Astilbin ameliorates pulmonary fibrosis via blockade of Hedgehog signaling pathway

Astilbin ameliorates pulmonary fibrosis via blockade of Hedgehog signaling pathway

Accepted Manuscript Astilbin ameliorates pulmonary fibrosis via blockade of Hedgehog signaling pathway Jie Zhang, Hanchen Liu, Chenguang Song, Jinjin ...

NAN Sizes 0 Downloads 89 Views

Accepted Manuscript Astilbin ameliorates pulmonary fibrosis via blockade of Hedgehog signaling pathway Jie Zhang, Hanchen Liu, Chenguang Song, Jinjin Zhang, Youlei Wang, Changjun Lv, Xiaodong Song PII:

S1094-5539(18)30030-0

DOI:

10.1016/j.pupt.2018.03.006

Reference:

YPUPT 1718

To appear in:

Pulmonary Pharmacology & Therapeutics

Received Date: 31 January 2018 Revised Date:

25 March 2018

Accepted Date: 31 March 2018

Please cite this article as: Zhang J, Liu H, Song C, Zhang J, Wang Y, Lv C, Song X, Astilbin ameliorates pulmonary fibrosis via blockade of Hedgehog signaling pathway, Pulmonary Pharmacology & Therapeutics (2018), doi: 10.1016/j.pupt.2018.03.006. 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

1

Astilbin ameliorates pulmonary fibrosis via blockade

2

of Hedgehog signaling pathway

5

a Department of Cellular and Genetic Medicine, School of Pharmaceutical Sciences,

9 10

11 12

SC

8

b Department of Respiratory Medicine, Affiliated Hospital to Binzhou Medical University, Binzhou 256602, China.

M AN U

7

Binzhou Medical University, Yantai 264003, China.

c Department of Medical Oncology, People`s Liberation Army 107th Hospital, Yantai,264003, China.

d Department of Respiratory Medicine, Zouping Chinese Medicine Hospital, Binzhou 256602, China.

TE D

6

RI PT

4

Jie Zhang△a,b, Hanchen Liu△c, Chenguang Song△d, Jinjin Zhanga,b, Youlei Wang a, Changjun Lv *a,b, Xiaodong Song* a,b

3

13

Running Title: Astilbin ameliorates pulmonary fibrosis

15

△These authors contributed equally to this work.

16

*Correspondence author: Prof. Xiaodong Song and Changjun Lv, School of

17

Pharmaceutical Sciences of Binzhou Medical University, No. 346, Guanhai Road,

18

Laishan District, Yantai City, 264003, China. Email: [email protected];

19

[email protected]

20

Tel: +86-535-6913985; Fax: +86-535-6913985

AC C

EP

14

1

ACCEPTED MANUSCRIPT 21

22

RI PT

23

ABSTRACT

25

Background and objective: The nature of pulmonary fibrosis involves inadequate

26

repair of the epithelial cell barrier accompanied by impaired regulation of the

27

fibroblast. Moreover, pulmonary fibrosis currently lacks an effective therapeutic drug.

28

This study targets the protection of the epithelial cell and fibroblast to identify a novel,

29

potentially therapeutic drug (i.e., astilbin).

30

Methods: In this study, the cytotoxicity of astilbin was firstly detected using CCK-8.

31

A real-time proliferation/migration analysis system was used to test the inhibitory

32

proliferation and migration of astilbin in vitro. The expression of mesenchymal

33

markers and the loss of epithelial cell markers were analyzed to evaluate the

34

antifibrotic activity of astilbin on TGF-β1-treated AEC-II and L929 cells and

35

bleomycin-treated mice. Then, in fibrosis-associated signaling pathways, the

36

regulation of astilbin was tested using RNA sequencing and Cignal Finder

37

45-Pathway system. Rescue and other experiments were used to confirm this pathway

38

regulation further.

39

Results: The data showed that astilbin inhibited proliferation and migration of cell

40

samples. Its treatment resulted in the reduction of pathological score and collagen

41

deposition, with a decrease in α-SMA and Snail and an increase in E-cadherin and

AC C

EP

TE D

M AN U

SC

24

2

ACCEPTED MANUSCRIPT SP-C in vivo and in vitro. The fibrosis-associated aberrant genes are some of the most

43

notable components of the Hedgehog signaling pathway.

44

Conclusions: Astilbin ameliorates pulmonary fibrosis via blockade of Hedgehog

45

signaling pathway and has potential therapeutic value for lung fibrosis treatment.

46

Key Words: Astilbin; Anti-pulmonary fibrosis; Hedgehog pathway

47

1. Introduction

SC

RI PT

42

Pulmonary fibrosis results from abnormal tissue repair and is associated with

49

persistent and/or severe tissue damage and cellular stress [1]. The nature of this

50

aberrance involves inadequate repair of the epithelial cell barrier accompanied by

51

impaired regulation of the fibroblast [2], which results in profound changes to

52

alveolar epithelial cell and fibroblast, leading to uncontrolled fibrosis [3]. Thus,

53

targeting the protection of the epithelial cell and fibroblast is an attractive approach to

54

treat pulmonary fibrosis [4]. Pirfenidone and nintedanib were approved for the

55

treatment of pulmonary fibrosis in 2014 [5,6]. However, differences in adverse event

56

frequency, such as photosensitivity, gastrointestinal symptoms, and liver function test

57

abnormalities, were observed [7,8]. Gene therapy is a potential treatment; however,

58

successfully applying it to clinical practice has so far been a formidable challenge

59

[9,10]. Therefore, the identification of a novel therapeutic drug in preclinical study is

60

highly desired.

