Received: 2017.11.7 Accepted: 2018.2.14 J

1 downloads 0 Views 1MB Size Report
Mar 8, 2018 - A. and Manunta, A. F. Subchondral bone remodeling: comparing nanofracture. 560 with microfracture. An ovine in vivo study. Joints.2016.4:.
Advance Publication Experimental Animals

Received: 2017.11.7 Accepted: 2018.2.14 J-STAGE Advance Published Date: 2018.3.8

1

1

Original paper: Bioresource

2

Subchondral Bone Derived Mesenchymal Stem Cells

3

Display Enhanced Osteo-Chondrogenic Differentiation,

4

Self-renewal and Proliferation Potentials

5

Hao Zhang1, 2, Zhong-Li Li1,*, Xiang-Zheng Su1, Li Ding3, Ji Li1, Heng Zhu2,*

6

1. Department of Orthopedics, Sports Medicine Center, People's Liberation

7

Army General Hospital, Beijing 100853, China

8

2. Department of Cell Biology, Institute of Basic Medical Sciences, Beijing

9

100850, P.R. China

10

3. Department of Hematology, General Hospital of Air Forces, PLA, Beijing,

11

China

12 13

Corresponding author: Zhong-Li Li, Department of Orthopedics, Sports

14

Medicine Center, People's Liberation Army General Hospital, Beijing 100853,

15

China. Tel: 86-10-66938206. Fax: 86-10-88219862. Email: [email protected]

16

Heng Zhu, Department of Cell Biology, Institute of Basic Medical Sciences,

17

Taiping Road 27, Beijing 100850, P.R. China, and Tel: 861066930913, Fax:

18

861068213039 E-mail: [email protected]

19 20 21

1

2

22

Abstract

23

Rabbit mesenchymal stem cells (MSCs) are important seed cells in regenerative

24

medicine research, particularly in translational research. In the current study, we

25

showed that rabbit subchondral bone is a reliable source of MSCs. First, we

26

harvested subchondral bone (SCB) from the rabbit knee-joint and initiated the

27

MSC culture by cultivating enzyme-treated SCB. Adherent fibroblast-like cells

28

that outgrew from SCB fulfill the common immuno-phenotypic criteria for

29

defining MSCs, but with low contamination of CD45+ hematopoietic cells.

30

Interestingly, differentiated SCB-MSCs expressed osteogenic and chondrogenic

31

markers at significantly higher levels than those in bone marrow cell

32

suspension-derived MSCs (BMS-MSCs) (P < 0.05). No differences in the

33

expression of adipogenic markers

34

0.05) were observed. Moreover, the results of the colony forming unit-fibroblast

35

assay and sphere formation assay demonstrated that the SCB-MSCs had

36

increased self-renewal potential. SCB-MSCs expressed higher levels of the

37

stemness markers Nanog, OCT4, and Sox-2 compared to in BMS-MSCs (P


2

3

44

properties of SCB-MSC are important for the potential treatment of tissue

45

damage resulting from disease and trauma.

46 47

Keywords: Bioresource; Experimental Animals; Mesenchymal Stem Cell;

48

Rabbit; Subchondral Bone

49

3

4

50

Introduction

51

Mesenchymal stem cells (MSCs), also known as multipotent stromal cells, were

52

first identified in the bone marrow[14]. In postnatal organisms, loosely woven

53

and highly vascularized bone marrow form a unique niche for stem cells[14,31].

54

Like hematopoietic stem cells, the multi-potency and self-renewal of MSCs are

55

tightly controlled by the bone marrow microenvironment[8,31,36]. According to

56

the requirements of the hosts, MSCs migrate out from connective tissue of bone

57

marrow and regenerate mesenchymal tissues[25]. Increasing data have shown

58

that MSCs play a role as promising seed cells for cellular replacement therapy

59

for diabetes, rheumatoid arthritis, and bone repair[8,17,18,31,38]. The rabbit is a

60

commonly used experimental animal for orthopedic application and tissue

61

engineering because of its easy accessibility and convenient maneuverability.

62

However, MSC-based therapies in rabbit models are limited because of

63

contamination by hematopoietic cells. Notably, the structure of the MSC niche

64

was typically destroyed, while rabbit MSCs were routinely cultured in bone

65

marrow cell suspension[2,16,36]. We previously isolated and characterized

66

MSCs and examined the potential application of these cells[24,45,48]. In our

67

previous studies, we found that collagenase digestion efficiently loosened the

68

tissue microstructure and facilitated MSC outgrowth from tissues without

69

reducing cell viability. In addition, enzymic treatment induced the release of

70

hematopoietic cells and made it easier to deplete them[19,47].

71

Because mesenchymal stem cells were first described in the 1970s, many types 4

5

72

of biological tissue have been developed as stem cell resources[15,30].

73

Particularly,

74

nonimmunogenic, and easily available stem cells[1,22,26,35]. However, many

75

studies have shown that the sources of origin and microenvironment greatly

76

impact the differentiation ability of MSCs[26]. Researchers revealed that

77

adipose tissue-derived MSCs show decreased osteogenic and chondrogenic

78

differentiation capacity compared to bone marrow-derived MSCs[9,42].

79

Therefore, attention should be given to subchondral bone (SCB), which is

80

accessible in orthopedics surgery and has a similar microenvironment, to

81

facilitate bone and cartilage injury regeneration.

