Genetically Encoded Protein Tyrosine Nitration in Mammalian Cells

May 22, 2019 - Strategies for installing site-specific tyrosine nitration in target proteins in ..... Colonies were scraped from the plate using a gla...
0 downloads 0 Views 880KB Size
Subscriber access provided by BOSTON UNIV

Article

Genetically encoded protein tyrosine nitration in mammalian cells Joseph J. Porter, Hyo Sang Jang, Elise M. Van Fossen, Duy P Nguyen, Taylor S. Willi, Richard B. Cooley, and Ryan A. Mehl ACS Chem. Biol., Just Accepted Manuscript • DOI: 10.1021/acschembio.9b00371 • Publication Date (Web): 22 May 2019 Downloaded from http://pubs.acs.org on May 30, 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 24 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

1

ACS Chemical Biology

Genetically encoded protein tyrosine nitration in mammalian cells

2 3

Joseph J. Porter, Hyo Sang Jang, Elise M. Van Fossen, Duy P. Nguyen, Taylor S. Willi, Richard

4

B. Cooley, Ryan A. Mehl*

5 6

Running title: Genetically encode protein tyrosine nitration in mammalian cells

7 8

*To whom correspondence should be addressed: Dr. Ryan A. Mehl, Department of Biochemistry

9

and Biophysics, 2135 ALS, Oregon State University, Corvallis OR 97331-7305 Telephone:

10

(541) 737-4429 Fax: (541) 737-0481 Email: [email protected]

11 12

Keywords: 3-nitrotyrosine, 3-nitrophenylalanine, genetic code expansion, oxidative stress, post-

13

translational modifications (PTMs), tyrosine modifications

14 15 16 17

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 24

18 19

Abstract Tyrosine nitration has served as a major biomarker for oxidative stress and is present in

20

high abundance in over 50 disease pathologies in humans. While data mounts on specific disease

21

pathways from specific sites of tyrosine nitration, the role of these modifications is still largely

22

unclear. Strategies for installing site-specific tyrosine nitration in target proteins in eukaryotic

23

cells, through routes not dependent on oxidative stress, would provide a powerful method to

24

address the consequences of tyrosine nitration. Developed here is a Methanosarcina barkeri

25

aminoacyl-tRNA synthetase/tRNA pair that efficiently incorporates nitrotyrosine site-

26

specifically into proteins in mammalian cells. We demonstrate the utility of this approach to

27

produce nitrated proteins identified in disease conditions by producing site-specific nitroTyr-

28

containing manganese superoxide dismutase and 14-3-3 proteins in eukaryotic cells.

29 30

ACS Paragon Plus Environment

2

Page 3 of 24 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

31 32

ACS Chemical Biology

Introduction Tyrosine nitration is an oxidative post-translational modification (Ox-PTM) that has

33

served as a biomarker of oxidative stress in a variety of human diseases including

34

neurodegeneration, atherosclerosis, and cancer1. While many proteins contain sites of nitration,

35

the consequences of Ox-PTM formation at most of these sites remain unexplored. Progress in

36

this field has been hampered because Ox-PTMs are installed chemically as opposed to standard

37

enzyme directed PTM installation. Ox-PTMs like nitrotyrosine (nitroTyr) are formed through the

38

reaction of cellular reactive oxygen species (ROS) and reactive nitrogen species (RNS) with

39

proteins. It is difficult to study the effects of site-specific Ox-PTMs since no chemical oxidation

40

method is specific to a single amino acid type or location2. To address these challenges, genetic

41

code expansion (GCE) is a promising technology that permits the programmable installation of

42

single or multiple PTMs site-specifically, thus enabling direct interrogation of the consequences

43

of site-specific Ox-PTMs.

44

Many reports have proposed the functional importance of tyrosine nitration on proteins

45

involved in cell signaling, metabolism, and cellular structure but no direct method is available to

46

evaluate which sites of tyrosine nitration have a cellular effect for eukaryotes3-8. Nitration of the

47

mitochondrial antioxidant enzyme manganese superoxide dismutase (MnSOD) is known to

48

compromise its function and has been implicated in several chronic inflammatory diseases

49

including chronic organ rejection, arthritis, and tumorigenesis9. Nitration is also found at key

50

regulatory binding sites on all 7 isoforms of the 14-3-3 family of phospho-binding proteins

51

which regulate most major cellular functions 10-13. The standard approach to identify these

52

modifications is to immunoblot cellular protein for nitroTyr modifications, then the site of