AC C

EP

TE D

M AN U

48

61

Astilbin, (2R,3R)-3,3′,4′,5,7-pentahydroxyflavanon-3-α-L-rhamnopyranoside, is a

62

dihydroflavonol rhamnoside, which is significant in the treatment of immunologically 3

ACCEPTED MANUSCRIPT related diseases. Several studies have demonstrated that astilbin ameliorates

64

experimental autoimmune myasthenia gravis and imiquimod-induced psoriasis-like

65

skin lesions [11,12]. Astilbin has also shown many other bioactivities, such as

66

anti-oxidative, anti-inflammation, and 3-hydroxy-3-methylglutaryl coenzyme A

67

reductase inhibitor [13,14]. Astilbin is currently used to ameliorate cisplatin-induced

68

nephrotoxicity through reducing oxidative stress and inflammation [15]. In the

69

treatment of lung injury, astilbin can alleviate sepsis-induced acute lung injury by

70

inhibiting the expression of macrophage inhibitory factor in rats [16]. However, the

71

anti-pulmonary fibrosis of astilbin remains unknown. Pulmonary fibrosis is actually a

72

kind of lung injury, except that this is a specific form of chronic, progressive injury.

73

This study aims to investigate the effects of astilbin against pulmonary fibrosis and its

74

potential anti-pulmonary fibrosis mechanism in vivo and in vitro.

75

2. Methods

76

2.1. Cell culture

EP

TE D

M AN U

SC

RI PT

63

Mouse type II alveolar epithelial cell (AEC-II) and mouse lung fibroblast (L929)

78

cell lines were purchased from Cell Bank of Chinese Academy of Sciences.

79

Transforming growth factor beta 1 (TGF-β1), the most important regulatory factor

80

among fibrogenic cytokines, was used to stimulate AEC-II and L929 cells to establish

81

the pulmonary fibrosis model in vitro [17,18]. Cells were maintained in advanced

82

minimum essential medium and supplemented with 10% newborn calf serum, 100

83

U/mL penicillin and 100 µg/mL streptomycin at 37℃ under a humidified atmosphere

AC C

77

4

ACCEPTED MANUSCRIPT of 5% CO2 and 95% air. AEC-II and L929 cells were first administered with 5 ng/mL

85

TGF-β1 for 6 h and then co-treated with astilbin (Dalian Meilun Biotech Co., Ltd) for

86

48 h.

87

2.2. Cell counting kit-8 assay (CCK-8)

RI PT

84

Cell proliferation was determined using CCK-8 kit (DOJINDO, Japan) in

89

accordance with the manufacturer’s instructions. In brief, 1 × 106 cells/well were

90

seeded in a 96-well and flat-bottomed plate and then grown at 37 °C for 24 h. After 10

91

µL of WST-8 dye was added to each well, the cells were incubated at 37 °C for 2 h.

92

Absorbance was determined at 450 nm by using a microplate reader. Cell proliferation

93

was calculated on the basis of the coloration depth by using the following formula:

94

Cell proliferation (%) = (Measurement tube absorbance − Absorbance of blank) /

95

(Standard pipe of absorbance − Absorbance of blank) × 100%.

96

2.3. Real-time cellular proliferation and migration analysis

EP

TE D

M AN U

SC

88

Cell proliferation was performed in E-Plate (Proliferation plate) and CIM-Plate

98

(Migration plate) in a real-time cellular proliferation/migration analysis DPlus

99

instrument (ACEA Biosciences, Inc., Hangzhou, China), which can automatically

100

record proliferation/migration curves. The cell index representing the amount of

101

proliferation/migration cells was calculated with RTCA Software from ACEA

102

Biosciences.

103

2.4. Animal model and ethics statement

AC C

97

5

ACCEPTED MANUSCRIPT Mice with mean weight of 25±2 g were purchased from the Model Animal

105

Research Center of Nanjing University (Nanjing, China). All animal experiments

106

were performed following regulations of Committee on the Ethics of Animal

107

Experiments of Binzhou Medical University. Mice were housed under 12 h light/dark

108

cycle, allowed free access to food and water, and randomly divided into four groups

109

(10 mice per group), namely, sham group, bleomycin-treated group (BLM group), 20

110

mg/kg astilbin (AST I group), and 40 mg/kg AST II group. BLM animal model was

111

administered with 5 mg/kg BLM dissolved in saline via single intratracheal

112

instillation under anesthesia as previously described. At day 14, astilbin was orally

113

administered every day. At day 30, all mice were killed, and lung tissue sections were

114

collected and immediately frozen in liquid nitrogen for further studies.

115

2.5. Western blot

TE D

M AN U

SC

RI PT

104

20 µg of protein sample was subjected to 10% sodium dodecyl sulfate

117

polyacrylamide gel electrophoresis, transferred onto polyvinylidene difluoride

118

membranes, and blocked with 7% non-fat milk in Tris-buffered saline and Tween-20

119

(TBST; 50 mM Tris-HCl [pH 7.6], 150 mM NaCl, 0.1% Tween-20). Membranes were

120

washed thrice with TBST buffer and incubated at 4°C overnight with specific

121

antibodies. After washing with TBST, membranes were incubated with horseradish

122

peroxidase-labeled IgG for 1.5 h. Membranes were then washed with TBST,

123

incubated with ECL reagent, and exposed. Then, membranes were subsequently

124

stripped and re-probed with glyceraldehyde 3-phosphate dehydrogenase antibody,

AC C

EP

116

6

ACCEPTED MANUSCRIPT 125

which served as loading control.

