Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

Postal Subscription Code 80-967

2018 Impact Factor: 1.847

Front. Med.    2019, Vol. 13 Issue (5) : 575-589    https://doi.org/10.1007/s11684-019-0693-9
RESEARCH ARTICLE
Glycosylation of dentin matrix protein 1 is critical for fracture healing via promoting chondrogenesis
Hui Xue1, Dike Tao1, Yuteng Weng1, Qiqi Fan1, Shuang Zhou1, Ruilin Zhang1, Han Zhang2, Rui Yue3, Xiaogang Wang4(), Zuolin Wang1(), Yao Sun1()
1. Department of Implantology, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200072, China
2. School & Hospital of Stomatology, Tongji University, Shanghai 200072, China
3. School of Life Sciences and Technology, Tongji University, Shanghai 200072, China
4. Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing 100083, China
 Download: PDF(12088 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Fractures are frequently occurring diseases that endanger human health. Crucial to fracture healing is cartilage formation, which provides a bone-regeneration environment. Cartilage consists of both chondrocytes and extracellular matrix (ECM). The ECM of cartilage includes collagens and various types of proteoglycans (PGs), which play important roles in maintaining primary stability in fracture healing. The PG form of dentin matrix protein 1 (DMP1-PG) is involved in maintaining the health of articular cartilage and bone. Our previous data have shown that DMP1-PG is richly expressed in the cartilaginous calluses of fracture sites. However, the possible significant role of DMP1-PG in chondrogenesis and fracture healing is unknown. To further detect the potential role of DMP1-PG in fracture repair, we established a mouse fracture model by using a glycosylation site mutant DMP1 mouse (S89G-DMP1 mouse). Upon inspection, fewer cartilaginous calluses and down-regulated expression levels of chondrogenesis genes were observed in the fracture sites of S89G-DMP1 mice. Given the deficiency of DMP1-PG, the impaired IL-6/JAK/STAT signaling pathway was observed to affect the chondrogenesis of fracture healing. Overall, these results suggest that DMP1-PG is an indispensable proteoglycan in chondrogenesis during fracture healing.

Keywords fracture      extracellular matrix      dentin matrix protein 1      proteoglycan      cartilage     
Corresponding Author(s): Xiaogang Wang,Zuolin Wang,Yao Sun   
Just Accepted Date: 04 April 2019   Online First Date: 09 May 2019    Issue Date: 14 October 2019
 Cite this article:   
Hui Xue,Dike Tao,Yuteng Weng, et al. Glycosylation of dentin matrix protein 1 is critical for fracture healing via promoting chondrogenesis[J]. Front. Med., 2019, 13(5): 575-589.
 URL:  
https://academic.hep.com.cn/fmd/EN/10.1007/s11684-019-0693-9
https://academic.hep.com.cn/fmd/EN/Y2019/V13/I5/575
Fig.1  Expression of DMP1-PG in cartilage callus during fracture healing. (A) H&E and Toluidine blue staining of unfractured femurs and fractured femurs is shown at days 7, 21, and 28 post-fracture in WT mice. Scale bars= 500 mm. (B) Weaker cartilaginous calluses were formed in the 12-month-old WT mice compared with those of the 3-month-old WT mice at day 7 post-fracture. Lower magnification, scale bars= 500 mm; higher magnification, scale bars= 200 mm. (C) RT-qPCR quantification of Acan, Bgn, Dcn, Vcan, and Dmp1 in the fracture calluses from normal mice and fracture model mice. Data are shown as mean±SEM. *P<0.05, n = 3 per time point. NS, not significant. (D) DMP1-F/DMP1-PG was expressed in the cartilaginous calluses of the fracture model mice. Lower magnification, scale bars= 100 mm; higher magnification, scale bars= 50 mm. (E) The expression level of DMP1-PG was downregulated in the fracture callus of the 12-month-old WT mice compared that of the 3-month-old WT mice at day 7 post-operation by Western immunoblotting. *P<0.05, n = 3 per group. 3m, 3-month-old; 12m, 12-month-old.