53

tyrosine modification is identified via mass spectrometry. It is common to mutate the site(s) of

54

tyrosine nitration to phenylalanine and the cells are exposed to the same conditions that caused

55

the original modification. The comparison of activity between the tyrosine- and the

56

phenylalanine-containing protein can show that removing a site of tyrosine nitration ablates the

57

cell phenotype originally seen. This approach is not always feasible for proteins like MnSOD

58

and 14-3-3 as conversion of key active-site tyrosine(s) to phenylalanine(s) adversely affect

59

protein function. A method to site-specifically install nitroTyr in eukaryotic cells is necessary in

60

order to directly demonstrate the impact of a particular site of nitration on protein and cellular

61

function.

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

62

Page 4 of 24

GCE uses orthogonal aminoacyl-tRNA synthetase/tRNACUA (aaRS/tRNA) pairs

63

engineered for specific non-canonical amino acids (ncAAs)14. This method allows for the

64

synthesis of recombinant proteins containing PTMs, providing insights into how these

65

modifications regulate protein structure and function15. A GCE system developed for the

66

incorporation of nitroTyr has been used to show the functional consequences of nitroTyr

67

modification on proteins in vitro16, 17. This tyrosyl-RS/tRNA pair derived from the methanogenic

68

archaeon Methanocaldococcus janaschii (Mj) is orthogonal in E. coli and has been used to

69

produce a number of site-specifically nitrated proteins for functional studies16, 18, 19. Studying the

70

effect of nitration in heat shock protein 90 (Hsp90) in vivo required the expression and

71

purification of site-specifically modified Hsp90 from E. coli followed by delivery to eukaryotic

72

cells via protein transfection reagent18. While transfection of cells with nitrated protein was

73

successful in evaluating the toxicity of site-specifically nitrated Hsp90, studies on cell

74

development, protein processing, or transmembrane proteins will require eukaryotic cell

75

expression. The Mj nitroTyr-RS/tRNA pair is not orthogonal in eukaryotic cells, necessitating a

76

new system for evaluating the effects of nitroTyr in eukaryotic systems. The pyrrolysine

77

RS/tRNA pair from several species of methanogenic archaea has emerged as a particularly useful

78

platform for GCE as it allows for the evolution of new aaRSs in E. coli and application of the

79

evolved aaRS/tRNA in bacterial and eukaryotic cells20.

80

Here we develop pyrrolysine aaRS/tRNA pairs that can encode nitroTyr, and the

81

structural analogue 3-nitroPhenylalanine (3-nitroPhe). We characterize the efficiency and fidelity

82

of these aaRS/tRNA pairs using ncAA-sfGFP expressed in E. coli and mammalian cells. We

83

show that functionally efficient pyrrolysine aaRS/tRNA pairs in E. coli do not always function in

84

mammalian cells but this limitation can be overcome when the efficiency of the pyrrolysine

85

aaRS/tRNA pairs is improved to function with lower concentrations of ncAA. We demonstrate

86

the utility of the optimized nitroTyr-RS/tRNA pairs by producing the physiologically nitrated

87

proteins, MnSOD and 14-3-3, in mammalian cells and verify the site-specific nitration in vivo.

88

The technology to site-specifically install nitroTyr and structural analogues directly into proteins

89

in mammalian cells will facilitate an understanding of the structural and functional consequences

90

of this Ox-PTM.

ACS Paragon Plus Environment

4

Page 5 of 24 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

91 92 93 94

Results and Discussion Selection of an aminoacyl-tRNA synthetase specific for nitroTyrosine and 3-nitroPhenylalanine. To enable studies of nitroTyr modifications on eukaryotic cellular function, we sought to

95

identify a Methanosarcina barkeri pyrrolysyl-aaRS/tRNA pair capable of site-specifically

96

incorporating nitroTyr into proteins in response to an amber stop codon (TAG). To do this, we

97

screened a library of MbPylRS variants in which five active-site residues were randomized to all

98

20 amino acids (L270, Y271, L274, N311, C313)21. After a single round of positive selection in

99

the presence of nitroTyr or 3-nitroPhe and a single round of negative selection against canonical

100

amino acids, 48 colonies were assessed for their efficiency in suppressing a TAG codon

101

interrupted sfGFP gene at amino acid site 150 (sfGFP-150TAG) in the presence of nitroTyr and

102

3-nitroPhe. Simultaneously, the ability of the selected synthetases to discriminate against

103

canonical amino acids was assessed by expressing the sfGFP-150TAG in the absence of ncAA.

