Synthetic artificial "long non-coding RNAs" targeting

4 downloads 0 Views 5MB Size Report
a sponge43. Based on these findings, we previously constructed a microRNA mower that can inhibit a ..... frontier in the study of human diseases. Cancer Lett ...
Accepted Manuscript Synthetic artificial "long non-coding RNAs" targeting oncogenic microRNAs and transcriptional factors inhibit malignant phenotypes of bladder cancer cells Haibiao Xie, Hengji Zhan, Qunjun Gao, Jianfa Li, Qun Zhou, Zhicong Chen, Yuhan Liu, Mengting Ding, Huizhong Xiao, Yuchen Liu, Weiren Huang, Zhiming Cai PII:

S0304-3835(18)30179-4

DOI:

10.1016/j.canlet.2018.02.038

Reference:

CAN 13788

To appear in:

Cancer Letters

Received Date: 20 December 2017 Revised Date:

13 February 2018

Accepted Date: 26 February 2018

Please cite this article as: H. Xie, H. Zhan, Q. Gao, Jianfa Li, Q. Zhou, Z. Chen, Y. Liu, M. Ding, H. Xiao, Y. Liu, W. Huang, Z. Cai, Synthetic artificial "long non-coding RNAs" targeting oncogenic microRNAs and transcriptional factors inhibit malignant phenotypes of bladder cancer cells, Cancer Letters (2018), doi: 10.1016/j.canlet.2018.02.038. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT

Abstract Both oncogenic transcription factors (TFs) and microRNAs (miRNAs) play important roles in human cancers, acting as transcriptional and post-transcriptional regulators, respectively. These phenomena raise questions about the ability of an artificial device to simultaneously regulate miRNAs

RI PT

and TFs. In this study, we aimed to construct artificial long non-coding RNAs, “alncRNAs”, and to investigate their therapeutic effects on bladder cancer cell lines. Based on engineering principles of synthetic biology, we combined tandem arrayed aptamer cDNA sequences for TFs with tandem

arrayed cDNA copies of binding sites for the miRNAs to construct alncRNAs. In order to prove the

SC

utility of this platform, we chose β-catenin and the miR-183-182-96 cluster as the functional targets and used the bladder cancer cell lines 5637 and SW780 as the test models. Dual-luciferase reporter

M AN U

assay, real-time quantitative PCR (qRT-PCR) and related phenotypic experiments were used to test the expression of related genes and the therapeutic effects of our devices. The result of dual-luciferase reporter assay and qRT-PCR showed that alncRNAs could inhibit transcriptional activity of TFs and expression of corresponding microRNAs. Using functional experiments, we observed decreased cell proliferation, increased apoptosis, and motility inhibition in alncRNA-infected bladder cancer cells.

TE D

What’s more, follow-up mechanism experiments further confirmed the anti-tumor effect of our devices. In summary, our synthetic devices indeed function as anti-tumor regulators, which synchronously accomplish transcriptional and post-transcriptional regulation in bladder cancer cells. Most importantly,

EP

anti-cancer effects were induced by the synthetic alncRNAs in the bladder cancer lines. Our devices, all in all, provided a novel strategy and methodology for cancer studies, and might show a great potential

AC C

for cancer therapy if the challenges of in vivo DNA delivery are overcome.

ACCEPTED MANUSCRIPT

Synthetic artificial "long non-coding RNAs" targeting oncogenic microRNAs

2

and transcriptional factors inhibit malignant phenotypes of bladder cancer cells

3

Haibiao Xie1, 2, 3*, Hengji Zhan1,3*, Qunjun Gao1, 3, 4*,Jianfa Li1, 2, 5*,Qun Zhou1,3, Zhicong Chen1,3,

4

Yuhan Liu1,3, Mengting Ding1,3,Huizhong Xiao1,3, Yuchen Liu1,2Ψ, Weiren Huang1Ψ, Zhiming Cai1, 2, 3Ψ

RI PT

1

5 1.

7

Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital,

SC

6

First Affiliated Hospital of Shenzhen University, Shenzhen 518039, China 2.

Shantou University Medical College, Shantou 515041, Guangdong Province, China;

9

3.

Department of Urology, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen

10

University, Shenzhen 518039, China

M AN U

8

11

4.

Guangzhou Medical University, Guangzhou, China,

12

5.

Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute

13

TE D

of Urology, Peking University Shenzhen Hospital, Peking University, Shenzhen 518036, China

14 15

Ψ

16

Zhiming Cai, Weiren Huang &Yuchen Liu

17

Emails: [email protected] (ZC) or [email protected](WH) & [email protected]

18

(YL)

19 20 21 22 23 24 25 26 27 28 29

*

AC C

EP

Authors for correspondence:

The three authors contributed this work equally.

ACCEPTED MANUSCRIPT 30 31

Abstract Both oncogenic transcription factors (TFs) and microRNAs (miRNAs) play important roles in human cancers, acting as transcriptional and post-transcriptional regulators, respectively. These

33

phenomena raise questions about the ability of an artificial device to simultaneously regulate miRNAs

34

and TFs. In this study, we aimed to construct artificial long non-coding RNAs, “alncRNAs”, and to

35

investigate their therapeutic effects on bladder cancer cell lines. Based on engineering principles of

36

synthetic biology, we combined tandem arrayed aptamer cDNA sequences for TFs with tandem

37

arrayed cDNA copies of binding sites for the miRNAs to construct alncRNAs. In order to prove the

38

utility of this platform, we chose β-catenin and the miR-183-182-96 cluster as the functional targets

39

and used the bladder cancer cell lines 5637 and SW780 as the test models. Dual-luciferase reporter

40

assay, real-time quantitative PCR (qRT-PCR) and related phenotypic experiments were used to test the

41

expression of related genes and the therapeutic effects of our devices. The result of dual-luciferase

42

reporter assay and qRT-PCR showed that alncRNAs could inhibit transcriptional activity of TFs and

43

expression of corresponding microRNAs. Using functional experiments, we observed decreased cell

44

proliferation, increased apoptosis, and motility inhibition in alncRNA-infected bladder cancer cells.

45

What’s more, follow-up mechanism experiments further confirmed the anti-tumor effect of our devices.

46

In summary, our synthetic devices indeed function as anti-tumor regulators, which synchronously

47

accomplish transcriptional and post-transcriptional regulation in bladder cancer cells. Most importantly,

48

anti-cancer effects were induced by the synthetic alncRNAs in the bladder cancer lines. Our devices, all

49

in all, provided a novel strategy and methodology for cancer studies, and might show a great potential

50

for cancer therapy if the challenges of in vivo DNA delivery are overcome.

51

Key words

52

Bladder cancer, synthetic biology, aptamer, miRNA, TFs.

AC C

EP

TE D

M AN U

SC

RI PT

32

53 54 55 56

Introduction Bladder cancer (BCa) is one of the most common urologic malignancies worldwide, and its high prevalence and recurrence rates have captured great attention in the past decades1-3. Although there

ACCEPTED MANUSCRIPT have been great advances in the therapy of BCa, such as chemotherapy, radiotherapy and surgery, the

58

five-year survival rate still remains unsatisfactory4-6. Although several novel antineoplastic drugs have

59

been developed and subjected to clinical trials to assess their use as cancer therapy, no major

60

breakthroughs have been achieved with these strategies7. Thus, there is an increasing need for the

61

development of new and efficient strategies to treat BCa.

62

RI PT

57

With the completion of the ENCODE project, more than three-quarters of the genome has been found to be transcribed into RNA, but only 1–2% of this is translated into proteins8. Such RNAs

64

without protein-coding function are classified as non-coding RNAs (ncRNAs). Among them,

65

microRNAs (miRNAs), a class of small non-coding RNAs, regulate gene expression by binding to

66

partially complementary sequences in the 3'UTR of mRNAs, participate in cellular differentiation and

67

homeostasis, and are consequently involved in the development of various diseases, including cancers9.

