Efficient Intracellular Delivery of CRISPR-Cas Ribonucleoproteins

7 days ago - The University of New South Wales Sydney, Faculty of Medicine, St. Vincent's Clinical School, Darlinghurst , New South Wales Australia...
0 downloads 0 Views 757KB Size
Subscriber access provided by MIDWESTERN UNIVERSITY

Letter

Efficient intracellular delivery of CRISPR-Cas ribonucleoproteins through receptor mediated endocytosis Romain Rouet, and Daniel Christ ACS Chem. Biol., Just Accepted Manuscript • DOI: 10.1021/acschembio.9b00116 • Publication Date (Web): 19 Feb 2019 Downloaded from http://pubs.acs.org on February 20, 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 21 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 Chemical Biology

1

Efficient intracellular delivery of CRISPR-Cas ribonucleoproteins

2

through receptor mediated endocytosis

3 4

Authors

5

Romain Rouet1,2* and Daniel Christ1,2

6 7

Affiliations

8

1

Garvan Institute of Medical Research, Darlinghurst, NSW, Australia.

9

2

The University of New South Wales Sydney, Faculty of Medicine, St Vincent's Clinical

10

School, Darlinghurst, NSW, Australia.

11 12

Correspondence

13

Romain Rouet, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia.

14

Email: [email protected]

15 16

Abstract

17

We recently reported a new delivery system harnessing surface receptors for targeted uptake

18

of CRISPR-Cas9 ribonucleoprotein into mammalian cells (Rouet et al., JACS 2018). For this

19

purpose, Cas9 protein was labeled with the small molecule ligand ASGRL, specific for the

20

asialoglycoprotein receptor, enabling endosomal uptake of the ribonucleoprotein into human

21

cells expressing the receptor. However, detailed mechanistic insights had remained unknown

22

and editing efficiency low. Here we investigate the mechanism of endosomal escape as

23

mediated by the ppTG21 endosomolytic peptide and outline the development of novel Cas9 or

24

Cas12a ribonucleoprotein complexes with increased editing efficiency.

25 26

Introduction

27

CRISPR-Cas systems have transformed biology by enabling rapid gene editing or modulation,

28

with broad application in basic research 1, 2 and medicine 3, 4. Optimizing delivery methods for

29

the CRISPR-Cas gene editing components (Cas9 endonuclease and single-guide RNA, forming

30

a ribonucleoprotein (RNP)) to ensure safe, specific and efficient delivery into cells have been

31

the focus of many recent research activities 5-7. Current methods generally rely on either virus-

32

encoded components or alternatively recombinant ribonucleoproteins 5. Among viral delivery

33

platforms, adeno-associated virus and lentivirus are highly efficient at delivering their genetic

ACS Paragon Plus Environment

1

ACS Chemical 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 2 of 21

34

material into mammalian cells, although exhibit limited cell tropism for specific targeting 8. In

35

addition, such viral platforms pose serious risks of immunogenicity, the possibility of random

36

genomic integration, and restrict the size of encoded transgenes (to around 4 to 6 kb)

37

Recombinant RNPs, on the other hand, are generally not immunogenic, characterized by low

38

levels of off-target editing activity and do not rely on potentially hazardous genomic integration

39

5, 11.

40

delivery platforms for cellular uptake have been developed

41

including electroporation (nucleofection), cell penetrating peptides and nanoparticle

42

encapsulation, are characterized by variable efficiencies and are generally not cell type specific

43

5.

44

We previously demonstrated that a recombinant Cas9 RNP with a single nuclear localization

45

signal (Cas9-1NLS), bearing a small molecule ligand (ASGRL) specific for the

46

asialoglycoprotein receptor (ASGR), is taken up selectively by hepatocarcinoma cell lines

47

expressing the receptor

48

peptide ppTG21, genomic insertions or deletions (indels) could be detected in approximately

49

5% of treated cells, as identified by targeted next generation sequencing (NGS, with no

50

detectable indels background in the absence of ppTG21 peptide). However, to achieve this

51

level of efficacy, the method relied on using relatively high amounts of RNP (250 pmol). In

52

addition, the mechanism of endosomal escape remains poorly understood.

53

We identified here a new Cas9 construct, bearing a mCherry and 3NLS tags, that increases the

54

ASGRL-mediated indels in human hepatocarcinoma cells (HepG2) by 2.5-fold compared to

55

the previously reported construct (Cas9-1NLS), while reducing the amount of RNP for

56

incubation by 5-fold (only 50 pmol). While some background editing is observed with this new

57

construct in HepG2, none is detectable in other cell lines. We then demonstrate that co-

58

incubation of the endosomolytic peptide ppTG21 and the ASGRL-Cas9 RNP is necessary to

59

observe gene editing. Using inhibitors of endocytosis pathways and ppTG21 peptide

60

mutagenesis, we also demonstrate that endosomal escape of the RNP with ppTG21 is pH-

61

dependent and mediated by histidine residues on the surface of ppTG21. Finally, we show that

62

the described targeted delivery approach is applicable to the Cas12a system.

9, 10.

However, unlike viruses, RNPs cannot readily enter mammalian cells, and thus multiple

12.

12, 13.

These delivery platforms,

When administered in the presence of the small endosomolytic

63 64

Results and Discussion

65

Increased editing efficiency with ASGRL conjugated Cas9-mCherry-3NLS

66

In order to further increase the efficacy of gene editing, we utilized a high-throughput method

67

based on T7 Endonuclease I DNA mismatch detection (T7E1 assay) 2

ACS Paragon Plus Environment

14-16.

We first evaluated

Page 3 of 21 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 Chemical Biology

68

the editing efficiency of the ASGRL-Cas9-1NLS RNP (250 pmol or 333 nM) used in our prior

69

study by co-incubation with ppTG21 endosomolytic peptide (7800 pmol or 10 µM) with the

70

hepatocarcinoma HepG2 cell line. As previously observed, we detected only a small proportion

71

of indels at the targeted EMX1 locus (1.2% compared to 5% when analyzed by NGS 12) (Supp.