104

The top 16 performing clones, as based on their efficiency (full-length protein yield) and fidelity

105

(level of canonical amino acid misincorporation), were further evaluated at a larger scale in the

106

presence of 1 mM nitroTyr and 1 mM 3-nitroPhe. Sequencing of these 16 clones revealed 14

107

unique MbPylRS sequences (Supporting Table 1). Of these clones, the selected mutant MbPylRS

108

“F4” efficiently incorporated nitroTyr and showed remarkable permissivity for 3-nitroPhe

109

compared to the other mutant MbPylRSs (Supporting Figure 1), and was chosen for use in

110

mammalian cells. While the F4 MbPylRS efficiently encoded nitroTyr and 3-nitroPhe in E. coli,

111

when we tested its ability to incorporate these ncAAs into sfGFP in HEK293T cells only 3-

112

nitroPhe was efficiently incorporated (Figure 1 and Supporting Figure 2). In order to select an

113

MbPylRS that would efficiently incorporate nitroTyr in eukaryotic cells, the entire pool of

114

library members left after the first round of positive and negative selection were subjected to two

115

more rounds each of positive and negative selections. The most efficient mutant MbPylRS from

116

these rounds of selection “A7” incorporated nitroTyr in E. coli but was not permissive to 3-

117

nitroPhe (Figure 1B). The A7 MbPylRS/tRNA pair possesses comparable efficiency and fidelity

118

for nitroTyr incorporation in E. coli as compared to the previously evolved Mj tyrosyl-tRNA

119

synthetase/tRNA pair (Mj-RS 5b)17.

120 121 122

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

Page 6 of 24

123 124

125 126 127 128 129 130 131 132 133 134 135 136 137

Figure 1. Evaluation of top selection hits by expression of TAG-interrupted sfGFP. (A) Structures of nitroTyr and the 3-nitroPhe incorporated via genetic code expansion in this study. (B) Assessment of fluorescence normalized to optical density at 600 nm for cells expressing the sfGFP150TAG gene along with each of the synthetase variants identified from the selection process. Cultures were expressed in the presence of 1 mM nitroTyr (black), 3-nitroPhe (light gray), in the absence of ncAA (grey). The top MbPylRS/tRNA pairs characterized in this study were compared against the previously developed Mj tyrosyl aminoacyl-tRNA/tRNA pair for nitroTyr (5b MjRS). The WT sfGFP positive control represents non-TAG-interrupted sfGFP expression levels achieved by purely natural translation. (C) Coomassie blue stained SDS-PAGE of sfGFP variants expressed and affinity purified from cultures shown in panel B.

We then evaluated the efficiency, fidelity, and permissivity (range of ncAAs

138

incorporated) of the F4 and A7 MbRSs in an E. coli sfGFP-150TAG suppression reporter assay.

139

The F4 MbPylRS efficiently suppressed sfGFP-150TAG in the presence of 1 mM nitroTyr and

140

3-nitroPhe-, while the A7 MbPylRS only suppressed sfGFP-150TAG in the presence of 1 mM

141

nitroTyr (Figure 1B). Modified sfGFP was purified to verify the ncAA incorporation did not

142

alter sfGFP fluorescence, alter sfGFP solubility and in vivo fluorescence resulted from full-

143

length expressed sfGFP containing a C-terminal 6xhis affinity tag. Approximately 340 mg

144

(nitroTyr – F4 MbPylRS), 750 mg (3-nitroPhe- F4 MbPylRS), and 140 mg (nitroTyr – A7

145

MbPylRS) of sfGFP containing ncAA at site 150 were purified to homogeneity per liter of media

146

(Figure 1C). For comparison, WT-sfGFP yielded 500 mg per liter culture under similar

147

conditions. No substantial amber suppression was observed in cultures not supplemented with

148

ncAA and in vivo sfGFP-fluorescence correlated with purified protein yields. (Figure 1C).

149

To confirm nitroTyr and 3-nitroPhe were accurately incorporated into recombinant

150

proteins by the F4 and A7 MbPylRSs, we measured the masses of WT-sfGFP, sfGFP-nitroTyr-

151

150, and sfGFP-3-nitroPhe-150 using ESI-Q-Tof mass analysis (Supporting Figure 3). Each of

ACS Paragon Plus Environment

6

Page 7 of 24

152

these variants had the expected masses associated with incorporation of their respective ncAA.

