Assaying RNA Localization in Situ with Spatially Restricted

Sep 27, 2017 - To localize fluorophores at specific subcellular regions, we hypothesized we could tether dyes to Halo-tag fusion proteins, which are l...
0 downloads 7 Views 1MB Size
Subscriber access provided by Purdue University Libraries

Letter

Assaying RNA localization in situ with spatially-restricted nucleobase oxidation Ying Li, Mahima B. Aggarwal, Kim Nguyen, Ke Ke, and Robert C. Spitale ACS Chem. Biol., Just Accepted Manuscript • DOI: 10.1021/acschembio.7b00519 • Publication Date (Web): 27 Sep 2017 Downloaded from http://pubs.acs.org on September 27, 2017

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 free 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 accessible to all readers and 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.

ACS Chemical Biology 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 16

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

Assaying RNA localization in situ with spatially-restricted nucleobase oxidation

2 3

Ying Li1, Mahima B. Aggarwal1, Kim Nguyen1, Ke Ke1, Robert C. Spitale*1,2

4

(1) Department of Pharmaceutical Sciences, (2) Department of Chemistry. University of

5

California, Irvine. Irvine, California. 92697

6 7

Correspondence: [email protected]

8 9

Abstract.

10

We report herein a novel chemical-genetic method for assaying RNA localization within living

11

cells. RNA localization is critical for normal physiology as well as the onset of cancer and

12

neurodegenerative disorders. Despite its importance there is a real lack of chemical methods to

13

directly assay RNA localization with high resolution in living cells. Our novel approach relies on

14

in situ nucleobase oxidation by singlet oxygen generated from spatially confined fluorophores.

15

We demonstrate that our novel method can identify RNA molecules localized within specific

16

cellular compartments. We anticipate that this platform will provide the community with a much-

17

needed methodology for tracking RNA localization within living cells, and sets the stage for

18

systematic large scale analysis of RNA localization in living systems.

19 20 21 22 23 24 25 26

ACS Paragon Plus Environment

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

27 28

Page 2 of 16

Text. Recent genetic and functional analysis has demonstrated that RNA molecules can

29

control many aspects of gene regulation: from transcription to translation.1,

2

30

organization of an RNA molecule can be intimately related to its function. For example,

31

chromatin-associated RNAs can regulate transcription.3 Cytosolic RNA molecules can interact

32

with messenger RNAs (mRNAs) to control translation and mRNA decay.4 mRNAs can also be

33

localized to the outer membrane of the mitochondria for import into the mitochondrial matrix.5

34

Each of these facets of RNA localization is critical for RNA function and cellular fitness.

The spatial

35

Analysis of RNA localization is critical for studying RNA functions. Current methods for

36

RNA localization study primarily focus on a small number of RNAs6 and typically require

37

intensive labor7 to achieve slightly larger scale analysis. In addition, these strategies rely highly

38

on existing knowledge about the primary sequence of RNAs (in situ hybridization) and limit the

39

potential to discover unknown or unconventional RNA localization. Cellular fractionation,

40

permitting relatively larger scale analysis (sequencing etc.), inevitably comes with many false-

41

positives. During fractionation, the original spatial organization of biomolecules is disrupted and

42

the falsely re-associated biomolecules prevent accurate analysis for further study.8,

43

development of a method that begins to address these challenges, while assaying RNA

44

localization in situ, would have a tremendous impact on our ability to analyze RNA localization.

9

The

45

Here we demonstrate that RNA localization can be assayed within intact living cells

46

through spatially restricted nucleobase oxidation. We employ localized fluorophores, which,

47

upon excitation with blue light (Supporting Information, SI), can utilize energy transfer to excite

48

nearby ground state triplet oxygen to singlet state, resulting in guanosine oxidation in RNA.

49

Nucleophiles in solution can intercept oxidized guanosine residues to tag them for downstream

50

study. We demonstrated that this oxidative approach provides subcellular, and even sub-

51

organellar resolution for RNA localization. Sub-cellularly tagged RNAs can be enriched and

52

profiled by RT-qPCR. The enrichment step significantly reduces background and focuses on

ACS Paragon Plus Environment

Page 3 of 16

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

53

genes of interest. We anticipate that many labs aiming to explore RNA localization will adopt our

54

method: from one gene to many in parallel.