126

2.6. RNA-Sequencing

A total amount of 2 µg RNA per sample was used as input material for the RNA

128

sample preparations. Sequencing libraries were generated using NEBNext® Ultra™

129

RNA Library Prep Kit for Illumina® (#E7530L, NEB, USA) following the

130

manufacturer’s recommendations and index codes were added to attribute sequences

131

to each sample. Briefly, mRNA was purified from total RNA using poly-T

132

oligo-attached magnetic beads. Fragmentation was carried out using divalent cations

133

under elevated temperature in NEBNext First Strand Synthesis Reaction Buffer (5X).

134

First strand cDNA was synthesized using random hexamer primer and RNase H.

135

Second strand cDNA synthesis was subsequently performed using buffer, dNTPs,

136

DNA polymerase I and RNase H. The library fragments were purified with QiaQuick

137

PCR kits and elution with EB buffer, then terminal repair、A-tailing and adapter added

138

were implemented. The aimed products were retrieved by agarose gel electrophoresis

139

and PCR was performed, then the library was completed. RNA concentration of

140

library was measured using Qubit® RNA Assay Kit in Qubit® 3.0 to preliminary

141

quantify and then dilute to 1 ng/µL. Insert size was assessed using the Agilent

142

Bioanalyzer 2100 system (Agilent Technologies, CA, USA), and qualified insert size

143

was accurately quantified using StepOnePlus™ Real-Time PCR System (Library valid

144

concentration>10 nM). The clustering of the index-coded samples was performed on

145

a cBot cluster generation system using HiSeq PE Cluster Kit v4-cBot-HS (Illumina)

AC C

EP

TE D

M AN U

SC

RI PT

127

7

ACCEPTED MANUSCRIPT according to the manufacturer`s instructions. After cluster generation, the libraries

147

were sequenced on an Illumina Hiseq 4000 platform and 150 bp paired-end reads

148

were generated.

149

2.7. Cignal Finder 45-Pathway reporter array

RI PT

146

The signaling pathway was evaluated using the Cignal Finder 45-Pathway

151

reporter array [19]. All the techniques were performed using standard protocols

152

established according to the manufacturer’s protocol (Qiagen Co Ltd, Germany).

153

Briefly, 50 µL of

154

Array plate. 0.6 µL of Attractene used in 50 µL of Opti-MEM® per well for each

155

individual transfection. After the 5 minute incubation, 50 µL of diuted Attractene was

156

added into each well containing 50 µL of the diluted nucleic acids. Then cells were

157

washed in a culture dish once with Dulbecco's PBS without calcium and magnesium,

158

and treated with 1-3 mL trypsin-EDTA for 2-5 minutes at 37℃ in a humidified

159

atmosphere containing 5% CO2. Cells were suspended in 7-9 mL of Opti-MEM®

160

containing 5% of fetal bovine serum, then centrifuged, removed the superatant, and

161

resuspended to 8 x 105 cells/mL in Opti-MEM® containing 10% of fetal bovine

162

serum and 1% NEAA. 50 µL of prepared cell suspension was added to each well

163

containing constructes-Attractene complexes. A final volume in each well was 150 µL.

164

After 16-24 h incubation, the medium was changed to complete growth medium.

165

50 µL of Dual-Glo Luciferase reagent was added and the fluorescence intensity was

166

determined using a microplate reader (SpectraMax M2).

added

to

each well of

the Cignal Finder

AC C

EP

TE D

M AN U

Opti-MEM® was

SC

150

8

ACCEPTED MANUSCRIPT 167

2.8. Patients and healthy volunteers

Lung tissue samples were obtained by open lung biopsy. IPF was diagnosed in

169

accordance with the American Thoracic Society/European Respiratory Society

170

consensus criteria [20], which include clinical, radiographic, and characteristic

171

histopathological features (n=4). Control non-pulmonary fibrosis lung tissue samples

172

were obtained from smokers who underwent thoracic surgery for localized primary

173

lung carcinoma. As stated in agreement, written informed consent was obtained by

174

doctors from each participant. This study was approved by ethics committee of

175

Binzhou Medical University.

176

2.9. Immunofluorescence, Immunohistochemistry, H&E and Masson staining

SC

M AN U

All the techniques were performed using standard protocols established in our

TE D

177

RI PT

168

laboratory [21].

179

2.10. Statistical analysis

EP

178

Statistical analysis was performed with SPSS 17 software. All data were

181

generated from minimum of three independent experiments and expressed as mean ±

182

standard deviation (SD). Different groups were compared using student’s t-test (for

183

two groups) or one-way ANOVA (for more than two groups). P < 0.05 was considered

184

statistically significant.

185

3. Results

186

3.1. Cytotoxicity of astilbin in vitro

AC C

180

9

ACCEPTED MANUSCRIPT First, the cytotoxicity of astilbin in vitro was confirmed using CCK-8. AEC-II

188

and L929 were incubated for 48 h under increasing concentrations of astilbin. Then,

189

astilbin toxicity was evaluated. As compared with untreated cells, the half maximal

190

inhibitory concentration (IC50) in AEC-II and L929 cell lines were 800 µg/mL <

191

IC50 < 1 mg/mL and 1 mg/mL < IC50 < 1.2 mg/mL, respectively (Fig. 1A and B).

192

Then, the inhibitory effects of astilbin on TGF-β1-treated cell viability were tested.

193

Astilbin inhibited cell viability in a dose-dependent manner when the cells were

194

exposed to graded doses (2.5–80 µg/mL) for 48 h. This inhibition was pronounced in

195

vitro. The most pronounced inhibition was at the beginning of 5 and 20 µg/mL

196

concentrations in AEC-II and L929, respectively. Thus, 5, 10, and 20 µg/mL in

197

AEC-II and 20 and 40 µg/mL in L929 were used for further in vitro studies (Fig. 1C

198

and D).