153

No mis-incorporation of natural amino acids was detected.

154

While E. coli media is generally supplemented with 1 mM ncAA, we wanted to assess

155

the concentration of ncAA required for efficient production of ncAA-containing sfGFP using the

156

F4 and A7 MbPylRSs for recombinant protein expression in cells. In order to assess this, we

157

titrated the amount of nitroTyr or 3-nitroPhe added to the media and measured the amount of

158

sfGFP-TAG-150 protein produced. The curves shown in Figure 2 are fit with parameters we

159

have termed UP50 (ncAA concentration at which half-maximal sfGFP-150-ncAA is produced)

160

and UPmax (maximum amount produced or the yield of sfGFP). These analyses revealed F4

161

MbPylRS UP50’s of 90 μM and 600 μM respectively for nitroTyr and 3-nitroPhe. The A7

162

MbPylRS UP50 was 3 μM for nitroTyr, while 3-nitroPhe is a very poor substrate for this

163

MbPylRS (Figure 2B). Based on the lower UP50 for nitroTyr, we expect that the A7 MbPylRS

164

will have a higher yield of nitroTyr-containing protein at lower nitroTyr concentrations in cells.

B

5000 4000 3000 2000

F4 MbRS A7 MbRS

1000 0 0

165

OD Adjusted Fluorescence (A.U.)

A

OD Adjusted Fluorescence (A.U.)

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

0.2

0.4 0.6 [nitroTyr] mM

0.8

1

1.2

16000 12000 8000

F4 MbRS A7 MbRS

4000 0 0

0.5

1 1.5 2 [3-nitroPhe] mM

2.5

3

166 167 168 169 170

In-cell fluorescence was measured to determine relative amounts of sfGFP-150-nitroTyr or sfGFP-150-3nitroPhe protein production in the presence of (A) nitroTyr, or (B) 3-nitroPhe. The curves indicated display the best fit from which the indicated UP50 values were derived.

Figure 2. UP50 determination for nitroTyr or 3-nitroPhe incorporated by the F4 or A7 MbPylRS.

171

We have found that nitroTyr sensitive antibodies used to monitor protein nitration show a

172

wide range of sensitivities dependent on the nitrated protein as well as the site of nitration. To

173

determine the best antibody to monitor site-specifically nitrated proteins in HEK cells we

174

prepared nitroTyr- and 3-nitroPhe-containing MnSOD and 14-3-3 proteins in E. coli. All of the

175

three antibodies tested were immunoreactive to MnSOD with nitroTyr incorporated at residue 34

176

while they displayed minimal immunoreactivity to 3-nitro-Phe installed at the same site

ACS Paragon Plus Environment

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

Page 8 of 24

177

(Supporting Figure 4A-C). For site-specifically modified human 14-3-3 β protein, only the

178

polyclonal nitroTyr antibodies were immunoreactive to 14-3-3 with nitroTyr at site 130, while

179

the monoclonal nitroTyr antibody 1A6 was not immunoreactive under the conditions tested

180

(Supporting Figure 4A-C). Based on the broad specificity and robust sensitivity we used the

181

Millipore polyclonal nitroTyr antibody in future studies to monitor nitroTyr incorporation into

182

proteins in eukaryotic cells. The substrates tested demonstrated marked differences in nitroTyr

183

antibody sensitivity indicating that those employing nitroTyr antibodies should be mindful to

184

determine the sensitivity of the various nitroTyr antibodies to their protein of interest. Finally,

185

the Cayman polyclonal nitroTyr antibody displayed a low level of immunoreactivity to human

186

14-3-3 β with 3-nitroPhe incorporated at site 130, indicating that the nitrophenol moiety is

187

generally necessary for efficient recognitions by the nitroTyr antibodies (Supporting Figure 4C).

188 189 190

Expression of site-specifically incorporated nitroTyr proteins in eukaryotic cells The MbPylRS/tRNA pair possesses the added utility of usage in eukaryotic cells14, 22

191

potentially allowing the study of nitroTyr in its native cellular context. To that end we set out to

192

apply this system to the first genetically encoded Ox-PTM in eukaryotic cells. Before

193

incorporating nitroTyr in eukaryotic cells, we first determined the maximum allowable

194

concentration of nitroTyr for HEK293T cell viability. We found that nitroTyr did not display

195

significant toxicity at concentrations up to 0.3 mM at 48 hours after treatment, but cell viability

196

was compromised when media was supplemented with 1 mM nitroTyr (Supporting Figure 5).