82

Therefore, we hypothesized that culturing the SCB and allowing MSCs to

83

migrate out from their stem cell niche may be an efficient strategy for obtaining

84

viable and homogeneous rabbit MSC populations. We digested SCB and

85

conducted MSC (SCB-MSC) culture using these cells. Our results showed that

86

SCB-MSCs display enhanced osteo-chondrogenic differentiation, self-renewal,

87

and proliferation potential compared to bone marrow suspension-derived MSCs

88

(BMS-MSCs).

adipose

tissue

is

an

optimal

source

of

proliferating,

89 90

Materials and methods

91

Isolation and culture of SCB-MSCs

92

MSCs were isolated from male New Zealand White rabbits (3–4 weeks of age,

93

from the Laboratory Animal Center of the Academy of Military Medical 5

6

94

Sciences of China, Beijing, China). All experiments in this study were

95

performed in accordance with the Academy of Military Medical Sciences Guide

96

for Laboratory Animals. To isolate SCB-MSCs, the knee joints of the rabbit

97

were carefully excised with scissors, and the subchondral bones were collected

98

using forceps. Subchondral bone fragments were cultured in α-MEM containing

99

10% (vol/vol) fetal bovine sum (FBS) (Solarbio, Hyclone, Logan, UT, USA) in

100

the presence of 1 mg mL−1 (wt/vol) of collagenase II (Gibco, Grand Island, NY,

101

USA) at 37°C for 20 min. The digestion medium and released cells were

102

discarded, and the enzyme-treated SCBs were seeded into a plastic culture dish

103

(250 mL) in the presence of α-MEM supplemented with 10% (vol/vol) FBS. The

104

culture medium was changed on the third day of culture, and the tissue debris

105

was maintained to allow more MSCs to outgrow. To isolate BMS-MSCs, the

106

bone marrow was flushed out from the marrow cavity in the tibiae and femurs

107

and mononuclear cells were isolated from the bone marrow suspensions by

108

routine density gradient centrifugation. The cells were then seeded on a plastic

109

dish (BD Biosciences, Franklin Lakes, NJ, USA, 100 × 15 mm), and the MSCs

110

were allowed to adhere for 72 h before the total volume of the culture medium

111

was changed.

112 113

Flow cytometry analysis

114

SCB-MSCs and BMS-MSCs were harvested at passages 3–6 by trypsin

115

digestion and stained individually with 6

phycoerythrin- or fluorescein

7

116

isothiocyanate-conjugated monoclonal antibodies against rabbit CD44, CD45,

117

CD14, CD79a, CD81, or CD90 (BD Biosciences and Abcam, Cambridge, UK)

118

for 30 min in the dark at 4°C. After two washes with PBS, the cells were

119

collected with a FACScan (BD Biosciences) and the data were analyzed using

120

WinMDI 2.9 software.

121 122

Multi-differentiation of MSCs

123

Multi-differentiation analysis of MSCs was performed as described previously

124

with minor modifications[19,47]. Briefly, for osteogenic differentiation, MSCs

125

at passage 3 were seeded into 24-well culture plates (1 mL/well) at a density of

126

5 × 103 cells/cm2, grown in osteogenic induction medium for 14 days, and

127

subjected to alkaline phosphatase (ALP) staining. The osteogenic induction

128

medium consisted of culture medium, 0.1 µM dexamethasone, 10 mM

129

β-glycerophosphate, and 50 µM ascorbic acid (Sigma-Aldrich, St. Louis, MO,

130

USA). The osteogenic differentiation of MSCs was assayed by in situ ALP

131

staining with a commercial kit (Sigma-Aldrich).

132

For adipogenic differentiation, MSCs at passage 3 were seeded into 24-well

133

culture plates at a density of 1 × 104 cells/cm 2, incubated in adipogenic induction

134

medium for 14 days, and subjected to Oil-Red-O staining. The adipogenic

135

induction medium consisted of culture medium, 1 µM dexamethasone, 0.2 mM

136

indomethacin, 0.5 mM 3-butyl-L-methylxanthine (IBMX), and 0.01 mg/mL

137

insulin (Sigma-Aldrich). The accumulation of lipid vacuoles in MSCs was 7

8

138

evaluated by in situ Oil-Red-O staining.

139

For chondrogenic differentiation, 4 × 105 MSCs were centrifuged in

140

polypropylene tubes to form a pelleted micromass and maintained in

141

chondrogenic induction medium consisting of α-MEM supplemented with 10−7

142

M dexamethasone, 1% (vol/vol) insulin-transferrin-sodium selenite, 50 µM

143

ascorbate-2 phosphate, 1 mM sodium pyruvate, 50 µg/mL (wt/vol) proline, and

144

20 ng/mL (wt/vol) TGF-β3. On day 21, the pellets were fixed and sectioned as

145

previously described[47]. The development of chondrocytes and accumulation

146

of the cartilage matrix were evaluated by toluidine blue staining.

147 148

Colony-forming unit-fibroblast (CFU-F) assay

149

The clonogenic potential of MSCs was tested in a colony-forming unit-fibroblast

150

(CFU-F) assay as described previously with minor revisions[11]. Briefly, MSCs

151

at passage 1 were seeded into a 6-well plate (Corning, Inc., Corning, NY, USA,

152

16.8 mL/well) at a density of 1 × 103/well and maintained in culture medium. To

153

detect the formation of CFU-F, the cultured cells in three replicates were stained

154

with 3% crystal violet in methanol for 10 min at days 5, 10, and 15. All visible

155

colonies larger than 5 mm in diameter were counted.

156 157

Sphere formation assay

158

The clonogenic potential of the MSCs was further tested in a sphere formation

159

assay[20,31]. MSCs at passage 1 were seeded at 2 × 105/cm 2 on an ultra-low 8

9

160

attachment dish (Corning) in α-MEM supplemented with 10% (vol/vol) FBS.

161

Primary cell spheres were counted after 3 days in culture, trypsinized, and

162

re-plated. Secondary spheres were counted on day 6.

163 164

CCK8 assay

165

MSC proliferation assays were performed using the Cell Counting Kit-8 (CCK-8;

166

Dojindo Laboratories, Kumamoto, Japan)[41]. Briefly, MSCs at passage 3 were

167

seeded into 96-well plates (Thermo Scientific, Waltham, MA, USA) at a density

168

of 1×103 cells/cm 2, cultured in α-MEM medium with 10% FBS (6 wells in each

169

group), added to CCK-8 solution at a ratio of 100 µL/µL, and incubated at 37°C

170

for 1 h. Absorbance was then measured at a wavelength of 450 nm using a

171

microplate reader (BMG LABTECH, Offenburg, Germany). In the current study,

172

CCK-8 experiments were performed on days 1, 5, 7, 10, and 13.

173 174

CFSE dilution assay

175

Moreover, the proliferation of SCB-MSCs and BMS-MSCs was also examined

176

in a CFSE dilution assay. Briefly, MSCs were suspended at a concentration of

177

107 cells/mL in PBS containing 2% FBS. MSCs were incubated in the presence

178

of 10 µM CFSE for 20 min in the dark, followed by blockage of CFSE

179

incorporation by FBS. The cells were then washed twice before they were

180

re-plated. MSCs were harvested on days 2 and 4. The dye dilution was assayed

181

with a FACSCalibur instrument and data were analyzed using WinMdi2.8 9

10

182

software.

