9

2014-Feng ZHANG-Book_Methods_Cas9
2014-Feng ZHANG-Book_Methods_Cas9

C hapter 17

R NA-Guided Genome Editing of Mammalian Cells

N eena K.P yzo cha,F.A nn R an,P atrick D.H su,and F eng Z hang

A bstract

T he microbial CRI SPR-Cas adaptive immune system can be harnessed to facilitate genome editing in eukaryotic cells (Cong L et al., Science 339, 819–823, 2013; Mali P et al., Science 339, 823–826, 2013). Here we describe a protocol for the use of the RNA-guided Cas9 nuclease from the S treptococcus pyogenes type II CRISPR system to achieve speci? c, scalable, and cost-ef? cient genome editing in mammalian cells.

K ey words C RISPR-Cas G enome editing D NA cleavage C as9 G uide RNA P AM sequence N HEJ

G ene knockout

1I ntro ductio n

T he ability to introduce targeted modi? cations into genomes and

engineer model organisms holds enormous promise for biomedical

and biotechnological applications. The development of program-

mable nucleases [ 1–9] has allowed targeting of speci? c genomic

loci to introduce double-strand breaks (DSBs) in the DNA. These

DSBs are subsequently repaired through either the error-prone

nonhomologous end-joining (NHEJ) pathway or the homology-

directed repair (HDR) pathway, allowing formation of indels or

precise editing of the genome, respectively [ 10]. These endonucle-

ases can be used for studies in basic biology, biotechnology, and

medicine, including the development of reporter cell lines [ 11],

transgenic organisms [ 12], disease models [ 13], and gene therapy

[ 14], among others. Although ZFNs and TALENs can be repro-

grammed to target speci? c DNA sequences, these tools still require

time-consuming engineering of proteins de novo for each target,

and there remains a de? cit for technologies that are easily custom-

izable, multiplexable, and affordable.

Francesca Storici (ed.), Gene Correction: Methods and Protocols, Methods in Molecular Biology, vol. 1114,

DOI 10.1007/978-1-62703-761-7_17, ? Springer Science+Business Media, LLC 2014

269

270 T

he microbial adaptive immune system CRI SPR (Clustered Regularly Interspaced Short Palindromic Repeats) consists of a set

of enzymes and noncoding RNA elements [

15 – 17 ]. Among the three types of CRISPR systems in bacteria and archaea [

15 , 16 ], type I I requires only a single protein, Cas9 (formerly Csn1), to

mediate DNA cleavage [

18 ]. Cas9 is targeted to speci? c DNA sequences by a pair of noncoding RNA elements: the CRI SPR

RNA (crRNA), which carries the target-specifying guide sequence

via Watson–Crick base pairing (Fig.

1 ), and the trans-activating crRNA (tracrRNA), which hybridizes with crRNA and is required

for loading onto Cas9 [

19 , 20 ]. T he type I I CRI SPR system of S treptococcus pyogenes can be

reconstituted in mammalian cells to mediate DNA cleavage with

three minimal components: Cas9, crRNA, and tracrRNA. The lat-

ter two components can further be truncated and fused into a single

chimeric guide RNA scaffold (Fig.

1 ) [ 18 ] for a target sequence selected from any genomic locus with its 3’ end followed by a NGG

trinucleotide motif [

19 ]. This protospacer-adjacent motif (PAM) is speci? c to each CRISPR system [

21 ]. Generation of speci? c guide RNAs for targeted genome editing only requires the purchase of

two short oligos and simple cloning that can take as little as two days.

T he wild-type S . pyogenes Cas9 (SpCas9) enzyme has multiple

endonuclease domains, two of which cleave DNA in a strand-

speci? c manner. Two catalytic residues, D10 or H840 [

18 ], can be mutated to convert the wild-type SpCas9 into a DNA-nicking

enzyme (SpCas9n) [ 1 , 18 ]. Given that single-stranded nicks in the F ig.1 T argeted DNA cleavage by SpCas9 in the human E MX1 locus. The SpCas9 enzyme ( y ellow ) interacts with its genomic target ( b lue ) with the help of a guide RNA. The genomic target is directly 5′ to the PAM sequence, which is -NGG- for SpCas9. The guide RNA is composed of the guide sequence ( b lue ), which anneals with the ge nomic targe t via Watson–Crick base pairing and a chime ric guide RNA scaffold consisting of a fusion between the crRNA ( g ray ) and the tracrRNA ( r ed )

Neena K. Pyzocha et al.