72

Fig. 1A), highlighting the limitations of the method for detection of low indel levels.

73

We next attempted to identify a construct with higher editing efficiency. We had previously

74

demonstrated that Cas9 RNP, bearing the ASGRL and a mCherry and 3NLS tags (ASGRL-

75

Cas9-mCherry-3NLS; Fig. 1A and Supp. Fig. 1C), displays faster ASGR-mediated uptake

76

than its 1NLS counterpart on HepG2 cells, although some level of non-specific uptake was

77

also observed upon prolonged incubation 12. We sought to investigate whether this construct

78

would also display higher editing levels. Indeed, we observed more than 3% editing efficiency

79

using ASGRL-Cas9-mCherry-3NLS RNP using the T7E1 assay and the previously reported

80

conditions (250 pmol of RNP and 7800 pmol of ppTG21) 12 (Supp. Fig. 1B).

81

We next screened for assay conditions that further improved editing efficiency at lower RNP

82

concentrations, by titrating ppTG21 in the presence of 50 pmol (66 nM) of ASGRL-Cas9-

83

mCherry-3NLS RNP. Intriguingly, we observed increased gene editing compared to ASGRL-

84

Cas9-1NLS, with up to 16% indels observed (Fig. 1B). Importantly, we retained the ASGR

85

selectivity previously analyzed by live cell imaging

86

editing efficiency observed in the presence of ASGRL-RNP compared to RNP only. Moreover,

87

the high background gene editing with the RNP only was not observed when analyzed in a

88

different hepatocarcinoma cell line (SkHep), lacking expression of the ASGR, or in HEK293

89

cells (Supp. Fig. 2). Thus, we consider the background editing to be an artefact of the HepG2

90

cell line with prolonged RNP incubation. To minimize non-ASGR mediated gene editing, we

91

initially chose 750 pmol (1 µM) of ppTG21 for our experiments (Fig. 1B). Further screening

92

of RNP concentrations did not yield further improvements (Supp. Fig. 3). However, our results

93

suggest that the ppTG21 to RNP ratio is key to successful endosomal escape, with optimal

94

ratios between 15:1 and 30:1 (Supp. Fig. 3). This observation is in excellent agreement with

95

our previously reported finding for Cas9-1NLS complexes (~30:1 ratio, with 250 pmol or 333

96

nM of RNP and 7800 pmol or 10 µM of ppTG21) 12.

97

Based on the optimizations carried out here, the following assay conditions were used for all

98

subsequent experiments: 50 pmol (66 nM) of RNP and 750 pmol (1 µM) of ppTG21, which

99

achieved gene levels of 9% for the ASGRL RNP and 4% for the background RNP (devoid of

100

the ASGRL) in HepG2 cells using the T7E1 assay (Fig. 1C). These results were confirmed by

101

targeted NGS, with approximately 13% editing efficiency observed for the ASGRL-Cas9-

12,

with approximately 2.5-fold higher

ACS Paragon Plus Environment

3

ACS Chemical 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

102

mCherry-3NLS compound, with background activities observed at 6% for the Cas9-mCherry-

103

3NLS and 0% for both ASGRL- and Cas9-mCherry-3NLS in the absence of ppTG21 (Supp.

104

Table 1 and Methods). These editing efficiencies were achieved using considerably lower

105

amount of RNP (5-fold less) and ppTG21 peptide (10-fold less) than reported in our previous

106

study 12. In fact, the 50 pmol RNP amount used in the optimized protocol developed here is the

107

same as used in nucleofection. Nucleofection is currently the gold standard in terms of editing

108

efficiency for RNP mediated genome editing in human cells 5. It thereby allows direct gene

109

editing efficiency comparisons between the two delivery technologies. Importantly, non

110

ASGR-mediated RNP uptake is not concerning as gene editing occurs only in the presence of

111

the ppTG21 peptide (Fig. 1B-C). Therefore, the mechanism of RNP endosomal escape must

112

be similar, regardless of the uptake pathway.

113

Simultaneous incubation of ASGRL-Cas9 RNP and ppTG21 is necessary for endosomal

114

escape

115

In order to confirm the requirement for co-incubation of the ASGRL-Cas9 RNP (Cas9

116

comprising mCherry and 3NLS tags) and the endosomolytic peptide ppTG21 to achieve

117

genome editing, we performed pulse/chase experiments. We incubated HepG2 cells with

118

ASGRL-Cas9 RNP for 3 h, then washed the cells with heparin to remove any membrane bound

119

RNP and subsequently incubated with ppTG21 for a further 40 h. Interestingly, no gene editing

120

was observed under these conditions (Supp. Fig. 4A). A similar assay where the ASGRL-Cas9

121

RNP and ppTG21 incubations were inversed also did not yield any gene editing (Supp. Fig.

122

4A). This demonstrates that co-incubation of the RNP and the peptide is required for intra-

123

cellular endosomal escape and suggests that the peptide is being internalized together with the

124

RNP via ASGR-mediated or non-specific endocytosis pathways. To further demonstrate the

125

requirement for simultaneous incubation of the two components, we co-incubated HepG2 cells

126

with the RNP and ppTG21 for only 3 h, then washed the cells and incubated them with media

127

for a further 40 h. This resulted in 2.6% of indels for the ASGRL-Cas9 RNP and only 0.9% for

128

the background Cas9 RNP, representing a 3-fold ASGR-specific increase in editing, and

129

correlated with the previous live cell imaging study (3-fold increase after 4 h) 12 (Supp. Fig.

130

4B).

131

ASGRL-Cas9 RNP is rapidly degraded upon receptor-mediated uptake

132

We next sought to estimate how rapidly endosomal escape occurs by monitoring intracellular

133

RNP degradation. It was previously demonstrated that the ASGR pathway directs internalized

134

glycosylated protein directly to the lysosome 17. We therefore monitored the intracellular levels

135

of RNP over time in HepG2 cells. Cells were incubated with 100 pmol (132 nM) of ASGRL4

ACS Paragon Plus Environment

Page 4 of 21

Page 5 of 21 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 Chemical Biology

136

Cas9 RNP (to allow for better quantification compared to 50 pmol) and at different time points

137

washed with heparin, harvested and total RNA extracted. We intentionally avoided using

138

ppTG21 in this assay to allow the RNP to migrate through the ASGR endocytic pathway.