183 184

Cell cycle assay

185

MSCs were seeded at 5 × 103 cells/cm 2 and cultured in α-MEM medium with 10%

186

FBS. At 80–90% confluence, the MSCs were collected for cell cycle analysis.

187

Briefly, the MSCs were washed and fixed overnight in 70% ethanol at -20°C in

188

1.5-mL microcentrifuge tubes (Biologix, Shandong, China). The fixed cells were

189

then washed and incubated in 100 µg/mL propidium iodide (Sigma-Aldrich) and

190

20 ng/mL RNase (Sigma-Aldrich) in PBS for 30 min. Cell cycle analysis was

191

then conducted by flow cytometry. Independent experiments were replicated at

192

least three times. The cell subpopulations in the G0/G1 and S phases were

193

calculated by gating analysis based on differences in DNA content.

194 195

Real-time polymerase chain reaction

196

Aliquots of MSCs (2 × 105) at passages 3–6 were seeded in 6-well culture plates

197

and maintained in osteogenic/adipogenic/chondrogenic induction medium for 7

198

days before they were harvested. Total RNA was extracted from MSCs with

199

TRIzol reagent (Invitrogen) and reverse-transcribed using the mRNA Selective

200

PCR Kit (TaKaRa, Shiga, Japan). Rabbit HPRT, Runx-2, osteopontin (OPN),

201

CEBP/α, PPARγ, Sox-9, collagen I, Nanog, OCT4, and Sox2 cDNA were

202

amplified by real-time PCR using the SYBR Green PCR kit (Sigma). The

203

primer sequences used for real-time PCR are shown in Table 1. 10

11

204 205

Western blotting

206

MSCs at passage 3 and 6 were plated in 6-well plates at a density of 1 × 105

207

cells/cm2 and starved in serum-free α-MEM medium for at least 6 h. Protein

208

lysis buffer (Bio-Rad, Hercules, CA, USA) was added, and thawed lysates were

209

vortexed and centrifuged. The proteins were separated by 10% sodium dodecyl

210

sulfate polyacrylamide gel electrophoresis and transferred onto nitrocellulose

211

membranes. The membranes were blocked by incubation with 5% wt/vol nonfat

212

dry milk. Membranes were then incubated with anti-ERK, anti-phospho-ERK,

213

and β-actin (Sigma) Abs at the appropriate dilutions overnight at 4°C. After

214

incubation, the membranes were washed in Tris-buffered saline containing

215

Tween-20 (TBST). Secondary antibody conjugated to horseradish peroxidase

216

was added to the membranes in 5% nonfat dry milk in TBST. The negative

217

control was used as described previously. The western blotting assay was

218

performed at least 3 times independently, representative results are shown.

219 220

Statistical analysis

221

The data were expressed as the mean values with the standard deviation.

222

Statistical significance was analyzed by Student’s t test and two-tailed p-values

223

were calculated, and P < 0.05 was considered statistically significant. The error

224

bars in all figures represent the standard deviation.

225 11

12

226

Results

227

SCB-MSC exhibit morphological features and surface antigens similar to

228

those of BMS-MSCs

229

Forty-eight hours after the primary culture, fibroblast-like cells migrated out

230

from the digested SCB fragments and adhered to the dish (Figure 1A-a),

231

whereas a few elongated adhesion cells were observed in the dish in which the

232

bone marrow cell suspension cells were seeded (Figure 1A-b). An adherent layer

233

of vortex-shaped cells developed within 6 days (Figure 1A-c), whereas a culture

234

confluence of only 30–40% was achieved when the nuclear cells were cultivated

235

(Figure 1A-d). Further, the results of immuno-phenotyping showed that both

236

SCB-MSCs and BMS-MSCs were homogenously positive for the mesenchymal

237

markers CD44 and CD81 but negative for the hematopoietic markers CD14 and

238

CD45 and co-stimulating molecule CD79α (Figure 1B). Unlike human MSCs, it

239

remains controversial whether rabbit MSCs are positive for CD90[2,28,36]. Our

240

results showed that SCB-MSCs and BMS-MSCs were negative for CD90

241

(Figure 1B). In addition, the percentage of CD45+ cells in the SCB-MSCs (3.31

242

± 0.78%) was significantly lower than that in BMS-MSCs (13.93 ± 1.63%) (**P

243

< 0.01), demonstrating that a homogeneous cell population was expanded from

244

the digested subchondral bone (Figure 1C).

245 246

SCB-MSCs display enhanced osteogenic and chondrogenic differentiation

247

potential 12

13

248

Although the SCB-MSCs and BMS-MSCs shared similar morphologic and

249

immuno-phenotypic features, SCB-MSCs display enhanced differentiation

250

capacity compared to BMS-MSCs. Analysis of osteogenic differentiation

251

showed higher ALP activity in SCB-MSCs than in BMS-MSCs after 14 days of

252

induction (Figure 2A). Additionally, the analysis of chondrogenic differentiation

253

showed that more SCB-MSCs developed into toluidine blue-positive

254

chondrocytes, indicating that the cells secreted sulfated proteoglycan at a higher

255

level to form a cartilage extracellular matrix (Figure 2A). However, no

256

significant differences were observed in the accumulation of intracellular

257

Oil-Red-O-stained lipids, indicating that SCB-MSCs and BMS-MSCs shared a

258

similar adipogenic differentiation capacity (Figure 2A). Complementing the

259

results of histochemical analysis, SCB-MSCs after induction exhibited high

260

levels of mRNA expression of osteogenic markers (Runx-2 and OPN) and

261

chondrogenic markers (Sox-9 and Collage I) (*P < 0.05; **P < 0.01, Figure 2B).

262

The mRNA expression of adipogenic transcription factor CEBP/α and PPARγ in

263

SCB-MSCs was similar to that in BMS-MSCs (Figure 2B).

264 265

SCB-MSCs display higher self-renewal potential

266

Functional MSCs were initially identified by their capacity to form clonogenic

267

cell clusters in vitro, a common feature different to other stromal cell

268

populations. In the current study, self-renewal potential was measured in a

269

CFU-F assay and sphere formation assay. As indicated in Figure 3A and 3B, the 13

14

270

CFU-F frequency remained relatively higher in SCB-MSCs than in BMS-MSCs

271

(SCB-MSCs versus BMS-MSCs: 6.33 ± 0.94 versus 3 ± 0.82, 11 ± 1.63 versus

272

5.67 ± 0.94, 17 ± 0.82 versus 10.67 ± 1.25 for days 5, 10, and 15, respectively.

273

*P < 0.05; **P < 0.01).

274

Sphere formation assays have long been used to evaluate progenitor/multipotent

275

cell populations in epithelial systems. Recent studies suggested that MSCs can

276

also produce spheres[20,31]. Three days after culture on ultra-low adherent

277

tissue culture plates, sphere formation was evident in the SCB-MSC group and

278

BMS-MSC group (Figure 3C). These spheres were disassociated and re-plated

279

on non-adherent plates. Fewer spheres developed after another 3 days of culture.

280

Interestingly, there was a noticeable difference in primary and secondary sphere

281

number in SCB-MSC culture compared with to in BMS culture (SCB-MSCs

282

versus BMS-MSCs: 38 ± 9.53 versus 22 ± 0.82, 24.67 ± 3.21 versus 10 ± 1.63

283

for primary spheres and secondary spheres, respectively. Figure 3D, *P < 0.05;

284

**P < 0.01).

285

The results of the CUF-F and sphere formation assays strongly suggest that

286

SCB-MSCs have an increased stem cell population that can self-renew. To

287

further explore the cause of enhanced self-renewal, we next measured the

288

mRNA expression of several stemness markers (Nanog, OCT4, and Sox-2) in

289

SCB-MSCs[4,7,27,34].

290

significantly higher transcription levels of Nanog, OCT4, and Sox-2 than in

291

BMS-MSCs (Figure 3E, *P < 0.05; **P < 0.01).

The

data

indicated

14

that

SCB-MSCs

displayed

15

292 293

SCB-MSCs display enhanced proliferative capacity

294

To investigate the proliferation ability of SCB-MSCs, a CCK-8 assay and CFSE

295

dilution assay were performed. The results of the CCK-8-based cell proliferation

296

assay (Figure 4A) showed that SCB-MSCs exerted stronger proliferative effects

297

than BMS-MSCs (*P < 0.05). Consistently, the CFSE data showed that a higher

298

proportion of SCB-MSCs underwent cell division on days 2 and 4 (Figure 4B),

299

indicating that these cells had an enhanced proliferation capacity.

300

Enhanced cell proliferation is also reflected by an increased number of cells in

301

the S phase and decreased number of cells arrested in the G0/G1 phase. A higher

302

percentage of SCB-MSCs (50 ± 1.41%) were in S phase compared to

303

BMS-MSCs (36.5 ± 3.55%) (Figure 4C), indicating that an increased number of

304

cells proceeded into G2/S phase (*P < 0.05).

305

Because

306

proliferation[6,13], we further examined the phosphorylation of ERK-MAPK in

307

the cells. The data in Figure 4D shows enhanced Erk1/2 phosphorylation in

308

passages 3 and 6 SCB-MSCs. The results support that SCB-MSC harbors an

309

enhanced proliferation capacity.

ERK-MAPK

signaling

is

involved

in

controlling

cell

310 311

Discussion

312

Rabbit MSCs are important seed cells in regenerative medicine research,

313

particularly in translational research. A variety of healthy tissues have been 15

16

314

developed as stem cell resources, including bone marrow, blood, umbilical cord,

315

placenta, fat, heart, brain, skin, muscle, liver, gonads, and teeth[37]. Many

316

studies have shown that the differentiation ability of MSCs varies greatly from

317

different resources. In the orthopedics field, SCB has received attention in

318

regeneration research[10,21,40,44,46].

319

To identify MSCs, surface antigen markers were tested. It remains controversial

320

weather rabbit MSC express CD90 based on previous studies. Tan et al. (2013)

321

characterized rabbit MSCs and found that they expressed CD90[36]. Bakhtina

322

(2014) and Lee (2014) compared the surface markers between human and rabbit

323

MSCs and found rabbit MSCs did not express CD90[2,28]. The results of flow

324

cytometry analysis in the present study showed that rabbit MSCs were

325

CD90-negative, which is in accordance with the previous reports. The adult

326

bone marrow contains niches that control the multi-differentiation potential and

327

self-renewal capacity of stem cells[3]. Several studies demonstrated that

328

implanted bone marrow could support long-term repopulating cells in vivo[5,39].

329

Therefore, maintaining the bone marrow niche in primary culture may be

330

beneficial for MSC properties. In the present study, we initiated MSC culture

331

using digested rabbit SCBs, which are mainly composed of adipose tissue and

332

vessel networks.

333

Our results suggest that SCB-MSCs meet the generally accepted criteria,[12]

334

including the fibroblast-like morphology, typical cell surface profile, and

335

multi-lineage differentiation capacity. It had been widely accepted that MSCs 16

17

336

cultured from different tissues share many common features, but the

337

differentiation potential vary[9,26]. In this study, the results showed that

338

SCB-MSCs gain enhanced osteogenic and chodrogenic differentiation potential

339

that is comparable to that of BMS-MSCs, which is important for the potential

340

treatment of tissue damage resulting from disease and trauma.

341

Several factors have been reported to influence MSC self-renewal capacity,

342

including cell passages, differentiation, and other factors[23,33,43]. In the

343

present

344

differentiation into osteoblasts and chondrocytes, maintain a higher self-renewal

345

capacity. The results of the CFU-F and sphere forming assays suggest that

346

SCB-MSCs contain more potent cells. Nanog, OCT4, and Sox-2 are crucial

347

stemness transcription factors, and lower expression of these proteins leads to a

348

deficiency of self-renewal[4,7,27,34]. Based on the results of the colony

349

formation assay, SCB-MSCs expressed high levels of Nanog, OCT4, and Sox-2.

