A signal-amplification circuit between miR-218 and Wnt/β-catenin signal promotes human adipose...

8

Click here to load reader

Transcript of A signal-amplification circuit between miR-218 and Wnt/β-catenin signal promotes human adipose...

Page 1: A signal-amplification circuit between miR-218 and Wnt/β-catenin signal promotes human adipose tissue-derived stem cells osteogenic differentiation

1

2

3

4

5Q1

678

9

101112131415161718192021222324252627

47

48

49

50

51

52

53

54

55

56

57

58

Bone xxx (2013) xxx–xxx

BON-10157; No. of pages: 8; 4C:

Contents lists available at ScienceDirect

Bone

j ourna l homepage: www.e lsev ie r .com/ locate /bone

Original Full Length Article

A signal-amplification circuit between miR-218 and Wnt/β-cateninsignal promotes human adipose tissue-derived stem cellsosteogenic differentiation

OO

F

Wei-Bing Zhang a,b,⁎, Wei-Jie Zhong c, Lin Wang a,b

a Institute of Stomatology, Nanjing Medical University, Nanjing, Chinab Department of Orthodontics, School of Stomatology, Nanjing Medical University, Nanjing, Chinac Department of Orthodontics, Shanghai Stomatological Disease Center, Shanghai, China

U

Abbreviations:miRNA, microRNA; UTR, untranslated rqRT-PCR, quantitative reverse transcription-polymerasemarrow-derived mesenchymal stem cells; hASCs, humOCN, osteocalcin; BSP, bone sialoprotein; ALP, alkaline phtranscription factor 2; SFRP2, secreted frizzled related proprotein 2.⁎ Corresponding author at: Institute of Stomatology

Nanjing, China.E-mail address: [email protected] (W.-B. Zh

8756-3282/$ – see front matter © 2013 Published by Elsehttp://dx.doi.org/10.1016/j.bone.2013.09.015

Please cite this article as: Zhang W-B, et al, Atissue-derived stem cells osteogenic differen

Ra b s t r a c t

a r t i c l e i n f o

28

29

30

31

32

33

34

35

36

37

38

39

40

Article history:Received 18 May 2013Revised 19 September 2013Accepted 23 September 2013Available online xxxx

Edited by: David Fyhrie

Keywords:MicroRNAHuman adipose-derived mesenchymal stemcellsOsteogenic differentiationmiR-218Wnt

41

42

43

44

ECTED PHuman adipose-derived stem cells (hASCs) have become a highly attractive source of seed cells in bone regener-

ative. It has become a key issue how to effectively improve osteogenic differentiation of hASCs in the bone tissueengineering.Numerous regulatory pathways dominate osteogenic differentiation of hASCs involve transcriptional factors andsignalingmolecules. However, how these factors combinewith each other to regulate hASCs osteogenic differen-tiation still remain to be illustrated. The identification of microRNAs will illuminate this and might permit finelytuning the osteogenic differentiation process. Here, we present evidence thatmiR-218 acts as a positive regulatorof hASCs osteogenesis. Real-time PCR shows that miR-218 was up-regulated during osteogenic differentiation.Overexpression of exogenous miR-218 enhanced osteogenic differentiation in vitro, whereas inhibition of miR-218 would suppress osteogenic differentiation. Furthermore, miR-218 directly targeted SFRP2 and DKK2,which is a WNT signaling pathway antagonist, and enhanced Wnt/β-catenin signaling activity. Finally, wefound that mimicking Wnt/β-catenin signal strengthened the expression level of miR-218, while blocking thesignal attenuated the expression level of miR-218. This feed-forward regulatory circuit provides additional in-sight into howmiRNAs acting as a signal amplifier interact with signal molecules during hASCs osteogenic differ-entiation. Taken together, we have established a regulatory networkwith a central role for themiR-218 in hASCsosteogenic differentiation.

© 2013 Published by Elsevier Inc.

4546

R

59

60

61

62

63

64

65

66

67

68

69

NCO

RIntroduction

Human adipose-derived stem cells (hASCs) can be isolated fromadipose tissues that supply an abundant and easily accessible pool ofstem cells [1,2]. hASCs are capable of differentiating into osteogenic line-ages and have become a highly attractive source of seed cells in skeletaltissue engineering [3–5]. It has become a key issue how to effectively im-prove osteogenic differentiation of hASCs in the bone tissue engineering.

MicroRNAs (miRNAs) refer to endogenous 22-nucleotide non-codingsingle-stranded RNA molecules that suppress their targets gene expres-sion by inhibiting translation, promoting mRNA decay [6,7]. miRNAs

70

71

72

73

74

75

76

77

78

79

egion; mRNA, messenger RNA;chain reaction; BMSCs, bone

an adipose-derived stem cells;osphatase; Runx2, runt-relatedtein 2; DKK2, dickkopf-related

, Nanjing Medical University,

ang).

vier Inc.

signal-amplification circuit btiation, Bone (2013), http://d

are involved in a broad spectrum of biologic processes, including cell dif-ferentiation, tissue development, and growth [7–9].