139

Levels of Cas9 RNP were determined using RT-qPCR to detect the sgRNA and compared to

140

endogenous house-keeping genes and a spike-in control of 100 pmol RNP for quantification.

141

Overall, we observed a sharp increase in RNP accumulation for the first 2 h, reaching up to

142

about 25 pmol (25% of total RNP); however, a slow decrease in RNP level occurred soon after,

143

dropping by 2.5-fold after 4 h (10 pmol - 40% of endocytosed RNP) and 25-fold after 8 h (1

144

pmol - 4% of endocytosed RNP), which may correspond to lysosomal degradation (Fig. 2).

145

We previously demonstrated a linear uptake of the ASGRL-Cas9-mCherry-3NLS RNP in

146

HepG2 cells for the first 12h following incubation 12, suggesting that lysosomal degradation

147

occurs rapidly. Overall, the results indicate that ppTG21-dependant RNP endosomal escape

148

may occur rapidly, most likely in the first 8h following uptake. It also indicates that a rapid

149

uptake may be necessary to achieve sufficient ppTG21 concentration in the endosome and thus

150

induce RNP escape, thereby potentially explaining the lower editing efficiency with the 1NLS

151

RNP.

152

Histidine residues on the surface of ppTG21 allow escape of the ASGRL-Cas9 RNP from

153

the late endosome

154

To understand the mechanism of endosomal escape mediated by the ppTG21 peptide, we first

155

performed alanine scanning mutagenesis of surface exposed residues and evaluated the mutant

156

peptides for gene editing by co-incubation with ASGRL-Cas9 RNP in HepG2 cells. We

157

observed a reduction in indel levels, between 30% and 80%, when single residues were mutated

158

to alanine (Fig. 3A). In particular, histidine residues at positions 11 and 19 and serine residue

159

at position 12 show the most dramatic decrease in gene editing (70-80%). To confirm the

160

importance of histidine residues in endosomal escape capability of ppTG21, we evaluated

161

mutant peptides with two histidine substitutions to alanine. Due to the increased

162

hydrophobicity of the peptides, only three peptides could be synthesized: 8/15AA, 8/19AA and

163

11/19AA. All three peptides dramatically reduced indel levels by 85 to 95%, thus confirming

164

the importance of histidine residues for escape from endocytic pathways (Fig. 3A). The results

165

are in agreement with previous findings showing that peptide variants of ppTG21, with

166

substitution of all histidine residues to glutamic acid (JTS-1) or lysine (ppTG1), were not

167

capable of endosomal escape 12, 18 (Fig. 3A). Interestingly, when we co-incubated the ASGRL-

168

Cas9 RNP in HepG2 cells with previously described histidine-rich endosomolytic peptides

169

such as H5WYG 19, LAH4 20, EB1 21, or the small chemical drug chloroquine 22, we did not

ACS Paragon Plus Environment

5

ACS Chemical 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

170

observe gene editing. This suggests a novel mechanism of endosomal escape using the ppTG21

171

peptide.

172

We finally decided to map the localization of the escape along the endocytic pathway. To

173

achieve this, we performed the same ASGRL-Cas9 RNP ppTG21 co-incubation experiments,

174

with HepG2 cells, this time the cells being treated with chemical inhibitors of endocytosis 23.

175

We used the following inhibitors: dynasore, a dynamin inhibitor that will arrest ASGR clathrin-

176

mediated endocytosis on the cell surface; N-ethyl-isopropyl amiloride (EIPA), an actin

177

remodeling inhibitor; wortmannin, a PI3K inhibitor involved in phagocytosis pathway;

178

bafilomycin A, a V-type ATPase inhibitor preventing endosome maturation to late endosome

179

and lysosome. EGTA, a calcium ion chelator, preventing ASGRL from binding to its receptor,

180

and asialo-orosomucoid (ASOR), an ASGRL competitor for its receptor, were used as controls

181

17, 24.

182

endocytic pathways without being toxic for cells 23. HepG2 cells were initially pre-incubated

183

with the inhibitors for 1 h, then the ASGRL-Cas9 RNP and ppTG21 added to allow for

184

immediate pathway inhibition upon RNP uptake. Cells were incubated with the inhibitors and

185

RNP/ppTG21 for only 24 h (instead of 48 h), to mitigate loss of inhibition over prolonged

186

incubation. We observed that all inhibitors decreased gene editing levels to various degrees

187

(Fig. 3B). More specifically, editing was reduced by over 90% with EGTA and ASOR, and

188

70% with dynasore, thus confirming the predominant ASGR dynamin-mediated RNP uptake

189

but also the presence of minor alternative uptake pathways. Almost complete inhibition with

190

EIPA (preventing actin remodeling) also confirmed RNP entry via endocytic pathways and not

191

through pore formation. Weak inhibition (~ 30%) by wortmannin suggested that this may

192

represent the alternative RNP uptake pathway. Indeed, it was recently demonstrated that

193

wortmannin blocks micropinocytosis, a non-specific uptake pathway, of nanoparticles in

194

HepG2 25. More striking was the complete loss of gene editing in the presence of bafilomycin

195

A. This suggests that inhibition of endosome maturation, from early to late stage, abrogates

196

RNP endosomal escape and thus implies that escape may be occurring in the late endosome.

197

Endosome maturation is characterized by acidification of the endosomal milieu, therefore

198

suggesting a pH-dependent escape mechanism of the RNP with the ppTG21 peptide. Combined

199

with the mutagenesis results on the ppTG21 peptide, this suggests that protonation of histidine

200

residues, during the maturation of endosomal vesicles, leads to activation of the endosomolytic

201

property of the peptide.

