Rapid Assembly of gRNA Arrays via Modular Cloning in Yeast

2 Imperial College Centre for Synthetic Biology, Imperial College London, London, ... expression of multiple gRNAs in S. cerevisiae has thus far been ...
0 downloads 0 Views 622KB Size
Subscriber access provided by OCCIDENTAL COLL

Technical Note

Rapid Assembly of gRNA Arrays via Modular Cloning in Yeast Nicholas S McCarty, William M Shaw, Tom Ellis, and Rodrigo Ledesma-Amaro ACS Synth. Biol., Just Accepted Manuscript • DOI: 10.1021/acssynbio.9b00041 • Publication Date (Web): 02 Apr 2019 Downloaded from http://pubs.acs.org on April 3, 2019

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 11 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Synthetic Biology

1 2

Rapid Assembly of gRNA Arrays via Modular Cloning in Yeast

3

Nicholas S. McCarty1,2, William M. Shaw2,3, Tom Ellis2,3 & Rodrigo Ledesma-Amaro2,3,4,*

4 5 6 7

1 Bioengineering, California Institute of Technology, Pasadena, CA 91125, USA

8

*Correspondence: [email protected]

2 Imperial College Centre for Synthetic Biology, Imperial College London, London, SW7 2AZ, UK 3 Department of Bioengineering, Imperial College London, London, SW7 2AZ, UK 4 Lead contact

9 10

ABSTRACT

11 12

CRISPR is a versatile technology for genomic editing and regulation, but the

13

expression of multiple gRNAs in S. cerevisiae has thus far been limited. We present

14

here a simple extension to the Yeast MoClo Toolkit, which enables the rapid assembly

15

of gRNA arrays using a minimal set of parts. Using a dual-PCR, Type IIs restriction

16

enzyme Golden Gate assembly approach, at least 12 gRNAs can be assembled and

17

expressed from a single transcriptional unit. We demonstrate that these gRNA arrays

18

can stably regulate gene expression in a synergistic manner via dCas9-mediated

19

repression. This approach expands the number of gRNAs that can be expressed in

20

this model organism and may enable the versatile editing or transcriptional regulation

21

of a greater number of genes in vivo.

22 23

Keywords: yeast, MoClo toolkit, CRISPR, multiplex, assembly method, Golden Gate

24

assembly

25 26

The expression of many gRNAs simultaneously enables multiplexed genome

27

engineering and transcriptional modulation of genetic targets with CRISPR-Cas

28

systems. There are numerous methods to accomplish this, such as by encoding each

29

gRNA under the control of a separate RNA polymerase III (Pol III) promoter1, cleaving

30

gRNAs from a longer RNA sequence by encoding them in introns2, by flanking gRNAs

31

with self-processing ribozyme sequences3, or via excision of gRNAs by other Cas

32

family endonucleases4–6. This last approach, in which gRNAs are excised by an

33

endonuclease, can be performed with Csy4, which processes pre-crRNA in native

34

CRISPR systems7. By flanking gRNAs with Csy4 recognition sites, multiple crRNAs or

ACS Paragon Plus Environment

ACS Synthetic Biology 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

35

fused gRNAs can be efficiently processed from a single transcript in a variety of cell

36

lines5,6.

37 38

Numerous methods have also been reported to assemble gRNA arrays for in vitro and

39

in vivo applications, typically by utilizing annealed oligonucleotides with modified

40

Gibson or Golden Gate assembly approaches6,8–12. These studies have largely

41

focused on mammalian cells but methods have recently emerged for gRNA

42

multiplexing in yeast. Efforts to multiplex gRNAs in S. cerevisiae have been limited to

43

7 gRNAs if expressed from individual promoters13 and 4 gRNAs if expressed and

44

processed from a single array, either by using gRNA-tRNA arrays with Pol III

45

promoters or by using Pol II promoters with the Csy4 enzyme to process the gRNA

46

arrays.5,14

47 48

Here, we expand the widely-used Yeast MoClo Toolkit1, a widely-used library of highly

49

characterized parts designed for Golden Gate assembly and integration into S.

50

cerevisiae, to enable the rapid, modular assembly and expression of gRNA arrays in

51

this model organism. This method is a dual-PCR, Type IIs restriction enzyme Golden

52

Gate approach that enables the construction of up to 12 gRNAs in a single array, each

53

flanked by recognition sites for Csy4, in two days. This method uses one plasmid to

54

produce gRNAs with desired spacer sequences and a second plasmid to assemble

55

the final arrays (Supplementary Figure S1). All other parts are available from the

56

Yeast MoClo Toolkit.1 This system will enhance the ease and utility of multiplexed

57

gRNA applications in S. cerevisiae.