55

Tagging RNA with high resolution inside living cells is a formidable challenge. We

56

envisioned that singlet oxygen induced guanosine oxidation could be useful in this regard as the

57

singlet oxygen has an extremely short half-life and thus a short diffusion radius (~268 nm).10

58

Recent analysis on singlet oxygen diffusion inside cells is consistent with this note.11

59

Fluorescent dyes are well known for their ability to perform triplet energy transfers to molecular

60

oxygen (in the triplet state).12 The resulting singlet oxygen can oxidize guanosine to 8-

61

oxoguanosine (8oxoG, Figure 1A). The higher oxidation state of 8oxoG is susceptible to attack

62

by a wide variety of nucleophiles in solution.13-15 We therefore hypothesized that restricted

63

localization of a fluorophore would induce a high local concentration of singlet oxygen, which

64

would permit tagging of guanine residues in RNA in the presence of a nucleophile (amine,

65

outlined in Figure 1 B).

66

We focused on the fluorophore eosin, as it has been demonstrated to afford singlet

67

oxygen efficiently.16 To first demonstrate the ability to form adducts with guanine, in a singlet-

68

oxygen dependent manner, we exposed single stranded synthetic DNA oligonucleotides (15 nt),

69

which contain four guanosine residues to blue light in the presence of eosin and amino PEG

70

(Figure 2A). We observed adducts formed only in the presence of blue light and only on an

71

oligonucleotide that contained guanine (Figure 2B). We also characterized the time-dependent

72

nature of the adduct formation and observed the reaction reaches a limit after ~15 minutes of

73

blue light exposure (Figure 2C). To demonstrate the formation of a biotin adduct we exposed

74

isolated total RNA to blue light with eosin and amino-PEG2-biotin and observed biotinylation by

75

transfer blot (Figure 2D). Biotinylation was inhibited in the presence of sodium azide, a known

76

singlet oxygen quencher (Figure 2E).17 Overall these data suggest that singlet oxygen,

77

generated from fluorophore excitation, can oxidize guanine and permit biotinylation for potential

78

downstream enrichment.

ACS Paragon Plus Environment

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

79

We next focused on demonstrating that RNA tagging could be accomplished within living

80

cells. HEK293T cells were incubated with eosin and propargyl amine (PA), a smaller amine

81

nucleophile, which should easily traverse into cells. Extracted RNA was appended with a biotin

82

tag through azide-biotin using conventional Cu(I)-catalyzed azide-alkyne cycloaddition

83

(CuAAC). We were able to observe significant amounts of tagged RNA when cells were

84

exposed to blue light in the presence of eosin and PA, even at concentrations as low as 100 µM

85

for eosin (Figure 2F). We also observed significant amounts of protein and DNA labeling under

86

these conditions, suggesting that our methodology could be used to track DNA and protein

87

localization in situ (SI, Figure S1). Our results demonstrate that our tagging approach is capable

88

of working inside cells.

89

As a starting point for verifying the resolution of RNA tagging, we focused on the nucleus

90

and cytoplasm. These two compartments are experimentally feasible and have several known

91

positive controls. To localize fluorophores at specific subcellular regions we hypothesized we

92

could tether dyes to Halo-tag fusion proteins, which are localized to specific regions of the cells

93

(Figure 3A).18 We synthesized a dibromofluorescein-halo ligand (DBF), a synthetically easier

94

eosin derivative that is more hydrophilic and has slightly lower yield of singlet oxygen generation

95

(SI).19 We reasoned DBF would have lower background and higher-resolution tagging than

96

eosin.

97

We utilized Halo-tag constructs localized in the nucleus (histone protein H2B) and

98

cytoplasm (NF-kappa-B p65) (Figure 3A; SI). Imaging experiments revealed very specific

99

localization of Halo-DBF. Exposure to blue light, followed by CuAAC to visualize propargyl

100

amine, afforded well-overlapping signals with Halo-DBF. (Figure 3B and 3C). Negative controls

101

(- hv; -PA; -DBF) showed lack of adduct formation with PA (Figure S2).