202

ppTG21 mediates escape of ASGRL conjugated Cas12a

6

All the inhibitors were used at concentrations that were previously shown to inhibit

ACS Paragon Plus Environment

Page 6 of 21

Page 7 of 21 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 Chemical Biology

203

We finally investigated whether Acidaminococcus sp. Cas12a could also be harnessed for

204

receptor-mediated gene editing using ppTG21 as endosomolytic peptide. Cas12a was shown

205

to be an RNA-guided DNA endonuclease 26 and active in eukaryotic organisms

206

Cas9, Cas12a recognizes a different PAM sequence (5’-TTTV), generates staggered double

207

stranded DNA breaks and is capable of processing its own pre-crRNA 29.

208

Following analogous methodology to Sp. Cas9 12, As. Cas12a (bearing two C-terminal NLS

209

tags) was conjugated with pyridyl-disulphide ASGRL onto surface exposed cysteine residues.

210

To avoid over-labeling of the protein (Cas12a contains 8 cysteine residues) and thus possibly

211

affect endonuclease or crRNA processing activities, we used limited conjugation with only 2-

212

fold molar excess of ASGRL compared to Cas12a. Native and tryptic digestion mass

213

spectrometry revealed that the ASGRL-Cas12a contained a mixture of single- and bis-

214

conjugated proteins, with only cysteine residue as positions 334, 379 and 1248 being labeled

215

with an ASGRL (Supp. Fig. 5A). In vitro pre-crRNA processing and DNA endonuclease

216

activities remained unaffected by the presence of the conjugated-ASGRL (Fig. 5A-B).

217

We next evaluated the gene editing activity of the Cas12a RNP. We used a pre-crRNA targeting

218

human PCSK9 locus, assembled it with Cas12a to generate RNP and incubated with HepG2

219

cells for 48 h. As previously observed with Cas9, no gene editing was observed in the absence

220

of the endosomolytic peptide ppTG21 (Fig. 4C and Supp. Fig. 5B). However, co-incubation

221

of 50 pmol of Cas12a RNP with only 375 pmol ppTG21 (7.5:1 molar ratio) successfully

222

achieved gene editing to about 4.5% for the ASGRL-Cas12a RNP, and 2% for the background

223

control Cas12a RNP. Titration of ppTG21 did not lead to improvement in gene editing levels

224

(Supp. Fig. 6). Overall, this confirms that ppTG21 is a universal endosomolytic peptide that

225

allows endocytosed CRISPR-Cas RNP, either through receptor-mediated endocytosis or non-

226

specific uptake pathways such as macropinocytosis, to escape to the cytoplasm, enabling

227

subsequent nuclear import for gene editing. The gene editing levels observed here are lower

228

than those observed when the RNP is physically introduced in HepG2 cells by nucleofection

229

(around 20%) but still substantial (Fig. 4C and Supp. Fig. 5B). Altogether, the results confirm

230

that Cas12a construct optimization may also be required to improve receptor-mediated gene

231

editing efficiencies, similar to what was performed for Cas9, with attachment of various tags.

232

Discussion

233

Our data highlights key mechanisms of CRISPR-Cas endonuclease RNP endosomal escape

234

employing co-incubation of the cell-specific targeting RNP with the small endosomolytic

235

peptide ppTG21. In particular, it was demonstrated that the RNP and the peptide must be co-

236

incubated to allow endosomal escape, as sequential incubations do not yield any gene editing.

ACS Paragon Plus Environment

27, 28.

Unlike

7

ACS Chemical 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

237

In addition, inhibition of actin remodeling almost completely prevented gene editing, thus

238

ruling out non-specific entry via pore formation on the cell membrane. Gene editing was also

239

abolished in the presence of bafilomycin A, which prevents maturation of the early endosome

240

to the late endosome and lysosome, thus suggesting that endosomal escape occurs at the late

241

endosome stage, before any protein degradation occurs in the lysosome. Moreover,

242

endosomolytic activity of ppTG21 was shown to be pH-dependent as substitutions of histidine

243

residues to alanine or charged amino acids (lysine or glutamic acid) reduced or abolished gene

244

editing capabilities. Altogether, this suggest a model where the ASGRL-RNP and the ppTG21

245

peptide are taken up together via ASGR-mediated endocytosis, and subsequently migrate along

246

the endocytic pathway. There, the intra-endosomal milieu progressively acidifies until it

247

reaches a point where ppTG21 allows disruption of the late endosome and facilitates escape of

248

the trapped RNP to the cytoplasm.

249

It is unclear why the Cas9 comprising the mCherry and 3NLS tags demonstrated superior gene

250

editing compared to the 1NLS tag counter-part. However, considering the fast RNP

251

degradation occurring in the absence of ppTG21 peptide, one may suggest that fast uptake is

252

necessary to achieve sufficient ppTG21 concentration in the late endosome and subsequent

253

escape. The slower uptake of the Cas9-1NLS RNP may not be enough to compensate for the

254

fast degradation, thereby allowing limited amount of RNP to escape 12.

255

Better understanding this mechanism will allow for the engineering of peptides with improved

256

endosomolytic properties. Indeed, the difference in gene editing levels between nucleofection

257

and receptor-mediated uptake (about 2-fold for Cas9-mCherry-3NLS and 5-fold for Cas12-

258

2NLS) and the previously demonstrated rapid and strong ASGR-mediated uptake by live cell

259

imaging

260

process. Further improvements may lead to gene editing efficiencies that may surpass that of

261

the current gold standard for CRISPR-Cas RNP delivery: nucleofection.

12

suggest that endosomal escape with ppTG21 is currently a relatively inefficient

262 263

Methods

264

Cell culture and reagents

265

Human HepG2 and SkHep hepatocarcinoma cell lines and HEK293 cell line were obtained

266

from ATCC and cultivated in EMEM media (ATCC) or DMEM media for HEK293 (Gibco)

267

supplemented with 10% FBS and penicillin/streptomycin at 37C with 5% CO2.