Fig.2  Delayed femur fracture healing in S89G-DMP1 mice. (A) A schematic of the DMP1-PG point mutation model is shown. (B) Decreased DMP1-PG expression in the fracture callus of the S89G-DMP1 mice is shown at day 7 post-fracture by Western immunoblotting. (C) Representative three-dimensional reconstruction of micro-CT images of fracture sites at days 7, 14, 21, and 28 post-fracture. Arrows show the fracture gaps and calluses. (D) The quantitative micro-CT analysis of fracture callus is shown as mean±SEM. *P<0.05, n = 5 per group per time point. BV/TV, callus bone volume; BMD, bone mineral density; Tb.N, trabecular number; Tb.Th, trabecular thickness; Tb.Sp, trabecular space. (E) Biomechanical testing of the fractured femurs displaying the impaired mechanical property of the new bone in S89G-DMP1 mice at day 28 post-fracture. *P<0.05, n = 5 per group per time point.
Fig.3  Histological analysis of the femur fracture healing in WT and S89G-DMP1 mice. (A) H&E, Toluidine blue, and Safranin O staining of fracture calluses showed small cartilaginous calluses and new bone calluses in S89G-DMP1 mice. Scale bars= 500 mm. (B) Quantification of cartilaginous callus areas from (A) is shown as mean±SEM. *P<0.05, n = 5 per group per time point. (C) Quantification of new bone callus areas from (A) is shown as mean±SEM. *P<0.05, n = 5 per group per time point.
Fig.4  Alterations of cartilage markers in the cartilaginous calluses of the WT and S89G-DMP1 mice. (A1–A6, B1–B6) Immunohistochemistry staining of COL-II, COL-X, SOX9, ACAN, DCN, and VCAN in cartilaginous callus at day 7 post-fracture. Samples from S89G-DMP1 mice showed remarkably decreased immunoreactivity. Scale bars= 100 mm. (C1–C6) Quantitative measurements of the positive zone area/total callus area or the number of positive cells/number of total cells from the WT and S89G-DMP1 mice are shown as mean±SEM. *P<0.05, n = 4 per group. (D1–D6) RT-qPCR quantification analysis of Col-II, Col-X, Sox9 Acan, Dcn, and Vcan is presented as mean±SEM. *P<0.05, n = 4–5 per group per time point.
Fig.5  BMSCs differentiation, proliferation, and migration of WT and S89G-DMP1 mice. (A) Comparison of cultured aggregates from WT and S89G-DMP1 mice is shown by Toluidine blue staining. Scale bars= 1500 mm. (B) The mRNA levels of chondrogenesis-related genes and proteoglycan genes were determined by RT-qPCR. *P<0.05, n = 3 samples per group. (C) A CCK8 assay was performed to analyze the proliferation ability of BMSCs. *P<0.05, n = 5 samples per group. (D) Representative images of Transwell migration assay of BMSCS from the WT group and the S89G-DMP1 group. (E) The quantification of invasive cells of Transwell migration assay is shown as mean±SEM. *P<0.05, n = 4 samples per group. No difference was found in the ability of migration of BMSCs between the WT and S89G-DMP1 mice.
Fig.6  Differentially expressed mRNA sequences between WT and S89G-DMP1 fracture calluses. (A) Heat map and (B) Volcano plot analysis displayed differentially expressed genes of fracture calluses between the WT and DMP1-S89G mice. KEGG analysis showed the top ten upregulated (C) and downregulated (D) signaling pathways of fracture calluses in S89G-DMP1 mice compared with WT mice.
Fig.7  Detection of IL-6/JAK/STAT signaling in the WT and S89G-DMP1 mice. (A) RT-qPCR quantification of inflammatory molecules during fracture healing is shown as mean±SEM. *P<0.05, n = 6 per group per time point. (B) IL-6 (green) immunofluorescence staining of fracture sites at days 1 and 3 post-fracture. Arrows showed the fracture ends. Lower magnification, scale bars= 500 mm; higher magnification, scale bars= 200 mm. (C) RT-qPCR quantification of JAK/STAT signaling molecules JAK-2 and STAT-3 in the fracture calluses at days 1 and 3 post-fracture is shown as mean±SEM. *P<0.05, n = 4 per group per time point. (D) Expression of phosphorylated STAT-3 was downregulated in the fracture site in S89G-DMP1 mice by Western immunoblotting. *P<0.05, n = 3 per group per time point. (E) Loss of DMP1-PG affected chondrogenesis in fracture healing.