Recent evidence has shown that some specific miRNAs enhance orinhibit osteogenic differentiation of hASCs by targeting some inhibitorsor activators of signaling cascades [10,11]. miR-196a enhances osteo-genic differentiation of hASCs through targeting HOXC8 [10]. miR-26ainhibits osteogenic differentiation in human adipose tissue stromalcells by targeting SMAD1 [11]. However, the regulatory mechanism be-tween miRNA and osteogenic differentiation of hASCs is more similarto a network than to a linear cascade. It is still unclear howmiRNAs or-chestrate their regulation of cellular signaling and networks during os-teogenic differentiation of hASCs. The elucidation of the molecularmechanisms governing hASC osteogenic differentiation will facilitatestem cell-mediated bone regeneration therapy.

miR-218 has been found as an enhancer of bone marrow-derivedmesenchymal stem cells (BMSCs) osteogenic differentiation [12].BMSCs and ASCs possess many similar biological characteristics; how-ever, there are some differences in their differentiation potential,transcriptome and proteome [13]. Thus it is necessary to investigatethe role of miR-218 during hASC osteogenic differentiation. We foundthatmiR-218 promoted hASC osteogenic differentiation. Overexpression

etween miR-218 and Wnt/β-catenin signal promotes human adiposex.doi.org/10.1016/j.bone.2013.09.015

Page 2: A signal-amplification circuit between miR-218 and Wnt/β-catenin signal promotes human adipose tissue-derived stem cells osteogenic differentiation

T

80

81

82

83

84

85

86

87

88

89

90

91

92

93

94

95

96

97

98

99

100

101

102

103

104

105

106

107

108

109

110

111

112

113

114

115

116

117

118

119

120

121

122

123

124

125

126

127

128

129

130Q4

131

132

133

134

135

136

137

138

139

140

141

142

143

144

145

146

147

148

149

150

151

152

153

154

155

156

157

158

159

160

161

162

163

164

165

166

167

168

169

170

171

172

173

174

175

Table 1 t1:1

t1:2Primers used for qPCR Q3.

t1:3Gene name Real time qPCR primers (5′–3′)

t1:4Runx2 Forward: GTC TCA CTG CCT CTC ACT TGt1:5Reverse: CAC ACA TCT CCT CCC TTC TGt1:6ALP Forward: CGG ACA TCA TGA GGG TAA GGt1:7Reverse: GAG ACA TTT TCC CGT TCA CCt1:8OCN Forward: ACA GAC AAG TCC CAC ACA GCA GCt1:9Reverse: TGA AGG CTT TGT CAG ACT CAG GGCt1:10BSP Forward: GCCAGAGGAGCAATCACCAAt1:11Reverse: CAGGCTGGAGGTTCACTGGTt1:12SFRP2 Forward: CGT GGG CTC TTC CTC TTC Gt1:13Reverse: ATG TTC TGG TAC TCG ATG CCGt1:14Lef-1 Forward: AATAAAGTCCCGTGGTGCt1:15Reverse: ATGGGTAGGGTTGCCTGAATCt1:16Tcf-4 Forward: GCCTCTCATCACGTACAGCAt1:17Reverse: GGATGGGGGATTTGTCCTACt1:18miR-218 TTG TGC T TG ATC TAA CCA TGTt1:19U6 Forward: CGC TTC GGC AGC ACA TAT ACt1:20Reverse: AAA ATA TGG AAC GCT TCA CGA

2 W.-B. Zhang et al. / Bone xxx (2013) xxx–xxx

UNCO

RREC

of miR-218 significantly increased hASCs osteogenic, whereas inhibitingmiR-218 had the opposite effects. Furthermore, we found that duringhASC osteogenic differentiation miR-218 directly targets 3′-UTR of theSFRP2 and DKK2 gene, which is WNT signaling pathway antagonist[14,15]. Upregulation of miR-218 decreased the expression of DKK2and SFRP2 thus activatedWnt signaling, then promoted hASC osteogenicdifferentiation; whereas inhibiting miR218 suppressed hASC osteogenicdifferentiation. MimickingWnt/β-catenin signal strengthens the expres-sion level of miR-218, while blocking the signal weakens the expressionlevel ofmiR-218. Our studies have shown a complex signal-amplificationregulatory circuit betweenmiR-218 andWnt/β-catenin signaling duringhASC osteogenic differentiation.

Materials and methods

Cell culture

All protocols involving human and animal subjects were approvedby the Ethics Committee of School of Stomatology NanjingMedical Uni-versity. Adipose tissueswere obtained frompatients undergoing tumes-cent. Bonemarrow stromal cells were isolated by flushingmarrow fromthe femurs and tibia of 6–8-weeks-old C57/BL mice. BMSCs were cul-tured as a positive control. The hASCs were isolated according to themethods as previously described [16]. To further establish that Wntsignaling could functionally affect endogenous miR-218, we examinedexpression levels of miR-218 in three biological conditions, i.e., Wnt3a,SFRP2 and DKK2 stimulated hASCs. Confluent hASCs cells were treatedwith 100 ng/ml SFRP2 (R&D), 100 ng/ml DKK2 (R&D) or 50 ng/mlWnt3a (R&D) in osteogenic medium.We used five different hASC sam-ples for each experiment and performed the duplicate experiment pereach sample.

Induction of differentiation

BMSCs or ASCswere plated at 3 × 105 cells/cm2 and cultured in com-plete culture media for 2–3 days in 6-well culture dishes. Osteogenic dif-ferentiation was induced via the culturing of the cells in osteogenicmedium (20% FBS, 0.1 μM dexamethasone, 10 mM β-glycerophosphate,and 50 μM ascorbic acid in α-MEM); the osteoblast phenotype wasevaluated by determining alkaline phosphatase (ALP) activity. The ALPactivity of hASCs was detected with a commercially available assay kit(Sigma).