268

Endosomolytic and cell-penetrating peptides (lyophilized) were purchased from Anaspec and

269

reconstituted in DMSO at high concentration (10 mM). Heparin, EGTA, Dynasore, EIPA,

8

ACS Paragon Plus Environment

Page 8 of 21

Page 9 of 21 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 Chemical Biology

270

Wortmannin and Bafilomycin A were purchased from Sigma. Pyridyl-disulphide ASGRL, and

271

ASOR (asialo-orosomucoid) was supplied by Pfizer.

272

Protein expression, purification, sgRNA/pre-crRNA synthesis and RNP formation

273

Cas9 and Cas12a proteins were expressed and purified as previously described 12. Single-guide

274

RNA and pre-crRNA were synthesized by T7 in vitro transcription, purified by PAGE and

275

refolded in 20 mM HEPES pH 7.5, 1 mM MgCl2, 150 mM KCl, 10% glycerol as previously

276

described 16. Pre-crRNA was synthesized with a self-cleaving hepatitis delta virus ribozyme at

277

its 3’-end to ensure homogeneous blunt ends. Pre-crRNA DNA template was generated using

278

Oligos 1 and 2 at 1 µM and Fwd 1 and Rev 1 at 10 µM as previously described 16 (Supp. Table

279

2). RNP was assembled by incubating Cas9 and sgRNA or Cas12a and pre-crRNA at a ratio

280

of 1:1.2 at 37C for 10 min.

281

Cas protein ASGRL conjugation

282

Pyridyl-disulfide ASGRL was dissolved to a concentration of 8 mM in DMSO. Cas9 or Cas12a

283

were filtered through 0.2 µm filter units prior to conjugation. Conjugation was carried out using

284

a 20:1 molar ratio of ASGRL (diluted 10-fold in 20 mM HEPES pH 7.5, 150 mM KCl, 10%

285

glycerol) to Cas9 or 2:1 for Cas12a at 4C over-night. The conjugated protein was further

286

purified by size-exclusion chromatography in 20 mM HEPES pH 7.5, 150 mM KCl, 10%

287

glycerol to remove unreacted ASGRL. Conjugation was assessed by native mass spectrometry

288

as previously described 12. Tryptic digestion was performed by denaturation in 6 M urea and

289

incubation at 55C for 20 min and over-night digestion at room temperature with mass spec

290

grade trypsin (Promega) after dilution in 50 mM Tris pH 7.0, 1 mM CaCl2 and ~ 0.5 M urea

291

final concentration. Digested fragments were analyzed by mass spectrometry.

292

Cell-based assays and T7E1 assay

293

For nucleofection assay, cells were harvested from culture plates, washed once with PBS and

294

resuspended at 10,000,000 cells/ml in SF buffer (Lonza). Ten microliters of Cas9 or Cas12a

295

RNP (containing the desired amount of RNP) were added to 20 µl of cells in SF buffer and

296

transferred to wells of a nucleofection plate (Lonza). Cells were electroporated using the 96-

297

well shuttle nucleofector (Lonza) and the HepG2 settings, incubated at room temperature for

298

10 min, resuspended in growth media and finally transferred onto a 12-well culture plate (1

299

ml/well).

300

For receptor-mediated uptake assay, cells were harvested, seeded in 24-well plates at 80,000

301

cells/well (700 µL) and incubated at 37C for 4-12 h. RNP was diluted into 50 µl of Opti-MEM

ACS Paragon Plus Environment

9

ACS Chemical 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

and incubated for 5 min. Endosomolytic peptide was then dispensed to the Cas9 RNP/Opti-

303

MEM mix, incubated for 5 min and dispensed to cells.

304

Cells were harvested in Quick Extract buffer (Epicenter) after 48 h co-incubation with RNP

305

and peptide (unless stated otherwise), incubated for 10 min at 65C then for 10 min at 95C.

306

Genomic DNA concentrations were estimated by measuring absorbance at 260 nm. Targeted

307

locus was amplified by PCR (Kapa Biosystems) and quantified on agarose gel stained with

308

SYBR Gold (Invitrogen) by comparison to a standard. 150 ng of PCR product was melted then

309

hybridized and subjected to cleavage with T7 Endonuclease I (NEB). The resulting reaction

310

was run on a 2% agarose gel stained with SYBR Gold and the cleavage bands quantified using

311

Image Lab (BioRad). The fraction of cleaved PCR product (f(cut)) was determined by the ratio

312

of cleaved DNA (intensity of cleaved bands) to total DNA (combined intensity of all bands).

313

The percentage of indels was determined as follows: 𝑖𝑛𝑑𝑒𝑙𝑠 (%) = 100 × (1 ― √(1 ― 𝑓(𝑐𝑢𝑡)).

314

Genomic DNA – Cas9 target sequences (PAM):

315

Human EMX1 exon 1: GTCACCTCCAATGACTAGGG (TGG)

316

Genomic DNA – Cas12a target sequences (PAM):

317

Human PCSK9 exon 4: (TTTC) CCGGTGGTCACTCTGTATGCTGG

318

Targeted next generation sequencing

319

Genomic DNA was amplified by PCR (Kapa Biosystems) with primers Fwd 2 and Rev 2

320

(Supp. Table 2) comprising a unique molecular identifier (N10) and stub sequence (5’-

321

GCTCTTCCGATCT - 3’) and purified using QIAquick PCR purification kit (Qiagen). Indexes

322

were subsequently added by PCR and the final libraries ran on an agarose gel and extracted.

323

Libraries were quantified by Qubit (Thermo Fisher), pooled and sequenced on a HiSeq 2500

324

(Illumina) using 250 bp paired-end sequencing. Paired-end reads were first merged using

325

PEAR 30, then unique molecular identifier duplicates were removed using PRINSEQ 31, and

326

finally indels were analyzed using Cas-Analyzer tool 32.