68

In our previous work, we used deep sequencing of genome-wide miRNA profiles in human BCa and

69

demonstrated that the miR-183-96-182 cluster and miR-17-92 are strongly up-regulated in BCa10. Thus,

70

miRNAs could be a potential target for BCa therapy11. To enhance the effect of inhibiting tumor

71

progression, concurrently knocking down or knocking out various miRNAs are common strategies.

72

However, accomplishing multiple miRNA knockouts simultaneously is difficult. Therefore, a device

73

for inhibiting these functional classes of paralogous miRNAs is needed.

TE D

M AN U

SC

63

Aptamers are small single-stranded oligonucleotides that can fold into unique tertiary structures and

75

bind to target molecules such as proteins and RNAs with high affinity and specificity. Therefore, they

76

are considered “chemical antibodies”12. Using a technique known as systematic evolution of ligands by

77

exponential enrichment (SELEX), aptamers binding to various proteins have been routinely selected

78

from synthetic combinatorial libraries13. Compared with monoclonal antibodies, aptamers can be

79

synthesized quickly and are structurally stable under a wide range of temperatures and storage

80

conditions. Most significantly, they exhibit low toxicity and non-immunogenicity in contrast to

81

conventional antibodies12. To date, various studies have demonstrated that RNA aptamers can inhibit

82

tumorigenesis by targeting oncogenic transcription factors (TFs) such as β-catenin14 and NF-κB15.

AC C

EP

74

83

Synthetic biology is a discipline that enables multiple signals to be cascaded and even to alter the

84

signal flow by designing and linking various elements16. It is flexible, interesting and programmable so

ACCEPTED MANUSCRIPT that scientists can use a variety of elements to verify their own ideas such as a logic gate17, signal

86

conductor18 or signal-connectors19. In this study, we constructed “artificial long non-coding RNAs”

87

(alncRNAs) based on the principles in the field of synthetic biology, by combining an RNA aptamer

88

for TFs with miRNA sponges targeting oncogenic miRNAs, and tested the utility of this device in BCa

89

cells. The results showed that alncRNA could inhibit the transcription of TF target genes and the

90

expression of targeted miRNAs, as expected. Importantly, our devices partly inhibited the malignant

91

phenotypes of BCa cells. These results showed that our tools could simultaneously accomplish both

92

transcriptional and posttranscriptional regulation of genes of interest, thus providing a novel strategy

93

for treating human BCa cells.

SC

RI PT

85

95

Materials and Methods

96

Cell lines and cell culture

97

M AN U

94

Human bladder transitional cell carcinoma cell lines (SW780, 5637) and normal human cell lines (SV-40-immortalized human uroepithelial cell line, SV-HUC1) were purchased from the American

99

Type Culture Collection (ATCC, Manassas, VA, USA). The 5637 cells were grown in RPMI-1640

TE D

98

Medium (Gibco, Thermo Fisher Scientific, Waltham, MA, USA) supplemented with 10% fetal bovine

101

serum (Gibco). The SW780 cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM;

102

Gibco), supplemented with 10% fetal bovine serum (Gibco). The SV-HUC-1 cells were cultured in

103

F12K (Gibco) plus 1% antibiotics (100 U/mL penicillin and 100 µg/mL streptomycin sulfate), with 10%

104

fetal bovine serum. All cells were cultured at 37°C, in a humidified atmosphere of 5% CO2.

105

Creation of the artificial lncRNAs and lentivirus infection

AC C

106

EP

100

Bulged miRNA binding sites for the miR-183, miR-182, miR96 and miR-17-5p and linkers

107

between them were designed and chemically synthesized. In addition, RNA-aptamers specifically

108

modulating β-catenin and NF-κB were applied from previous studies14,15. The combination DNA parts

109

for the miR-183-182-96 cluster-β-catenin and miR-17-5p-NF-κB were driven by CMV or U6. These

110

sequences and untargeted-control binding sites (negative control; NC) were cloned into a lentiviral

111

vector (PLV-EGFP-N). Detailed descriptions of the related sequences are presented in Table 1, and

112

detailed maps of alncRNA vectors are shown in supplementary Figure 1A.

ACCEPTED MANUSCRIPT 113

To produce lentivirus, lentivirus expression vectors (PLV-EGFP-N) were transfected into 293T cells. Supernatants were collected 48 and 72 hours after transfection, and cell debris was removed by

115

centrifugation at 1500 × g for 30 minutes then passing through a syringe filter. Titers of the

116

concentrated viruses were determined by infecting 293T cells with seral dilutions. The titers of the

117

virus stocks were calculated to be 5 × 107–1 × 108 transduction units (TU)/mL based on the formula:

118

(%GFP + cells) × (1/dilution). To effectively construct a stable cell line, three parts of the viral

119

supernatant were mixed with one part fresh medium and 10 µg/mL of polybrene, and the mixture was

120

applied to freshly-seeded cells. MOI (multiplicities of infection) of 10 pfu/cell were used, as shown in

121

supplementary Figure 2. Stable cell lines were selected by flow cytometric assay by observing GFP

122

expression.

123

Enzyme-linked immunosorbent assay (ELISA)

SC

M AN U

124

RI PT

114

The concentration of β-catenin and NF-κB was measured by ELISA, which was carried out according to the manufacturer’s instructions. Briefly, 106 sample cells (SW780, 5637 and SW780) were

126

harvested and resuspended in 200 µL of lysis buffer. The supernatants of lysates were collected by

127

centrifugation and used for the subsequent procedures. The OD values were then measured using an

128

automatic microplate reader (Bio-Rad, Hercules, CA, USA) and converted into protein concentrations

129

using standard calibration curves.

130

Dual-luciferase reporter assay

To measure the transcriptional activity of β-catenin and NF-κB, a dual-luciferase reporter assay was

EP

131

TE D

125

performed using β-catenin or NF-κB reporter constructs (Supplementary Figure 1B). The β-catenin

133

reporter contained copies of Tcf-binding elements (responsive elements), minimal promoter and

134

dual-reporter vector (Beijing Syngenetech CO., Ltd, China). In addition, the NF-κB reporter contained

135

multiple binding sites for NF-κB, minimal promoter and dual-reporter vector (Beijing Syngenetech Co.,

136

Ltd, China). cDNAs of the binding sites of β-catenin and NF-κB are shown in Table 1. Cells were

137

seeded into six-well plates (5 × 105 per well) and transfected with the β-catenin or NF-κB

138

dual-luciferase reporter vectors. Luciferase activity was measured using the dual luciferase assay

139

system (Promega, Madison, WI, USA) as per the manufacturer’s instructions at 48 hours after

140

transfection. Firefly luciferase activities were normalized to Renilla luciferase activities. All assays

AC C

132

ACCEPTED MANUSCRIPT 141

were performed in duplicate, and all data shown are representative of at least two independent

142

experiments.

143

RNA extraction and real-time quantitative PCR

144

Total RNAs were isolated from SW780, 5637 and SV-HUC1 cells using TRIzol reagent (Invitrogen, Carlsbad, CA, USA) according to the manufacturer’s protocol. The concentration and purity of total

146

RNA were measured using UV spectrophotometric analysis at 260 nm. cDNAs of coding RNA and

147

microRNA were synthesized using a Revertra Ace qPCR RT Kit (Toyobo, Osaka, Japan) and a

148

miScript II RT Kit (Qiagen, Hilden, Germany). Real time PCR was carried out using real time PCR

149

Master Mix (Toyobo) and the miScript® SYBR® Green PCR Kit (Qiagen). GAPDH or U6 small

150

nuclear RNA (snRNA) was selected as the endogenous control. The miRNA qRT-PCR primers were

151

ordered from GeneCopoeia, Inc. (Rockville, MD, USA). The qPCR primers are listed in Table 2. The

152

PCR mixtures were prepared according to the manufacturer’s protocols and amplification was

153

performed under PCR conditions of 40 cycles of 15 sec at 95°C, 20 sec at 55°C, and 30 sec at 70°C on

154

a ABI PRISM 7300 Fluorescent Quantitative PCR System (Applied Biosystems, Foster City, CA,

155

USA). Expression fold changes were calculated using the 2-△△ct method.