Viral vector construction

Control (PMIRH000PA-1), miR-218 (PMIRH218-2PA-1) and anti-miRLenti virus (miR-Zip-218 antimiR, MZIP218-PA-1) (mature miR218sequence: 5′-TTG TGC T TG ATC TAA CCA TGT; anti miR-218: 5′-AACCACATGGTTAGATCAAGCACAA-3′; control: 5′-CCTCTTACCTCAGTTACAATTTATA-3′)were purchased from SystemBioscience. Lenti virus plasmidswere transfected by using Fugene (Promega). We used virus titers rang-ing from 5 × 105 up to 1 × 107 transducing units (TU)/ml.

miRNA extraction and qPCR analysis

Total RNA andmiRNAwere purified fromhASCs cell cultures treatedwith or without 50 μg/ml of ascorbic acid for d0, d2, d10 and d21daysusing miRNeasy mini kit (Qiagen).

Total RNA (0.5–1 μg) was reverse transcribed into cDNA using Oligo(dT) primers and was analyzed by real time qPCR. SYBR Green MasterMix (Applied Bio Systems Inc.) was used to detect the expression ofgene markers. For miRNA detection, the isolation of small species-enriched RNA was performed as per the manufacturer's instructions(mirVana miRNA isolation kit, Ambion). miRNA was reverse-transcribedwith an Ncode miRNA first-strand cDNA synthesis kit (Invitrogen)according to the manufacturer-specified guidelines. Forward primer

Please cite this article as: Zhang W-B, et al, A signal-amplification circuit btissue-derived stem cells osteogenic differentiation, Bone (2013), http://d

ED P

RO

OF

sequences were designed as the corresponding mature miRNA se-quences, and U6 was used as a normalizing control. qPCR wasperformed with the Light Cycler assay, using a fluorogenic SYBR GreenI reaction mixture for PCR carried out in a Light Cycler Instrument(Roche). Data were corrected for GAPDH expression and expressed aslog (2−ΔΔCt). Primers were synthesized from Invitrogen Inc. and se-quences are listed in Table 1.

Western blot analysis

Confluent hASCs were treated under the appropriate conditions andlysed. The proteins were loaded on 10% SDS polyacrylamide gels, trans-ferred to PVDFmembrane, and probed with primary antibody for Sfrp2(Abcam), DKK2 (Abcam), actin (Santacruz), β-catenin (cell signaling),active-β-catenin (Millipore), SP1(Millipore), Axin2 (Abcam), Runx2(R&D), cyclinD1 (Abcam) and β-actin (Santacruz),. Immunoreactivebands were detected with secondary antibodies and visualized via en-hanced chemiluminescence (Amersham Pharmacia Biotech).

Nuclear and cytoplasmic fractionation

For preparation of nuclear protein, hASCs were collected andwashed twice with cold PBS. Nuclear and cytoplasmic protein wasprepared using NE-PER Nuclear and Cytoplasmic Extraction Reagents,according to the protocols specified by the manufacturer (Pierce,Rockford, IL).

3′UTR reporter construction and reporter gene assay

The predicted consequential pairing of the target region of SFRP2,DKK2 and miR-218 sequences are in Table 2. The 3′UTRs of the SFRP2or DKK2 genes were amplified and cloned into the SacI/HindIII sites ofthe pMIR-Report luciferase vector. The seed region of the miR-218 tar-get sites in the DKK2 and SFRP2 3′UTRs were mutated using theQuickchange II site directedmutagenesis kit (Stratagene). The Luciferaseassay was conducted by co-transfecting miR-218 miRNA, NS miR andanti-miR-218 with WT and mutant pMIR-REPORT-Luc DNA construct.The transfections were performed in duplicate and all experimentswere repeated several times. Luciferase expression was normalized toβ-galactosidase activity in all cases.

Statistics

Data are expressed asmean ± SD. Student's t-test or one-way anal-ysis of variance (ANOVA) was made for multiple comparisons. All ex-periments were repeated 3 times, and representative experiments areshown. P-values b 0.05 will be considered to be statistically significant.

etween miR-218 and Wnt/β-catenin signal promotes human adiposex.doi.org/10.1016/j.bone.2013.09.015

Page 3: A signal-amplification circuit between miR-218 and Wnt/β-catenin signal promotes human adipose tissue-derived stem cells osteogenic differentiation

176

177

178

179

180

181

182

183

184

185

186

187

188

189

190

191

192

193

194

195

196

197

198

199Q5

200

201

202

203

204

Table 2t2:1

t2:2 Predicted 3′-UTR binding sites by TargetScan Human.

t2:3 Putative Target Predicted consequential pairing of target region (top) andmiRNA (bottom)

t2:4 SFRP2 SFRP2 3′UTR 5′ …ACUUCCAUUUAUUAGAAGCACAU…t2:5 hsa-miR-218 3′ UGUACCAAUCUAGUUCGUGUUt2:6 DKK2 DKK2 3′UTR 5′ …UAUUAUUAAGAGAACAAGCACAC.t2:7 hsa-miR-218 3′ UGUACCAAUCUAGUUCGUGUU

3W.-B. Zhang et al. / Bone xxx (2013) xxx–xxx

Result

Expression level of miR218 and its target genes (SFRP2 and DKK2) duringosteogenic differentiation of hASCs

To better understand the role of miR-218 in regulating the differen-tiation potential of hASCs, we investigated its expression profile duringshort- and long-term osteoblastic induction of hASCs by qPCR. Afterstandard osteoblastic induction, we determined miR-218 expressionduring the osteogenic differentiation of hASCs by qPCR. From thisstudy, we found that miR-218 expression was gradually increasedafter the induction of osteogenic differentiation of hASCs (Fig. 1A).

UNCO

RRECT

Fig. 1. Expression level of miR218 and target genes (SFRP2 and DKK2) during osteogenic differetime PCR analysis of SFRP2 normalized to GAPDH during osteogenic differentiation of hASCs. C.hASCs. D. The expression of miR-218 in hASCs transfectedwith miR 218 was assayed by real-timoverexpressing miR 218. F. Real-time PCR analysis of DKK2 normalized to GAPDH by overexprassayed by real-time and normalized to U6. H. The expression of SFRP2 in hASCs transfected wmean ± SD of 3 independent experiments. **P b 0.01.

Please cite this article as: Zhang W-B, et al, A signal-amplification circuit btissue-derived stem cells osteogenic differentiation, Bone (2013), http://d

We induced hASCs to differentiate into osteoblasts by pre- or anti-miR-218. Transduced hASC were grown to confluence and osteogenicdifferentiation was induced in differentiation media.

qPCR analysis confirmed that endogenous miR-218 level was in-creased more than 300-fold compared to the control cells after twoday transfection, and remained 300-, 150-fold of the controls in 10and 21 days after pre-miR-218 infected, respectively (Fig. 1D). Thetransfected hASCs with anti-miR-218 had 4-, 3-, and 2.5-fold decreaseof miR-218 expression at 2, 10 and 21 days respectively after transfec-tion (Fig. 1G).

When miR-218 was up-regulated or down-regulated during osteo-genic differentiation, the expression levels of its targets – SFRP2, andDKK2 – also were concomitantly down-regulated or up-regulated(Figs. 1B, C, E, F, H, I).

OO

F

miR-218 promotes osteogenic differentiation of hASCs

To elucidate the impact of miR-218 on hASCs osteogenic differentia-tion,we infected hASCs by pre- or anti-miR-218. Transduced hASCweregrown to confluence and osteogenic differentiation was induced in dif-ferentiation media.

ED P

R

ntiation of hASCs. A. Quantitation ofmiR-218 in indicated hASCs normalized to U6. B. Real-Real-time PCR analysis of DKK2 normalized to GAPDH during osteogenic differentiation ofe PCR and normalized to U6. E. Real-time PCR analysis of SFRP2 normalized to GAPDH by

essing miR 218. G. The expression of miR-218 in hASCs transfected with antimiR 218 wasith antimiR 218 was assayed by real-time PCR and normalized to GAPDH. The data are the

etween miR-218 and Wnt/β-catenin signal promotes human adiposex.doi.org/10.1016/j.bone.2013.09.015

Page 4: A signal-amplification circuit between miR-218 and Wnt/β-catenin signal promotes human adipose tissue-derived stem cells osteogenic differentiation

205

206

207

208

209

210

4 W.-B. Zhang et al. / Bone xxx (2013) xxx–xxx

Fig. 2A showed BMSCs' osteogenic differentiation as a positive con-trol group. Overexpression ofmiR-218 significantly increased ALP activ-ity, whereas inhibiting miR-218 had the opposite effects.

UNCO

RRECT

Fig. 2.miR-218 promotes osteogenic differentiation of hASCs. A. AP staining at day 10 of BMSCsstaining at day 10 of hASCs' osteogenic differentiation by pre miR 218 and inhibitionmiR218 (mto GAPDH. D. Real-time PCR analysis of osteogenic-specific genes—OCNnormalized to GAPDH.time PCR analysis of osteogenic-specific genes — Runx2 normalized to GAPDH. The data are th

Please cite this article as: Zhang W-B, et al, A signal-amplification circuit btissue-derived stem cells osteogenic differentiation, Bone (2013), http://d

Up-regulating the expression of miR-218 obviously promotedhASCs osteogenic differentiation. Overexpression of miR-218 en-hanced osteogenic differentiation, which was indicated by the increase

ED P

RO

OF

osteogenic differentiation by premiR 218 and inhibitionmiR218 (magnification 4×). B. APagnification 4×). C. Real-time PCR analysis of osteogenic-specific genes— ALP normalizedE. Real-time PCR analysis of osteogenic-specific genes— BSP normalized to GAPDH. F. Real-e mean ± SD of 3 independent experiments. **P b 0.01.

etween miR-218 and Wnt/β-catenin signal promotes human adiposex.doi.org/10.1016/j.bone.2013.09.015

Page 5: A signal-amplification circuit between miR-218 and Wnt/β-catenin signal promotes human adipose tissue-derived stem cells osteogenic differentiation

211

212

213

214

215

216

217

218

219

220

221Q6

222

223

224

225

226

227

228

229

230

231

232

233

234

235

236

237

238

239

240

5W.-B. Zhang et al. / Bone xxx (2013) xxx–xxx

of ALP activity (Fig. 2B). Inhibition of miR-218 has an opposite effect(Fig. 2B).

The effect was accompanied with the increased mRNA levels of sev-eral key factors that are used as osteoblastic-specific markers.

Expression of mRNAs for ALP, the osteogenic transcription factorRunx2, and the mineralization marker (bone sialoprotein (BSP) andOCN) were all significantly increased with exogenous miR-218 but de-creased with antimiR-218 (Figs. 2CDEF).

Together, these experiments demonstrate the essential role ofmiR218 for hASCs osteogenic differentiation.