327

RT-qPCR

328

HepG2 cells were seeded in individual 3cm petri dishes at a density of 100,000 cells/well. 100

329

pmol of ASGRL-Cas9 RNP were dispensed onto cells and incubated for different times at 37C

330

with 5% CO2. Cells were washed once with PBS and twice with heparin (200 U/ml in PBS),

331

and lysed in Trizol (Life Technologies). Total RNA was extracted from the Trizol, treated with

332

DNase I (NEB) and purified by phenol/chloroform extraction. As a control, 100 pmol of RNP

333

was spiked into Trizol lysed untreated cells. cDNA was generated using 1 µg of RNA and the

334

Superscript Vilo kit (Invitrogen), spiked with sgRNA Rev primer (Supp. Table 2). qPCR was

10

ACS Paragon Plus Environment

Page 10 of 21

Page 11 of 21 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 Chemical Biology

335

performed using the Dynamo SYBR green mix (Thermo Scientific) and the relative quantities

336

of sgRNA determined by double ΔCT analysis between sgRNA (using Fwd 3 and Rev 3) and

337

GAPDH (using Fwd 4 and Rev 4) house-keeping gene PCR (sequences in Supp. Table 2).

338

Cas12a in vitro pre-crRNA processing and DNA cleavage

339

For processing assay, in vitro transcribed pre-crRNA at 20 µM was mixed with Cas12a at 20

340

µM in 20 mM HEPES pH 7.5, 10 mM MgCl2, 10 mM CaCl2, 150 mM KCl and incubated at

341

37C for 15 min followed by 95C for 5 min. Half of the reaction was ran on a PAGE gel and

342

visualized by staining with SYBR Gold. 0.4 pmol (about 150 ng) of human PCSK9 PCR

343

product (amplified using Fwd 5 and Rev 5 primers; sequences in Supp. Table 2) was mixed

344

with 4 pmol of RNP (in 20 mM HEPES pH 7.5, 10 mM MgCl2, 10 mM CaCl2, 150 mM KCl)

345

and incubated at 37C for 30 min followed by 95C for 5 min. Cleavage products were

346

visualized on agarose gel stained with SYBR Gold.

347 348

References

349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377

1. Knott, G. J., and Doudna, J. A. (2018) CRISPR-Cas guides the future of genetic engineering, Science 361, 866-869. 2. Shi, J., Wang, E., Milazzo, J. P., Wang, Z., Kinney, J. B., and Vakoc, C. R. (2015) Discovery of cancer drug targets by CRISPR-Cas9 screening of protein domains, Nat Biotechnol 33, 661-667. 3. Normile, D. (2017) China sprints ahead in CRISPR therapy race, Science 358, 20-21. 4. Yin, H., Xue, W., Chen, S., Bogorad, R. L., Benedetti, E., Grompe, M., Koteliansky, V., Sharp, P. A., Jacks, T., and Anderson, D. G. (2014) Genome editing with Cas9 in adult mice corrects a disease mutation and phenotype, Nat Biotechnol 32, 551-553. 5. Yin, H., Kauffman, K. J., and Anderson, D. G. (2017) Delivery technologies for genome editing, Nat Rev Drug Discov 16, 387-399. 6. Fellmann, C., Gowen, B. G., Lin, P. C., Doudna, J. A., and Corn, J. E. (2017) Cornerstones of CRISPR-Cas in drug discovery and therapy, Nat Rev Drug Discov 16, 89-100. 7. Rouet, R., de Onate, L., Li, J., Murthy, N., and Wilson, R. C. (2018) Engineering CRISPRCas9 RNA–Protein Complexes for Improved Function and Delivery, The CRISPR Journal 1, 367-378. 8. Yang, Y., Wang, L., Bell, P., McMenamin, D., He, Z., White, J., Yu, H., Xu, C., Morizono, H., Musunuru, K., Batshaw, M. L., and Wilson, J. M. (2016) A dual AAV system enables the Cas9-mediated correction of a metabolic liver disease in newborn mice, Nat Biotechnol 34, 334-338. 9. Wu, Z., Yang, H., and Colosi, P. (2010) Effect of genome size on AAV vector packaging, Mol Ther 18, 80-86. 10. Chew, W. L., Tabebordbar, M., Cheng, J. K., Mali, P., Wu, E. Y., Ng, A. H., Zhu, K., Wagers, A. J., and Church, G. M. (2016) A multifunctional AAV-CRISPR-Cas9 and its host response, Nat Methods 13, 868-874. 11. Vakulskas, C. A., Dever, D. P., Rettig, G. R., Turk, R., Jacobi, A. M., Collingwood, M. A., Bode, N. M., McNeill, M. S., Yan, S., Camarena, J., Lee, C. M., Park, S. H., Wiebking, V., Bak, R. O., Gomez-Ospina, N., Pavel-Dinu, M., Sun, W., Bao, G., Porteus, M. H., and Behlke, M. A. (2018) A high-fidelity Cas9 mutant delivered as a ribonucleoprotein