102

To test the specificity of enrichment we profiled tagged RNAs by RT-qPCR. We chose

103

the following controls for nuclear-localized RNAs: (1) 7SK, a ncRNA transcribed from RNA

104

polymerase III (pol. 3) that remains tightly associated with chromatin;20 (2) U6 nuclear snRNA,

ACS Paragon Plus Environment

Page 4 of 16

Page 5 of 16

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

105

which is transcribed from pol. 3 and remains in the nucleus;21 (3) U1 snRNA, which is

106

transcribed by pol. 2, exported into the cytoplasm and recycled back into the nucleus.22 For the

107

cytoplasm the following control RNAs were profiled: (1) GAPDH and actin mRNA, which are

108

transcribed by pol. 2 and exported into the cytoplasm;23 (2) 18S rRNA, which is transcribed by

109

pol. 1 in the nucleolus and exported into the cytoplasm.24

110

We first focused on 7SK for enrichment from Halo-H2B expressing cells. Increased

111

exposure time to blue light irradiation, in the presence of PA, resulted in an exponential increase

112

in enrichment over background, with 5-minute short exposure giving reasonable enrichment

113

(Figure S3). Enrichment experiments based on fixed and permeabilized cells were also

114

performed but afford less spatial resolution despite high yields of adduct formation (Figure S4).

115

As such, performing the singlet oxygen induced tagging in living cells results in lower yield of

116

tagging, but much better resolution. These results are consistent with the observation that many

117

singlet oxygen quenchers exist in the cells, which would decrease the labeling radius.25 We

118

therefore proceeded with these conditions in living cells for all the subsequent experiments.

119

We then performed more systematic analysis of enrichment patterns for both nuclear

120

and cytoplasmic localizations. As shown in Figure 3D, RNAs predicted to be in the nucleus were

121

highly enriched in the Halo-H2B expressing cells. RNAs predicted to be in the cytoplasm were

122

highly enriched in the Halo-P65 expressing cells. Notably, the two RNAs that do not exit the

123

nucleus (7SK and U6) have very striking differences between the two conditions. 7SK, which is

124

tightly bound to chromatin, had the largest difference for fold enrichment. U1 snRNA, which is

125

exported then re-imported, still had low enrichment in Halo-p65 expressing cells. Consistent

126

with its known localization it was highly enriched in the Halo-H2B cells.

127

As predicted, GAPDH and actin mRNAs had substantial enrichment in the Halo-P65

128

cells and very little enrichment in the Halo-H2B cells (Figure 3E). Perhaps the most remarkable

129

result is the lack of 18S rRNA tagging in the Halo-H2B construct: this is impressive because the

130

18S rRNA is a highly abundant RNA and it is transcribed within the nucleolus, a sub

ACS Paragon Plus Environment

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

131

compartment in the nucleus not associated with chromatin (where H2B is localized). This RT-

132

qPCR result is consistent with the lack of DBF and Cy5 signal in the nucleolus (Figure 3B).

133

These results strongly suggest that localized RNA tagging can be achieved in living cells with

134

locally induced singlet oxygen in the presence of propargyl amine.

135

The results above suggested that singlet oxygen induced tagging is a high-resolution

136

method for analyzing RNA localization in situ. We next sought to further test the resolution limits

137

of our approach. We focused on the nucleolus and wondered if localization of DBF inside the

138

nucleolus would permit selective tagging of RNAs therein. We focused on three RNAs: (1) 18S

139

rRNA which is transcribed within the nucleolus and then exported into the cytoplasm; (2) U13

140

snRNA, which is specifically localized within the nucleolar compartment;26 (3) 7SK which is

141

known to remain tightly bound to chromatin.

142

We constructed a Halo-tag protein fusion with fibrillarin (Fib), a component of the small

143

nucleolar ribonucleoprotein (snRNP) that is highly enriched in the nucleolus.27 Imaging of the

144

Halo-Fib fusion showed that DBF is highly accumulated within the nucleolus, in sharp contrast

145

to Halo-H2B fusion (Figure 4A). The PA signal also suggested confined RNA tagging within the

146

nucleolus for Halo-Fib (Figure 4A). We compared the RT-qPCR profiles for the three RNAs from

147

cells transfected with Halo-Fib and Halo-H2B. As predicted, U13 and 18S are highly enriched in

148

Halo-Fib expressing cells and 7SK is highly enriched in the Halo-H2B expressing cells (Figure

149

4B). Together, these observations strongly support that the resolution of our approach is at sub-

150

organellar level.