271

target DNA can also stimulate HDR, SpCas9n reduces the

l ikelihood of error-prone repair by NHEJ. Furthermore, both catalytic domains of SpCas9 can be mutated to convert SpCas9

into a RNA- g uided DNA-binding protein [

18 , 22 ]. This chapter describes a set of protocols for using the SpCas9 system for genome

editing in mammalian cells.

2M aterials

1. C loning plasmids: p X330 (CBh::SpCas9 + U6::chimeric guide

RNA) (Addgene) or p X335 (CBh::SpCas9n (D10A) + U6::

chimeric guide RNA) (Addgene) (Fig.

2 ). 2. O ligos for target sequence. S ee Subheading 3.1 for discussion

regarding locus selection and Subheading

3.2 on oligo design (Integrated DNA Technologies).

3. R estriction enzymes and phosphatase: FastDigest B bs I

(Fermentas), FastAP (Fermentas), 10× FastDigest Buffer

(Fermentas) ( s ee N ote 1 ).

4. Q IAquick Gel Extraction Kit (QIAGEN).

5. P hosphorylation, annealing, and ligation reagents: 10× T4

Ligation Buffer (NEB), T4 Polynucleotide Kinase (NEB), 2×

Quick Ligation Reaction Buffer (NEB), Quick Ligase (NEB).

6. P lasmid-Safe exonuclease (Epicentre Biotechnologies).

7. C ompetent cells and bacterial growth reagents. 2.1 M olecular Cloning Components F ig. 2 B icistronic expression vector for guide RNA and SpCas9 (or SpCas9n). A genomic target directly upstream to the PAM sequence can be cloned into the expression vector. After a target is selected, two DNA oligos can be designed based on the schematic showing the guide sequence insert. One oligo ( t op strand, written 5′–3′) contains ligation adapter sequences for cloning into the expression vector and G(N) 19

, which is the selected genomic target sequence. The other oligo ( b ottom strand, written 3′–5′) also contains ligation adapter sequences for cloning into the expression vector and the complementary bases to the genomic target sequence. Once annealed and phosphorylated, the oligos can be inserted into the vector digested with B bsI

RNA-Guided Genome Editing of Mammalian Cells

272 8. Q IAGEN Plasmid Midi Kit (QIAGEN).

9. S tandard gel electrophoresis reagents.

1. C ell line: For validation, human embryonic kidney (HEK) cell line 293FT (Life Technologies). For additional discussions on working with other cell lines, s ee N ote 2 .

2. C ell culture reagents for maintenance of 293FT cells:

Dulbecco’s Modi? ed Eagle’s Medium (DMEM) (Life

Technologies), 10 % fetal bovine serum (HyClone), 2 mM

GlutaMAX (Life Technologies), 100 U/mL penicillin, and

100 μg/mL streptomycin.

3. D issociation reagent: TrypLE? (Life Technologies).

4. T ransfection reagent: Lipofectamine 2000 (Life Technologies)

for HEK293FT or Neuro-2a cells (Sigma Aldrich) ( s ee N ote 3 ).

5. 24-well tissue culture plates (Corning).

6. T ransfection Control Plasmid: pMaxGFP (Lonza).

7. Q uickExtract? DNA extraction kit (Epicentre

Biotechnologies).

1. S URVEYOR Mutation Detection Kit (Transgenomic).

2. 4–20 % Novex TBE polyacrylamide gels (Life Technologies).

3. A mpli? cation primers speci? c to the targeted locus (Integrated

DNA Technologies).

4. H erculase II High Fidelity Polymerase (Agilent).

3M etho ds

F or use with the SpCas9 system, target sites must be followed by a NG

G trinucleotide motif on the 3′ end ( s ee N otes 4 and 5 ).

W e designed cloning vectors (pX330 for SpCas9 or pX335 for SpCas9n, a D10A nickase) to aid co-expression of SpCas9 and

guide RNA in mammalian cells (Fig.

2 ). In this vector, SpCas9 is driven by the CBh promoter [

23 ], and the guide RNA is driven by the human PolIII promoter U6. Phosphorylated and annealed oli-

gos (design indicated in Fig.

2 ) can be cloned into the B bsI digested plasmid containing the entire guide RNA scaffold. The oligos are

designed based on the target site sequence (20 bp sequence cor-

responding to the target site). The G(N) 19 refers to the sequence

selected upstream of the PAM sequence in the genomic DNA

( s ee N ote 6 ). Create oligos using the schematic in Fig. 2 . 2.2 T issue Culture,

Transfection, and DNA

Extraction

Components 2.3 C omponents for the Analysis of Genome Modi? cation 3.1 T arget Selection 3.2 C onstruct Design

Neena K. Pyzocha et al.

273

1. D igest 1 μg of pX330 or pX335 with B bs I

for 30 min at 37 °C: 2. G el purify digested pX330 or pX335 using QI Aquick Gel

Extraction Kit and elute in EB.

3. P

hosphorylate and anneal each pair of oligos for the insert piece:

A

nneal in a thermocycler using the following parameters: 4. S et up ligation reaction and the negative control. Incubate at

room temperature for 10 min:

3.3 M olecular

Cloning: Oligo

Annealing and Cloning

into Backbone Vectors RNA-Guided Genome Editing of Mammalian Cells

274 5. (Optional but highly recommended) Treat ligation reaction with Plasmid-Safe exonuclease ( s ee N ote 7 )

: I ncubate reaction at 37 °C for 30 min. 6. T ransform 2 μL of reaction from s tep 5 into competent cells and plate on ampicillin selection plates. 7. P ick two colonies the following day and analyze for correct insertion of the target sequence oligos. T he CRISPR-Cas DNA cleavage system has been validated for use in a variety of mammalian cell lines [ 1 , 2 , 24 ] ( s ee N ote 2 ). The protocol below is for HEK239FT cells. 1. H EK293FT cells are maintained in DMEM supplemented with 10 % fetal bovine serum and passaged before reaching 70 % con? uency. Cells are maintained in an incubator set at 37 °C supplemented with 5 % CO 2 . 2. H EK293FT cells can be transfected using Lipofectamine 2000 according to the manufacturer’s protocol. 3. F or each well of a 24-well plate, a total of 500 ng of plasmid is transfected. One well should be a control to see the relative transfection ef? ciency using a plasmid such as pmaxGFP . 4. A fter 12 h of transfection, replace the medium with pre-warmed maintenance medium. After 72 h, genomic DNA can be iso-lated using the QuickExtract DNA extraction kit following the manufacturer’s protocol. Brie? y, cells are resuspended in QuickExtract solution (50 μL per 24 well) and incubated at 65 °C for 15 min followed by 98 °C for 10 min. 1. T he ef? ciency of cleavage can be detected by assessing the per-centage of cells containing indels in the target region ( s ee N ote 8 ). In order to detect indels in the DNA, follow the instructions provided in the SURVEYOR Mutation Detection Kit manual

( s ee N ote 9 ).

2. I t is recommended that the SURVEYOR Nuclease digestion

products are analyzed on a PAGE gel.

3.4 C ell Culture and

Transfection

3.5 A nalysis of

Genomic Modi? cation:

SURVEYOR and

Sequencing Neena K. Pyzocha et al.

275

3. T o calculate the percent cutting ef? ciency of a CRISPR locus,

use the following formula: %indel =-?è????÷÷1100 where a and b refer to the relative concentrations of the cut

bands and c equals the relative concentration of the full-length

PCR template. A representative SURVEYOR gel image and

quantitation is shown in Fig.

3 .4N o tes

1. C onventional restriction enzymes can be substituted for FastDigest

restriction enzymes. I n this case, adjust digestion reagents and

digestion times according to manufacturer’s protocol.

2. E xperimental conditions may need to be optimized for each

cell line. F ig.3 S URVEYOR assay comparing SpCas9-mediated DNA cleavage at two different targets in the same gene. ( a ) The third exon of the human E MX1 locus was targeted using guide RNAs at two unique sites. ( b )A re pre -sentative SURVEYOR assay gel image comparing the targeted cleavage ef? ciency by SpCas9 at the two targets in the human E MX1locus

a

b

RNA-Guided Genome Editing of Mammalian Cells

276

3. F or other cell lines, we suggest doing an initial comparison of

different transfection reagents (e.g., FuGENE HD, nucleofec-

tion, and TransIT).

4. A free computational resource maintained by the Zhang lab

(https://www.wendangku.net/doc/f310916832.html,) contains the most

up-to- d ate information relevant for Cas9 systems.

5. I t is ideal for these targets to be unique within the genome. We

also recommend testing multiple target sites for each gene and

selecting the most effective target.

6. S electing a target site with a 5′ G allows for ef? cient transcrip-

tion of the guide RNA from the U6 promoter.

7. P lasmid-Safe treatment is recommended because it degrades

linear dsDNA, helping to prevent unwanted recombination

products.

8. S pCas9-induced double-strand breaks in the target DNA are

usually repaired through the error-prone NHEJ process in

HEK293FT cells.

9. I t is important to make sure that the genomic PCR primers

yield a single amplicon for reliable quanti? cation of the percent

cutting ef? ciency. In the case that primers do not yield a single

amplicon, the PCR product needs to be gel puri?ed or new

primers should be designed.