199

3.2. Anti-pulmonary fibrosis of astilbin in vitro

TE D

M AN U

SC

RI PT

187

Fundamental pathogenic hallmarks of pulmonary fibrosis exhibited uncontrolled

201

proliferation and high migration rates. Thus, a real-time proliferation/migration

202

analysis system was used to test the inhibitory proliferation and high migration of

203

astilbin on TGF-β1-treated AEC-II and L929 cells. As compared with TGF-β1-treated

204

cells, astilbin inhibited proliferation and migration in a dose-dependent manner when

205

the cells were exposed to graded doses (Fig. 2A and B). Then, lung fibrosis and

206

epithelial cell markers were assessed using Western blot analysis. The analysis

207

showed that astilbin increased the epithelial cell marker expressions of E-cadherin and

AC C

EP

200

10

ACCEPTED MANUSCRIPT SP-C and decreased the mesenchymal marker expressions of α-SMA, vimentin, Snail,

209

and collagen I and III compared with the TGF-β1-treated group (Fig. 2C and D).

210

3.3. Anti-pulmonary fibrosis of astilbin in vivo

RI PT

208

The anti-pulmonary fibrosis of astilbin in mice was tested to evaluate the

212

therapeutic value of astilbin further. First, astilbin increased the force vital capacity

213

(FVC) compared with the BLM-treated group (Fig. 3A). Thin alveolar walls were

214

observed in tissue sections, and the number of fibroblasts and collagen matrices

215

decreased significantly in astilbin-treated groups (Fig. 3B and C). The indicators of

216

pulmonary fibrosis, such as E-cadherin, SP-C, vimentin, α-SMA, Snail, and collagen I

217

and III, were investigated using Western blot analysis and immunofluorescence

218

staining (Fig 4A and B). The data showed that astilbin increased the epithelial cell

219

markers of E-cadherin and SP-C and decreased the mesenchymal markers of vimentin,

220

α-SMA, Snail, and collagen I and III.

221

3.4. Regulation of astilbin in pulmonary fibrosis-associated aberrant mRNAs

EP

TE D

M AN U

SC

211

The differentially expressed RNAs were evaluated using RNA sequencing to

223

elucidate the anti-pulmonary fibrosis mechanism of astilbin. From the RNA

224

sequencing data, the mRNA expression profiles of the astilbin-treated group were

225

compared with that of the BLM-treated group. Enrichment analysis was applied to

226

explore the functions of the mRNAs identified in this study. Genes were organized

227

into hierarchical categories to uncover gene regulatory networks on the basis of

228

biological processes, cellular components, and molecular functions. The analysis

AC C

222

11

ACCEPTED MANUSCRIPT showed that many dysregulated mRNAs were enriched in the Hedgehog signaling

230

pathway (Fig. 5), such as Boc, Lrp, Rab23, Gli, Gsk3b, Csnk, Prkaca, Sufu, Stk, Zic2

231

and so on.

232

3.5. Confirmation of the Hedgehog pathway involved in the antifibrotic activity of

233

astilbin

RI PT

229

The Cignal Finder 45-Pathway Reporter Array confirmed the RNA sequencing

235

results which showed that astilbin significantly inhibited glioma-associated oncogene

236

homolog (Gli) reporter luciferase activities compared with other reporters (Fig. 6A).

237

Then, Gli1 was selected as a representative gene to confirm the in vivo and in vitro

238

results using Western blot analysis and immunofluorescence staining. The data

239

showed that Gli1 increased in the pulmonary fibrosis model in vivo and in vitro, and

240

astilbin remarkably decreased Gli1 expression (Fig. 6B-E). Gli1 expression in patients

241

with pulmonary fibrosis was tested using immunohistochemistry to evaluate

242

therapeutic value of astilbin. Gli1 expression was also increased in patients with

243

pulmonary fibrosis, supporting the in vivo and in vitro results (Fig. 6F). The rescue

244

experiment was used to evaluate and verify that the Hedgehog pathway is involved in

245

the antifibrotic activity of astilbin further. Chemical activator smoothened agonist

246

(SAG) and inhibitor vismodegib (GDC-0449) that are specific to the Hedgehog

247

pathway were used, which showed that the activator can promote the expression of

248

α-SMA and collagen I and III, but the inhibitor can block the expression of these

249

fibrotic indicators. And astilbin reversed these changes caused by SAG and

AC C

EP

TE D

M AN U

SC

234

12

ACCEPTED MANUSCRIPT 250

GDC-0449 (Fig. 7).

251

3.6. Analysis of co-expressed lncRNA with Gli1

Long noncoding RNA (lncRNA) is the upstream factor of mRNA that regulates

253

its function. From the RNA sequencing data, Gli1 and its co-expressed lncRNA were

254

analyzed in the astilbin-treated group compared with the BLM-treated group. The

255

degree of mRNA–lncRNA co-expression was analyzed. An increased degree indicated

256

an

257

lncRNA-NUNMM028949.2 has the highest degree (Figures 8A and 8B).

258

lncRNA-NUNMM028949.2 was selected to determine its expression levels through

259

qRT-PCR to validate the data. As shown in Figure 8C, the lncRNA decreased in the

260

astilbin-treated group compared with the BLM-treated group.

261

4. Discussion

in

their

SC

co-expression

interaction.

Among

these

lncRNAs,

In

the

TE D

M AN U

increased

present

study,

we

investigated

the

effect

of

astilbin

on

EP

262

RI PT

252

TGF-β1/BLM-induced lung fibrogenesis in vivo and in vitro, and its mechanism of

264

Hedgehog pathway signaling pathway in astilbin treatment. Our results showed that

265

astilbin significantly decreased alveolar wall thickness and collagen fiber formation,

266

and these anti-fibrotic effects of astilbin may be mediated through blockade of

267

Hedgehog signaling pathway.