350

High proliferation is a fundamental property of MSCs and is important for the

351

potential treatment of tissue damage resulting from disease and trauma. The

352

CCK-8 assay and CFSE dilution assay are widely used to analyze the

353

proliferation of stem cells[29,32]. Because ERK-MAPK signaling is involved in

354

controlling cell proliferation, phosphorylation of ERK-MAPK in MSCs was also

355

detected in this present study. The results showed SCB-MSCs grew at a higher

356

rate than their marrow counterparts. These results demonstrate that the

357

proliferation of MSCs was improved in SCB culture.

study,

we

demonstrated

that

17

SCB-MSCs,

when

undergoing

18

358

There were also many limitations in our study. First, the most widely used MSC

359

resource in regenerative medicine domain is fat tissue, umbilical cord, and

360

placenta. We only compared bone marrow-derived MSCs and SCB-derived

361

MSCs in the present study. Second, all tests were performed in vitro in this study,

362

and an animal joint injury model would be useful in further studies to explore

363

the differences between different source origin-derived MSCs in vivo. Third, the

364

mechanism of differentiation and proliferation potential changes should be

365

evaluate in further studies.

366 367

Conclusion

368

In conclusion, our results support that maintaining the bone marrow niche in

369

MSC culture minimizes the negative impact on cell yield and purity while

370

retaining enhanced multi-potency, self-renewal, and proliferation potential of

371

MSCs. However, the precise mechanism regulating the fate of SCB-MSCs

372

requires further investigation. The results also suggest that SCB is a novel

373

resource for rabbit MSCs and may provide helpful information for

374

understanding MSC niches.

375 376

Acknowledgements

377

This study was supported by the National Natural Science Foundation

378

(81572159, and 81371945) and the Beijing Natural Sciences Grants (No.

379

7182123). 18

19

380

The authors declare no competing financial interests.

381 382

References

383

[1]. Bajek, A., Gurtowska, N., Olkowska, J., Kazmierski, L., Maj, M. and Drewa,

384

T. Adipose-Derived Stem Cells as a Tool in Cell-Based Therapies. Archivum

385

immunologiae

386

443-454.doi:10.1007/s00005-016-0394-x

387

[2]. Bakhtina, A., Tohfafarosh, M., Lichtler, A. and Arinzeh, T. L.

388

Characterization and differentiation potential of rabbit mesenchymal stem cells

389

for translational regenerative medicine. In vitro cellular & developmental

390

biology Animal.2014.50: 251-260.doi:10.1007/s11626-013-9702-5

391

[3]. Bardelli, S. and Moccetti, M. Remodeling the Human Adult Stem Cell

392

Niche

393

international.2017.2017: 6406025.doi:10.1155/2017/6406025

394

[4]. Basu-Roy, U., Ambrosetti, D., Favaro, R., Nicolis, S. K., Mansukhani, A.

395

and Basilico, C. The transcription factor Sox2 is required for osteoblast

396

self-renewal.

397

1345-1353.doi:10.1038/cdd.2010.57

398

[5]. Bigildeev, A. E., Zhironkina, O. A., Lubkova, O. N. and Drize, N. J.

399

Interleukin-1

400

Cytokine.2013.64: 131-137.doi:10.1016/j.cyto.2013.07.003

401

[6]. Cao, Y., Xia, D. S., Qi, S. R., Du, J., Ma, P., Wang, S. L. and Fan, Z. P.

for

et

therapiae

Regenerative

Cell

beta

is

Medicine

death

an

experimentalis.2016.64:

Applications.

and

irradiation-induced

19

Stem

cells

differentiation.2010.17:

stromal

growth

factor.

20

402

Epiregulin can promote proliferation of stem cells from the dental apical papilla

403

via MEK/Erk and JNK signalling pathways. Cell proliferation.2013.46:

404

447-456.doi:10.1111/cpr.12039

405

[7]. Chambers, I., Silva, J., Colby, D., Nichols, J., Nijmeijer, B., Robertson, M.,

406

Vrana, J., Jones, K., Grotewold, L. and Smith, A. Nanog safeguards pluripotency

407

and

408

1230-1234.doi:10.1038/nature06403

409

[8]. da Silva Meirelles, L., Caplan, A. I. and Nardi, N. B. In search of the in vivo

410

identity

411

2287-2299.doi:10.1634/stemcells.2007-1122

412

[9]. Danisovic, L., Varga, I., Polak, S., Ulicna, M., Hlavackova, L., Bohmer, D.

413

and Vojtassak, J. Comparison of in vitro chondrogenic potential of human

414

mesenchymal stem cells derived from bone marrow and adipose tissue. General

415

physiology and biophysics.2009.28: 56-62

416

[10]. de Girolamo, L., Bertolini, G., Cervellin, M., Sozzi, G. and Volpi, P.

417

Treatment of chondral defects of the knee with one step matrix-assisted

418

technique enhanced by autologous concentrated bone marrow: in vitro

419

characterisation of mesenchymal stem cells from iliac crest and subchondral

420

bone. Injury.2010.41: 1172-1177.doi:10.1016/j.injury.2010.09.027

421

[11]. Ding, L., Zhu, H., Yang, Y., Wang, Z. D., Zheng, X. L., Yan, H. M., Dong,

422

L., Zhang, H. H., Han, D. M., Xue, M., Liu, J., Zhu, L., Guo, Z. K. and Wang, H.

423

X. Functional mesenchymal stem cells remain present in bone marrow

mediates

of

germline

mesenchymal

development.

stem

20

cells.

Nature.2007.450:

Stem

cells.2008.26:

21

424

microenvironment of patients with leukemia post-allogeneic hematopoietic stem

425

cell

426

1635-1644.doi:10.3109/10428194.2013.858815

427

[12]. Dominici, M., Le Blanc, K., Mueller, I., Slaper-Cortenbach, I., Marini, F.,

428

Krause, D., Deans, R., Keating, A., Prockop, D. and Horwitz, E. Minimal

429

criteria for defining multipotent mesenchymal stromal cells. The International

430

Society

431

315-317.doi:10.1080/14653240600855905

432

[13]. Eom, Y. W., Oh, J. E., Lee, J. I., Baik, S. K., Rhee, K. J., Shin, H. C., Kim,

433

Y. M., Ahn, C. M., Kong, J. H., Kim, H. S. and Shim, K. Y. The role of growth

434

factors in maintenance of stemness in bone marrow-derived mesenchymal stem

435

cells.