ACS Paragon Plus Environment

11

ACS Chemical 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

378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427

complex enables efficient gene editing in human hematopoietic stem and progenitor cells, Nat Med 24, 1216-1224. 12. Rouet, R., Thuma, B. A., Roy, M. D., Lintner, N. G., Rubitski, D. M., Finley, J. E., Wisniewska, H. M., Mendonsa, R., Hirsh, A., de Onate, L., Compte Barron, J., McLellan, T. J., Bellenger, J., Feng, X., Varghese, A., Chrunyk, B. A., Borzilleri, K., Hesp, K. D., Zhou, K., Ma, N., Tu, M., Dullea, R., McClure, K. F., Wilson, R. C., Liras, S., Mascitti, V., and Doudna, J. A. (2018) Receptor-Mediated Delivery of CRISPR-Cas9 Endonuclease for Cell-Type-Specific Gene Editing, J Am Chem Soc 140, 6596-6603. 13. Mout, R., Ray, M., Yesilbag Tonga, G., Lee, Y. W., Tay, T., Sasaki, K., and Rotello, V. M. (2017) Direct Cytosolic Delivery of CRISPR/Cas9-Ribonucleoprotein for Efficient Gene Editing, ACS Nano 11, 2452-2458. 14. Vouillot, L., Thelie, A., and Pollet, N. (2015) Comparison of T7E1 and surveyor mismatch cleavage assays to detect mutations triggered by engineered nucleases, G3 (Bethesda) 5, 407-415. 15. Sentmanat, M. F., Peters, S. T., Florian, C. P., Connelly, J. P., and Pruett-Miller, S. M. (2018) A Survey of Validation Strategies for CRISPR-Cas9 Editing, Sci Rep 8, 888. 16. Lin, S., Staahl, B. T., Alla, R. K., and Doudna, J. A. (2014) Enhanced homology-directed human genome engineering by controlled timing of CRISPR/Cas9 delivery, Elife 3, e04766. 17. Stockert, R. J. (1995) The asialoglycoprotein receptor: relationships between structure, function, and expression, Physiol Rev 75, 591-609. 18. Rittner, K., Benavente, A., Bompard-Sorlet, A., Heitz, F., Divita, G., Brasseur, R., and Jacobs, E. (2002) New basic membrane-destabilizing peptides for plasmid-based gene delivery in vitro and in vivo, Mol Ther 5, 104-114. 19. Midoux, P., Kichler, A., Boutin, V., Maurizot, J. C., and Monsigny, M. (1998) Membrane permeabilization and efficient gene transfer by a peptide containing several histidines, Bioconjug Chem 9, 260-267. 20. Kichler, A., Leborgne, C., Marz, J., Danos, O., and Bechinger, B. (2003) Histidine-rich amphipathic peptide antibiotics promote efficient delivery of DNA into mammalian cells, Proc Natl Acad Sci U S A 100, 1564-1568. 21. Lundberg, P., El-Andaloussi, S., Sutlu, T., Johansson, H., and Langel, U. (2007) Delivery of short interfering RNA using endosomolytic cell-penetrating peptides, FASEB J 21, 2664-2671. 22. Mellman, I., Fuchs, R., and Helenius, A. (1986) Acidification of the endocytic and exocytic pathways, Annu Rev Biochem 55, 663-700. 23. Appelbaum, J. S., LaRochelle, J. R., Smith, B. A., Balkin, D. M., Holub, J. M., and Schepartz, A. (2012) Arginine topology controls escape of minimally cationic proteins from early endosomes to the cytoplasm, Chem Biol 19, 819-830. 24. Sanhueza, C. A., Baksh, M. M., Thuma, B., Roy, M. D., Dutta, S., Preville, C., Chrunyk, B. A., Beaumont, K., Dullea, R., Ammirati, M., Liu, S., Gebhard, D., Finley, J. E., Salatto, C. T., King-Ahmad, A., Stock, I., Atkinson, K., Reidich, B., Lin, W., Kumar, R., Tu, M., Menhaji-Klotz, E., Price, D. A., Liras, S., Finn, M. G., and Mascitti, V. (2017) Efficient Liver Targeting by Polyvalent Display of a Compact Ligand for the Asialoglycoprotein Receptor, J Am Chem Soc 139, 3528-3536. 25. Zhu, R., Zhang, C. G., Liu, Y., Yuan, Z. Q., Chen, W. L., Yang, S. D., Li, J. Z., Zhu, W. J., Zhou, X. F., You, B. G., and Zhang, X. N. (2015) CD147 monoclonal antibody mediated by chitosan nanoparticles loaded with alpha-hederin enhances antineoplastic activity and cellular uptake in liver cancer cells, Sci Rep 5, 17904. 26. Zetsche, B., Gootenberg, J. S., Abudayyeh, O. O., Slaymaker, I. M., Makarova, K. S., Essletzbichler, P., Volz, S. E., Joung, J., van der Oost, J., Regev, A., Koonin, E. V., and

12

ACS Paragon Plus Environment

Page 12 of 21

Page 13 of 21 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

428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446

ACS Chemical Biology

Zhang, F. (2015) Cpf1 is a single RNA-guided endonuclease of a class 2 CRISPR-Cas system, Cell 163, 759-771. 27. Moreno-Mateos, M. A., Fernandez, J. P., Rouet, R., Vejnar, C. E., Lane, M. A., Mis, E., Khokha, M. K., Doudna, J. A., and Giraldez, A. J. (2017) CRISPR-Cpf1 mediates efficient homology-directed repair and temperature-controlled genome editing, Nat Commun 8, 2024. 28. Kim, Y., Cheong, S. A., Lee, J. G., Lee, S. W., Lee, M. S., Baek, I. J., and Sung, Y. H. (2016) Generation of knockout mice by Cpf1-mediated gene targeting, Nat Biotechnol 34, 808-810. 29. Fonfara, I., Richter, H., Bratovic, M., Le Rhun, A., and Charpentier, E. (2016) The CRISPR-associated DNA-cleaving enzyme Cpf1 also processes precursor CRISPR RNA, Nature 532, 517-521. 30. Zhang, J., Kobert, K., Flouri, T., and Stamatakis, A. (2014) PEAR: a fast and accurate Illumina Paired-End reAd mergeR, Bioinformatics 30, 614-620. 31. Schmieder, R., and Edwards, R. (2011) Quality control and preprocessing of metagenomic datasets, Bioinformatics 27, 863-864. 32. Park, J., Lim, K., Kim, J. S., and Bae, S. (2017) Cas-analyzer: an online tool for assessing genome editing results using NGS data, Bioinformatics 33, 286-288.

447 448

Acknowledgments

449

R.R. thanks J. Doudna, C. Fellmann and members of the Doudna lab at UC Berkeley for helpful

450

discussions. R.R. was a member of the Doudna lab (2015-2017) as part of his early career

451

postdoctoral fellowship from the Australian National Health and Medical Research Council.

452

R.R. acknowledges support from the NHMRC (APP1090875). R.R. thanks members of Pfizer

453

for reagents.

454 455

Author contributions

456

RR designed and performed research; RR analyzed data; RR & DC wrote the manuscript.

457 458

Competing interests

459

The authors declare that they have no competing interests.