156

Cell proliferation assay

SC

M AN U

TE D

157

RI PT

145

The CCK-8 assay (TransGen, Beijing, China) was used to examine cell proliferation. Aliquots of 5 × 103 cells were seeded into 96-well plates and cultured at 37°C. After 24, 48, or 72 h, 10 µL CCK-8

159

was added to each well and incubated at 37°C for 1 h. The absorbance at 450 nm was read by an

160

automatic microplate reader (Bio-Rad). To directly observe the proliferating cells, Edu incorporation

161

assay was performed using an Edu assay kit (Ribobio, Guangzhou, China) according to the

162

manufacturer’s instructions. Each experiment was repeated at least three times.

163

Apoptosis assay and cell cycle analysis

AC C

164

EP

158

Apoptosis was determined by caspase 3 and cleaved-PARP ELISA assay. Aliquots of 1 × 106 cells

165

were counted to measure the activity of caspase 3 and PARP ELISA using caspase 3 and PARP ELISA

166

assay kits respectively (Biovision, Milpitas, CA, USA and American). A microplate reader was used to

167

measure the OD values at 405 nm. Each experiment was repeated at least three times.

ACCEPTED MANUSCRIPT For the cell cycle assay, DATS (Diallyl trisulfide)-treated cells were harvested, washed twice in

169

phosphate-buffered saline (PBS) and fixed in 75% cold alcohol overnight at 4°C. After washing in cold

170

PBS three times, cells were incubated with 1 × PI/RNase staining buffer for 15 min in the dark at room

171

temperature. Samples were then analyzed for their DNA content using flow cytometry. All assays were

172

performed in duplicate, and all data shown are representative of at least two independent experiments.

RI PT

168

173 174 175

Cell motility assay

Cell motility was determined by wound-healing assay. A wound field was created using a sterile 200 µL pipette tip when cells reached approximately 90% confluence. The migration of cells was

177

monitored with a digital camera system. The cell migration distance (µm) was calculated using the

178

software program HMIAS-2000, at 24 hours after the wound was created. These experiments were

179

repeated at least three times.

M AN U

SC

176

Additionally, the cell motility assay was also performed using a transwell insert (8 µm, Dow Corning

181

Corp., Midland, MI, USA). After 24 h, 5 × 10 4 SW780 cells and 15 × 105 5637 cells were first starved

182

in 200 uL serum-free medium and then placed in the uncoated dishes. Concurrently, the lower

183

chambers were filled with 500 µL medium containing 10% fetal bovine serum. Cells were cultured at

184

37°C in a 5% CO2 atmosphere for 24 hours. Cells under the surface of the lower chamber were washed

185

with 1 × PBS, fixed with 4% paraformaldehyde for 20 min, stained with 0.1% crystal violet for 25 min,

186

and then washed three times. Invaded cells were observed under the inverted microscope and imaged.

187

Afterwards, each chamber with the invaded cells was soaked in 1 mL 33% acetic acid for 10 min to

188

wash out the crystal violet. Then 100 µL of 33% acetic acid was added into each well of a 96-well plate,

189

and the absorbance was measured at a wavelength of 570 nm using a microplate reader (Bio-Rad).

190

Experiments were performed in triplicate.

191

Statistical analysis

AC C

EP

TE D

180

192

Each experiment was performed in triplicate. All quantitative data are expressed as mean ± standard

193

deviation (SD). Statistical analyses were performed using SPSS8.0 software (IBM Corp., Armonk, NY,

194

USA). Statistical significance was tested by Student’s t-test, Chi square test or ANOVA. P < 0.05 was

195

considered to be statistically significant.

ACCEPTED MANUSCRIPT 196 197

Results

198

Design and construction of the alncRNAs.

199

Inspired by two observations that some RNA molecules could regulate miRNAs by complementary base pairing in eukaryotic cells20, and that some RNA aptamers are capable of capturing proteins and

201

consequently inhibiting their function by protein–RNA interactions15,21,22, we set out to construct an

202

RNA tool, “alncRNA”, which could inhibit the malignant phenotype of BCa by regulating oncogenic

203

signal pathways at transcriptional and post-transcriptional levels simultaneously. This proposed tool

204

contains two modules, encoded on plasmids (Figure 1A). The first module, the RNA aptamer, can

205

capture proteins including transcription factors (TFs). The second module, the microRNA sponge,

206

causes miRNA loss-of-function by partially-complementary binding to target miRNAs. We connected

207

these modules by a linker of a few nucleotides to allow correct folding of aptamers and sponges23. To

208

ensure effective expression of alncRNAs, we used a powerful promoter (CMV) or U6 promoter, which

209

expresses nuclear localized RNAs24, to drive expression of alncRNAs.

M AN U

SC

RI PT

200

To test the utility of the designed device, we combined two tandem arrayed cDNA sequences of

211

aptamers for β-catenin with six tandem arrayed cDNA copies of binding sites for the miR-183-96-182

212

cluster (two copies for each miRNA of the cluster) to construct alncRNA1. As previously reported,

213

β-catenin is a well-known multifunctional protein, which always activates the transcription of several

214

oncogenic TFs25. To demonstrate the modularity of the circuit design, we also used another two cDNA

215

copies of the RNA aptamer for NF-κB, and six cDNA copies of binding sites for miR-17-5p to replace

216

the corresponding locations in alncRNA1, producing alncRNA2. Afterwards, we inserted these

217

alncRNAs and corresponding control cDNA, a repeated binding element not targeting any protein or

218

microRNA, into lentivirus vectors and used lentivirus to deliver the devices into the target cells for use

219

in subsequent experiments. As shown in Supplementary Figure 2, we used a MOI of 10 pfu/cell to

220

construct the stable cell lines. The mechanism diagram is shown in Figure 1B.

AC C

EP

TE D

210

221 222

Repression of dual-luciferase reporters by alncRNA in BCa cell lines

ACCEPTED MANUSCRIPT To validate the performance of our alncRNAs, we constructed a reporter that can sense β-catenin or

224

NF-κB transcriptional activity by specifically subcloning the β-catenin and NF-κB responsive promoter

225

sequences into a dual-luciferase vector, named β-catenin and NF-κB reporter (Figure 2A). In order to

226

verify reporter detection efficiency, we first measured the protein expression of β-catenin and NF-κB in

227

SW780, 5637 and SV-HUC1 cells by ELISA. The results showed that both β-catenin and NF-κB were

228

overexpressed in BCa cell lines (SW780 and 5637) compared with the normal human uroepithelial cell

229

line SV-HUC1 (Figure 2B). Next, we respectively transfected our β-catenin or NF-κB reporter into

230

BCa cell lines and SV-HUC1, and measured activity by luciferase assay. As expected, the ratio of

231

firefly luciferase versus Renilla luciferase expression was consistent with the ELISA results, indicating

232

that our reporter did indeed sense the transcriptional activity of β-catenin and NF- κB (Figure 2C).

SC

Next, in an effort to verify the effect of alncRNA on TF transcription, we introduced the β-catenin

M AN U

233

RI PT

223

and NF-κB reporter into BCa cell lines stably expressing alncRNA or NC, respectively (SW780, 5637).