AC C

263

268

Similar to cancer, pulmonary fibrosis show functional features such as

269

low adhesion, uncontrolled proliferation and high migration rates [22,23]. Snail is the

270

transcriptional repressors of adheren that is a direct repressor of E-cadherin 13

ACCEPTED MANUSCRIPT expression and critical for intercellular adhesions [24,25]. Astilbin decreased its

272

expression. Meanwhile, astilbin also could inhibit the proliferation and migration of

273

TGF-β1-treated AEC-II and L929 cells. One of the phenomena during the process of

274

pulmonary fibrosis is the manifestation of mesenchymal characteristics, including the

275

expression of mesenchymal markers and the loss of epithelial cell markers [26]. Our

276

data showed that astilbin increased levels of epithelial cell markers such as E-cadherin

277

and SP-C, and decreased levels of mesenchymal markers such as a-SMA, collagen

278

and vimentin. All these finding showed that the anti-pulmonary fibrosis of astilbin is

279

significant.

M AN U

SC

RI PT

271

The pathogenesis of pulmonary fibrosis indicates that the fibrotic response is

281

driven by abnormally activated alveolar epithelial cells, which produce mediators that

282

induce the formation of fibroblast and myofibroblast foci through the proliferation of

283

resident mesenchymal cells. The fibroblast and myofibroblast foci secrete excess

284

amounts of extracellular matrix, including collagen, α-SMA, and vimentin, resulting

285

in scarring and destruction of the lung architecture. However, the pathways leading to

286

the secretion of excess amounts of extracellular matrix that produce fibroblasts are

287

uncertain [27]. To address these uncertainties, RNA sequencing was used to explore

288

the pathways in this study. RNA sequencing is a powerful technique to investigate the

289

complexity of gene expression [28]. The findings showed that astilbin can ameliorate

290

pulmonary fibrosis through multigene, multi-pathway. These genes are some of the

291

most notable components of the Hedgehog signaling pathway, such as Hedgehog

292

ligand (Boc), Hedgehog receptors and cofactors (Lrp and Rab23), transcription factors

AC C

EP

TE D

280

14

ACCEPTED MANUSCRIPT and regulators (Gli, Gsk3b, Csnk, Prkaca, Sufu, Stk, and Zic2), Hedgehog target

294

genes (Ptch, Bcl-2, Vegfa, Disp, Mapk, Runx2, and Trp53), and pathways crosstalking

295

with Hedgehog (Wnt, Bmp, Mob1b, Nf2, and Wif1) [29]. Based on these results, the

296

influence of astilbin on Hedgehog pathways was further detected.

RI PT

293

Aberrant activation of the Hedgehog signaling pathway, which has a crucial role

298

in the development and homeostasis of many organs and tissues, has been linked to

299

tumorigenesis [30]. The role of the Hedgehog signaling pathway in pulmonary

300

fibrosis pathogenesis has not been thoroughly investigated. In this study, we analyzed

301

the function and mechanism of this pathway in pulmonary fibrosis. Gli is one of

302

components of the Hedgehog signaling pathway including Gli1, Gli2, and Gli3

303

[31,32], and its expression level is widely accepted to reflect Hedgehog pathway

304

activity [33]. It acts as a zinc finger transcription factor initially characterized in

305

human glioblastoma to control cell differentiation and proliferation [34]. Although the

306

involvement of Gli in a variety of cancers is well established [35], its role in

307

pulmonary fibrosis remains poorly understood. Genetic lineage tracing analysis

308

indicates that Gli1+ cells proliferate after kidney, lung, liver, or heart injury to

309

generate myofibroblasts. Genetic ablation of these cells substantially ameliorates

310

kidney and heart fibrosis and preserves ejection fraction in a model of induced heart

311

failure [36]. Our results showed that Gli1 increased in lung fibrosis, which supports

312

the notion that Gli1 is a key contributor to organ fibrosis and indicates the possible

313

pathways implicated in lung fibrosis as targets of therapeutic attempts. Astilbin can

314

block the progression of pulmonary fibrosis via blockade of Gli expression.

AC C

EP

TE D

M AN U

SC

297

15

ACCEPTED MANUSCRIPT 315

High-throughput technologies such as RNA sequencing have revealed that only 2% of the transcribed genome codes are attributed to proteins. The remaining part of the

317

transcribed genome is known as noncoding RNA. Among the various types of

318

noncoding RNA, a class referred to as long noncoding RNA (lncRNA) has attracted

319

increasing attention [37]. Emerging evidence suggests lncRNA plays important roles

320

in disease development. These noncoding RNAs are the upstream factors of mRNAs

321

that directly or indirectly regulate the biological function of mRNAs [38,39]. Here,

322

we demonstrate the lncRNAs in targeting Gli1 to regulate the anti-pulmonary fibrosis

323

mechanism of astilbin. We will be designing experiments to determine the

324

relationship between astilbin and lncRNAs -mediated pulmonary fibrosis for future

325

research.

326

5. Conclusions

TE D

M AN U

SC

RI PT

316

In summary, our study showed that astilbin can attenuate lung fibrosis by

328

suppressing the Hedgehog pathway, which provided a rationale for exploring the

329

therapeutic and chemoprotective potentials of astilbin for the treatment and prevention

330

of pathologic conditions involving pulmonary fibrosis in future studies.