197

Interestingly, 3-nitroPhe showed no apparent effect on the cell density when evaluated up to 1

198

mM, indicating that the 3-nitroPhe is notably less toxic than nitroTyr. Based on the HEK293T

199

cells toxicity profile for nitroTyr and 3-nitroPhe, future experiments were conducted using 0.3

200

mM ncAA in the media.

201

We hypothesized that we could take advantage of the highly efficient F4-MbPylRS/tRNA

202

pair selected for incorporation of nitroTyr in E. coli and use it for incorporation of nitroTyr in

203

mammalian cells. To test this, we cloned a human codon optimized version of the F4 synthetase

204

and tRNA into a pAcBac1 mammalian expression vector and in a separate pAcBac1 vector we

205

cloned a sfGFP-TAG150 (Supporting Figure 6). To our surprise, the F4-MbPylRS/tRNA pair

206

could efficiently incorporate 3-nitroPhe but not nitroTyr in HEK293T cells, as mentioned above

207

(Supporting Figure 2). Since the F4-MbPylRS/tRNA pair functioned efficiently with 0.1-1.0 mM

208

3-nitroPhe in the media, the GCE machinery is being functionally produced in eukaryotic cells.

ACS Paragon Plus Environment

8

Page 9 of 24 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

209

Comparing efficiencies of 3-nitroPhe vs nitroTyr UP50 data, the maximum efficiency of the F4-

210

MbPylRS/tRNA pair in E. coli for 3-nitroPhe was three times that compared to nitroTyr (Figure

211

2). If there is an ncAA uptake difference between E. coli and HEK cells, this efficiency

212

difference at low concentrations of ncAA could be exacerbated. We reasoned that a synthetase

213

able to more efficiently incorporate nitroTyr at lower nitroTyr concentrations was needed for

214

mammalian cell expression. After selecting and characterizing additional Mb synthetases (see

215

Materials and Methods), the A7-MbPylRS was found to be more efficient at lower nitroTyr

216

concentrations than the F4-MbPylRS (Figure 2A), and so the gene for it was codon optimized for

217

mammalian expression and incorporated into the pAcBac1 plasmid. Indeed, the A7-MbPylRS

218

did support robust incorporation of nitroTyr in mammalian cells in the presence of 0.3 mM

219

nitroTyr, but not in the absence of ncAA, as shown by fluorescence microscopy and fluorescence

220

of crude cell lysate (Figure 3 B-F).

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

Page 10 of 24

221 222 223 224 225 226 227 228 229 230 231

Figure 3. Expression and optimization of nitroTyr-containing protein in HEK293T cells. (A) Two plasmids were co-transfected for expression of nitroTyr-containing protein in HEK293T cells. The first plasmid (P1) contains two copies of MmPylTc downstream of U6 promoter (U6-Pyl tRNA1), two copies of DhPyl3M downstream of H1 RNA polymerase III promoter (H1-Pyl tRNA2), and sfGFP150TAG downstream of the CMV promoter (CMV). pA indicates bovine growth hormone gene polyadenylation signal. The second plasmid (P2) two copies of MmPylTc downstream of U6 promoter (U6-Pyl tRNA1), two copies of DhPyl3M downstream of H1 RNA polymerase III promoter (H1-Pyl tRNA2), and A7-MbPylRS, downstream of the CMV promoter. HEK293T cells were transfected for 48 h with P1 and P2 using Lipofectamine 2000 (Thermofisher) in the presence (C and E) or absence (B and D) of 0.3 mM nitroTyrosine. Fluorescence images (B and C) and phase contrast images (D and E) were

ACS Paragon Plus Environment

10

Page 11 of 24 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

232 233 234 235 236 237 238 239 240 241 242 243

captured using EVOS FL cell imaging system (Thermofisher). Scale bar, 50 μm. (F) Cell lysates were prepared in modified RIPA buffer and sfGFP fluorescence was measured using 485 nm excitation and 528 emission. (G) Assessment of production of nitroTyr-containing sfGFP based on the ratio of plasmid containing tRNA and sfGFP-150TAG (P1) to plasmid containing tRNA and A7-MbPylRS (P2). Increasing the ratio of tRNA and sfGFP-150TAG to A7-MbPylRS increases nitroTyr-containing sfGFP production shown here by flow cytometry. The MFI for (+)nitroTyr P2 population was calculated using CytExpert software. One-way ANOVA with Bonferroni post-test. mean±SEM. #, not significant; *, P