235

After 48 hours, the results of the dual-fluorescein assay confirmed that our alncRNA did inhibit

236

transcription of β-catenin and NF-κB reporter, and that the CMV and the U6 promoter-driven effects

237

were similar (Figure 2D). In addition, we found that alncRNA did not conduct crosstalk between the

238

β-catenin and NF-κB reporter pathways. These results confirmed that alncRNA could directly inhibit

239

transcriptional activity of both β-catenin and NF-κB without crosstalk. Since we previously verified the

240

effect of microRNA sponges by the luciferase assay, we did not repeat it here26.

241

244

The alncRNA achieved effective suppression of oncogenic signals and target miRNA levels in BCa cell lines.

AC C

242 243

EP

TE D

234

To further investigate the functionality of alncRNAs, we examined the relative expression levels of

245

the target miRNAs and the downstream genes of target TFs in the human BCa cell lines SW780 and

246

5637 treated with our device. The qRT-PCR results demonstrated that expression of the corresponding

247

alncRNAs induced a remarkable decrease in expression levels of the corresponding downstream genes

248

and the targeted miRNAs in the two BCa cell lines (Figure 3). The expression levels of the downstream

249

genes of NF-κB (Bcl-XL, TRAF1) and miR-17-5p could not be suppressed by alncRNA1. At the same

250

time, the expression level of the miR-183-182-96 cluster also could not be changed by alncRNA2.

251

Interestingly, we found that the expression levels of two well-known downstream genes (c-myc, cyclin

ACCEPTED MANUSCRIPT 252

D1) of β-catenin could be decreased by alncRNA2 (Figure 3A, B, C and D). After searching the

253

literature, we found that c-myc and cyclin D1 can also be transcriptionally activated by NF-κB27.

254

256

Inhibition of cell proliferation induced by alncRNAs in BCa cell lines Stable BCa cell lines (SW780 and 5637) expressing alncRNAs or corresponding NC vector were

RI PT

255

seeded into 96-well plates to test the cancer cell proliferation ability. Using the CCK-8 assay, we

258

demonstrated that the alncRNAs could effectively reduce cell growth compared with the NC group in

259

both BCa cell lines (Figure 4A, B, C, and D). The Edu incorporation assay was then used to determine

260

the effects of alncRNAs on proliferation of BCa cell lines (Figure 4E, F and Supplementary Figure3).

261

The results showed that alncRNAs could markedly inhibit cancer cell growth.

SC

257

Since alncRNA could induce the down-regulation of myc and cyclin D1, we hypothesized that

263

alncRNA acted to inhibit tumor proliferation by affecting the cell cycle. To verify our hypothesis, we

264

performed cell cycle analysis of SW780 cells stably expressing alncRNA and NC. The results showed

265

that BCa cells expressing alncRNAs in G0 / G1 phase increased significantly, while those in G2 phase

266

decreased significantly (Figure 5A). The result was consistent with the down-regulation of myc and

267

cyclinD1. These data suggest that alncRNA may cause cell cycle arrest by affecting the transcriptional

268

activity of TFs, thus inhibiting the expression of cell cycle regulators.

271

TE D

270

Induction of apoptosis by alncRNAs in BCa cell lines.

EP

269

M AN U

262

According to cell cycle experiments, we found that alncRNA can increase the sub-G1 population, indicating the induction of apoptosis (Figure 5A). To verify and explore the mechanism by which

273

alncRNAs induce apoptosis, BCa cell lines (SW780, 5637) stably expressing alncRNAs or the

274

corresponding NC vectors were seeded into 6-well plates to investigate the extent of apoptosis and the

275

underlying mechanisms. We used ELISA to analyze the expression of caspase-3 protein and

276

cleaved-PARP-apoptotic proteins. We found an obvious increase in the expression of caspase-3 and

277

cleaved-PARP in the cells expressing alncRNA2, but only the expression of caspase3 was increased in

278

the cells expressing alncRNA1 (Figure 5B and Supplementary Figure 4). Previous studies suggested

279

that NF-κB exerts anti-apoptotic effects by inhibiting caspase3 and cleaved-PARP, whereas β-catenin

280

acts only to inhibit caspase3, rarely affecting the expression of PARP in BCa28,29. These results indicate

AC C

272

ACCEPTED MANUSCRIPT 281

that alncRNA can induce apoptosis, with alncRNA2 eliciting a higher rate of apoptosis than alncRNA1

282

(Figure 5A), which is consistent with the expression of apoptosis-related proteins.

283

285

Inhibition of cell migration induced by alncRNAs in BCa cell lines. Finally, we further investigated whether alncRNAs could inhibit cell migration in BCa. Stable BCa

RI PT

284

cell lines (SW780 and 5637) expressing alncRNAs and corresponding NC vector were seeded into

287

24-well plates to test their cell migration ability using the wound healing assay. Cell migration arrest

288

was observed in SW780 (Figure 6A) and 5637 cells (Supplementary Figure 5A) as expected. Similarly,

289

the transwell assay was used to further confirm our results. Decreased cell motility was observed in

290

stable cell lines expressing alncRNAs compared with NC cell groups (Figure 6B, Supplementary

291

Figure 5B). These results confirmed that alncRNAs suppress cell migration in BCa.

M AN U

SC

286

To elucidate the mechanism of inhibitory effects of alncRNAs on cell migration, we conducted a

293

literature search, and found that β-catenin and NF-κB, as well as the targeted microRNAs, could induce

294

epithelial-mesenchymal transition (EMT) and sequentially induce cancer cell migration and

295

invasion30-33. We hypothesized that our alncRNAs might reverse EMT and resulted in decreased cell

296

migration. To confirm our hypothesis, we used qRT-PCR to analyze the expression of several common

297

epithelial markers (E-cadherin) and mesenchymal markers (vimentin, slug) in SW780 cells stably

298

expressing alncRNAs and NC. Compared with control group, the expression levels of slug and

299

vimentin in SW780 cells stably expressing alncRNAs were significantly decreased, and the expression

300

of E-cadherin was significantly increased(Figure 4D). Additionally, we found that alncRNA-infected

301

cells underwent a phenotypic change from a spindle-shaped morphology to a cuboidal, epithelial-like

302

morphology (Figure 4C). These data suggested that our alncRNAs were likely to suppress migration by

303

reversing the EMT process.

305 306

EP

AC C

304

TE D

292

Discussion Medical synthetic biology is an interdisciplinary branch of medical biology and engineering34. One

307

of the major purposes of medical synthetic biology research is to connect synthetic genetic devices

308

together to control complex biological phenotypes16. In our present study, we connected the cDNA of

ACCEPTED MANUSCRIPT RNA aptamers and miRNA sponges to construct an artificial anti-tumor device, named alncRNA. The

310

reason for this name is that this device can accomplish simultaneous transcriptional and

311

post-transcriptional regulation, functioning just like a natural lncRNA35. To confirm the utility of our

312

device, we chose oncogenic transcription factors (TFs) and miRNAs as our targets. Based on the results

313

of previous studies36-40, we chose two tandem copies of aptamers and six binding sites of microRNAs

314

to construct our device. The results of qRT-PCR showed that the BCa cell lines (5637, SW780) stably

315

expressing alncRNA indeed decreased expression of the TF target genes and the corresponding

316

miRNAs. Lastly, phenotypic experiments revealed that alncRNA could inhibit cell proliferation,

317

suppress cell motility and induce apoptosis at the same time. These results demonstrate that our device

318

can effectively inhibit oncogenic signaling transduction to suppress the malignant phenotypes of BCa

319

cells.