436

16-22.doi:10.1016/j.bbrc.2014.01.084

437

[14]. Friedenstein, A. J., Chailakhyan, R. K., Latsinik, N. V., Panasyuk, A. F. and

438

Keiliss-Borok,

439

microenvironment of

440

retransplantation in vivo. Transplantation.1974.17: 331-340

441

[15]. Friedenstein, A. J., Petrakova, K. V., Kurolesova, A. I. and Frolova, G. P.

442

Heterotopic of bone marrow. Analysis of precursor cells for osteogenic and

443

hematopoietic tissues. Transplantation.1968.6: 230-247

444

[16]. Galindo, S., Herreras, J. M., Lopez-Paniagua, M., Rey, E., de la Mata, A.,

445

Plata-Cordero, M., Calonge, M. and Nieto-Miguel, T. Therapeutic Effect of

transplant.

for

Cellular

Leukemia

Therapy

Biochemical and

I.

V.

position

biophysical

Stromal

&

cells

lymphoma.2014.55:

statement.

research

communications.2014.445:

responsible

the hemopoietic tissues.

21

Cytotherapy.2006.8:

for

transferring

Cloning in

vitro

the and

22

446

Human Adipose Tissue-Derived Mesenchymal Stem Cells in Experimental

447

Corneal Failure Due to Limbal Stem Cell Niche Damage. Stem cells.2017.35:

448

2160-2174.doi:10.1002/stem.2672

449

[17]. Ge, Y., Gomez, N. C., Adam, R. C., Nikolova, M., Yang, H., Verma, A., Lu,

450

C. P., Polak, L., Yuan, S., Elemento, O. and Fuchs, E. Stem Cell Lineage

451

Infidelity

452

636-650.e614.doi:10.1016/j.cell.2017.03.042

453

[18]. Griffin, M. D., Elliman, S. J., Cahill, E., English, K., Ceredig, R. and Ritter,

454

T. Concise review: adult mesenchymal stromal cell therapy for inflammatory

455

diseases:

456

2033-2041.doi:10.1002/stem.1452

457

[19]. Guo, Z., Li, H., Li, X., Yu, X., Wang, H., Tang, P. and Mao, N. In vitro

458

characteristics and in vivo immunosuppressive activity of compact bone-derived

459

murine

460

992-1000.doi:10.1634/stemcells.2005-0224

461

[20]. Gutierrez, G. M., Kong, E., Sabbagh, Y., Brown, N. E., Lee, J. S., Demay,

462

M. B., Thomas, D. M. and Hinds, P. W. Impaired bone development and

463

increased mesenchymal progenitor cells in calvaria of RB1-/- mice. Proceedings

464

of the National Academy of Sciences of the United States of America.2008.105:

465

18402-18407.doi:10.1073/pnas.0805925105

466

[21]. Ilas, D. C., Churchman, S. M., McGonagle, D. and Jones, E. Targeting

467

subchondral bone mesenchymal stem cell activities for intrinsic joint repair in

Drives

how

well

Wound

are

mesenchymal

we

Repair

joining

progenitor

22

and

the

Cancer.

dots?

cells.

Stem

Stem

Cell.2017.169:

cells.2013.31:

cells.2006.24:

23

468

osteoarthritis. Future science OA.2017.3: Fso228.doi:10.4155/fsoa-2017-0055

469

[22]. Im, G. I. Bone marrow-derived stem/stromal cells and adipose

470

tissue-derived stem/stromal cells: Their comparative efficacies and synergistic

471

effects.

472

2640-2648.doi:10.1002/jbm.a.36089

473

[23]. Ishii, M., Kino, J., Ichinohe, N., Tanimizu, N., Ninomiya, T., Suzuki, H.,

474

Mizuguchi, T., Hirata, K. and Mitaka, T. Hepatocytic parental progenitor cells of

475

rat small hepatocytes maintain self-renewal capability after long-term culture.

476

Scientific reports.2017.7: 46177.doi:10.1038/srep46177

477

[24]. Jiang, X. X., Zhang, Y., Liu, B., Zhang, S. X., Wu, Y., Yu, X. D. and Mao,

478

N. Human mesenchymal stem cells inhibit differentiation and function of

479

monocyte-derived

480

4120-4126.doi:10.1182/blood-2004-02-0586

481

[25]. Kfoury, Y. and Scadden, D. T. Mesenchymal cell contributions to the stem

482

cell niche. Cell stem cell.2015.16: 239-253.doi:10.1016/j.stem.2015.02.019

483

[26]. Kocan, B., Maziarz, A., Tabarkiewicz, J., Ochiya, T. and Banas-Zabczyk, A.

484

Trophic Activity and Phenotype of Adipose Tissue-Derived Mesenchymal Stem

485

Cells as a Background of Their Regenerative Potential.

486

international.2017.2017: 1653254.doi:10.1155/2017/1653254

487

[27]. Lavial, F., Acloque, H., Bertocchini, F., Macleod, D. J., Boast, S.,

488

Bachelard, E., Montillet, G., Thenot, S., Sang, H. M., Stern, C. D., Samarut, J.

489

and Pain, B. The Oct4 homologue PouV and Nanog regulate pluripotency in

Journal

of

biomedical

materials

dendritic

research

cells.

23

Part

A.2017.105:

Blood.2005.105:

Stem cells

24

490

chicken embryonic stem cells. Development (Cambridge, England).2007.134:

491

3549-3563.doi:10.1242/dev.006569

492

[28]. Lee, T. C., Lee, T. H., Huang, Y. H., Chang, N. K., Lin, Y. J., Chien, P. W.,

493

Yang, W. H. and Lin, M. H. Comparison of surface markers between human and

494

rabbit

495

e111390.doi:10.1371/journal.pone.0111390

496

[29]. Ma, X. H., Xu, X., Zou, C. Y., Zhao, Y., Wang, Z. J., Wang, H. Y., Wang, Y.

497

F. and Hu, Z. B. [Effect of Human Umbilical Cord-derived Mesenchymal Stem

498

Cells on Proliferation and Differentiation of Leukemia Cells]. Zhongguo shi yan

499

xue

500

1710-1715.doi:10.7534/j.issn.1009-2137.2016.06.017

501

[30]. McKee, C. and Chaudhry, G. R. Advances and challenges in stem cell

502

culture.

503

62-77.doi:10.1016/j.colsurfb.2017.07.051

504

[31]. Mendez-Ferrer, S., Michurina, T. V., Ferraro, F., Mazloom, A. R.,

505

Macarthur, B. D., Lira, S. A., Scadden, D. T., Ma'ayan, A., Enikolopov, G. N.

506

and Frenette, P. S. Mesenchymal and haematopoietic stem cells form a unique

507

bone marrow niche. Nature.2010.466: 829-834.doi:10.1038/nature09262

508

[32]. Ochoa-Gonzalez, F., Cervantes-Villagrana, A. R., Fernandez-Ruiz, J. C.,

509

Nava-Ramirez, H. S., Hernandez-Correa, A. C., Enciso-Moreno, J. A. and

510

Castaneda-Delgado, J. E. Metformin Induces Cell Cycle Arrest, Reduced

511

Proliferation, Wound Healing Impairment In Vivo and Is Associated to Clinical

mesenchymal

stem

ye

Colloids

cells.

xue

and

PloS

za

surfaces

24

B,

one.2014.9:

zhi.2016.24:

Biointerfaces.2017.159:

25

512

Outcomes

in

Diabetic

513

e0150900.doi:10.1371/journal.pone.0150900

514

[33]. Saunders, A., Faiola, F. and Wang, J. Concise review: pursuing

515

self-renewal and pluripotency with the stem cell factor Nanog. Stem

516

cells.2013.31: 1227-1236.doi:10.1002/stem.1384

517

[34]. Seo, E., Basu-Roy, U., Zavadil, J., Basilico, C. and Mansukhani, A.

518

Distinct functions of Sox2 control self-renewal and differentiation in the

519

osteoblast

520

4593-4608.doi:10.1128/mcb.05798-11

521

[35]. Strioga, M., Viswanathan, S., Darinskas, A., Slaby, O. and Michalek, J.

522

Same or not the same? Comparison of adipose tissue-derived versus bone

523

marrow-derived mesenchymal stem and stromal cells. Stem cells and

524

development.2012.21: 2724-2752.doi:10.1089/scd.2011.0722

525

[36]. Tan, S. L., Ahmad, T. S., Selvaratnam, L. and Kamarul, T. Isolation,

526

characterization and the multi-lineage differentiation potential of rabbit bone

527

marrow-derived mesenchymal stem cells. Journal of anatomy.2013.222:

528

437-450.doi:10.1111/joa.12032

529

[37]. Tatullo, M., Codispoti, B., Pacifici, A., Palmieri, F., Marrelli, M., Pacifici,

530

L. and Paduano, F. Potential Use of Human Periapical Cyst-Mesenchymal Stem

531

Cells (hPCy-MSCs) as a Novel Stem Cell Source for Regenerative Medicine

532

Applications.

533

103.doi:10.3389/fcell.2017.00103

lineage.

Frontiers

Foot

Ulcer

Molecular

in

cell

and

and

25

Patients.

PloS

cellular

developmental

one.2016.11:

biology.2011.31:

biology.2017.5:

26

534

[38]. Udalamaththa, V. L., Jayasinghe, C. D. and Udagama, P. V. Potential role

535

of herbal remedies in stem cell therapy: proliferation and differentiation of

536

human mesenchymal stromal cells. Stem cell research & therapy.2016.7:

537

110.doi:10.1186/s13287-016-0366-4

538

[39]. Varas, F., Grande, T., Ramirez, A. and Bueren, J. A. Implantation of bone

539

marrow beneath the kidney capsule results in transfer not only of functional

540

stroma but also of hematopoietic repopulating cells. Blood.2000.96: 2307-2309

541

[40]. Wang, Y., Xu, J., Zhang, X., Wang, C., Huang, Y., Dai, K. and Zhang, X.

542

TNF-alpha-induced LRG1 promotes angiogenesis and mesenchymal stem cell

543

migration in the subchondral bone during osteoarthritis. Cell death &

544

disease.2017.8: e2715.doi:10.1038/cddis.2017.129

545

[41]. Xu, F. F., Zhu, H., Li, X. M., Yang, F., Chen, J. D., Tang, B., Sun, H. G.,

546

Chu, Y. N., Zheng, R. X., Liu, Y. L., Wang, L. S. and Zhang, Y. Intercellular

547

adhesion molecule-1 inhibits osteogenic differentiation of mesenchymal stem

548

cells and impairs bio-scaffold-mediated bone regeneration in vivo. Tissue

549

engineering Part A.2014.20: 2768-2782.doi:10.1089/ten.TEA.2014.0007

550

[42]. Xu, L., Liu, Y., Sun, Y., Wang, B., Xiong, Y., Lin, W., Wei, Q., Wang, H.,

551

He, W., Wang, B. and Li, G. Tissue source determines the differentiation

552

potentials of mesenchymal stem cells: a comparative study of human

553

mesenchymal stem cells from bone marrow and adipose tissue. Stem cell

554

research & therapy.2017.8: 275.doi:10.1186/s13287-017-0716-x

555

[43]. Yu, S. J., Kim, H. J., Lee, E. S., Park, C. G., Cho, S. J. and Jeon, S. H. 26

27

556

beta-Catenin Accumulation Is Associated With Increased Expression of Nanog

557

Protein

558

transplantation.2017.26: 365-377.doi:10.3727/096368916x693040

559

[44]. Zedde, P., Cudoni, S., Giachetti, G., Manunta, M. L., Masala, G., Brunetti,

560

A. and Manunta, A. F. Subchondral bone remodeling: comparing nanofracture

561

with

562

87-93.doi:10.11138/jts/2016.4.2.087

563

[45]. Zhang, Y., Li, C., Jiang, X., Zhang, S., Wu, Y., Liu, B., Tang, P. and Mao,

564

N. Human placenta-derived mesenchymal progenitor cells support culture

565

expansion of long-term culture-initiating cells from cord blood CD34+ cells.