331

Acknowledgements

AC C

EP

327

332

This study was supported by National Natural Science Foundation of China

333

(31470415, 31670365, 81670064, 81741170), Natural Science Foundation of

334

Shandong Province (ZR2016HP34).

335

Conflicts of Interest: The authors declare no conflict of interest. 16

ACCEPTED MANUSCRIPT 336

References

338

[1] A.L. Mora1, M. Rojas, A. Pardo, M. Selman, Emerging therapies for idiopathic

339

pulmonary fibrosis, a progressive age-related disease, Nature Reviews Drug

340

Discovery 170 (2017) 1-18.

Reviews Drug Discovery 9 (2010) 129-140.

SC

342

[2] R.M. du Bois, Strategies for treating idiopathic pulmonary fibrosis, Nature

M AN U

341

RI PT

337

343

[3] M. Selman, A. Pardo, Revealing the pathogenic and aging-related mechanisms of

344

the enigmatic idiopathic pulmonary fibrosis. an integral model, Am. J. Respir. Crit.

345

Care Med. 189 (2014) 1161–1172.

347

[4] B.D. Uhal, The role of apoptosis in pulmonary fibrosis, Eur. Respir. Rev. 17 (2008) 138–144.

TE D

346

[5] T.E. King Jr, W.Z. Bradford, S. Castro-Bernardini, E. A. Fagan, I. Glaspole, M. K.

349

Glassberg, et al., A phase 3 trial of pirfenidone in patients with idiopathic

AC C

350

EP

348

pulmonary fibrosis, N. Engl. J. Med. 370 (2014 ) 2083–2092.

351

[6] L. Richeldi, R.M. du Bois , G. Raghu, A. Azuma, K.K. Brown, U. Costabel , et al.,

352

Efficacy and safety of nintedanib in idiopathic pulmonary fibrosis, N. Engl. J.

353

Med. 370 (2014) 2071–2082.

354

[7] P.W. Noble, C. Albera, W. Z. Bradford, U. Costabel , R.M. du Bois, E.A. Fagan, et

355

al., Pirfenidone for idiopathic pulmonary fibrosis: analysis of pooled data from 17

ACCEPTED MANUSCRIPT 356

three multinational phase 3 trials, Eur. Respir. J. 47 (2016) 243–253.

[8] L. Richeldi, V. Cottin, R.M. du Bois, M. Selman, T. Kimura, Z. Bailes, et al.,

358

Nintedanib in patients with idiopathic pulmonary fibrosis: Combined evidence

359

from the TOMORROW and INPULSIS® trials, Respiratory Medicine 113 (2016)

360

74-79.

RI PT

357

[9] L. Naldini, Gene therapy returns to centre stage, Nature 526 (2015) 351–360.

362

[10] N. Somia, I.M. Verma, Gene therapy: trials and tribulations, Nat. Rev. Genet. 1

M AN U

363

SC

361

(2000) 91–99.

[11] Q.F. Meng, Z. Zhang, Y.J. Wang, W. Chen, F.F. Li, L.T. Yue, et al., Astilbin

365

ameliorates experimental autoimmune myasthenia gravis by decreased Th17

366

cytokines and up-regulated T regulatory cells, J. Neuroimmunol. 298 (2016)

367

138-45.

TE D

364

[12] T.T. Di, Z.T. Ruan, J.X. Zhao, Y. Wang, X. Liu, Y. Wang, et al., Astilbin inhibits

369

Th17 cell differentiation and ameliorates imiquimod-induced psoriasis-like skin

370

lesions in BALB/c mice via Jak3/Stat3 signaling pathway, Int. Immunopharmacol.

AC C

371

EP

368

32 (2016) 32-38.

372

[13] Q.F. Zhang, Y.J. Fu, Z.W. Huang, X.C. Shangguang, Y.X. Guo, Aqueous Stability

373

of Astilbin: Effects of pH, Temperature, and Solvent, J. Agric. Food Chem. 61

374

(2013) 12085−12091.

375

[14] Q.F. Zhang, Z.R. Zhang, H.Y. Cheung, Antioxidant activity of Rhizoma Smilacis 18

ACCEPTED MANUSCRIPT 376

Glabrae extracts and its key constituent − astilbin, Food Chem. 115 (2009)

377

297−303.

[15] S.W. Wang, Y. Xu, Y.Y. Weng, X.Y. Fan, Y.F. Bai, X.Y. Zheng, et al., Astilbin

379

ameliorates cisplatin-induced nephrotoxicity through reducing oxidative stress and

380

inflammation, Food Chem. Toxicol. 114 (2018) 227-236.

RI PT

378

[16] H.B. Zhang, L.C. Sun, L.D. Zhi, Q.K. Wen, Z.W. Qi, S.T. Yan, et al., Astilbin

382

alleviates sepsis-induced acute lung injury by inhibiting the expression of

383

macrophage inhibitory factor in rats, Arch. Pharm. Res. 40 (2017) 1176-1185.

384

[17] Y. Shi, B.R. Gochuico, G. Yu, X. Tang, J.C. Osorio, I.E. Fernandez, et al.,

385

Syndecan-2 exerts antifibrotic effects by promoting caveolin-1-mediated

386

transforming growth factor-β receptor I internalization and inhibiting transforming

387

growth factor-β1 signaling, Am. J. Respir. Crit. Care Med. 188 (2013) 831–841.

388

[18] H. Liu, B. Wang, J. Zhang, S. Zhang, Y. Wang, J. Zhang, et al., A novel lnc-PCF

389

promotes the proliferation of epithelial cell activated with TGF-β1 by targeting

390

miR-344a-5p to regulate map3k11 in pulmonary fibrosis, Cell death and disease 8

M AN U

TE D

EP

AC C

391

SC

381

(2017) e3137.