SC

M AN U

320

RI PT

309

It is widely acknowledged that miRNAs degrade mRNA and non-coding RNA by complementary base pairing with 3’UTR of these RNAs, achieving post-transcriptional regulation41. Interestingly,

322

recent studies have revealed that mRNA–miRNAs interaction can also block the function of

323

miRNAs20,42. For instance, the pseudogene PTENP1 was found to regulate cellular levels of PTEN by

324

interacting with miRNAs targeting PTEN. A similar result was shown in the reciprocal relationship

325

between MALAT1 and miR-125b. MALAT1 is an lncRNA which inhibits miR-125b by functioning as

326

a sponge43. Based on these findings, we previously constructed a microRNA mower that can inhibit a

327

miRNA of interest by integrating the specific recognition sites into a mower RNA sequence26. The

328

mower indeed performed as expected. However, a single protein can be involved in more than one step

329

of gene expression, such as transcription, post-transcriptional processing, and translation. Our previous

330

device focused only on transcription and did not show an absolute effect. Therefore, we aimed to

331

construct a more effective device which could achieve both transcriptional and post-transcriptional

332

regulation.

333

AC C

EP

TE D

321

An interesting tool–an RNA aptamer specifically targeting proteins–came to our attention. Its high

334

specificity and high-level expression in cells make it possible to specifically recognize proteins and

335

even modulate their activity. Previous studies revealed that DNA or RNA molecules can compete with

336

genes for TFs, thus affecting the transcription of these genes44,45. For example, nucleophosmin (NPM)

ACCEPTED MANUSCRIPT is a multi-functional protein that plays a pivotal role in cell proliferation and apoptosis46. Jian et al.

338

took advantage of an RNA aptamer to target the central acidic region of NPM and inhibit its

339

oligomerization resulting in disruption of the downstream signaling pathway47. Based on these

340

observations, we, first constructed alncRNA by connecting the sequences of miRNA sponges and RNA

341

aptamers to accomplish simultaneous transcriptional and post-transcriptional regulation. For maximum

342

expression of alncRNA, we used the strong promoters CMV or U6 to drive our devices and inserted

343

them into cells using a lentivirus to achieve stable and highly-efficient expression. Based on these

344

designs, we hypothesized that our devices would target oncogenic signals and subsequently inhibit

345

malignant phenotypes of BCa cells.

SC

RI PT

337

To test our hypothesis, we first chose the TFs β-catenin and the miR-183-182-96 cluster as targets.

347

β-catenin is a multifunctional TF involved in cell adhesion and the Wnt pathway. It is also well known

348

to activate transcription of various oncogenes, such as c-myc, cyclinD1 and Cox-225,48. Another factor,

349

the miR-183-182-96 cluster, has been considered to play an important role in tumor apoptosis and the

350

AKT–mTOR signaling pathway 49. The results of qRT-PCR confirmed that our tool also successfully

351

inhibited the expression of the miRNA of interest and the function of β-catenin. The next step was to

352

conduct a functional experiment to demonstrate that our design strongly suppressed bladder urothelial

353

carcinoma cell proliferation, increased the apoptosis rate and decreased cell motility by modulating the

354

corresponding genes.

TE D

Another feature of our device is its modularity, with tandem binding elements for protein and

EP

355

M AN U

346

miRNA that could in theory be replaced by other similar elements. Here, we replaced the binding sites

357

of β-catenin and the mir-183-182-96 cluster with sequences of NF-κB and mir-17-5p to test the

358

modularity. NF-κB is also a cancer-associated TF involved in transcription of multiple oncogenes, such

359

as Bcl-xl, vascular endothelial growth factor (VEGF) and HIF-1α50. MiR-17-5p has been shown to

360

regulate basic cellular processes such as proliferation and apoptosis. In a wide variety of cancers,

361

miR-17-5p is highly expressed and always associates with cancer development and aggressiveness51.

362

As we expected, alncRNA2 also played an anti-tumor role in BCa.

AC C

356

363

The highlight of our work is the simultaneous regulation of transcription and post-transcriptional

364

regulation by designing an RNA-based device. Compared with traditional single elements, our device

ACCEPTED MANUSCRIPT exerts a more significant effect in the regulation of signal flow. Due to our tandem and programmable

366

design, we can change the TFs we want to target as well as the miRNAs. However, there remain a few

367

limitations that need to be addressed. Firstly, as an anti-tumor devices, alncRNAs really need to be

368

effectively controlled to avoid damage to normal cells. There are a variety of ways previously shown to

369

control expression, such as riboswitches52, logic gates 17 and light-induced switches53. However the

370

availability of these tools for alncRNA will need verification. Secondly, one notable limitation to the

371

application of this methodology is that functional aptamers still need to be screened from randomized

372

libraries, which currently limits the widespread application of alncRNAs. Finally, before any such

373

anti-tumor device can be used in the clinic, it will need to be tested in vivo. Our group used lentivirus to

374

transport alncRNA into cancer cells. Thus, in vivo experiments would be needed to address the key

375

question of identifying the best concentration (MOI) of alncRNA that will inhibit tumor cells but not

376

normal cells. Even so, we still believe that our novel device will be widely applied in biology and

377

medicine in future. Here, we show a future scheme for application of alncRNA (Figure 7).

SC

M AN U

378

RI PT

365

In summary, our team constructed anti-tumor devices, alncRNAs, which had the ability to capture oncogenic TFs and miRNAs of interest and to effectively inhibit malignant phenotypes of BCa cells.

380

Although there is a long way to go before our tool can be used for clinical diagnosis and therapy,

381

alncRNAs still provide a novel strategy for cancer therapy and could be useful “weapons” against

382

cancer cells when the challenges of in vivo DNA delivery are overcome.

EP

383

TE D

379

Author contributions

385

H.X., Q.G, H.Z., J.L. and Y.L. performed experiments and data analysis. Y.L. and H.X. designed the

386

project and wrote the paper. Z.C., W.H. and Y.L. supervised the project. W.H. and Z.C. provided

387

financial support for the project.

AC C

384

388 389

Acknowledgments

390 391 392 393

We are indebted to the donors whose names were not included in the author list, but who participated in this program. This work was supported by the National Key Basic Research Program of China (973 Program) (2014CB745201), the Chinese High-Tech (863) Program (2014AA020607), National Natural Science Foundation of China (81773257), International S&T Cooperation program of China (ISTCP)

ACCEPTED MANUSCRIPT 394 395 396 397 398

(2014DFA31050),

The

National

Science

Foundation

Projects

of

Guangdong

Province

(2014A030313717), the Shenzhen Municipal Government of China (ZDSYS201504301722174, JCYJ20150330102720130, GJHZ20150316154912494), and Special Support Funds of Shenzhen for Introduced High-Level Medical Team. What’s more, we thank International Science Editing (http://www.internationalscienceediting.com) for editing this manuscript.

399

RI PT

400

AC C

EP

TE D

M AN U

SC

401

ACCEPTED MANUSCRIPT 402 403

Figure legends Figure 1. Design and construction of the alncRNAs in cancer cells. (A) General compositional

404

framework and design strategy for constructing alncRNAs. (B) Thorough description of the engineered

405

alncRNAs for inducing cancer cell inhibition.

407

RI PT

406

Figure 2. Repression of dual-luciferase reporters by alncRNA in bladder cancer (BCa) cell lines. (A) Description of the working mechanism of TF reporters. (B) The expression of β-catenin and NF-κB

409

protein were detected by ELISA in SW780, 5637 and SV-HUC1 cells. (C) Luciferase expression levels

410

of several cell lines transfected with β-catenin or NF-κB reporters. (D) Luciferase expression levels of

411

BCa cell lines (SW780 and 5637) expressing alncRNA and NC transfected with β-catenin or NF-κB

412

reporters. Results are shown as mean ± SD. ***P < 0.001 compared with control. ** P < 0.01

413

compared with control. * P < 0.05 compared with control.