241

242

243

244

245

246

247

248

249

250

251

252

253

miR-218 targets Wnt inhibitors — SFRP2, DKK2 and mRNA and enhancesWnt/β-catenin signaling activity

To clarify the mechanism by which miR-218 controls hASCs osteo-genic differentiation, we focused on the identification of the potentialtargets of miR-218. To confirm the relationship between miR-218 andSFRP2/DKK2, we investigated the mRNA and protein levels of SFRP2/DKK2 in osteogenic differentiation of hASCs overexpressing miR-218or antimiR218. qPCR and Western blot analyses displayed that overex-press of miR-218 downregulated SFRP2/DKK2 expression and inhibi-tion of miR-218 upregulated SFRP2/DKK2 expression (Figs. 3A, C).

UNCO

RRECT

Fig. 3. Targeting of DKK2and SFRP2 mRNA by miR-218 and regulation of Wnt signaling pathwtransfection usingmiR-C,miR-218 and anti-miR-218. Relative expressionswere normalized to Gand anti-miR-218 was analyzed by qPCR. Relative expressions were normalized to GAPDH leveWestern blot inmiR-C-,miR-218 and anti-miR-218 transfected hASCs. The protein profiles weretarget SFRP2 sequence transfected with wild type (WT) and mutated (MT) UTR constructs aloactivity was normalized with Renilla luciferase activity and expressed in relative luminescencwild type (WT) and mutated (MT) UTR constructs along with nonspecific miR control (miR-C)ciferase activity and expressed in relative luminescence units. The data represent the mean ±

Please cite this article as: Zhang W-B, et al, A signal-amplification circuit btissue-derived stem cells osteogenic differentiation, Bone (2013), http://d

OO

F

To identify a mechanism for miR-218 and Wnt signaling in hASCs,WemonitoredWnt/β-catenin signaling activity.We examinedwhethertranscriptionalmediators of activatedWnt signaling, Lef-1 and Tcf-4, re-spond to miR-218 (Fig. 3B). Over expression of miR-218 robustly in-creased endogenous Tcf-4 and Lef-1 expression, while treatment ofanti-miR-218 markedly decreased Lef-1 and Tcf-4 expression. Stimula-tion ofWnt signaling bymiR-218 through down-regulation of inhibitorsof Wnt signaling that is reflected by elevated Lef1 and Tcf4. Proteinlevels of active β-catenin and nucleus β-catenin were drastically en-hanced by miR218 treatment but decreased by anti-miR-218, concomi-tant with elevated or diminished RUNX2 as an indicator of osteoblastdifferentiation (Fig. 3C). We also examined the expression of CyclinD1 and Axin2 the classical β-catenin target gene. Cyclin D1 was in-creased by miR-218 and decreased by the anti-miR218. Axin2 was de-creased by miR-218 and increased by the anti-miR218. Axin2 is aninhibitor and well documented feedback regulator of Wnt signaling.miR-218 doesn't target Axin2 directly. miR-218 may regulate Axin2mRNA indirectly by targeting a transcription factor which inducesAxin2 [12].

A luciferase reporter assay was used to demonstrate that miR-218directly decreased DKK2 and SFRP2 expression. We aligned the miR-218 sequence with DKK2 or SFRP2 3′UTR insert and used the resultingconstruct to transfect hASCs. Transfection of miR218-infected hASCs

ED P

R

ay. A. Analysis of DKK2/SFRP2 mRNA expression by real time qRT-PCR in hASCs 72 h postAPDH levels. B. Expression of Lef-1mRNA, Tcf-4mRNA inhASCs transducedwithmiR-218ls. C. SFRP2, DKK2, β-catenin, CyclinD1, Axin2 and Runx2 protein levels were analyzed bynormalizedwith anti-β-actin or anti SP1antibody. D. activity of the 3-UTR Luc reporter forng with nonspecific miR control (miR-C), miR-218, and anti-miR-218. Relative luciferasee units. E. activity of the 3-UTR Luc reporter for target DKK2 sequence transfected with, miR-218, and anti-miR-218. Relative Luciferase activity was normalized with Renilla lu-SD of 3 independent experiments. **P b 0.01.

etween miR-218 and Wnt/β-catenin signal promotes human adiposex.doi.org/10.1016/j.bone.2013.09.015

Page 6: A signal-amplification circuit between miR-218 and Wnt/β-catenin signal promotes human adipose tissue-derived stem cells osteogenic differentiation

TED P

RO

OF

254

255

256

257

258

259

260Q7

261

262

263

264

265

266

267

268

269

270

271

272

273Q8

274

275

276

277

278

279

280

281

282

283

284

285

286

287

288

289

290

291

292

293

294

295

296

297

298

299

300

301

302

303

304

305

306

307

308

309

310

311

312

313

314

315

316

317

318

319

320

321

322

323

324

325

326

327

328

329

330

331

332

333

Fig. 4.miR218 is regulated by Wnt3a/DKK2/SFRP2. A. miR218 levels were determined inhASCs treated with Wnt3a or DKK2 or SFRP2 under osteogenic conditions. B. Real-timePCR analysis of the osteogenic gens ALP and Runx2 in hASCs treated with Wnt3a orDKK2 or SFRP2 under osteogenic conditions. C. Key transcriptional factor β-catenin andRunx2 expression levels in hASCs treatedwithWnt3a or DKK2 or SFRP2 under osteogenicconditions were analyzed byWestern blot. The data represent the mean ± SD of 3 inde-pendent experiments. **P b 0.01.

6 W.-B. Zhang et al. / Bone xxx (2013) xxx–xxx

UNCO

RREC

with the parental luciferase construct (without the DKK2 or SFRP2 3′UTR) or a scrambled construct did not significantly change the expres-sion of the reporter. Cells transfected with a luciferase construct in themiR-218 target site from where the DKK2 or SFRP2 3′UTR (pMIR-DKK2 or pMIR-SFRP2) was inserted exhibited significantly lower lucif-erase activity in miR218-transfected hASCs than in control cells(Figs. 3D, E). No down regulation was observed when we mutated themiR binding sites for all potential target UTRs. Co-transfection with anantimiR218 oligo increased the luciferase activity of pMIR-DKK2 orpMIR-SFRP2 relative to co-transfection with a control oligo. These re-sults suggested that DKK2 and SFRP2 are potential direct targets ofmiR-218.

miR-218 and Wnt/β-catenin signaling are involved in a self-reinforcedregulatory circuit to regulate hASCs osteogenic differentiation

To further establish thatWnt/β-catenin signaling functionally affectsendogenous miR-218, we examined expression levels of miR-218 inthree biological conditions, i.e., Wnt3a-, DKK2 and SFRP2 stimulatedhASCs.

Our results showed that treatment of Wnt3a resulted in a nearly2.5-fold increase of miR-218 expression at 24 h. Furthermore, treat-ment of DKK-2/SFRP2 for 24 h decreased the expression of miR-218more than 2-fold.

Mimicking Wnt/β-catenin signal will strengthen the expressionlevel of miR-218, while blocking the signal weakens the expressionlevel of miR-218 (Fig. 4A). Wnt3a activated Wnt/β-catenin signal thusthe miR-218 molecule is up-regulated simultaneously and up-regulateALP, β-catenin and Runx2. DKK2/SFRP2 block Wnt/β-catenin signalthus down regulated miR-218 expression and then down regulateALP, β-catenin and Runx2 (Figs. 4B and C). Fig. 3B shows that the levelof active β-catenin proteinwas increasedwhen themiR-218 expressionwas upregulated, whereas inhibition of miR-218 leads to inhibition ofactive β-catenin. AntimiR-218 attenuated Wnt3a-induced hASC osteo-genic differentiation (Figs. 5A and B). These results have shown theassociation of miR-218 and Wnt signaling is necessary and sufficientfor osteogenesis to proceed in hASC cultures. A complex signal-amplification regulatory circuit between miR-218 and Wnt/β-cateninsignaling promotes hASC osteogenic differentiation (Fig. 6). This feed-forward regulatory circuit provides additional insight into howmiRNAsacting as a signal amplifier interact with signal molecules during hASCsosteogenic differentiation.

Discussion

Human adipose-derived stem cells (hASCs) which could differenti-ate into osteoblasts under appropriate conditions have a great applica-tion prospect in skeletal tissue engineering [3–5]. How to effectivelyimprove osteogenic differentiation of hASCs has become a key issue inthe bone tissue engineering. Osteoblast differentiation is regulated bycomplex pathways at both transcriptional and posttranscriptional levels[17,18]. MicroRNA influence osteogenic differentiation, by repressingprotein expression of their targets [18–20]. Therefore, we focused ourinvestigation of microRNA on human adipose-derived stem cells osteo-genic differentiation.

miR-218 is a potent osteo-miR expressed and continuously up-regulated during ASCs' osteogenic differentiation. The aim of this studywas to elucidate the effect and the mechanism of miR218 on hASCsosteogenic differentiation. Overexpression of miR-218 enhanced hASCsosteogenic differentiation. Inhibition ofmiR218 suppressed hASCs osteo-genic differentiation. Therefore, our data suggest thatmiR-218 positivelyregulates hASCs osteogenic differentiation.

How does miR-218 enhance hASCs' osteogensis? Our data pointstrongly to a mechanism involving the repression of Wnt antagonistsDKK2 and SFRP2, although other targets are certainly possible. miR-218overexpression downregulated the expression of DKK2 and SFRP2 at

Please cite this article as: Zhang W-B, et al, A signal-amplification circuit btissue-derived stem cells osteogenic differentiation, Bone (2013), http://d

themRNAandprotein levels. Conversely, inhibition ofmiR-218 increasedDKK2 and SFRP2 expression. These data, collected with the use of aluciferase reporter plasmid containing the DKK2/SFRP2-predicted targetgene sequences, indicated that miR-218 overexpression decreased lucif-erase activity, and inhibition of miR-218 increased luciferase activity,confirming that DKK2 and SFRP2 is a direct target of endogenous miR-218 in hASCs. Wnt signaling pathway was concordantly regulated byoverexpression and knockdown of miR-218 and has obvious link tohASCs osteogenic differentiation. Our data present evidence that miR-218 directly activates Wnt signaling promoting hASCs osteogensis. As aresult of down regulation of theDKK2/SFRP2protein level by overexpres-sion of miR-218, Wnt signaling is enhanced significantly and increasedβ-catenin transcriptional activity. Upregulation of miR-218 caused accu-mulation of active β-catenin in the cytoplasm and its translocation to thenucleus. Consequently, expression levels of Wnt target genes cyclinD1,Runx2, BSP and OCN are significantly upregulated. It is well knownthat Wnt promotes osteogenic differentiation of MSCs [21,22]. In-vivostudies have shown that constitutive active Wnt signaling leads to

etween miR-218 and Wnt/β-catenin signal promotes human adiposex.doi.org/10.1016/j.bone.2013.09.015