392

[19] Z. Xing, A. Lin, C. Li, K. Liang, S. Wang, Y. Liu, et al., lncRNA directs

393

cooperative epigenetic regulation downstream of chemokine signals, Cell 159

394

(2014) 1110-1125.

395

[20] G. Raghu, H.R. Collard, J.J. Egan, F.J. Martinez, J. Behr, K.K. Brown, et al., An 19

ACCEPTED MANUSCRIPT 396

Official

397

Evidence-based Guidelines for Diagnosis and Management, Am. J. Respir. Crit.

398

Care Med. 183 (2011) 788-824.

Statement:

Idiopathic

Pulmonary

Fibrosis:

[21] X.D. Song, W.L. Liu, S.Y. Xie, M.R. Wang, G.H. Cao, C.P. Mao, et al.,

400

All-transretinoic

ameliorates

bleomycin-induced

lung

fibrosis

by

401

downregulating the TGF-b1/Smad3 signaling pathway in rats, Lab. Invest. 93

402

(2013) 1219–1231.

SC

acid

RI PT

399

ATS/ERS/JRS/ALAT

[22] C. Vancheri, M. Failla, N. Crimi, G. Raghu, Idiopathic pulmonary fibrosis: a

404

disease with similarities and links to cancer biology, Eur. Respir. J. 35 (2010)

405

496-504.

407

[23] C. Vancheri, Idiopathic pulmonary fibrosis: An altered fibroblast proliferation

TE D

406

M AN U

403

linked to cancer biology, Proc. Am. Thorac. Soc. 9 (2012) 153-157.

[24] A. Cano, M.A. Pérez-Moreno, I. Rodrigo, A. Locascio, M.J. Blanco, M.G. del

409

Barrio,et al., The transcription factor Snail controls epithelial–mesenchymal

410

transitions by repressing E-cadherin expression, Nat. Cell Biol. 2 (2000) 76-83.

411

[25] E. Batlle, E. Sancho, C. Francí, D. Domínguez, M. Monfar, J. Baulida, et al., The

412

transcription factor Snail is a repressor of E-cadherin gene expression in epithelial

413

AC C

EP

408

tumour cells, Nat. Cell Biol. 2 (2000) 84-89.

414

[26] K.V. Pandit, D. Corcoran, H. Yousef, M. Yarlagadda, A. Gibson, K.F.

415

Tzouvelekis, et al., Inhibition and Role of let-7d in Idiopathic Pulmonary Fibrosis, 20

ACCEPTED MANUSCRIPT 416

Am. J. Respir. Crit. Care Med. 182 (2010) 220–229.

[27] K.K. Kim, M.C. Kugler, P.J. Wolters, L. Robillard, M.G. Galvez, A.N. Brumwell,

418

et al., Alveolar epithelial cell mesenchymal transition develops in vivo during

419

pulmonary fibrosis and is regulated by the extracellular matrix, PNAS 103 (2006 )

420

13180–13185.

RI PT

417

[28] T.K. Doktor, Y. Hua, H.S. Andersen, S. Brøner, Y.H. Liu, A. Wieckowska, et al.,

422

RNA-sequencing of a mouse-model of spinal muscular atrophy reveals

423

tissue-wide changes in splicing of U12-dependent introns, Nucleic Acids Research

424

45 (2017) 395-416.

M AN U

425

SC

421

[29] S.J. Scales, F. J. de Sauvage, Mechanisms of Hedgehog pathway activation in cancer and implications for therapy, Trends in Pharmacological Sciences 30 (2009)

427

303-312.

430 431

EP

429

[30] D. Amakye, Z. Jagani, M. Dorsch, Unraveling the therapeutic potential of the Hedgehog pathway in cancer, Nature Medicine 19 (2013) 1410-1422.

[31] L. Lum, P.A. Beachy, The Hedgehog response network: sensors, switches, and

AC C

428

TE D

426

routers, Science 304 (2004) 1755–1759.

432

[32] A. Ruiz i Altaba, C. Mas , B. Stecca, The Gli code: an information nexus

433

regulating cell fate, stemness and cancer, Trends Cell Biol. 17 (2007) 438–447.

434

[33] S.J. Scales, F.J. de Sauvage, Mechanisms of Hedgehog pathway activation in

435

cancer and implications for therapy, Trends Pharmacol. Sci. 30 (2009) 303–312. 21

ACCEPTED MANUSCRIPT 436

[34] S. Sheybani-Deloui, L. Chi, M.V. Staite, J.E. Cain, B.J. Nieman, R.M.

437

Henkelman, et al., Activated Hedgehog-GLI Signaling Causes Congenital

438

Ureteropelvic Junction Obstruction, J. Am. Soc. Nephrol. (2017).

[35] L. Huang, V. Walter, D.N. Hayes, M. Onaitis, Hedgehog–GLI Signaling

440

Inhibition Suppresses Tumor Growth in Squamous Lung Cancer, Clin. Cancer Res.

441

20 (2014) 1566-1575.

SC

RI PT

439

[36] R. Kramann, R.K. Schneider, D.P. DiRocco, F. Machado, S. Fleig, P.A. Bondzie,

443

et al., Perivascular Gli1+ Progenitors Are Key Contributors to Injury-Induced

444

Organ Fibrosis, Cell Stem Cell 16 (2015) 51–66.

446

[37] F. Kopp, J.T. Mendell, Functional Classification and Experimental Dissection of Long Noncoding RNAs, Cell 172 (2018) 393-407.