A ckno wledgments

W e thank Randall Platt for comments and members of the Zhang

Lab for discussion, support, and advice. N.P. is supported by the

National Science Foundation Graduate Research Fellowship,

Primary Award #1122374. P.D.H. is a James Mills Pierce Fellow.

F.Z. is supported by the N H Transformative R01 Award

(R01-NS073124); the N I H Director’s Pioneer Award

(DP1-MH100706); the Keck, McKnight, Gates, Damon Runyon,

Searle Scholars, Merkin, Klingenstein, and Simons Foundations;

Bob Metcalfe; Mike Boylan; and Jane Pauley. Sequence and reagent

information are available through

h ttp://www.genome-

https://www.wendangku.net/doc/f310916832.html, .

R eferences

1. C ong L et al (2013) Multiplex genome engi-

neering using CRI SPR/Cas systems. Science 339:819–823

2. M ali P et al (2013) RNA-guided human

genome engineering via Cas9. Science 339:823–826 3. U rnov FD et al (2005) Highly ef? cient endog-

enous human gene correction using designed zinc-? nger nucleases. Nature 435:646–651 4. P orteus MH, Baltimore D (2003) Chimeric

nucleases stimulate gene targeting in human cells. Science 300:763

Neena K. Pyzocha et al.

277

5. M iller JC et al (2007) An improved zinc-? nger

nuclease architecture for highly speci? c genome

editing. Nat Biotechnol 25:778–785

6. C hristian M et al (2010) Targeting DNA

double- s trand breaks with TAL effector nucle-

ases. Genetics 186:757–761

7. M iller JC et al (2011) A TALE nuclease archi-

tecture for ef?cient genome editing. Nat Biotechnol 29:143–148

8. S anjana NE et al (2012) A transcription

activator- l ike effector toolbox for genome engineering. Nat Protoc 7:171–192

9. S toddard BL (2005) Homing endonuclease

structure and function. Q Rev Biophys 38:

49–95

10. R ouet P, Smih F, Jasin M (1994) Expression of

a site-speci? c endonuclease stimulates homolo-

gous recombination in mammalian cells. Proc Natl Acad Sci U S A 91:6064–6068

11. H ockemeyer D et al (2011) Genetic engineer-

ing of human pluripotent cells using TALE nucleases. Nat Biotechnol 29:731–734

12. C ui X et al (2011) Targeted integration in rat

and mouse embryos with zinc-? nger nucleases.

Nat Biotechnol 29:64–67

13. D ing Q et al (2013) A TALEN genome- e diting

system for generating human stem cell-based disease models. Cell Stem Cell 12:238–251 14. M aier D et al (2013) Ef?cient clinical scale

gene modi? cation via zinc ? nger nuclease tar-

geted disruption of the H I V co-receptor CCR5. Hum Gene Ther 24:245–258

15. D eveau H, Garneau JE, Moineau S (2010)

CRI SPR/Cas system and its role in phage-

bacteria interactions. Annu Rev Microbiol 64:475–493

16. G arneau JE et al (2010) The CRI SPR/Cas

bacterial immune system cleaves bacteriophage

and plasmid DNA. Nature 468:67–71

17. H orvath P, Barrangou R (2010) CRI SPR/

Cas, the immune system of bacteria and archaea. Science 327:167–170

18. J inek M et al (2012) A programmable dual-

RNA- g uided DNA endonuclease in adaptive bacterial immunity. Science 337:816–821

19. D eltcheva E et al (2011) CRISPR RNA matu-

ration by trans-encoded small RNA and host factor RNase III. Nature 471:602–607

20. J ore MM et al (2011) Structural basis for

CRI SPR RNA-guided DNA recognition by Cascade. Nat Struct Mol Biol 18:529–536 21. M ojica FJ, Diez-Villasenor C, Garcia-Martinez

J, Almendros C (2009) Short motif sequences

determine the targets of the prokaryotic CRI SPR defence system. Microbiology 155:

733–740

22. Q i LS et al (2013) Repurposing CRISPR as an

RNA-guided platform for sequence-speci? c con-

trol of gene expression. Cell 152:1173–1183 23. G ray SJ et al (2011) Optimizing promoters for

recombinant adeno-associated virus-mediated gene expression in the peripheral and central nervous system using self-complementary vectors. Hum Gene Ther 22:1143–1153

24. C ho SW, Kim S, Kim JM, Kim JS (2013)

Targeted genome engineering in human cells

with the Cas9 RNA-guided endonuclease. Nat

Biotechnol 31:230–232

RNA-Guided Genome Editing of Mammalian Cells

相关文档