151

In this Letter, we have developed a framework for assaying RNA localization within intact

152

living cells. We have utilized localized fluorophores to convert triplet oxygen to singlet oxygen

153

for spatially-restricted RNA oxidation. We have demonstrated that oxidized RNAs can be tagged

154

with nucleophiles for subsequent enrichment and profiling by RT-qPCR. Our approach is of high

155

resolution as RNAs can be tagged within sub-organeller compartments.

ACS Paragon Plus Environment

Page 6 of 16

Page 7 of 16

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

156

We have designed our approach to interface with the Halo tag proteins, which are now

157

commercially available for many human proteins. As such, it can be widely applied for

158

systematic analysis of RNA localization. In addition, the approach of RT-qPCR paves the way to

159

RNA sequencing for potential discovery of RNAs localized within specific subcellular

160

compartments. We anticipate the results herein will provide a roadmap for a wide-scale and

161

systematic analysis of RNA localization (live cell differentiation, etc). Such studies are currently

162

underway in our lab.

163 164

Acknowledgements

165

We thank members of the Spitale lab for their careful reading and critique of the

166

manuscript. Spitale lab is supported by start up funds from the University of California, Irvine,

167

and the NIH (1DP2GM119164 RCS). RCS is a Pew Biomedical Scholar.

168 169 170

Supporting Information Supporting information accompanying this manuscript is available online. These include

171

NMR spectra and additional biochemical methods. This material is available free of charge via

172

the internet at http://pubs.acs.org.

173

ACS Paragon Plus Environment

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

174 175

Figures.

176 177 178 179 180

Figure 1. Fluorophore-mediated RNA oxidation. (A) Schematic of fluorophore-mediated oxidation of guanosine, followed by nucleophilic tagging. (B) Schematic of localized RNA oxidation, followed by enrichment and profiling.

ACS Paragon Plus Environment

Page 8 of 16

Page 9 of 16

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

181 182 183 184 185 186 187 188 189 190

ACS Chemical Biology

Figure 2. Fluorophore-mediated RNA oxidation tagging. (A) Schematic of eosin-mediated oxidation and RNA tagging by an alkyl amine. B = biotin. (B) Phosphoroimaging gel shift demonstrating guanosine-dependent oxidation and nucleophilic adduct formation with amino PEG (1 kDa). G = guanosine (C) Phosphoroimaging gel of time-dependent guanosine tagging (0-20 min). (D) Demonstration of eosin-mediated tagging with amino-PEG2-biotin, in vitro. (E) Sodium azide, a singlet oxygen quencher, decreases the signal of eosin-mediated RNA tagging. (F) Demonstration of eosin-mediated RNA tagging inside living cells (final concentration of eosin: 100, 200 500 µM). EB = ethidium bromide loading control. Biotin = streptavidin blot.

ACS Paragon Plus Environment

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

191 192 193 194 195 196 197 198

Page 10 of 16

Figure 3. Spatially restricted tagging of RNA inside living cells. (A) Schematic of localized DBF to Halo fusion proteins. (B) Imaging of Halo-H2B fusions. DBF is denoted by green signal. PA is denoted by red signal. (C) Imaging of Halo-P65 fusions. DBF is denoted by green signal. PA is denoted by red signal. (D & E) RT-qPCR from Halo-H2B and Halo-P65 experiments. Enrichment was calculated against a no-hv negative control with ∆∆Ct method. N=3; biological duplicates.

ACS Paragon Plus Environment

Page 11 of 16

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

199 200 201 202 203 204

ACS Chemical Biology

Figure 4. Spatially restricted tagging of RNA inside the nucleolus. (A & B) Imaging of HaloFib and Halo-H2B constructs. Yellow arrows mark the nucleolus. (C) RT-qPCR of U13, 7SK, and 18S RNA. Enrichment was calculated against a no-hvnegative control with ∆∆Ct method. N=3; biological duplicates.