58 59

RESULTS

60 61

We build and express arrays of tandem gRNAs in S. cerevisiae by adding just two

62

parts to the Yeast MoClo Toolkit: a Guide-Generating Vector (GGV) and a Destination

63

Vector (DV) (Figure 1a and Supplementary Figures S2, S3). An initial round of PCR

64

using the GGV as a template adds desired spacer sequences via primer overhangs.

65

The GGV contains a Csy4 recognition site, a selection/screening marker and a

66

sequence encoding the gRNA scaffold of interest (which associates the gRNA with a

67

chosen Cas endonuclease). Once the linear PCR products are ligated, the designed

68

spacers are positioned next to the scaffold to produce functional gRNAs.

ACS Paragon Plus Environment

Page 2 of 11

Page 3 of 11 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Synthetic Biology

69 70

A second round of PCR adds Type IIs restriction enzyme recognition sites with unique,

71

four-nucleotide overhangs via primer overhangs15. The generated, linear PCR

72

fragments can then be assembled into the DV via Golden Gate assembly, which

73

consists of a promoter of interest, followed by another screening/selection marker, a

74

terminal copy of the Csy4 recognition sequence, and the terminator of interest (Figure

75

1a). Target sequences of gRNAs in the array must be devoid of BsmBI, BsaI and NotI

76

recognition sites. For this study, we use a DV with a start codon-deficient variant of

77

PTDH3, a Pol II promoter derived from the Yeast MoClo Toolkit1. The DV also contains

78

designed XhoI and BglII sites after the promoter and before the final Csy4 site,

79

respectively, which enables assembled gRNA arrays to be swapped between

80

promoters and terminators from the Yeast MoClo Toolkit.

81

82 83 84 85 86 87 88 89 90

Figure 1: Assembly of gRNA arrays and screening of gRNA candidates. (a) Schematic overview of the system for assembling gRNA arrays. Primers are indicated by arrows, with slanted lines indicating primer overhangs. (b) Spatial positions of the spacer sequences tested on their corresponding genomic targets. Each gRNA contains a 20-nucleotide spacer sequence complementary to the PALD6, PHHF1 or PTEF1 promoter and is adjacent to an NGG PAM sequence. Numbers in the gray boxes indicate each unique gRNA tested and corresponds to the results plotted in panel (c). Numbers marked with an asterisk indicate gRNAs which were used in the construction of gRNA arrays. A minimal number of PAM sites

ACS Paragon Plus Environment

ACS Synthetic Biology 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

91 92 93 94 95 96 97 98

in the PTEF1 promoter limited the number of gRNAs which could be tested for mTagBFP2. (c) Normalized fluorescence for each individual gRNA tested. Each sample was normalized to the fluorescence measured for a yeast strain expressing a null spacer sequence (100% offtarget score using Doench et al. algorithm)16. The fluorescence of yeast expressing the null gRNA was set to a value of 1.0 for each fluorescent reporter. All other groups were normalized to the null gRNA mean. Error bars represent the standard deviation between the n=3 replicates within each group.

99

To assess the efficiency of gRNA array assembly using this system, arrays containing

100

between 3 to 12 gRNAs within a single transcriptional unit were constructed. Colonies

101

were screened for the absence of the screening marker (GFP) (Supplementary

102

Figure S4) and the insert length of each construct was evaluated by diagnostic

103

restriction digest with BsaI (Supplementary Figure S5). Restriction digests indicated

104

that 100% of colonies contained an array of the expected size (for 3 or 6 gRNAs),

105

followed by a decrease in efficiency for 9 and 12 gRNA arrays (66.6% and 50%,

106

respectively) (Supplementary Figure S6). Purified DNA isolated from colonies that

107

yielded bands of the expected sizes from restriction digests were additionally sent

108

for Sanger sequencing (n=3 each) for the 3, 6, 9 and 12 gRNA arrays. All samples

109

were sequence-verified without any observed mutations (Supplementary Methods).

110 111

To demonstrate gRNA multiplexing capacity, an experiment was implemented that

112

enabled us to test the transcriptional repression mediated by multiple gRNAs in

113

budding yeast simultaneously using fluorescent reporters and flow cytometry. S.

114

cerevisiae strain BY4741 was engineered to express three fluorescent reporters, each

115

driven by a unique constitutive promoter: PTEF1-mTagBFP2, PHHF1-mRuby2, and PALD6-

116

Venus. These promoters are constitutive, strong promoters from the Yeast MoClo

117

Toolkit derived from genes in S. cerevisiae that are either nonessential or have

118

paralogs in the genome. These promoter-reporter pairs were cloned into a single

119

cassette and genome-integrated at the HO locus. A DNA construct expressing dCas9

120

(with N- and C-termini nuclear localization signals) and Csy4 endonuclease, under

121

control of PPGK1 and PHHF2, respectively, were also genome-integrated at the LEU2

122

locus17.

123 124

Individual gRNAs were next designed to repress fluorescent protein expression by

125

targeting each of the three constitutive promoters and their individual efficiencies were

126

assessed. Spacer sequences to repress PTEF1, PHHF1, or PALD6 were targeted to bind

ACS Paragon Plus Environment

Page 4 of 11

Page 5 of 11 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Synthetic Biology

127

between positions -300 and +1 bp upstream of the start codon of each fluorescent

128

reporter (Figure 1b). The BY4741-derived strain expressing these fluorescent

129

reporters, dCas9 and Csy4 was then transformed with individual gRNAs possessing

130

the designed spacer sequences integrated at the URA3 locus. Changes in

131

fluorescence of each reporter following integration of the gRNA were assessed by flow

132

cytometry (Figure 1c).

133 134

Using these data, four gRNAs were selected for dCas9-mediated transcriptional

135

repression for each of the fluorescent reporters. They were selected based on weak

136

repression of fluorescent output, so that synergistic effects could be visualized when

137

multiplexing many gRNAs to a promoter, and distributed targeting within the promoter

138

regions so that gRNA recognition sites do not overlap, as we speculated that this would

139

occlude our ability to observe synergistic dCas9-mediated repression of multiple

140

targets simultaneously. gRNAs V1, V4, V6, V8 for Venus repression, gRNAs R2, R8,

141

R6, R4 for mRuby2 repression, and gRNAs B1, B2, B3, B4 for mTagBFP2 repression

142

(Figure 1b) were selected.

143 144

Arrays encoding 3, 6, 9, or 12 gRNAs within a single transcriptional unit were

145

constructed. As arrays increase in size, more gRNAs are targeted to repress

146

fluorescent protein expression; the 3-gRNA array targets one gRNA per promoter,

147

whereas the 12-gRNA array targets four gRNAs to each promoter. The 3-gRNA array

148

consists of V1, R2 and B1. The 6-gRNA array consists of V1, R2, B1, V4, R8 and B2.

149

The 9-gRNA array consists of V1, R2, B1, V4, R8, B2, V6, R6 and B3. The 12-gRNA

150

array consists of all twelve selected gRNAs. All arrays were genome-integrated at the

151

URA3 locus (Figure 2a). We observed a slight growth defect in the strains expressing

152

gRNA arrays compared to controls, potentially due to a side targeting of the three

153

native promoters.18

154 155

Characterization of per cell fluorescence by flow cytometry revealed synergistic

156

repression of all three fluorescent proteins when an increasing number of gRNAs were

157

expressed from the arrays (Figure 2b). Co-expression of dCas9 with four gRNAs

158

targeting each fluorescent reporter, in the case of the 12-gRNA array, showed a

159

repression of 92% in the mVenus channel, 81% in the mRuby2 channel and 95% in

160

the mTagBFP2 channel. These repression efficiencies are significantly greater than

ACS Paragon Plus Environment

ACS Synthetic Biology 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 6 of 11

161

with any of the individual gRNAs and indicated synergistic repression of three separate

162

genes by a tandem array of up to 12 gRNAs using this multiplexed system.