1 A Schindeler, MM McDonald, P Bokko, DG Little. Bone remodeling during fracture repair: the cellular picture. Semin Cell Dev Biol 2008; 19(5): 459–466
https://doi.org/10.1016/j.semcdb.2008.07.004 pmid: 18692584
2 ZS Ai-Aql, AS Alagl, DT Graves, LC Gerstenfeld, TA Einhorn. Molecular mechanisms controlling bone formation during fracture healing and distraction osteogenesis. J Dent Res 2008; 87(2): 107–118
https://doi.org/10.1177/154405910808700215 pmid: 18218835
3 L Claes, S Recknagel, A Ignatius. Fracture healing under healthy and inflammatory conditions. Nat Rev Rheumatol 2012; 8(3): 133–143
https://doi.org/10.1038/nrrheum.2012.1 pmid: 22293759
4 JN Williams, AV Kambrath, RB Patel, KS Kang, E Mével, Y Li, YH Cheng, AJ Pucylowski, MA Hassert, MJ Voor, MA Kacena, WR Thompson, SJ Warden, DB Burr, MR Allen, AG Robling, U Sankar. Inhibition of CaMKK2 enhances fracture healing by stimulating indian hedgehog signaling and accelerating endochondral ossification. J Bone Miner Res 2018; 33(5): 930–944
https://doi.org/10.1002/jbmr.3379 pmid: 29314250
5 GS Baht, L Vi, BA Alman. The role of the immune cells in fracture healing. Curr Osteoporos Rep 2018; 16(2): 138–145
https://doi.org/10.1007/s11914-018-0423-2 pmid: 29508143
6 R Dimitriou, E Tsiridis, PV Giannoudis. Current concepts of molecular aspects of bone healing. Injury 2005; 36(12): 1392–1404
https://doi.org/10.1016/j.injury.2005.07.019 pmid: 16102764
7 Y Sun, Y Weng, C Zhang, Y Liu, C Kang, Z Liu, B Jing, Q Zhang, Z Wang. Glycosylation of dentin matrix protein 1 is critical for osteogenesis. Sci Rep 2015; 5(1): 17518
https://doi.org/10.1038/srep17518 pmid: 26634432
8 LE Bertassoni, MV Swain. The contribution of proteoglycans to the mechanical behavior of mineralized tissues. J Mech Behav Biomed Mater 2014; 38: 91–104
https://doi.org/10.1016/j.jmbbm.2014.06.008 pmid: 25043659
9 Y Weng, Y Liu, H Du, L Li, B Jing, Q Zhang, X Wang, Z Wang, Y Sun. Glycosylation of DMP1 is essential for chondrogenesis of condylar cartilage. J Dent Res 2017; 96(13): 1535–1545
https://doi.org/10.1177/0022034517717485 pmid: 28759313
10 JI Furukawa, K Okada, Y Shinohara. Glycomics of human embryonic stem cells and human induced pluripotent stem cells. Glycoconj J 2017; 34(6): 807–815
https://doi.org/10.1007/s10719-017-9800-9 pmid: 29150735
11 Y Gao, S Liu, J Huang, W Guo, J Chen, L Zhang, B Zhao, J Peng, A Wang, Y Wang, W Xu, S Lu, M Yuan, Q Guo. The ECM-cell interaction of cartilage extracellular matrix on chondrocytes. BioMed Res Int 2014; 2014: 648459
https://doi.org/10.1155/2014/648459 pmid: 24959581
12 MC Embree, TM Kilts, M Ono, CA Inkson, F Syed-Picard, MA Karsdal, A Oldberg, Y Bi, MF Young. Biglycan and fibromodulin have essential roles in regulating chondrogenesis and extracellular matrix turnover in temporomandibular joint osteoarthritis. Am J Pathol 2010; 176(2): 812–826
https://doi.org/10.2353/ajpath.2010.090450 pmid: 20035055
13 M Myren, DJ Kirby, ML Noonan, A Maeda, RT Owens, S Ricard-Blum, V Kram, TM Kilts, MF Young. Biglycan potentially regulates angiogenesis during fracture repair by altering expression and function of endostatin. Matrix Biol 2016; 52-54: 141–150
https://doi.org/10.1016/j.matbio.2016.03.008 pmid: 27072616
14 AD Berendsen, EL Pinnow, A Maeda, AC Brown, N McCartney-Francis, V Kram, RT Owens, PG Robey, K Holmbeck, LF de Castro, TM Kilts, MF Young. Biglycan modulates angiogenesis and bone formation during fracture healing. Matrix Biol 2014; 35: 223–231