Page 7: A signal-amplification circuit between miR-218 and Wnt/β-catenin signal promotes human adipose tissue-derived stem cells osteogenic differentiation

T

334

335

336

337

338

339

340

341

342

343

344

345

346

347

348

349

350

351

352

353

354

355

356

357

358

359

360

361

362

363

364

365

366

367

368

369

370

371

372

373

374

375

376

377

378

379

380Q9

381382383384385386387388389390391392393394395396397398

Q2 Fig. 5. AntimiR-218 attenuated Wnt3a-induced hASC osteogenic differentiation. A. ALPand Runx2 mRNA expression levels by knock down miR-218 expression in the presenceof Wnt3a treatment with hASCs. B. Active β-catenin and Runx2 expression levels byknock down miR-218 expression in the presence of Wnt3a treatment with hASCs.

7W.-B. Zhang et al. / Bone xxx (2013) xxx–xxx

CO

RRECincreased bone mass and increased expression of bone marker genes

[23–25]. Conversely, deactivation of Wnt signaling by mutating LRP 5/6produces a low bonemass phenotype and inhibited osteogenic differen-tiation [14,26]. Negative regulators of Wnt signaling such as SFRPs haveall been shown to cause increased bone mass when mutated [27,28].DKK2 downregulation is a necessary in matures osteoblasts in order formineralization to proceed [27]. Recent studies also have shown thatWnt pathways are active during fracture repair and increasing the activ-ities of Wnt pathway components accelerates bone regeneration[28–31]. miR-218 modulation of hASCs osteogenic differentiation mayplan an important role in skeletal tissue regeneration. These suggestthe possibility of developing a new therapeutic approach targeting theWnt/beta-catenin pathway for improving bone regeneration whenusing hASCs as seed cells. In future work, it will be important to investi-gate whether miR-218 effectively improves hASCs' osteogensis for bone

UN 399

400401402403404405406407408409410411412413414415416417418419Fig. 6. The signal-reinforced regulatory circuit betweenmiR-218 and canonicalWnt signal.

Please cite this article as: Zhang W-B, et al, A signal-amplification circuit btissue-derived stem cells osteogenic differentiation, Bone (2013), http://d

ED P

RO

OF

regeneration in vivo. This information may prove valuable in usingmiR-218 as a therapeutic targeting molecule to regulate for hASCs ap-plications in bone regeneration.

The present study has uncovered a signal amplification circuit regu-lating hASCs osteogenesis betweenmiR-218 andWnt/β-catenin signal-ing. miR-218 stimulated osteogenesis signaling by negatively regulatinginhibitors of Wnt signaling. Inhibition of Wnt/β-catenin pathwaysdecreased expression of miR-218. Activation of Wnt/β-catenin signalincreased expression of endogenous miR-218. miR-218 acts as a signal-ing amplifier for positively regulating the osteogenic differentiation ofhASCs. When osteoblastic differentiation is initiated canonical Wnt sig-nal activated. The activation of Wnt signal further enhances miR-218expression, whereas the attenuation of DKK2/SFRP2 to promote hASCsosteogenic differentiation. The signal-reinforced regulatory circuit be-tweenmiR-218 andWnt/β-catenin signal leads to the prominent effectof modulating β-catenin and Runx2 expression, which fine tunes thepace of progression in hASCs osteogenic differentiation (Fig. 6).

In conclusion, we have characterized the roles of miR-218 in osteo-genic differentiation of hASCs and elucidated the mechanisms of miR-218 action in this process. Thesefindings presentmiR-218 as a potentialtarget in the management osteogenic differentiation of hASCs for boneregeneration.

Acknowledgments

This work was supported by grants from the Natural Science Foun-dation for Colleges and Universities in JiangSu Province (Grant Number:10kJB320004), and the Priority Academic Program Development ofJiangsu Higher Education Institutions (PAPD).

Disclosure of potential conflict of interest

The authors declare no conflict of interest.

References

[1] Tremolada C, Palmieri G, Ricordi C. Adipocyte transplantation and stem cells: plasticsurgery meets regenerative medicine. Cell Transplant 2010;19(10):1217–23.

[2] Zuk PA, ZhuM, Mizuno H, et al. Multilineage cells from human adipose tissue: impli-cations for cell-based therapies. Tissue Eng 2001;7(2):211–28.

[3] Levi B, Longaker MT. Concise review: adipose-derived stromal cells for skeletal re-generative medicine. Stem Cells 2011;29(4):576–82.

[4] Izadpanah R, Trygg C, Patel B, et al. Biologic properties of mesenchymal stem cellsderived from bone marrow and adipose tissue. J Cell Biochem 2006;99(5):1285–97.

[5] Rada T, Reis RL, Gomes ME. Distinct stem cells subpopulations isolated from humanadipose tissue exhibit different chondrogenic and osteogenic differentiation poten-tial. Stem Cell Rev 2011;7(1):64–76.

[6] Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell2004;116(2):281–97.

[7] Ebert MS, Sharp PA. Roles for microRNAs in conferring robustness to biological pro-cesses. Cell 2012;149(3):515–24.

[8] Lakshmipathy U, Hart RP. Concise review: microRNA expression in multipotentmesenchymal stromal cells. Stem Cells 2008;26(2):356–63.

[9] Filipowicz W, Bhattacharyya SN, Sonenberg N. Mechanisms of post-transcriptionalregulation bymicroRNAs: are the answers in sight? Nat Rev Genet 2008;9(2):102–14.