163 164

Since homologous recombination in budding yeast is more active in regions containing

165

repetitive DNA sequences19, we also assessed the stability of these repetitive gRNA

166

arrays over generations of growth by performing flow cytometry on yeast expressing

167

each gRNA array after five consecutive, 1:100 dilutions with twelve hours of growth

168

between dilutions; 72 hours of growth in total (Supplementary Figure S7). Flow

169

cytometry

170

transcriptional repression of fluorescent protein expression. Colony PCR on the gRNA

171

arrays at the URA3 locus confirmed stable genomic integration of all gRNA arrays after

172

the dilution series (Supplementary Figure S7).

after

five

consecutive

dilutions

indicated

sustained,

synergistic

173 174 175 176 177 178 179 180 181 182 183 184 185

Figure 2: Genome-integrated gRNA arrays in S. cerevisiae enable synergistic, dCas9mediated repression of gene expression. (a) General method to quantify the repression efficiency of tandem gRNA arrays in S. cerevisiae. gRNA arrays were driven by PTDH3 on a plasmid integrated at the URA3 locus. Arrays contain 3, 6, 9, or 12 gRNAs in tandem, with each successive increase in tandem length adding an additional gRNA targeting each of the promoters driving fluorescent protein expression. gRNA arrays were transformed into yeast expressing Csy4, dCas9 and Venus, mRuby2 and mTagBFP2 and fluorescence was measured by flow cytometry to quantify the repression efficiency of each gRNA array. (b) Normalized repression of fluorescence by multiplexed expression of gRNA arrays. All experimental samples were normalized to the fluorescence measured for this yeast strain also expressing an array with 3 ‘null’ gRNAs. The ‘3 Null gRNAs’ control was set to a value of 1.0 for all three fluorescence channels. All values plotted are averages from n=8 (3, 6, 9, 12

ACS Paragon Plus Environment

Page 7 of 11 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

186 187 188

ACS Synthetic Biology

gRNA arrays) or n=4 (3 Null gRNAs) samples. Error bars represent one standard deviation from the geometric mean. Asterisks denote two-tail p-value as determined by two-sample ttest, with **p ≤ 0.01, and ***p ≤ 0.0001.

189 190

This assembly method is rapid, inexpensive, and fully compatible with the existing

191

Yeast MoClo Toolkit1. It can be used to express at least 12 gRNAs from one promoter,

192

which may prove useful in multiplexed CRISPR applications, such as the

193

transcriptional regulation of multiple genes in a pathway, or to construct more complex,

194

gRNA-based logic circuits13.

195 196

MATERIALS AND METHODS

197 198

Assembly and Validation of gRNA Arrays

199

A detailed protocol, including primer sequences and thermocycler settings, can be

200

found in the Supporting Information (Supplementary Tables S1-S13). 5 ng of GGV

201

template was PCR amplified with a phosphorylated reverse primer and a forward

202

primer with 20 nucleotide overhang to add spacer sequences. PCR mixtures were

203

then digested with 0.3 µL DpnI at 37°C for at least 30 minutes and the entire mixture

204

loaded in an agarose gel for electrophoresis. Fragments corresponding to the

205

expected band sizes were excised and purified with a Zymoclean Gel DNA Recovery

206

kit (Zymo Research) and then ligated with T4 ligase. A second round of PCR to add

207

BsmBI cut sites and four-base overhangs was performed with 10 ng of each ligated

208

vector as template. PCR reactions were digested with 0.3 µL of BsmBI and DpnI for

209

30 minutes at 37°C and 30 minutes at 55°C, then gel purified again. Golden Gate

210

assembly was then performed, with all fragments and the DV diluted to 50 nM before

211

use. 0.15 µL DV, 0.5 µL each gRNA part, 1 µl T4 DNA ligase (NEB), 1 µl 10X T4 ligase

212

buffer (NEB) and 1.5 µl of BsmBI enzyme (NEB) were added to a 10 µl total reaction

213

volume with ultrapure water. Assembly reactions were incubated for thirty cycles, with

214

5 min at 37°C and 5 min at 16°C, followed by a 5 min, 55°C final incubation step prior

215

to transformation into TurboComp cells (NEB). Transformed cells were plated on LB

216

+ chloramphenicol and screened visually for colonies lacking GFP under blue light.

217

Colonies lacking GFP upon visual inspection were picked, incubated at 37°C and their

218

DNA isolated. Restriction digests were performed with 0.5µl BsaI (NEB) on 100 ng of

219

DNA from each colony at 37°C for 1 hour and gel electrophoresis performed to verify

ACS Paragon Plus Environment

ACS Synthetic Biology 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 8 of 11

220

insert sizes. Of those colonies that yielded DNA bands of expected sizes, DNA from

221

three colonies with each array (3, 6, 9, 12) were sent for additional validation by Sanger

222

sequencing.