M AN U

SC

408

414

Figure 3. The artificial lncRNAs simultaneously reduced the expression levels of mRNA of various

416

downstream genes of the targeted TFs and corresponding miRNAs. alncRNA1 reduced the expression

417

of myc and cyclinD1 mRNAs (A, B, C, and D) and miR-183-182-96 (E, F, G, H) in BCa cell lines

418

(SW780, 5637) whether it was driven by CMV or U6 promoters. alncRNA2 reduced the expression of

419

myc, cyclinD1, Bcl-XL, and TRAF1 mRNAs (A, B, C, D) and miR-17-5p (E, F, G, H) in BCa cell

420

lines (SW780, 5637) whether it was driven by CMV or U6 promoters. Results are shown as mean ± SD.

421

***P < 0.001 compared with control. ** P < 0.01 compared with control. * P < 0.05 compared with

422

control.

424

EP

AC C

423

TE D

415

Figure 4. The effects of artificial lncRNAs on cell proliferation in BCa cells. The growth curves of

425

SW780 or 5637 cells infected with alncRNA driven by CMV (A, B) and U6 (C, D) promoters were

426

determined using CCK-8 assay. Data are shown as mean ± SD. The proliferation of 5637 (E) and

427

SW780 (F) cells infected with alncRNA were also determined using the EDU incorporation assay.

428 429

Figure 5. The effects of artificial lncRNAs on apoptosis in BCa cells. (A) AlncRNA induce

ACCEPTED MANUSCRIPT 430

apoptosis in SW780 cell. (B) The relative activity of caspase 3 was calculated in SW780 and 5637 cells

431

infected with alncRNA driven by CMV, and U6 promoters using ELISA.

432 Figure 6. The effects of artificial lncRNAs on migration of BCa cells. (A) The relative rate of cell

434

migration was calculated in SW780 cells infected with alncRNAs using the wound-healing assay. (B)

435

Cell migration was determined by the transwell assay. Inhibition of cell migration following infection

436

with alncRNAs was observed in SW780 cells. Data are shown as mean ± SD. (C) Cell morphology of

437

SW780 cells expressing alncRNAs and NC. (D) The expression of E-cadherin, vimentin and slug in

438

SW780 expressing alncRNA and NC.

SC

RI PT

433

439

Figure 7. The future optimization and application of alncRNA.

M AN U

440 441 442 443

Supplementary Figure 1. The detailed profile of the vectors. A. Map of alncRNA lentivirus vectors. B. Map of NF-κB and β-catenin responsive-reporter vector.

TE D

444

Supplementary Figure 2. Cell viability and transduction efficiency of SW780 cells after infection

446

with various MOI of lentivirus. A. Cell viability of SW780 cells infected with alncRNA and NC driven

447

by CMV was determined by CCK8 assay. B. GFP positive SW780 cells infected with alncRNA and

448

NC driven by CMV were evaluated by manual counting after 72 hours.

449

Supplementary Figure 3. The effects of artificial lncRNAs on proliferation of BCa cells. The

453

AC C

450

EP

445

454

Supplementary Figure 4. The effects of artificial lncRNAs on apoptosis in BCa cells. The relative

451

proliferation of 5637 (A) and SW780 (B) cells infected with alncRNA were also determined using the

452

EDU incorporation assay.

455

activity of cleaved-PARP was calculated in SW780 cells infected with alncRNA driven by CMV (A)

456

and U6 (B) promoters using ELISA. Results are shown as mean ± SD. ***P < 0.001 compared with

457

control. ** P < 0.01 compared with control. * P < 0.05 compared with control.

ACCEPTED MANUSCRIPT 458 459

Supplementary Figure 5. The effects of artificial lncRNAs on cell migration in BCa cells. (A) The relative rate of cell migration was calculated in 5637 cells infected with alncRNAs using the

461

wound-healing assay. (B) Cell migration was determined by transwell assay. Inhibition of cell

462

migration following transfection with alncRNAs was observed in 5637 cells. Data are shown as mean ±

463

SD.

RI PT

460

464

Table 1. cDNA sequences of the protein-binding elements used in the vectors.

466 467

Table 2. Relative primers used in this research.

M AN U

468

SC

465

AC C

EP

TE D

469

ACCEPTED MANUSCRIPT 470

Reference

471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510

1 2

RI PT

3

Siegel, R. L., Miller, K. D. & Jemal, A. Cancer statistics, 2016. CA: a cancer journal for clinicians 66, 7-30, doi:10.3322/caac.21332 (2016). Kluth, L. A. et al. Prognostic and Prediction Tools in Bladder Cancer: A Comprehensive Review of the Literature. European urology 68, 238-253, doi:10.1016/j.eururo.2015.01.032 (2015). Kamat, A. M. et al. Bladder cancer. Lancet 388, 2796-2810, doi:10.1016/S0140-6736(16)30512-8 (2016). Marta, G. N. et al. The role of radiotherapy in urinary bladder cancer: current status. International braz j urol : official journal of the Brazilian Society of Urology 38, 144-153; discussion 153-144 (2012). Racioppi, M. et al. Value of current chemotherapy and surgery in advanced and metastatic bladder cancer. Urologia internationalis 88, 249-258, doi:10.1159/000335556 (2012). Sofra, M. et al. Immunomodulatory effects of total intravenous and balanced inhalation anesthesia in patients with bladder cancer undergoing elective radical cystectomy: preliminary results. Journal of experimental & clinical cancer research : CR 32, 6, doi:10.1186/1756-9966-32-6 (2013). Wong, Y. N. S. et al. Evolving adoptive cellular therapies in urological malignancies. The Lancet. Oncology 18, e341-e353, doi:10.1016/S1470-2045(17)30327-3 (2017). Djebali, S. et al. Landscape of transcription in human cells. Nature 489, 101-108, doi:10.1038/nature11233 (2012). Bracken, C. P., Scott, H. S. & Goodall, G. J. A network-biology perspective of microRNA function and dysfunction in cancer. Nature reviews. Genetics 17, 719-732, doi:10.1038/nrg.2016.134 (2016). Han, Y. et al. MicroRNA expression signatures of bladder cancer revealed by deep sequencing. PloS one 6, e18286, doi:10.1371/journal.pone.0018286 (2011). Franzen, C. A. et al. Urinary Exosomes: The Potential for Biomarker Utility, Intercellular Signaling and Therapeutics in Urological Malignancy. The Journal of urology 195, 1331-1339, doi:10.1016/j.juro.2015.08.115 (2016). Keefe, A. D., Pai, S. & Ellington, A. Aptamers as therapeutics. Nat Rev Drug Discov 9, 537-550, doi:10.1038/nrd3141 (2010). Ellington, A. D. & Szostak, J. W. In vitro selection of RNA molecules that bind specific ligands. Nature 346, 818-822, doi:10.1038/346818a0 (1990). Lee, H. K., Choi, Y. S., Park, Y. A. & Jeong, S. Modulation of oncogenic transcription and alternative splicing by beta-catenin and an RNA aptamer in colon cancer cells. Cancer research 66, 10560-10566, doi:10.1158/0008-5472.CAN-06-2526 (2006). Culler, S. J., Hoff, K. G. & Smolke, C. D. Reprogramming cellular behavior