205 206 207 208 209 210 211 212 213 214

ACS Paragon Plus Environment

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

215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261

Page 12 of 16

Methods. Additional methods can be found within the supplementary information. Synthesis. Synthesis and spectra for all compounds is found within the supplementary information. Cell lines and culture conditions. Hek293T cells were cultured in DMEM supplemented with 10% FBS, 1% penicillin and streptomycin and grown at 37ºC, 5% CO2. Cloning and plasmids. H2B-Halo plasmid (a kind gift from Dr. Luke D. Lavis laboratory; Jenelia Research Campus, HHMI, Virginia, USA) was used as a template for amplifying Halo tag sequence by polymerase chain reaction (PCR) with primers oKN283F (5’-aattt ACCGGT ATGGGATCCG AAATCGGTAC TGGCTTTCCA TTCGACCC-‘3) and oKN284R (5’TCAATGGTAC CGCCGGAAAT TTCTAGCGTC GACAGCCA-‘3) using KOD Hotstart DNA polymerase (EMD Millipore Corp.) Halo tag fragment was cloned into pTagRFP-C1-Fibrillarin backbone (Addgene ID # 70649) and replaced RFP sequence via AgeI and KpnI resulting pKN326 or Halo-Fib plasmid. The plasmid sequence was verified by Sanger sequencing analysis (Genwiz, Inc.) Photo-oxidation labeling of cellular RNA. The cell culture dishes were coated with poly-Dlysine (50 µg/ml) for 6 h at 37ºC and washed three times with autoclaved water to remove the excessive amount of poly-D-lysine. HEK293T cells were seeded at equal density (106 cells/plate in 10-cm plates). HEK293T cells were transiently transfected for 24-36 h with appropriate halo fusion protein plasmid (5 µg) on the following day using jetPRIME transfection reagent according to manufacturer’s manual (Polyplus Transfection, France). Cell media was replaced with 5 µM Halo-DBF (compound 7) in HBSS, diluted from 5 mM DMSO stocks. Cells were incubated with the ligand for 15 min at 37ºC, 5% CO2 and washed twice with full media for 25 min. Cells were then incubated with 1 mM propargyl amine (PA) in fresh HBSS media for 5 min at 37ºC, 5% CO2, irradiated for 15 min with 500 nm light at room temperature and incubated for 5 min in dark. RNA fluorescence imaging via CuAAC. The cell culture dishes and glass cover slips were coated with poly-D-lysine (50 µg/ml) for 6 h at 37ºC and washed three times with autoclaved water to remove the excessive amount of poly-D-lysine. HEK293T cells were seeded at equal density (2.5×105 cells/well in 6-well plates) and were transiently transfected with 1 µg appropriate halo fusion protein plasmid on the following day using jetPRIME transfection reagent according to manufacturer’s manual (Polyplus Transfection, France) for 24-36 h. Cells were incubated with the ligand (5 µM in HBSS from 5 mM stock in DMSO) for 15 min at 37ºC, 5% CO2 and replaced with fresh full media for 25 min twice. Cells were then incubated with 1 mM propargyl amine (PA) in fresh HBSS media for 5 min at 37ºC, 5% CO2, irradiated for 15 min with 500 nm light at room temperature and incubated for 5 min in dark. Cells were washed three times with DPBS, fixed and permeabilized for 30 min at room temperature with 3.7% paraformaldehyde and 0.1% Triton-X100. Cells were then washed three times (7 min/each) on orbital shaker with DPBS, blocked with BSA (1 mg/ml in DPBS, 0.45% NaCl and 0.025% NaN3) for 30 min at room temperature, washed twice for 5 min with DPBS, and incubated with 200 µL of click solution (1 mM CuSO4, 2 mM THPTA ligand, 10 mM NaAsc, and 15 µM azide-Cy5) for 1 hour at 37ºC in the dark. Cells were washed three times for 5 min/each on an orbital shaker with DPBS-0.1% Triton-X100 and one with DPBS. Cells were stained with Hoechst 333242 (1:2000,