460 461

Data availability

462

All data generated or analyzed during this study are included in this published article (and its

463

Supporting Information files).

464 465 466 467 468

Nomenclature CRISPR Clustered Regularly Interspaced Short Palindromic Repeats RNP RiboNucleoProtein ASGR ASialoGlycoprotein Receptor

ACS Paragon Plus Environment

13

ACS Chemical 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

469 470 471 472 473 474

ASGRL NLS T7E1 ASOR EIPA PAGE

14

ASialoGlycoprotein Receptor Ligand Nuclear Localization Signal T7 Endonuclease I ASialo-ORosomucoid N-ethyl-isopropyl amiloride PolyAcrylamide Gel Electrophoresis

ACS Paragon Plus Environment

Page 14 of 21

Page 15 of 21 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

475

ACS Chemical Biology

Figure legends

476 477

Figure 1. Editing efficiency of Cas9-mCherry-3NLS RNP in HepG2 cells. (A) Cartoons

478

representing the previous construct Cas9-1NLS and new one Cas9-mCherry-3NLS (Cas9 in

479

blue, ASGRL in pink, NLS in yellow and mCherry in red). (B) Comparison of human EMX1

480

gene editing efficiency between ASGRL-Cas9-mCherry-3NLS RNP and Cas9-mCherry-3NLS

481

RNP upon co-incubation with ppTG21 endosomolytic peptide. 50 pmol of RNP was incubated

482

with HepG2 cells for 48h with an increasing amount of ppTG21 (0, 50, 200, 800 and 1600

483

pmol or 0, 66, 266, 1067, 2133 nM) and indels analyzed by T7E1 assay (shown in one

484

representative of n=2 independent experiments). (C) Validation of gene editing efficiency with

485

Cas9-mCherry-3NLS RNP only in the presence of ppTG21 or by nucleofection. 50 pmol of

486

RNP was either incubated with or without 750 pmol of ppTG21 or nucleofected in HepG2 cells

487

for 48h and indels analyzed by T7E1 assay (n=6 independent experiments; unpaired t-test).

488 489

Figure 2. Quantification of ASGRL-Cas9 RNP half-life in HepG2 cells upon receptor-

490

mediated endocytosis. 100 pmol (132 nM) of ASGRL-Cas9 RNP was incubated with HepG2

491

cells and the cells washed and harvested at different time points. Total RNA was extracted, and

492

RT-qPCR performed on sgRNA and GAPDH house-keeping gene. Amount of sgRNA was

493

compared to a spike-in control of 100 pmol (n=2 independent experiments).

494 495

Figure 3. Mechanism and localization of endosomal escape with ppTG21 peptide. (A)

496

Effect of substitutions of surface exposed residues to alanine on the editing efficiency of the

497

peptide. 50 pmol (66 nM) of ASGRL-Cas9 RNP was incubated with HepG2 cells and with 750

498

pmol (1 µM) of peptide for 48h and indels analyzed by T7E1 assay. Single mutations are shown

499

in blue and double mutations in purple (n=3 independent experiments). (B) Effect of endocytic

500

pathway inhibitors on the editing efficiency of the Cas9 RNP co-incubated with ppTG21

501

peptide. HepG2 cells were pre-incubated with EGTA at 0.5 mM, ASOR at 80 µM, dynasore at

502

80 µM, EIPA at 50 µM, bafilomycin A at 200 nM, or wortmannin at 200 nM for 1h before the

503

RNP and ppTG21 were added. Cells were harvested after 24h incubation and indels analyzed

504

by T7E1 assay (n=3 independent experiments).

505 506

Figure 4. PCSK9 gene editing with As Cas12a RNP and ppTG21. (A) In vitro pre-crRNA

507

processing with ASGRL-Cas12a and Cas12. Cas12a at 20 µM and pre-crRNA at 20 µM were

508

mixed, incubated at 37C for 15 min followed by 95C for 5 min and resolved on PAGE gel

ACS Paragon Plus Environment

15

ACS Chemical 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

509

(shown is one representative of n=2 independent experiments). (B) In vitro DNA cleavage with

510

ASGRL-Cas12a and Cas12a RNP. 200 ng (~ 0.4 pmol) of PCSK9 PCR product was incubated

511

with 2 pmol of RNP at 37C for 30 min followed by 95C for 5 min and resolved on agarose

512

gel (shown is one representative of n=2 independent experiments). (C) Validation of gene

513

editing efficiency with Cas12a RNP only in the presence of ppTG21 or by nucleofection. 50

514

pmol (66 nM) of RNP was either incubated with or without 375 pmol (500 nM) of ppTG21 or

515

nucleofected in HepG2 cells for 48h and indels analyzed by T7E1 assay (n=4 independent

516

experiments except for nucleofection where n=2, unpaired t-test).

517

16

ACS Paragon Plus Environment

Page 16 of 21

Page 17 of 21 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 Chemical Biology

518 519

ACS Paragon Plus Environment

17

ACS Chemical 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

Figure 2

520 521

18

ACS Paragon Plus Environment

Page 18 of 21

Page 19 of 21 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 Chemical Biology

Figure 3

A 1 5 10 15 20 (ppTG21) GLFHALLHLLHSLWHLLLHA

GLFAALLHLLHSLWHLLLHA GLFHALLALLHSLWHLLLHA GLFHALLHLLASLWHLLLHA GLFHALLHLLHALWHLLLHA GLFHALLHLLHSLAHLLLHA GLFHALLHLLHSLWALLLHA GLFHALLHLLHSLWHLLL AA GLFHALLALLHSLWALLLHA GLFHALLALLHSLWHLLLAA GLFHALLHLLASLWHLLLAA (JTS-1) GLFEALLELLESLWELLLEA (ppTG1) GLFKALLKLLKSLWKLLLKA

B

522 523

ACS Paragon Plus Environment

19

ACS Chemical 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

524

20

ACS Paragon Plus Environment

Page 20 of 21

Page 21 of 21 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 Chemical Biology

80x40mm (300 x 300 DPI)

ACS Paragon Plus Environment