4

SC

5

8 9

10

AC C

11

EP

7

TE D

M AN U

6

12 13 14

15

ACCEPTED MANUSCRIPT

21

22

23

24

25 26

RI PT

20

SC

19

M AN U

18

TE D

17

EP

16

with RNA controllers responsive to endogenous proteins. Science 330, 1251-1255, doi:10.1126/science.1192128 (2010). Brophy, J. A. & Voigt, C. A. Principles of genetic circuit design. Nature methods 11, 508-520, doi:10.1038/nmeth.2926 (2014). Liu, Y. et al. Synthesizing AND gate genetic circuits based on CRISPR-Cas9 for identification of bladder cancer cells. Nat Commun 5, 5393, doi:10.1038/ncomms6393 (2014). Liu, Y. et al. Directing cellular information flow via CRISPR signal conductors. Nature methods 13, 938-944, doi:10.1038/nmeth.3994 (2016). Liu, Y., Li, J., Chen, Z., Huang, W. & Cai, Z. Synthesizing artificial devices that redirect cellular information at will. eLife 7, doi:10.7554/eLife.31936 (2018). Poliseno, L. et al. A coding-independent function of gene and pseudogene mRNAs regulates tumour biology. Nature 465, 1033-1038, doi:10.1038/nature09144 (2010). Kwak, H. et al. Modulation of transcription by the peroxisome proliferator-activated receptor delta--binding RNA aptamer in colon cancer cells. Molecular cancer therapeutics 8, 2664-2673, doi:10.1158/1535-7163.MCT-09-0214 (2009). Liu, Y. et al. Synthesizing oncogenic signal-processing systems that function as both "signal counters" and "signal blockers" in cancer cells. Mol Biosyst 9, 1909-1918, doi:10.1039/c3mb70093c (2013). Weigand, J. E., Wittmann, A. & Suess, B. RNA-based networks: using RNA aptamers and ribozymes as synthetic genetic devices. Methods Mol Biol 813, 157-168, doi:10.1007/978-1-61779-412-4_9 (2012). Paul, C. P., Good, P. D., Winer, I. & Engelke, D. R. Effective expression of small interfering RNA in human cells. Nat Biotechnol 20, 505-508, doi:10.1038/nbt0502-505 (2002). Tetsu, O. & McCormick, F. Beta-catenin regulates expression of cyclin D1 in colon carcinoma cells. Nature 398, 422-426, doi:10.1038/18884 (1999). Liu, Y. et al. Synthetic miRNA-mowers targeting miR-183-96-182 cluster or miR-210 inhibit growth and migration and induce apoptosis in bladder cancer cells. PloS one 7, e52280, doi:10.1371/journal.pone.0052280 (2012). Barre, B. & Perkins, N. D. A cell cycle regulatory network controlling NF-kappaB subunit activity and function. EMBO J 26, 4841-4855, doi:10.1038/sj.emboj.7601899 (2007). Yang, T. et al. Huachansu suppresses human bladder cancer cell growth through the Fas/Fasl and TNF- alpha/TNFR1 pathway in vitro and in vivo. Journal of experimental & clinical cancer research : CR 34, 21, doi:10.1186/s13046-015-0134-9 (2015). Jeon, H. G. et al. Induction of caspase mediated apoptosis and down-regulation of nuclear factor-kappaB and Akt signaling are involved in

AC C

511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552

27

28

29

ACCEPTED MANUSCRIPT

34

35

36

37 38

RI PT

SC

33

M AN U

32

TE D

31

EP

30

the synergistic antitumor effect of gemcitabine and the histone deacetylase inhibitor trichostatin A in human bladder cancer cells. The Journal of urology 186, 2084-2093, doi:10.1016/j.juro.2011.06.053 (2011). Sun, Y. et al. NF-kappaB signaling plays irreplaceable roles in cisplatin-induced bladder cancer chemoresistance and tumor progression. International journal of oncology 48, 225-234, doi:10.3892/ijo.2015.3256 (2016). Mao, X. W., Xiao, J. Q., Li, Z. Y., Zheng, Y. C. & Zhang, N. Effects of microRNA-135a on the epithelial-mesenchymal transition, migration and invasion of bladder cancer cells by targeting GSK3beta through the Wnt/beta-catenin signaling pathway. Experimental & molecular medicine 50, e429, doi:10.1038/emm.2017.239 (2018). Kundu, S. T. et al. The miR-200 family and the miR-183~96~182 cluster target Foxf2 to inhibit invasion and metastasis in lung cancers. Oncogene 35, 173-186, doi:10.1038/onc.2015.71 (2016). Zhong, W. et al. Doxycycline inhibits breast cancer EMT and metastasis through PAR-1/NF-kappaB/miR-17/E-cadherin pathway. Oncotarget 8, 104855-104866, doi:10.18632/oncotarget.20418 (2017). Lienert, F., Lohmueller, J. J., Garg, A. & Silver, P. A. Synthetic biology in mammalian cells: next generation research tools and therapeutics. Nat Rev Mol Cell Biol 15, 95-107, doi:10.1038/nrm3738 (2014). Shi, X., Sun, M., Liu, H., Yao, Y. & Song, Y. Long non-coding RNAs: a new frontier in the study of human diseases. Cancer Lett 339, 159-166, doi:10.1016/j.canlet.2013.06.013 (2013). Ebert, M. S., Neilson, J. R. & Sharp, P. A. MicroRNA sponges: competitive inhibitors of small RNAs in mammalian cells. Nature methods 4, 721-726, doi:10.1038/nmeth1079 (2007). Breaker, R. R. Prospects for riboswitch discovery and analysis. Molecular cell 43, 867-879, doi:10.1016/j.molcel.2011.08.024 (2011). Catuogno, S., Rienzo, A., Di Vito, A., Esposito, C. L. & de Franciscis, V. Selective delivery of therapeutic single strand antimiRs by aptamer-based conjugates. Journal of controlled release : official journal of the Controlled Release Society 210, 147-159, doi:10.1016/j.jconrel.2015.05.276 (2015). Ilgu, M. et al. Light-up and FRET aptamer reporters; evaluating their applications for imaging transcription in eukaryotic cells. Methods 98, 26-33, doi:10.1016/j.ymeth.2015.12.009 (2016). Ruff, K. M., Muhammad, A., McCown, P. J., Breaker, R. R. & Strobel, S. A. Singlet glycine riboswitches bind ligand as well as tandem riboswitches. Rna 22, 1728-1738, doi:10.1261/rna.057935.116 (2016). Farazi, T. A., Spitzer, J. I., Morozov, P. & Tuschl, T. miRNAs in human cancer. J Pathol 223, 102-115, doi:10.1002/path.2806 (2011). Pasquinelli, A. E. MicroRNAs and their targets: recognition, regulation and an