ACS Paragon Plus Environment

Page 13 of 16

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

262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290

ACS Chemical Biology

Trihydrochloride, Trihydrate - 10 mg/mL solution in water from Thermo Fisher Scientific) for 10 min, washed twice with DPBS for 5 min and mounted using VectaShield (Vector Labs). Slides were imaged via fluorescence confocal microscopy using a 63x oil immersion objective on a Leica 700 Carl Zeiss microscope. RT-qPCR for biotinylated RNA. 2 µg of biotinylated RNA from each sample were subjected to cDNA synthesis with reverse primer for each gene using the PrimeScriptTM Reverse Transcriptase from Takara. 3 µl out of a 20-µl reaction was diluted with 87-µl nucleus free water and set aside as INPUT. To the remaining cDNA reaction, Dynabeads MyOne Streptavidin C1 (Invitrogen, Cat# 65002) that had been blocked with BSA (1 mg/mL) and yeast tRNA (1 mg/mL) at 4 ºC overnight.[5] The beads were washed three times with 4 M wash buffer (100 mM Tris, pH7.0, 4 M NaCl, 10 mM EDTA, 0.2% (vol/vol) Tween-20) and re-suspended in bead binding buffer (100 mM Tris, pH7.0, 1 M NaCl, 10 mM EDTA, 0.2% (vol/vol) Tween-20).[6] Blocked beads were added to enrich biotinylated RNA-cDNA hybrids. The mixture was incubated at room temperature for 30 min on shaker. Using DynaMag side magnet to retain the streptavidin beads, the flow-through was removed. The beads were washed twice with 4 M wash buffer at room temperature, twice with 4 M wash buffer incubated at 50 ºC for 3 min, and twice with PBS (1X, pH7.4). The beads were then treated with RNase H mixture [1X RNAse H buffer, 12.5 mM D-biotin (Life Technologies, Cat# B20656), 0.1 U/µl RNAse H (New England Biolabs, Cat# M0297S), RNAse A/T1 cocktail at 40 ng/µL (Thermal Scientific, Cat# EN0551)] for 30 min at 37ºC on shaker. Each RNAse reaction was added 1 µL of DMSO and incubated at 95-98ºC for 5 min. Using DynaMag side magnet to retain the beads, released cDNA was collected and purified using DNA Clean & Concentration columns from Zymo Research (Cat# D4003) according to the manufacturer instructions. Purified cDNA was eluted with 34µl nucleus free water, further underwent a 1:20 dilution and set aside as ENRICH. INPUT (2 µl) and ENRICH (2 µl) were subjected to RT-qPCR using SYBR® Advantage® qPCR Premix from Takara and Biorad CFX connectTM real time system. The fold of enrichment was calculated against a no-hv negative control: 2^((CtENRICH-nohv –CtINPUT-nohv)-(CtENRICH-CtINPUT)).

ACS Paragon Plus Environment

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 14 of 16

291

References.

292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332 333 334 335 336 337 338