566

Experimental hematology.2004.32: 657-664.doi:10.1016/j.exphem.2004.04.001

567

[46]. Zhen, G., Wen, C., Jia, X., Li, Y., Crane, J. L., Mears, S. C., Askin, F. B.,

568

Frassica, F. J., Chang, W., Yao, J., Carrino, J. A., Cosgarea, A., Artemov, D.,

569

Chen, Q., Zhao, Z., Zhou, X., Riley, L., Sponseller, P., Wan, M., Lu, W. W. and

570

Cao, X. Inhibition of TGF-beta signaling in mesenchymal stem cells of

571

subchondral

572

704-712.doi:10.1038/nm.3143

573

[47]. Zhu, H., Guo, Z. K., Jiang, X. X., Li, H., Wang, X. Y., Yao, H. Y., Zhang, Y.

574

and Mao, N. A protocol for isolation and culture of mesenchymal stem cells

575

from

576

550-560.doi:10.1038/nprot.2009.238

577

[48]. Zhu, H., Jiang, X. X., Guo, Z. K., Li, H., Su, Y. F., Yao, H. Y., Wang, X. Y.,

and

Predicts

microfracture.

bone

mouse

Maintenance

An

attenuates

ovine

of

in

MSC

vivo

osteoarthritis.

compact

bone.

27

Self-Renewal.

study.

Nature

Nature

Cell

Joints.2016.4:

medicine.2013.19:

protocols.2010.5:

28

578

Li, X. S., Wu, Y., Liu, Y. L., Zhang, Y. and Mao, N. Tumor necrosis factor-alpha

579

alters the modulatory effects of mesenchymal stem cells on osteoclast formation

580

and

581

1473-1484.doi:10.1089/scd.2009.0021

function.

Stem

cells

582 583

28

and

development.2009.18:

29

584

Figure Legends

585

Figure 1. Morphologic and immuno-phenotypic features of SCB-MSCs and

586

BMS-MSCs

587

588 589

A: The morphologic characteristics of MSCs in two groups. B: The

590

immuno-phenotypic features of two groups. Both groups were homogenously

591

positive for mesenchymal markers but negative for hematopoietic markers. C:

592

The comparison of CD45+ cells between the SCB-MSC group and BMS-MSC

593

group (3.31 ± 0.78% vs 13.93 ± 1.63%) (**P < 0.01). The bar represents 200 µm

594

in Figure 1A. SCB-MSCs, subchondral bone-derived MSCs; BMS-MSCs, bone

595

marrow suspension-derived MSCs.

596 597

Figure 2. Results of multi-differentiation induction and RT-PCR assay

598 29

30

599 600

A: ALP and Oil-Red-O staining showed higher osteogenic and chondrogenic

601

potential in the SCB-MSC group after induction. There were no significant

602

differences in adipogenic potential between the two groups. B: Comparison of

603

mRNA expression levels of osteogenic (Runx-2 and OPN), chondrogenic (Sox-9

604

and collagen I) and adipogenic (CEBP/α and PPARγ) markers between the two

605

groups. The bar represents 200 µm in Figure 2A. SCB-MSCs, subchondral

606

bone-derived MSCs; BMS-MSCs, bone marrow suspension-derived MSCs.

607 608 609

Figure 3. CFU-F assay, sphere formation assay, and stemness markers

610 30

31

611 612

A, B: CFU-F frequency remained relatively higher in the SCB-MSC group than

613

in the BMS-MSC group. The bar represents 1 cm in Figure 3A. SCB-MSCs,

614

subchondral

615

suspension-derived MSCs. C, D: The results of primary and secondary sphere

616

culture revealed a significant difference between the two groups. The bars

bone-derived

MSCs;

31

BMS-MSCs,

bone

marrow

32

617

represent 100 µm in Figure 3C upper and 200 µm in Figure 3C low, respectively.

618

E: Comparison of mRNA expression of several stemness markers (Nanog,

619

OCT4, and Sox-2) between two groups.

620

Figure 4. SCB-MSCs display enhanced proliferative capacity

621

622 623

A: CCK-8-based cell proliferation assay indicated that the SCB-MSC group

624

harbors stronger proliferative potential than the BMS-MSC group (*P < 0.05). B:

625

CFSE data on days 2 and 4 showed that a greater proportion of SCB-MSCs

626

underwent cell division, indicating enhanced proliferation potential. C: The

627

results of cell cycle analysis showed a higher percentage of SCB-MSCs (50 ±

628

1.41%) were in the S phase compared to BMS-MSCs (36.5 ± 3.55%). D: The 32

33

629

data showed enhanced Erk1/2 phosphorylation in passages 3 and 6 SCB-MSCs.

630

SCB-MSCs, subchondral bone-derived MSCs; BMS-MSC, bone marrow

631

suspension-derived MSCs.

632 633

Table 1. Table 1: Primer sequences

634

genes HPRT Runx2 OPN

CEBP/α

PPARγ

primer sequences forward, reverse, forward, reverse, forward, reverse,

5′5′5′5′5′5′-

forward,

5′- GGGACGCTAGGTGACAGAAT -3′

reverse,

5′- GAAAGGACGCTGGCTGAAAA -3′

forward,

5′- TTGCTGTGGGGATGTCTCAT-3′

reverse,

5′- TTTCCTGTCAAGATCGCCCT-3′

Annealing temperature

GACCAGTCAACAGGGGACAT -3′ ACACTTCGAGGGGTCCTTTT -3′ ATTTCTCACCTCCTCAGCCC -3′ TCCCAAGTTTCCCTCATCCC -3′ TTTTGTCTCTTGGGCATGGC -3′ GCATTCTGCGGTGTTAGGAG-3′

60 °C Sox9

Collage I

Nanog

4-Oct

forward,

5′- ATGAAGATGACCGA CGAGCA -3′

reverse, forward,

5′- ACTTGTCCTCTTCGCTCTCC -3′ 5′- CCAAGGGAGAGCAAGGAGAA-3′

reverse,

5′- CCTTTGGGGCCTTCTTTTCC- 3′

forward,

5′-AAAACTCCCGACTCTGCAGA -3′

reverse,

5′-AGGCTGGAGAGTTCTTGCAT -3′

forward,

5′-CGGAAGAGAAAGCGAACGAG -3′ 33

34

Sox2

reverse,

5′-TGGCCTCAAAATCCTCTCGT -3′

forward,

5′-AAGGGAAATGGGGAGAGGTG - 3′

reverse,

5′-TGGATGGGATTGGTGGTCTC -3′

635

34