AC C

553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594

39

40

41 42

ACCEPTED MANUSCRIPT

629 630 631

47

48 49

50

51

52

RI PT

SC

46

M AN U

45

TE D

44

EP

43

emerging reciprocal relationship. Nature reviews. Genetics 13, 271-282, doi:10.1038/nrg3162 (2012). Xie, H. et al. LncRNA MALAT1 Inhibits Apoptosis and Promotes Invasion by Antagonizing miR-125b in Bladder Cancer Cells. J Cancer 8, 3803-3811, doi:10.7150/jca.21228 (2017). Jariwalla, R. J. Microcompetition and the origin of cancer. Eur J Cancer 41, 15-19, doi:10.1016/j.ejca.2004.09.027 (2005). Wurster, S. E., Bida, J. P., Her, Y. F. & Maher, L. J., 3rd. Characterization of anti-NF-kappaB RNA aptamer-binding specificity in vitro and in the yeast three-hybrid system. Nucleic acids research 37, 6214-6224, doi:10.1093/nar/gkp670 (2009). Gjerset, R. A. & Bandyopadhyay, K. Regulation of p14ARF through subnuclear compartmentalization. Cell Cycle 5, 686-690, doi:10.4161/cc.5.7.2623 (2006). Jian, Y. et al. RNA aptamers interfering with nucleophosmin oligomerization induce apoptosis of cancer cells. Oncogene 28, 4201-4211, doi:10.1038/onc.2009.275 (2009). He, T. C. et al. Identification of c-MYC as a target of the APC pathway. Science 281, 1509-1512 (1998). Weeraratne, S. D. et al. Pleiotropic effects of miR-183~96~182 converge to regulate cell survival, proliferation and migration in medulloblastoma. Acta Neuropathol 123, 539-552, doi:10.1007/s00401-012-0969-5 (2012). Lin, Y., Bai, L., Chen, W. & Xu, S. The NF-kappaB activation pathways, emerging molecular targets for cancer prevention and therapy. Expert Opin Ther Targets 14, 45-55, doi:10.1517/14728220903431069 (2010). Bobbili, M. R., Mader, R. M., Grillari, J. & Dellago, H. OncomiR-17-5p: alarm signal in cancer? Oncotarget 8, 71206-71222, doi:10.18632/oncotarget.19331 (2017). Hallberg, Z. F., Su, Y., Kitto, R. Z. & Hammond, M. C. Engineering and In Vivo Applications of Riboswitches. Annual review of biochemistry 86, 515-539, doi:10.1146/annurev-biochem-060815-014628 (2017). Lin, F. et al. An Efficient Light-Inducible P53 Expression System for Inhibiting Proliferation of Bladder Cancer Cell. International journal of biological sciences 12, 1273-1278, doi:10.7150/ijbs.16162 (2016).

AC C

595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628

53

ACCEPTED MANUSCRIPT Table1. cDNA sequences of vectors. Synthetic Sequences

CMV-alncRNA1

AGGCCGATCTATGGACGCTATAGGCACACCGGATACTTTAACGATTG GCTATATTCAGGCCGATCTATGGACGCTATAGGCACACCGGATACTTT AACGATTGGCTATATTCAGTGTGAGTTCTGAAGTTGCCAAACTTCAG TGAATTCTCAACGTGCCATACTTCAGCAAAAATGTTAGCGTGCCAAA CTTCAGTGTGAGTTCTGAAGTTGCCAAACTTCAGTGAATTCTCAACG TGCCATACTTCAGCAAAAATGTTAGCGTGCCAAA

CMV-alncRNA2

GATCTTGAAACTGTTTTAAGGTTGGCCGATCATATTCGATCTTGAAA CTGTTTTAAGGTTGGCCGATCATATTCCTACCTGCACTACGTGCACTT GCTTCCTACCTGCACTACGTGCACTTGCTTCCTACCTGCACTACGTG CACTTGCTTCCTACCTGCACTACGTGCACTTGCTTCCTACCTGCACT ACGTGCACTTGCTTCCTACCTGCACTACGTGCACTTG

CMV-NC

TTCTCCGAACGTGTCACGTCACGTTTCAAGAGAACGTGACACGTTC GGAGAAATATTCTTCTCCGAACGTGTCACGTCACGTTTCAAGAGAA CGTGACACGTTCGGAGAAATATTCAAGTTTTCAGAAAGCTAACAAA GTTTTCAGAAAGCTAACAAAGTTTTCAGAAAGCTAACAAAGTTTTC AGAAAGCTAACAAAGTTTTCAGAAAGCTAACAAAGTTTTCAGAAA GCTAACA

U6-alncRNA1

GAGGCCGATCTATGGACGCTATAGGCACACCGGATACTTTAACGATT GGCTATATTCAGGCCGATCTATGGACGCTATAGGCACACCGGATACTT TAACGATTGGCTATATTCAGTGTGAGTTCTGAAGTTGCCAAACTTCA GTGAATTCTCAACGTGCCATACTTCAGCAAAAATGTTAGCGTGCCAA ACTTCAGTGTGAGTTCTGAAGTTGCCAAACTTCAGTGAATTCTCAAC GTGCCATACTTCAGCAAAAATGTTAGCGTGCCAAATTTTTT

U6-alncRNA2

GATCTTGAAACTGTTTTAAGGTTGGCCGATCATATTCGATCTTGAAA CTGTTTTAAGGTTGGCCGATCATATTCCTACCTGCACTACGTGCACTT GCTTCCTACCTGCACTACGTGCACTTGCTTCCTACCTGCACTACGTG CACTTGCTTCCTACCTGCACTACGTGCACTTGCTTCCTACCTGCACT ACGTGCACTTGCTTCCTACCTGCACTACGTGCACTTGTTTTTT

AC C

EP

TE D

M AN U

SC

RI PT

Vector Names

U6-NC

GTTCTCCGAACGTGTCACGTCACGTTTCAAGAGAACGTGACACGTT CGGAGAAATATTCTTCTCCGAACGTGTCACGTCACGTTTCAAGAGA ACGTGACACGTTCGGAGAAATATTCAAGTTTTCAGAAAGCTAACAA AGTTTTCAGAAAGCTAACAAAGTTTTCAGAAAGCTAACAAAGTTTT CAGAAAGCTAACAAAGTTTTCAGAAAGCTAACAAAGTTTTCAGAAA GCTAACATTTTTT

Beta-catenin luciferase responsive elements

AAAGGGGGTAAGATCAAAGGGGGTAAGATCAAAGGGGCGCGAGAT CAAAGGGGGTAAGATCAAAGGGGGTAAGATCAAAGGGGGTAAGAT C

NF-KB luciferase

GGGAATTTCCGGGGACTTTCCGGGAATTTCCGGGGACTTTCCGGGA ATTTCC

ACCEPTED MANUSCRIPT responsive elements Note: Yellow and green parts are the cDNA sequences of RNA aptamers for β-catenin and NF-κB p50 respectively. Pink and blue parts are the cDNA sequences of miRNAs sponges for miR-183/96/182 cluster and miR-17-5p.

Sequences

GAPDH-F

CGCTCTCTGCTCCTCCTGTTC

GAPDH-R

ATCCGTTGACTCCGACCTTCAC

MYC-F

GGCTCCTGGCAAAAGGTCA

MYC-R

CTGCGTAGTTGTGCTGATGT

Cyclin D1-F

GCTGCGAAGTGGAAACCATC

Cyclin D1-R

CCTCCTTCTGCACACATTTGAA

BCL-XL-F

GAGCTGGTGGTTGACTTTCTC

BCL-XL-R

TCCATCTCCGATTCAGTCCCT

Vimentin-F

GACGCCATCAACACCGAGTT

Vimentin-R

CTTTGTCGTTGGTTAGCTGGT

E-cadherin-F

CGAGAGCTACACGTTCACGG

E-cadherin-R

GGGTGTCGAGGGAAAAATAGG

Slug-F

CGAACTGGACACACATACAGTG

Slug-R

CTGAGGATCTCTGGTTGTGGT

TRAF1-F

TCCTGTGGAAGATCACCAATGT

TE D

M AN U

SC

Name

RI PT

Table2. Relative primers used in this research.

TRAF1-R

GCAGGCACAACTTGTAGCC

Has-miR-96~HmiRQP0852

Has-miR-182~HmiRQP0239

The company did not provide sequence information of these Primers.

EP

Has-miR-183~HmiRQP0244 Hsa-miR-17-5P~ HmiRQP0230

AC C

SnRNA U6~HmiRQP9001

Note: F, forward primer; R, reverse primer.

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIPT

Highlights

RI PT SC M AN U TE D



EP



An anti-tumor device, artificial lncRNA (alncRNA), was constructed by connecting microRNA sponge and TFs aptamer. The alncRNA could synchronously accomplish transcriptional and post-transcriptional regulations in bladder cancer cells. Anti-cancer effects were induced by the synthetic alncRNAs in the bladder cancer lines.

AC C



Suggest Documents