TE D

445

M AN U

442

[38] L. Meng, A.J. Ward, S. Chun, C.F. Bennett, A.L. Beaudet, F. Rigo, Towards a

448

therapy for Angelman syndrome by targeting a long non-coding RNA, Nature 518

449

(2015) 409–412.

EP

447

[39] Y. Liu, R. Guo, G. Hao, J. Xiao, Y. Bao, J. Zhou, et al., The expression profiling

451

and ontology analysis of noncoding RNAs in peritoneal fibrosis induced by

452

AC C

450

peritoneal dialysis fluid, Gene 564 (2015) 210–219.

453

454

455 22

ACCEPTED MANUSCRIPT 456

457

RI PT

458

459

SC

460

M AN U

461

462

463

TE D

464

465

468

AC C

467

EP

466

469

Figure legends

470

Fig. 1. Astilbin cytotoxicity in AEC-II and L929 using CCK-8. (A) Astilbin

471

cytotoxicity in the normal AEC-II cells (800 µg/mL < IC50 < 1 mg/mL). (B) Astilbin

472

cytotoxicity in the normal L929 cells (1 mg/mL < IC50 < 1.2 mg/mL). (C) and (D)

473

AEC-II and L929 cells were initially administered with 5 ng/mL TGF-β1 for 6 h and 23

ACCEPTED MANUSCRIPT subsequently co-treated with astilbin for 48 h. As compared with TGF-β1-treated cells,

475

the most pronounced inhibition was at the beginning of 5 and 20 µg/mL

476

concentrations in AEC-II and L929, respectively. Each bar represents the mean ± SD,

477

with n = 6 and *p < 0.05, **p < 0.01.

478

Fig. 2. Anti-pulmonary fibrosis of astilbin in AEC-II and L929 cells. (A) The

479

real-time proliferation analysis system showed that astilbin inhibited TGF-β1-treated

480

cell proliferation. (B) The real-time migration analysis system showed that astilbin

481

inhibited TGF-β1-treated cell migration. (C) Astilbin increased the expressions of

482

E-cadherin and SP-C and decreased the expressions of vimentin, α-SMA, Snail, and

483

collagen I and III in AEC-II. (D) Astilbin decreased the expressions of vimentin,

484

α-SMA, Snail, and collagen I and III in L929. *p < 0.05, **p < 0.01 vs. the normal

485

group.

486

Fig. 3. Anti-pulmonary fibrosis of astilbin in mice. (A) Astilbin increased FVC

487

compared with the BLM-treated group. (B) Astilbin improved the alveolar structure

488

through H&E staining. (C) Astilbin inhibited the collagen fibers through Masson’s

489

staining. The color blue represents collagen fibers.

490

Fig. 4. Astilbin inhibited the indicators of pulmonary fibrosis in mice. (A) Western

491

blot analysis showed that astilbin promoted E-cadherin expression and inhibited

492

vimentin, α-SMA, Snail, and collagen I and III expressions. (B) Immunofluorescence

493

staining showed that astilbin promoted SP-C expression and inhibited α-SMA

494

expression. The nuclei and SP-C/α-SMA were counterstained with DAPI (blue) and

AC C

EP

TE D

M AN U

SC

RI PT

474

24

ACCEPTED MANUSCRIPT SP-C/α-SMA (green) antibodies.

496

Fig. 5. Heatmap of the expression profiles of the differentially expressed mRNAs

497

in vivo. The colors blue to yellow indicate the low to high expression levels. The

498

column labels at the right represent the differentially expressed mRNAs in the

499

Hedgehog signaling pathway. M represents the BLM-treated group; R represents the

500

astilbin-treated group.

501

Fig. 6. The aberrant signaling pathway of astilbin was tested in vivo and in vitro,

502

and in patients with pulmonary fibrosis. (A) Identification of signaling pathways

503

affected by astilbin in AEC-II cells. The x- and y-axes represent the normalized ratio

504

of firefly/Renilla luciferase activities, respectively. Most significant changes were

505

observed in the Hedgehog signaling pathways. (B) Western blot analysis showed that

506

astilbin

507

Immunofluorescence staining in L929 showed that astilbin blocked Gli1 expression.

508

The nuclei and Gli1 were counterstained with DAPI (blue) and Gli1 (green)

509

antibodies. (D) Western blot analysis showed that astilbin inhibited Gli1 expression in

510

mice. (E) Immunohistochemistry in mice showed that astilbin blocked Gli1

511

expression. (F) Gli1 expression increased in patients with pulmonary fibrosis.

512

Fig. 7. Rescue experiment was used to detect the Hedgehog pathway involved in

513

the antifibrotic mechanism of astilbin with chemical activator smoothened

514

agonist (SAG) and inhibitor vismodegib (GDC-0449) in L929. Astilbin further

515

reduced the expression of α-SMA and collagen I and III which increase or decrease

the

expression

levels

of

Gli1

protein

in

cells.

(C)

AC C

EP

inhibited

TE D

M AN U

SC

RI PT

495

25

ACCEPTED MANUSCRIPT caused by SAG and GDC-0449, respectively.

517

Fig. 8. Analysis of co-expressed lncRNAs with Gli1. (A) Co-expression degrees of

518

lncRNAs with Gli1. The list only showed the top 10 degrees of lncRNAs with Gli1.

519

(B) lncRNA-NUNMM028949.2 expression decreased in the astilbin-treated group

520

compared with the BLM-treated group using RNA sequencing. (C) qRT-PCR was

521

used to validate that lncRNA-NUNMM028949.2 expression was inhibited in the

522

astilbin-treated group compared with the BLM-treated group. *p < 0.05, **p < 0.01.

SC

RI PT

516

M AN U

523

524

EP AC C

526

TE D

525

26

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

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT