MiR-20a-5p suppressed TGF-β1-triggered apoptosis of human bronchial epithelial BEAS-2B cells by targeting STAT3

MiR-20a-5p suppressed TGF-β1-triggered apoptosis of human bronchial epithelial BEAS-2B cells by targeting STAT3

Journal Pre-proof MiR-20a-5p suppressed TGF-β1-triggered apoptosis of human bronchial epithelial BEAS-2B cells by targeting STAT3 Xiuyun Lv, Lihong Wa...

966KB Sizes 0 Downloads 8 Views

Journal Pre-proof MiR-20a-5p suppressed TGF-β1-triggered apoptosis of human bronchial epithelial BEAS-2B cells by targeting STAT3 Xiuyun Lv, Lihong Wang, Tianji Zhu PII:

S0890-8508(19)30489-X

DOI:

https://doi.org/10.1016/j.mcp.2019.101499

Reference:

YMCPR 101499

To appear in:

Molecular and Cellular Probes

Received Date: 16 December 2019 Accepted Date: 24 December 2019

Please cite this article as: Lv X, Wang L, Zhu T, MiR-20a-5p suppressed TGF-β1-triggered apoptosis of human bronchial epithelial BEAS-2B cells by targeting STAT3, Molecular and Cellular Probes (2020), doi: https://doi.org/10.1016/j.mcp.2019.101499. This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. 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. © 2019 Published by Elsevier Ltd.

1

MiR-20a-5p suppressed TGF-β1-triggered apoptosis of human bronchial epithelial

2

BEAS-2B cells by targeting STAT3

3

Xiuyun Lv, Lihong Wang*, Tianji Zhu

4

Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Inner

5

Mongolia Medical University, Hohhot, 010050, P. R. China

6 7

* Corresponding author: Lihong Wang

8

Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Inner

9

Mongolia Medical University, No. 1 Tongdao North Street, Hohhot, 010050, P. R. China

10

Tel & Fax: 0471-3451168

11

E-mail: [email protected]

12 13

Running title: MiR-20a-5p inhibits apoptosis of BEAS-2B cells

14

1

15

Abstract

16

Apoptosis of bronchial epithelial cells contributes to lung diseases, including asthma.

17

Although miR-20a-5p is reportedly downregulated in the bronchial epithelia of asthmatic

18

patients, its function and mechanism still need to be explored. Here, we explored how

19

miR-20a-5p affects human bronchial epithelial cells stimulated with transforming growth

20

factor (TGF)-β1. Using qRT-PCR, we observed downregulated miR-20a-5p levels in these

21

cells. After transfecting miR-20a-5p mimics or inhibitors into human bronchial epithelium

22

BEAS-2B cells, a Cell Counting Kit-8 assay and flow cytometry analysis showed that the

23

mimics mitigated suppression of cell viability and acceleration of apoptosis that was triggered

24

by TGF-β1, whereas the inhibitors exerted the opposite effects. TGF-β1 induced a decrease in

25

expression of Bcl-2 and an increase in expression of Bax, both of which were inhibited by

26

miR-20a-5p mimics and further enhanced by miR-20a-5p inhibitors. Further study verified

27

that miR-20a-5p targeted the signal transducer and activator of transcription 3 (STAT3) and

28

the STAT3 level was inversely related to the miR-20a-5p level. Furthermore, STAT3

29

overexpression partly counteracted the miR-20a-5p-induced anti-apoptotic effect in

30

TGF-β1-treated BEAS-2B cells. In summary, this study suggested that miR-20a-5p restrained

31

apoptosis in TGF-β1-stimulated BEAS-2B cells by targeting STAT3. MiR-20a-5p thus may

32

be a novel therapeutic target for asthma treatment.

33

Key words: miR-20a-5p; asthma; proliferation; apoptosis; BEAS-2B cells; STAT3

34 35

2

36

Introduction

37

Bronchial asthma is a common and frequent respiratory disorder and among the most

38

important chronic respiratory diseases endangering public health worldwide. Airway

39

epithelial cells are essential as a first line of defense against harmful stimuli and external

40

damage in the respiratory tract. These cells therefore serve as the initial link to and

41

pathological basis of many respiratory diseases, including asthma [1]. A series of studies have

42

associated the pathogenesis and clinical manifestations of asthma with changes in the physical

43

and biological barriers of airway epithelial cells [2]. Excessive apoptosis of airway epithelial

44

cells has been observed in severe asthma [3], suggesting an important mechanism leading to

45

airway injury. Therefore, protecting these cells from abnormal apoptosis may prevent the

46

development and progression of asthma.

47

Transforming growth factor (TGF)-β1 participates in cell growth, proliferation, migration,

48

and apoptosis. TGF-β1 promotes apoptosis in podocytes [4], human mesangial cells [5],

49

human gingival epithelial cells [6], and airway epithelial cells [7, 8]. Increased TGF-β1 levels

50

in the lung tissue have been observed in asthmatic rats [9] and in patients with asthma [10].

51

Gagliardo et al. show how TGF-β1 contributes to airway inflammation in childhood asthma

52

[11]. Establishing innovative therapeutic tactics to target TGF-β1-enhanced airway epithelial

53

cell apoptosis thus is vital for treating asthma.

54

Numerous studies have shown that microRNAs (miRNAs) help regulate many biological

55

processes, including cell growth and apoptosis [12]. Because MiRNAs have roles in asthma

56

progression, they may represent potential therapeutic targets for this condition [13-15].

57

Accumulating reports reveal abnormal expression of many miRNAs in the airway epithelial

58

cells of asthmatic patients [16, 17]. MiRNAs also regulate the biological function of bronchial

59

epithelial cells. For example, in a study of patients with severe asthma, Haj-Salem et al. reveal

60

that miR-19a is upregulated in bronchial epithelial cells and that it accelerates cell

61

proliferation by targeting TGFβR2 [18]. Huo et al. show that decreased miR-181b-5p in

62

epithelial cells is associated with inflammation of airway eosinophils in asthma [19].

63

MiR-221 participates in bronchial epithelial cell injury in asthma by targeting SIRT1 [20]. 3

64

MiR-20a-5p is decreased in the bronchial epithelia of patients with severe asthma [21]. Yet,

65

the potential influence of miR-20a-5p in asthma remains unclear. Thus, this study aimed to

66

investigate the effects of miR-20a-5p on TGF-β1-triggered apoptosis of human bronchial

67

epithelial cells and to explore its mechanisms, thus providing a theoretical and experimental

68

basis for effective asthma prevention strategies.

69 70

Materials and methods

71

Cell culture

72

Human embryonic kidney (HEK) 293T cells and human bronchial epithelial BEAS-2B cells

73

(ATCC, Rockville, MD, USA) were grown in Dulbecco's Modified Eagle's medium

74

(Invitrogen, Carlsbad, CA, USA) with 10% fetal bovine serum and maintained in a 5% CO2

75

atmosphere at 37 °C.

76

TGF-β1 treatment

77

Upon 90% confluence, TGF-β1 (10 ng/mL; Sigma, St. Louis, MO, USA) was added to the

78

BEAS-2B cell medium and incubated for 24 hours [8, 22].

79

Transfection

80

Before transfection, BEAS-2B cells were seeded into 96-well plates. The miR-20a-5p mimics,

81

miR-20a-5p inhibitors, and their respective negative controls (GenePharma, Shanghai, China)

82

each were transfected into the BEAS-2B cells using Lipofectamine™ 2000 (Invitrogen). After

83

24 hours, transfection efficacy was assessed using qRT-PCR. To construct the signal

84

transducer and activator of transcription 3 (STAT3) overexpression vectors (pcDNA/STAT3),

85

the pcDNA 3.1 vectors containing the full sequence of STAT3 were synthesized by

86

GenePharma. The pcDNA/STAT3 then was transfected into BEAS-2B cells using

87

Lipofectamine 2000.

88

Cell Counting Kit-8 assay

89

BEAS-2B cells were seeded into 96-well plates (2×103 cells per well) and incubated for 48

90

hours. Then, 10 µL of Cell Counting Kit-8 solution (Beyotime Technology, Jiangsu, China)

91

was added to each well. After 2 hours, absorbance at 450 nm was tested. 4

92

Flow cytometry assay

93

BEAS-2B cells (1×106/mL) were reacted with 5 µL of propidium iodine and 5 µL of

94

FITC-linked Annexin-V, both from BD Biosciences (Bedford, MA, USA). Following 15

95

minutes of incubation in the dark, apoptotic cells were detected using a flow cytometer (BD

96

Biosciences, San Jose, CA, USA).

97

Quantitative real-time PCR

98

The total RNA of BEAS-2B cells was extracted using TRIzol (Invitrogen). For miR-20a-5p

99

quantification, cDNA synthesis was performed using an M-MLV reverse transcriptase kit

100

(Promega, Madison, WI, USA). MiRNA was quantified with SYBR Premix ExTaq (Takara,

101

Dalian,

102

5’-UAAAGUGCUUAUAGUGCAGGUAG-3’

103

5’-CUACCUGCACUAUAAGCACUUUA-3’

104

5’-TGCGGGTGCTCGCTTCGGCAGC-3’ and reverse 5’-CCAGTGCAGGGTCCGAGGT-3’.

105

For STAT3 mRNA quantification, RNA samples were reverse-transcribed into template

106

cDNA and then quantified using a SYBR Green Master Mix Kit (Qiagen, Hilden, Germany).

107

The

108

5’-ATCACGCCTTCTACAGACTGC-3’

109

5’-CATCCTGGAGATTCTCTACCACT-3’

110

5’-CAGGGCTGCTTTTAACTCTGGTAA-3’

111

5’-ACTTGATTTTGGAGGGATCTCGCT-3’. Relative gene expression was calculated using

112

the 2-∆∆Ct method.

113

Western blot

114

Proteins were extracted from BEAS-2B cells and quantified using the BCA method. First, 20

115

µg of protein were separated onto SDS-PAGE gels (Invitrogen), transferred to a

116

polyvinylidene difluoride membrane (Millipore, Schwalbach, Germany), and then blocked in

117

5% fat-free milk. After 1 hour, the membrane was incubated overnight at 4 ºC with the

118

primary antibodies STAT3 (1:1000), Bcl-2 (1:1000), Bax (1:1000), and β-actin (1:2000), all

119

from Abcam (Cambridge, UK), followed by incubation for 1 hour with secondary antibodies

China).

PCR

primer

The

primers

sequence

were

as

follows:

miR-20a-5p

and and

sequences

were

as

reverse U6

follows: and

and

forward

STAT3

forward reverse

GAPDH and

5

forward

forward reverse

120

(1:4000), all from Abcam. Enhanced chemiluminescence (Amersham, Little Chalfont, UK)

121

was used to detect the blots, and Image Quant software was used to quantify the blots.

122

Dual-luciferase reporter assay

123

The wild or mutant sequence of the 3’ untranslated region (UTR) of STAT3 was amplified and

124

cloned into the pGL3 luciferase reporter vector (Promega) to create the wide-type plasmid

125

(Wt STAT3) or mutant-type plasmid (Mut STAT3), respectively. HEK293T cells were plated

126

into 96-well plates, and Wt STAT3 or Mut STAT3 was co-transfected with miR-20a-5p

127

mimics or mimic controls into HEK293 cells using Lipofectamine 2000 (Invitrogen). A

128

dual-luciferase reporter assay kit (Promega) was used to detect luciferase activity after 48

129

hours of transfection.

130

Statistical analysis

131

All statistical analyses were repeated at least three times for each experiment to collect valid

132

data. Data are represented as the mean ± standard deviation and were analyzed using SPSS

133

22.0 software (SPSS Inc., Chicago, IL, USA). Differences were determined via one-way

134

ANOVA or t test. P<0.05 was considered statistically significant.

135 136

Results

137

MiR-20a-5p was downregulated in TGF-β1-treated human bronchial epithelial cells

138

MiR-20a-5p expression was 36% lower in BEAS-2B cells stimulated with TGF-β1, compared

139

to unstimulated cells. (P<0.05; Fig. 1A), implying that miR-20a-5p might have a crucial role

140

in asthma. After transfecting the miR-20a-5p mimics or miR-20a-5p inhibitors into

141

TGF-β1-treated BEAS-2B cells (P<0.05; Fig. 1B), we observed that the mimics significantly

142

inhibited downregulation of miR-20a-5p and that the inhibitors further downregulated

143

miR-20a-5p in these cells (P<0.05; Fig. 1C).

144

MiR-20a-5p mediated anti-apoptotic effects in TGF-β1-stimulated BEAS-2B cells

145

To elucidate the contribution of miR-20a-5p in BEAS-2B cells, cell viability and apoptosis

146

were detected. TGF-β1 significantly decreased cell viability, which was mitigated by the

147

miR-20a-5p mimics and aggravated by the miR-20a-5p inhibitors (P<0.05; Fig. 2A). The 6

148

mimics suppressed apoptosis of TGF-β1-treated BEAS-2B cells, whereas the inhibitors

149

markedly enhanced this effect (P<0.05; Fig. 2B). MiR-20a-5p mimics restrained the

150

TGF-β1-induced reduction in Bcl-2 and increase in Bax in BEAS-2B cells (P<0.05; Fig. 2C),

151

and the miR-20a-5p inhibitors displayed the opposite effects (Fig. 2C). Our data suggested

152

that miR-20a-5p mediated the anti-apoptotic effects in TGF-β1-treated bronchial epithelial

153

cells.

154

MiR-20a-5p targeted the STAT3 gene

155

To identify potential targets of miR-20a-5p for the functional studies in asthma, we performed

156

bioinformatic analyses and found binding sites of miR-20a-5p in the 3’-UTR of STAT3 (Fig.

157

3A). Co-transfection with the Wt 3’-UTR of STAT3 and miR-20a-5p mimics led to a

158

reduction in relative luciferase activity, whereas luciferase activity was unaffected in cells that

159

were co-transfected with the Mut 3’-UTR of STAT3 and miR-20a-5p mimics (Fig. 3B).

160

Moreover, the miR-20a-5p mimics downregulated STAT3 expression, whereas transfection

161

with the miR-20a-5p inhibitors upregulated STAT3 expression in BEAS-2B cells under

162

TGF-β1 exposure (Fig. 3C and 3D). As shown in Fig. 3E, miR-20a-5p negatively regulated

163

STAT3 protein expression in these cells. Together, these data suggest that miR-20a-5p directly

164

modulated STAT3 expression in BEAS-2B cells under TGF-β1 stimulation.

165

STAT3 was essential for miR-20a-5p to modulate apoptosis in TGF-β1-treated BEAS-2B

166

cells

167

To assess whether miR-20a-5p derives its function in TGF-β1-induced apoptosis via STAT3,

168

BEAS-2B cells were transfected with the STAT3 overexpression vector and either the

169

miR-20a-5p mimics or mimic control. Notably, transfection with the STAT3 overexpression

170

vector alleviated the downregulation of STAT3 that was induced by the miR-20a-5p mimics

171

(P<0.05, Fig. 4A). STAT3 overexpression also markedly reversed the function of the

172

miR-20a-5p mimics on cell viability (Fig. 4B) and apoptosis (Fig. 4C) in TGF-β1-treated

173

BEAS-2B cells (P<0.05). Similarly, STAT3 overexpression also counteracted the levels of

174

Bax and Bcl-2 that were regulated by the miR-20a-5p mimics (P<0.05; Fig. 4D), indicating

7

175

that miR-20a-5p regulated the biofunction of TGF-β1-treated BEAS-2B cells by targeting

176

STAT3.

177 178

Discussion

179

In this study, we found downregulation of miR-20a-5p in the bronchial epithelial cells of

180

patients with asthma and in TGF-β1-stimulated BEAS-2B cells. MiR-20a-5p increased cell

181

viability and decreased apoptosis in these cells. Subsequently, our study corroborated that

182

miR-20a-5p directly regulated its target gene, STAT3. As a recent study has revealed that

183

hsa-miR-20a-5p attenuates allergic inflammation [23], it is worth investigating miR-20a-5p as

184

a novel gene target for asthma treatment.

185

Asthma is a common chronic illness of the respiratory system. Apoptosis of bronchial

186

epithelial cells is a main cause of airway epithelial injury in asthma [24, 25]. Abundant

187

evidence implicates miRNAs, which are epigenetic regulators, in the pathophysiology of

188

asthma [15, 26, 27]. Thus, targeting specific miRNAs may have therapeutic potential in

189

asthma treatment [14].

190

Abnormal changes in miRNA expression have been found in airway epithelial cells of

191

asthmatic patients [16]. Downregulated miR-20a-5p occurs in the bronchial epithelia of

192

patients with severe asthma [21]. TGF-β1, a pro-apoptosis factor, can induce apoptosis in

193

human bronchial epithelial cells [7]. Our study analyzed miR-20a-5p expression in

194

TGF-β1-stimulated BEAS-2B cells and found that it was markedly decreased, implying a

195

crucial role of miR-20a-5p in asthma. Moreover, TGF-β1 triggered apoptosis in BEAS-2B

196

cells, which is consistent with Sun et al. [28].

197

The Bcl-2 gene family has an essential role in the signal transduction pathway of

198

apoptosis, among which Bcl-2 and Bax are the most representative genes that inhibit and

199

promote apoptosis in airway epithelial cells [29, 30]. Our study showed that miR-20a-5p

200

overexpression increased cell viability and suppressed TGF-β1-triggered apoptosis in

201

BEAS-2B cells, accompanied by upregulation of Bcl-2 and reductions in Bax. MiR-20a-5p

202

inhibition showed the opposite effects. 8

203

To understand the molecular mechanism responsible for the effects of miR-20a-5p in

204

TGF-β1-triggered apoptosis, bioinformatic analyses were conducted that identified STAT3 as

205

a potential target of miR-20a-5p. Consistent with this prediction, miR-20a-5p was confirmed

206

to directly target the STAT3 3’-UTR. STAT3 participates in various cellular processes, such as

207

growth and apoptosis [31, 32]. STAT3 also is reported to be a crucial regulator in the

208

development and pathogenesis of asthma. STAT3 expression in airway epithelia contributes to

209

allergic inflammation in asthmatic mice [33]. Gavino et al. reveal that STAT3 repression

210

decreases lung inflammation, suppresses airway remodeling, and reduces Th2/Th17-type cell

211

accumulation in a mouse model of asthma [34].

212

In this research, the expression of STAT3 was elevated in BEAS-2B cells after TGF-β1

213

stimulation, and miR-20a-5p negatively regulated its expression. To verify whether

214

miR-20a-5p exerts its effects in BEAS-2B cells by targeting STAT3, a STAT3 overexpression

215

vector was transfected into cells. STAT3 overexpression markedly abrogated the regulatory

216

function of miR-20a-5p on TGF-β1-triggered apoptosis in BEAS-2B cells. These data suggest

217

that miR-20a-5p inhibited TGF-β1-induced apoptosis by downregulating STAT3.

218

Our data confirmed miR-20a-5p as an important regulator of TGF-β1-initiated apoptosis

219

in human bronchial epithelial cells. Overexpression of miR-20a-5p markedly restrained

220

TGF-β1-triggered apoptosis, downregulated Bax levels, and upregulated Bcl-2 levels.

221

Furthermore, miR-20a-5p alleviated TGF-β1-triggered apoptosis in BEAS-2B cells by

222

targeting STAT3. Limitations of our study included the use of only one cell line and the use of

223

an in vitro experiment to explore the role of miR-20a-5p. It is necessary to investigate these

224

effects in an in vivo asthma model to support our conclusions. Nevertheless, our results

225

indicate that miR-20a-5p might be a novel target for treating asthma.

226

9

227

Author’s Contributions

228

Xiuyun Lv and Lihong Wang disigned the study; Xiuyun Lv conducted experiments and wrote

229

the manuscript; Tianji Zhu analyzed experiments results; Lihong Wang revised the paper. All

230

authors contributed to and approved the final version of the manuscript.

231

Funding

232

This work was supported by the Natural science foundation of Inner Mongolia autonomous

233

region (grant numbers 2014MS08114).

234

Acknowledgments

235

None.

236

Declaration of interests

237

None.

10

238

References

239

[1] J.V. Fahy, R.M. Locksley, The airway epithelium as a regulator of Th2 responses in

240

asthma, American journal of respiratory and critical care medicine 184(4) (2011) 390-2.

241

[2] S.T. Holgate, The sentinel role of the airway epithelium in asthma pathogenesis,

242

Immunological reviews 242(1) (2011) 205-19.

243

[3] A. Trautmann, K. Kruger, M. Akdis, D. Muller-Wening, A. Akkaya, E.B. Brocker, K.

244

Blaser, C.A. Akdis, Apoptosis and loss of adhesion of bronchial epithelial cells in asthma,

245

International archives of allergy and immunology 138(2) (2005) 142-50.

246

[4] R. Das, S. Xu, T.T. Nguyen, X. Quan, S.K. Choi, S.J. Kim, E.Y. Lee, S.K. Cha, K.S. Park,

247

Transforming Growth Factor beta1-induced Apoptosis in Podocytes via the Extracellular

248

Signal-regulated Kinase-Mammalian Target of Rapamycin Complex 1-NADPH Oxidase 4

249

Axis, The Journal of biological chemistry 290(52) (2015) 30830-42.

250

[5] D.P. Jardim, P.C.E. Poco, A.H. Campos, Dact1, a Wnt-Pathway Inhibitor, Mediates

251

Human Mesangial Cell TGF-beta1-Induced Apoptosis, Journal of cellular physiology 232(8)

252

(2017) 2104-2111.

253

[6] T. Yoshimoto, T. Fujita, M. Kajiya, S. Matsuda, K. Ouhara, H. Shiba, H. Kurihara,

254

Involvement of smad2 and Erk/Akt cascade in TGF-beta1-induced apoptosis in human

255

gingival epithelial cells, Cytokine 75(1) (2015) 165-73.

256

[7] X. Jiang, Silencing of heart and neural crest derivatives expressed transcript 2 attenuates

257

transforming growth factor-beta1-enhanced apoptosis of human bronchial epithelial cells,

258

Oncology letters 16(4) (2018) 4997-5005.

259

[8] G. Cheng, Z. Shao, B. Chaudhari, D.K. Agrawal, Involvement of chloride channels in

260

TGF-beta1-induced apoptosis of human bronchial epithelial cells, American journal of

261

physiology. Lung cellular and molecular physiology 293(5) (2007) L1339-47.

262

[9] Y. Feng, C. Yang, W. Yang, T. Jiang, Effect of Dexamethasone on TGF-beta1/Smad3

263

Signalling Pathway in Airway Remodelling Model of Asthmatic Rats, Journal of the College

264

of Physicians and Surgeons--Pakistan : JCPSP 29(6) (2019) 537-540.

265

[10] M. Panek, T. Pietras, A. Fabijan, J. Ziolo, L. Wieteska, B. Malachowska, W. Fendler, J. 11

266

Szemraj, P. Kuna, Identification and association of the single nucleotide polymorphisms,

267

C-509T, C+466T and T+869C, of the TGF-beta1 gene in patients with asthma and their

268

influence on the mRNA expression level of TGF-beta1, International journal of molecular

269

medicine 34(4) (2014) 975-86.

270

[11] R. Gagliardo, P. Chanez, M. Gjomarkaj, S. La Grutta, A. Bonanno, A.M. Montalbano, C.

271

Di Sano, G.D. Albano, D. Gras, G. Anzalone, L. Riccobono, M. Profita, The role of

272

transforming growth factor-beta1 in airway inflammation of childhood asthma, International

273

journal of immunopathology and pharmacology 26(3) (2013) 725-38.

274

[12] H.O. Iwakawa, Y. Tomari, The Functions of MicroRNAs: mRNA Decay and

275

Translational Repression, Trends in cell biology 25(11) (2015) 651-665.

276

[13] O.A. Svitich, V.V. Sobolev, L.V. Gankovskaya, P.V. Zhigalkina, V.V. Zverev, The role of

277

regulatory RNAs (miRNAs) in asthma, Allergologia et immunopathologia 46(2) (2018)

278

201-205.

279

[14] C.M. Greene, K.P. Gaughan, microRNAs in asthma: potential therapeutic targets, Current

280

opinion in pulmonary medicine 19(1) (2013) 66-72.

281

[15] M.M. Perry, I.M. Adcock, K.F. Chung, Role of microRNAs in allergic asthma: present

282

and future, Current opinion in allergy and clinical immunology 15(2) (2015) 156-62.

283

[16] O.D. Solberg, E.J. Ostrin, M.I. Love, J.C. Peng, N.R. Bhakta, L. Hou, C. Nguyen, M.

284

Solon, C. Nguyen, A.J. Barczak, L.T. Zlock, D.P. Blagev, W.E. Finkbeiner, K.M. Ansel, J.R.