[10] Kim YJ, Bae SW, Yu SS, et al. miR-196a regulates proliferation and osteogenic differ-entiation in mesenchymal stem cells derived from human adipose tissue. J BoneMiner Res 2009;24(5):816–25.

[11] Luzi E, Marini F, Sala SC, et al. Osteogenic differentiation of human adipose tissue-derived stem cells is modulated by the miR-26a targeting of the SMAD1 transcrip-tion factor. J Bone Miner Res 2008;23(2):287–95.

[12] HassanMQ, Maeda Y, Taipaleenmaki H, et al. miR-218 directs aWnt signaling circuitto promote differentiation of osteoblasts and osteomimicry of metastatic cancercells. J Biol Chem 2012;287:42084–92.

[13] Strioga M, Viswanathan S, Darinskas A, et al. Same or not the same? Comparison ofadipose tissue-derived versus bone marrow-derived mesenchymal stem and stro-mal cells. Stem Cells Dev 2012;21(14):2724–52.

[14] Cui Y, Niziolek PJ, MacDonald BT, et al. Lrp5 functions in bone to regulate bonemass.Nat Med 2011;17(6):684–91.

[15] Oshima T, Abe M, Asano J, et al. Myeloma cells suppress bone formation by secretinga soluble Wnt inhibitor, sFRP-2. Blood 2005;106(9):3160–5.

[16] Boquest AC, Shahdadfar A, Brinchmann JE, et al. Isolation of stromal stem cells fromhuman adipose tissue. Methods Mol Biol 2006;325:35–46.

[17] Franceschi RT, Ge C, Xiao G, et al. Transcriptional regulation of osteoblasts. Ann N YAcad Sci 2007;1116:196–207.

etween miR-218 and Wnt/β-catenin signal promotes human adiposex.doi.org/10.1016/j.bone.2013.09.015

Page 8: A signal-amplification circuit between miR-218 and Wnt/β-catenin signal promotes human adipose tissue-derived stem cells osteogenic differentiation

420421422423424425426427428429430431432433434435436

437438439440441442443444445446447448449450451452453

455

8 W.-B. Zhang et al. / Bone xxx (2013) xxx–xxx

[18] Taipaleenmaki H, Bjerre L, Chen L, et al. Mechanisms in endocrinology: micro-RNAs:targets for enhancing osteoblast differentiation and bone formation. Eur J Endocrinol2012;166:359–71.

[19] Inose H, Ochi H, Kimura A, et al. AmicroRNA regulatorymechanism of osteoblast dif-ferentiation. Proc Natl Acad Sci U S A 2009;106:20794–9.

[20] Hu R, Li H, LiuW, et al. Targeting miRNAs in osteoblast differentiation and bone for-mation. Expert Opin Ther Targets 2010;14:1109–20.

[21] LiuG, Vijayakumar S, Grumolato L, et al. CanonicalWnts function as potent regulators ofosteogenesis by human mesenchymal stem cells. J Cell Biol 2009;185(1):67–75.

[22] Yang F, Yang D, Tu J, et al. Strontium enhances osteogenic differentiation of mesen-chymal stem cells and in vivo bone formation by activating Wnt/catenin signaling.Stem Cells 2011;29(6):981–91.

[23] Krishnan V, Bryant HU, Macdougald OA. Regulation of bone mass by Wnt signaling.J Clin Invest 2006;116(5):1202–9.

[24] Collette NM, Genetos DC, Economides AN, et al. Targeted deletion of Sost distalenhancer increases bone formation and bone mass. Proc Natl Acad Sci U S A2012;109(35):14092–7.

UNCO

RRECT

454

Please cite this article as: Zhang W-B, et al, A signal-amplification circuit btissue-derived stem cells osteogenic differentiation, Bone (2013), http://d

[25] Day TF, Guo X, Garrett-Beal L, et al.Wnt/beta-catenin signaling inmesenchymal pro-genitors controls osteoblast and chondrocyte differentiation during vertebrateskeletogenesis. Dev Cell 2005;8(5):739–50.

[26] Kokubu C, Heinzmann U, Kokubu T, et al. Skeletal defects in ringelschwanz mutantmice reveal that Lrp6 is required for proper somitogenesis and osteogenesis. Devel-opment 2004;131(21):5469–80.

[27] Li X, Liu P, Liu W, et al. Dkk2 has a role in terminal osteoblast differentiation andmineralized matrix formation. Nat Genet 2005;37:945–52.

[28] Bodine PV, Zhao W, Kharode YP, et al. The Wnt antagonist secreted frizzled-relatedprotein-1 is a negative regulator of trabecular bone formation in adult mice. MolEndocrinol 2004;18(5):1222–37.

[29] Leucht P, Kim JB, Helms JA. Beta-catenin-dependent Wnt signaling in mandibularbone regeneration. J Bone Joint Surg Am 2008;90(Suppl. 1):3–8.

[30] Komatsu DE, Mary MN, Schroeder RJ, et al. Modulation of Wnt signaling influencesfracture repair. J Orthop Res 2010;28:928–36.

[31] Minear S, Leucht P, Jiang J, et al. Wnt proteins promote bone regeneration. Sci TranslMed 2010;2:29ra30. http://dx.doi.org/10.1126/scitranslmed.3000231.

ED P

RO

OF

etween miR-218 and Wnt/β-catenin signal promotes human adiposex.doi.org/10.1016/j.bone.2013.09.015