223 224

Construction of Single gRNA Vectors for Quantification of Repression

225

To construct gRNAs for targeting dCas9 to one of the three promoters driving the

226

expression of a fluorescent protein, we used the pYTK050 part from the Yeast MoClo

227

Toolkit1. pYTK050 contains a phenylalanine tRNA, HDV ribozyme, the Cas9/dCas9

228

scaffold (sgRNA) sequence and a SNR52 terminator. Complementary oligos were

229

ordered (IDT) and annealed, with two additional ‘T’ nucleotides added before the

230

spacer sequence to enable its ligation into pYTK050. Annealed oligos were then

231

ligated into pYTK050 via BsmBI-mediated Golden Gate assembly. pYTK050 plasmids

232

containing designed spacer sequences were genome-integrated at the URA3 locus.

233 234

Strains and Cell Cultures

235

All Saccharomyces cerevisiae strains generated in this study are derivatives of

236

BY4741 (S288C MATa his3∆1 leu2∆0 met15∆0 ura3∆0) and are listed in

237

Supplementary Table S14. All experiments were performed in synthetic complete

238

media with 2% (w/v) glucose (VWR), 0.67% (w/v) Yeast Nitrogen Base without amino

239

acids (Sigma), 0.14% (w/v) Yeast Synthetic Drop-out Medium supplements without

240

histidine, leucine, tryptophan, and uracil (Sigma), 20 mg/L histidine (Sigma), 100 mg/L

241

leucine (Sigma), 20 mg/L tryptophan (Sigma), 20 mg/L uracil (Sigma).

242 243

Yeast colonies were inoculated into liquid Synthetic Complete media with all amino

244

acids supplemented apart from the corresponding, auxotrophic amino acids (Uracil,

245

Leucine and Histidine) and incubated in 96-well, 2.2 mL deep well plates at 30°C and

246

700 rpm.

247 248

Flow

Cytometry

249

Cell fluorescence was measured by a LSRFortessa X-20 flow cytometer (BD

250

Biosciences) using the following settings: FSC 330 V, SSC 250 V, mTagBFP2 - 405

251

nm line (450/50) 378 V, sfGFP - 488 nm line (530/30) 450 V, mRuby2 - 640 nm line

252

(685/35) 590 V. Fluorescence data was collected from 10000 viable cells for each

253

experiment and analyzed using FlowJo software.

ACS Paragon Plus Environment

Page 9 of 11 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Synthetic Biology

254 255

SUPPORTING INFORMATION

256

Detailed protocol of the method to assemble gRNA arrays, including all DNA

257

sequences, primers and parts used in this study, supporting data and additional

258

methods.

259 260

ACKNOWLEDGMENTS

261

This work was supported by BBSRC grant (BB/R01602X/1) (to RLA), BBSRC grant

262

BB/K019791/1 (to TE) and a Fulbright – Imperial College London Partnership award

263

(to N.S.M).

264 265

DECLARATION OF INTERESTS

266

A patent application related to this work has been filed by the authors.

267 268

REFERENCES

269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301

(1) Lee, M. E., DeLoache, W. C., Cervantes, B., and Dueber, J. E. (2015) A Highly Characterized Yeast Toolkit for Modular, Multipart Assembly. ACS Synth. Biol. 4, 975–986. (2) Nissim, L., Perli, S. D., Fridkin, A., Perez-Pinera, P., and Lu, T. K. (2014) Multiplexed and Programmable Regulation of Gene Networks with an Integrated RNA and CRISPR/Cas Toolkit in Human Cells. Mol. Cell 54, 698–710. (3) Gao, Y., and Zhao, Y. (2014) Self-processing of ribozyme-flanked RNAs into guide RNAs in vitro and in vivo for CRISPR-mediated genome editing. J. Integr. Plant Biol. 56, 343–349. (4) Nowak, C. M., Lawson, S., Zerez, M., and Bleris, L. (2016) Guide RNA engineering for versatile Cas9 functionality. Nucleic Acids Res. 44, 9555–9564. (5) Ferreira, R., Skrekas, C., Nielsen, J., and David, F. (2018) Multiplexed CRISPR/Cas9 Genome Editing and Gene Regulation Using Csy4 in Saccharomyces cerevisiae. ACS Synth. Biol. 7, 10–15. (6) Kurata, M., Wolf, N. K., Lahr, W. S., Weg, M. T., Kluesner, M. G., Lee, S., Hui, K., Shiraiwa, M., Webber, B. R., and Moriarity, B. S. (2018) Highly multiplexed genome engineering using CRISPR/Cas9 gRNA arrays. PLoS One 13, e0198714. (7) Haurwitz, R. E., Jinek, M., Wiedenheft, B., Zhou, K., and Doudna, J. A. (2010) Sequence- and structure-specific RNA processing by a CRISPR endonuclease. Science 329, 1205–1210. (8) Breunig, C. T., Durovic, T., Neuner, A. M., Baumann, V., Wiesbeck, M. F., Köferle, A., Götz, M., Ninkovic, J., and Stricker, S. H. (2018) One step generation of customizable gRNA vectors for multiplex CRISPR approaches through string assembly gRNA cloning (STAgR). PLoS One 13, e0196015. (9) Xie, K., Minkenberg, B., and Yang, Y. (2015) Boosting CRISPR/Cas9 multiplex editing capability with the endogenous tRNA-processing system. Proc. Natl. Acad. Sci. 112, 3570–3575. (10) Sakuma, T., Nishikawa, A., Kume, S., Chayama, K., and Yamamoto, T. (2014) Multiplex genome engineering in human cells using all-in-one CRISPR/Cas9 vector system. Sci. Rep. 4, 5400. (11) Vad-Nielsen, J., Lin, L., Bolund, L., Nielsen, A. L., and Luo, Y. (2016) Golden Gate Assembly of CRISPR gRNA expression array for simultaneously targeting multiple genes. Cell. Mol. Life Sci. 73, 4315–4325. (12) Cress, B. F., Toparlak, O. D., Guleria, S., Lebovich, M., Stieglitz, J. T., Englaender, J. A., Jones, J. A., Linhardt, R. J., and Koffas, M. A. G. (2015) CRISPathBrick: Modular Combinatorial Assembly of Type II-A CRISPR Arrays for dCas9-Mediated Multiplex Transcriptional Repression in E. coli. ACS Synth. Biol. 4, 987–1000. (13) Gander, M. W., Vrana, J. D., Voje, W. E., Carothers, J. M., and Klavins, E. (2017) Digital logic circuits in yeast with CRISPR-dCas9. Nat. Commun. 8, 1–11. (14) Zhang, Y., Wang, J., Wang, Z., Zhang, Y., Shi, S., Nielsen, J., and Liu, Z. (2019) A gRNA-tRNA

ACS Paragon Plus Environment

ACS Synthetic Biology 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321

array for CRISPR-Cas9 based rapid multiplexed genome editing in Saccharomyces cerevisiae. Nat. Commun. 10, 1–10. (15) Vladimir, P., Ong, J. L., Kucera, R. B., Langhorst, B. W., Bilotti, K., Pryor, J. M., Cantor, E. J., Canton, B., Knight, T. F., Evans, T. C., and Lohman, G. J. S. (2018) Comprehensive profiling of four base overhang ligation fidelity by T4 DNA Ligase and application to DNA assembly. ACS Synth. Biol. 8b00333. (16) Doench, J. G., Fusi, N., Sullender, M., Hegde, M., Vaimberg, E. W., Donovan, K. F., Smith, I., Tothova, Z., Wilen, C., Orchard, R., Virgin, H. W., Listgarten, J., and Root, D. E. (2016) Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR-Cas9. Nat. Biotechnol. 34, 184–191. (17) Lian, J., Hamedirad, M., Hu, S., and Zhao, H. (2017) Combinatorial metabolic engineering using an orthogonal tri-functional CRISPR system. Nat. Commun. 8, 1688. (18) Jensen, E. D., Ferreira, R., Jakočiunas, T., Arsovska, D., Zhang, J., Ding, L., Smith, J. D., David, F., Nielsen, J., Jensen, M. K., and Keasling, J. D. (2017) Transcriptional reprogramming in yeast using dCas9 and combinatorial gRNA strategies. Microb. Cell Fact. 16, 1–16. (19) Argueso, J. L., Westmoreland, J., Mieczkowski, P. A., Gawel, M., Petes, T. D., and Resnick, M. A. (2008) Double-strand breaks associated with repetitive DNA can reshape the genome. Proc. Natl. Acad. Sci. 105, 11845–11850.

322

ACS Paragon Plus Environment

Page 10 of 11

Page 11 of 11

ACS Synthetic Biology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Paragon Plus Environment