285

Arron, D.J. Erle, P.G. Woodruff, Airway epithelial miRNA expression is altered in asthma,

286

American journal of respiratory and critical care medicine 186(10) (2012) 965-74.

287

[17] R.T. Martinez-Nunez, V.P. Bondanese, F. Louafi, A.S. Francisco-Garcia, H. Rupani, N.

288

Bedke, S. Holgate, P.H. Howarth, D.E. Davies, T. Sanchez-Elsner, A microRNA network

289

dysregulated in asthma controls IL-6 production in bronchial epithelial cells, PloS one 9(10)

290

(2014) e111659.

291

[18] I. Haj-Salem, R. Fakhfakh, J.C. Berube, E. Jacques, S. Plante, M.J. Simard, Y. Bosse, J.

292

Chakir, MicroRNA-19a enhances proliferation of bronchial epithelial cells by targeting

293

TGFbetaR2 gene in severe asthma, Allergy 70(2) (2015) 212-9. 12

294

[19] X. Huo, K. Zhang, L. Yi, Y. Mo, Y. Liang, J. Zhao, Z. Zhang, Y. Xu, G. Zhen, Decreased

295

epithelial and plasma miR-181b-5p expression associates with airway eosinophilic

296

inflammation in asthma, Clinical and experimental allergy : journal of the British Society for

297

Allergy and Clinical Immunology 46(10) (2016) 1281-90.

298

[20] H. Zhang, Y. Sun, W. Rong, L. Fan, Y. Cai, Q. Qu, Y. Gao, H. Zhao, miR-221 participates

299

in the airway epithelial cells injury in asthma via targeting SIRT1, Experimental lung research

300

44(6) (2018) 272-279.

301

[21] R.T. Martinez-Nunez, H. Rupani, M. Plate, M. Niranjan, R.C. Chambers, P.H. Howarth,

302

T. Sanchez-Elsner, Genome-Wide Posttranscriptional Dysregulation by MicroRNAs in

303

Human Asthma as Revealed by Frac-seq, Journal of immunology 201(1) (2018) 251-263.

304

[22] Y. Itoigawa, N. Harada, S. Harada, Y. Katsura, F. Makino, J. Ito, F. Nurwidya, M. Kato, F.

305

Takahashi,

306

epithelial-mesenchymal transition in human bronchial epithelial cells, Respiratory research 16

307

(2015) 48.

308

[23] Y. Lu, Z. Li, B. Xie, Y. Song, X. Ye, P. Liu, hsa-miR-20a-5p attenuates allergic

309

inflammation in HMC-1 cells by targeting HDAC4, Molecular immunology 107 (2019)

310

84-90.

311

[24] S.R. White, D.R. Dorscheid, Corticosteroid-induced apoptosis of airway epithelium: a

312

potential mechanism for chronic airway epithelial damage in asthma, Chest 122(6 Suppl)

313

(2002) 278S-284S.

314

[25] C. Zhou, G. Yin, J. Liu, X. Liu, S. Zhao, Epithelial apoptosis and loss in airways of

315

children with asthma, The Journal of asthma : official journal of the Association for the Care

316

of Asthma 48(4) (2011) 358-65.

317

[26] W. Kai, X.U. Qian, W.U. Qun, MicroRNAs and Asthma Regulation, Iranian journal of

318

allergy, asthma, and immunology 14(2) (2015) 120-5.

319

[27] K. Maneechotesuwan, Role of microRNA in severe asthma, Respiratory investigation

320

57(1) (2019) 9-19.

321

[28] X. Sun, W. Zhang, Silencing of Gal-7 inhibits TGF-beta1-induced apoptosis of human

R.

Atsuta,

K.

Takahashi,

13

TWEAK

enhances

TGF-beta-induced

322

airway epithelial cells through JNK signaling pathway, Experimental cell research 375(2)

323

(2019) 100-105.

324

[29] B. Antonsson, Mitochondria and the Bcl-2 family proteins in apoptosis signaling

325

pathways, Molecular and cellular biochemistry 256-257(1-2) (2004) 141-55.

326

[30] C.H. Lin, Y.C. Hong, S.H. Kao, Aeroallergen Der p 2 induces apoptosis of bronchial

327

epithelial BEAS-2B cells via activation of both intrinsic and extrinsic pathway, Cell &

328

bioscience 5 (2015) 71.