1. Mehler, M. F., and Mattick, J. S. (2007) Noncoding RNAs and RNA editing in brain development, functional diversification, and neurological disease, Physiol Rev 87, 799823. 2. Pek, J. W., and Okamura, K. (2015) Regulatory RNAs discovered in unexpected places, Wiley Interdiscip Rev RNA 6, 671-686. 3. Rinn, J., and Guttman, M. (2014) RNA Function. RNA and dynamic nuclear organization, Science 345, 1240-1241. 4. Geisler, S., and Coller, J. (2013) RNA in unexpected places: long non-coding RNA functions in diverse cellular contexts, Nat Rev Mol Cell Biol 14, 699-712. 5. Lesnik, C., Golani-Armon, A., and Arava, Y. (2015) Localized translation near the mitochondrial outer membrane: An update, RNA Biol 12, 801-809. 6. Crosetto, N., Bienko, M., and van Oudenaarden, A. (2015) Spatially resolved transcriptomics and beyond, Nat Rev Genet 16, 57-66. 7. Shaffer, S. M., Wu, M. T., Levesque, M. J., and Raj, A. (2013) Turbo FISH: a method for rapid single molecule RNA FISH, PLoS One 8, e75120. 8. Riley, K. J., Yario, T. A., and Steitz, J. A. (2012) Association of Argonaute proteins and microRNAs can occur after cell lysis, Rna 18, 1581-1585. 9. Mili, S., and Steitz, J. A. (2004) Evidence for reassociation of RNA-binding proteins after cell lysis: implications for the interpretation of immunoprecipitation analyses, Rna 10, 16921694. 10. Skovsen, E., Snyder, J. W., Lambert, J. D., and Ogilby, P. R. (2005) Lifetime and diffusion of singlet oxygen in a cell, J Phys Chem B 109, 8570-8573. 11. To, T. L., Fadul, M. J., and Shu, X. K. (2014) Singlet oxygen triplet energy transfer-based imaging technology for mapping protein-protein proximity in intact cells, Nature Communications 5. 12. Amatguerri, F., Lopezgonzalez, M. M. C., Martinezutrilla, R., and Sastre, R. (1990) Singlet Oxygen Photogeneration by Ionized and Un-Ionized Derivatives of Rose-Bengal and Eosin Y in Diluted Solutions, Journal of Photochemistry and Photobiology a-Chemistry 53, 199-210. 13. Xu, X. Y., Muller, J. G., and Burrows, C. J. (2007) DNA-protein cross-links between guanine and lysine depend on the mechanism of oxidation, Chemical Research in Toxicology 20, 2002-2002. 14. An, N., Fleming, A. M., White, H. S., and Burrows, C. J. (2015) Nanopore detection of 8oxoguanine in the human telomere repeat sequence, ACS Nano 9, 4296-4307. 15. Hosford, M. E., Muller, J. G., and Burrows, C. J. (2004) Spermine participates in oxidative damage of guanosine and 8-oxoguanosine leading to deoxyribosylurea formation, J Am Chem Soc 126, 9540-9541. 16. Knox, J. P., and Dodge, A. D. (1985) The Photodynamic-Action of Eosin, a Singlet-Oxygen Generator - the Inhibition of Photosynthetic Electron-Transport, Planta 164, 30-34. 17. Bancirova, M. (2011) Sodium azide as a specific quencher of singlet oxygen during chemiluminescent detection by luminol and Cypridina luciferin analogues, Luminescence 26, 685-688. 18. England, C. G., Luo, H. M., and Cai, W. B. (2015) HaloTag Technology: A Versatile Platform for Biomedical Applications, Bioconjugate Chemistry 26, 975-986. 19. Ngo, J. T., Adams, S. R., Deerinck, T. J., Boassa, D., Rodriguez-Rivera, F., Palida, S. F., Bertozzi, C. R., Ellisman, M. H., and Tsien, R. Y. (2016) Click-EM for imaging metabolically tagged nonprotein biomolecules, Nature Chemical Biology 12, 459-U128.

ACS Paragon Plus Environment

Page 15 of 16

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

339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360

ACS Chemical Biology

20. Wassarman, D. A., and Steitz, J. A. (1991) Structural-Analyses of the 7sk Ribonucleoprotein (Rnp), the Most Abundant Human Small Rnp of Unknown Function, Molecular and Cellular Biology 11, 3432-3445. 21. Gerbi, S. A., and Lange, T. S. (2002) All small nuclear RNAs (snRNAs) of the [U4/U6.U5] trisnRNP localize to nucleoli; Identification of the nucleolar localization element of U6 snRNA, Molecular Biology of the Cell 13, 3123-3137. 22. Palacios, I., Hetzer, M., Adam, S. A., and Mattaj, I. W. (1997) Nuclear import of U snRNPs requires importin beta, Embo Journal 16, 6783-6792. 23. Kochan, J., Wawro, M., and Kasza, A. (2015) Simultaneous detection of mRNA and protein in single cells using immunofluorescence-combined single-molecule RNA FISH, Biotechniques 59, 209-221. 24. Henras, A. K., Plisson-Chastang, C., O'Donohue, M. F., Chakraborty, A., and Gleizes, P. E. (2015) An overview of pre-ribosomal RNA processing in eukaryotes, Wiley Interdisciplinary Reviews-Rna 6, 225-242. 25. Moan, J. (1990) On the Diffusion Length of Singlet Oxygen in Cells and Tissues, Journal of Photochemistry and Photobiology B-Biology 6, 343-347. 26. Tyc, K., and Steitz, J. A. (1989) U3, U8 and U13 Comprise a New Class of Mammalian Snrnps Localized in the Cell Nucleolus, Embo Journal 8, 3113-3119. 27. Baran, V., Vesela, J., Rehak, P., Koppel, J., and Flechon, J. E. (1995) Localization of Fibrillarin and Nucleolin in Nucleoli of Mouse Preimplantation Embryos, Molecular Reproduction and Development 40, 305-310.

ACS Paragon Plus Environment

ACS Chemical Biology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15

ACS Paragon Plus Environment

Page 16 of 16