329

[31] F. He, H. Liu, J. Guo, D. Yang, Y. Yu, J. Yu, X. Yan, J. Hu, Z. Du, Inhibition of

330

MicroRNA-124 Reduces Cardiomyocyte Apoptosis Following Myocardial Infarction via

331

Targeting STAT3, Cellular physiology and biochemistry : international journal of

332

experimental cellular physiology, biochemistry, and pharmacology 51(1) (2018) 186-200.

333

[32] N. Fathi, G. Rashidi, A. Khodadadi, S. Shahi, S. Sharifi, STAT3 and apoptosis challenges

334

in cancer, International journal of biological macromolecules 117 (2018) 993-1001.

335

[33] M.C. Simeone-Penney, M. Severgnini, P. Tu, R.J. Homer, T.J. Mariani, L. Cohn, A.R.

336

Simon, Airway epithelial STAT3 is required for allergic inflammation in a murine model of

337

asthma, Journal of immunology 178(10) (2007) 6191-9.

338

[34] A.C. Gavino, K. Nahmod, U. Bharadwaj, G. Makedonas, D.J. Tweardy, STAT3 inhibition

339

prevents lung inflammation, remodeling, and accumulation of Th2 and Th17 cells in a murine

340

asthma model, Allergy 71(12) (2016) 1684-1692.

341 342

14

343

Figure legends

344

Figure 1. MiR-20a-5p level in TGF-β1-stimulated BEAS-2B cells. (A) MiR-20a-5p level in

345

BEAS-2B cells after TGF-β1 exposure. After exposure to TGF-β1 (10 ng/ml) for 24 hours, the

346

miR-20a-5p level in BEAS-2B cells was detected. N=3, #P<0.05 vs. control. (B) MiR-20a-5p

347

expression after transfecting BEAS-2B cells with miR-20a-5p mimics or miR-20a-5p

348

inhibitors. N=3, #P<0.05 vs. control. (C) Expression of miR-20a-5p in TGF-β1-stimulated

349

BEAS-2B cells after transfection with miR-20a-5p mimics or miR-20a-5p inhibitors. N=3,

350

#

351

Figure 2. MiR-20a-5p inhibited TGF-β1-induced apoptosis of BEAS-2B cells. (A) Cell

352

viability was measured using a Cell Counting Kit-8 assay. (B) Apoptosis was assessed using

353

flow cytometry. (C) The protein levels of Bax and Bcl-2 were detected using western blot.

354

N=3, #P<0.05 vs. control; *P<0.05 vs. TGF-β1.

355

Figure 3. MiR-20a-5p directly targeted the 3’ untranslated region (UTR) of STAT3. (A)

356

Predicted interaction between miR-20a-5p and the STAT3 3’-UTR. (B) A dual-luciferase

357

reporter assay identified STAT3 as a direct target of miR-20a-5p. #P<0.05 vs. mimic control.

358

(C) STAT3 relative mRNA level. N=3, #P<0.05 vs. control. (D) STAT3 relative protein level.

359

N=3, #P<0.05 vs. control. (E) Effect of miR-20a-5p on the protein level of STAT3 in

360

TGF-β1-induced BEAS-2B cells. N=3, #P<0.05 vs. control; *P<0.05 vs. TGF-β1.

361

Figure 4. Restoration of STAT3 expression in BEAS-2B cells counteracted the repression

362

of miR-20a-5p on TGF-β1-induced apoptosis. (A) Relative protein level of STAT3. Cells

363

were co-transfected with a STAT3 overexpression vector and miR-20a-5p mimics. (B) Cell

364

viability in TGF-β1-treated cells. (C) Apoptosis in TGF-β1-treated cells. (D) The protein levels

365

of Bax and Bcl-2. N=3, #P<0.05 vs. TGF-β1; *P<0.05 vs. TGF-β1+miR-20a-5p mimics.

P<0.05 vs. control; *P<0.05 vs. TGF-β1.

366 367

15

Highlights 1.MiR-20a-5p is downregulated in in TGF-β1-treated BEAS-2B cells 2.MiR-20a-5p mediates anti-apoptotic effects in BEAS-2B cells 3.MiR-20a-5p targetes STAT3 to modulate apoptosis