https://doi.org/10.1016/j.matbio.2013.12.004 pmid: 24373744
15 A George, B Sabsay, PA Simonian, A Veis. Characterization of a novel dentin matrix acidic phosphoprotein. Implications for induction of biomineralization. J Biol Chem 1993; 268(17): 12624–12630
pmid: 8509401
16 RN D’Souza, A Cavender, G Sunavala, J Alvarez, T Ohshima, AB Kulkarni, M MacDougall. Gene expression patterns of murine dentin matrix protein 1 (Dmp1) and dentin sialophosphoprotein (DSPP) suggest distinct developmental functions in vivo. J Bone Miner Res 1997; 12(12): 2040–2049
https://doi.org/10.1359/jbmr.1997.12.12.2040 pmid: 9421236
17 C Qin, JC Brunn, RG Cook, RS Orkiszewski, JP Malone, A Veis, WT Butler. Evidence for the proteolytic processing of dentin matrix protein 1. Identification and characterization of processed fragments and cleavage sites. J Biol Chem 2003; 278(36): 34700–34708
https://doi.org/10.1074/jbc.M305315200 pmid: 12813042
18 C Qin, R D’Souza, JQ Feng. Dentin matrix protein 1 (DMP1): new and important roles for biomineralization and phosphate homeostasis. J Dent Res 2007; 86(12): 1134–1141
https://doi.org/10.1177/154405910708601202 pmid: 18037646
19 Y Lu, B Yuan, C Qin, Z Cao, Y Xie, SL Dallas, MD McKee, MK Drezner, LF Bonewald, JQ Feng. The biological function of DMP-1 in osteocyte maturation is mediated by its 57-kDa C-terminal fragment. J Bone Miner Res 2011; 26(2): 331–340
https://doi.org/10.1002/jbmr.226 pmid: 20734454
20 Y Lu, C Qin, Y Xie, LF Bonewald, JQ Feng. Studies of the DMP1 57-kDa functional domain both in vivo and in vitro. Cells Tissues Organs 2009; 189(1-4): 175–185
https://doi.org/10.1159/000151727 pmid: 18728349
21 Y Sun, S Ma, J Zhou, AK Yamoah, JQ Feng, RJ Hinton, C Qin. Distribution of small integrin-binding ligand, N-linked glycoproteins (SIBLING) in the articular cartilage of the rat femoral head. J Histochem Cytochem 2010; 58(11): 1033–1043
https://doi.org/10.1369/jhc.2010.956771 pmid: 20679519
22 Y Sun, L Chen, S Ma, J Zhou, H Zhang, JQ Feng, C Qin. Roles of DMP1 processing in osteogenesis, dentinogenesis and chondrogenesis. Cells Tissues Organs 2011; 194(2-4): 199–204
https://doi.org/10.1159/000324672 pmid: 21555863
23 C Qin, B Huang, JN Wygant, BW McIntyre, CH McDonald, RG Cook, WT Butler. A chondroitin sulfate chain attached to the bone dentin matrix protein 1 NH2-terminal fragment. J Biol Chem 2006; 281(12): 8034–8040
https://doi.org/10.1074/jbc.M512964200 pmid: 16421105
24 A Gericke, C Qin, Y Sun, R Redfern, D Redfern, Y Fujimoto, H Taleb, WT Butler, AL Boskey. Different forms of DMP1 play distinct roles in mineralization. J Dent Res 2010; 89(4): 355–359
https://doi.org/10.1177/0022034510363250 pmid: 20200415
25 F Loi, LA Córdova, J Pajarinen, TH Lin, Z Yao, SB Goodman. Inflammation, fracture and bone repair. Bone 2016; 86: 119–130
https://doi.org/10.1016/j.bone.2016.02.020 pmid: 26946132
26 V Bradaschia-Correa, AM Josephson, D Mehta, M Mizrahi, SS Neibart, C Liu, OD Kennedy, AB Castillo, KA Egol, P Leucht. The selective serotonin reuptake inhibitor fluoxetine directly inhibits osteoblast differentiation and mineralization during fracture healing in mice. J Bone Miner Res 2017; 32(4): 821–833
https://doi.org/10.1002/jbmr.3045 pmid: 27869327
27 GS Baht, P Nadesan, D Silkstone, BA Alman. Pharmacologically targeting β-catenin for NF1 associated deficiencies in fracture repair. Bone 2017; 98: 31–36
https://doi.org/10.1016/j.bone.2017.02.012 pmid: 28254468
28 A Balic, HL Aguila, MJ Caimano, VP Francone, M Mina. Characterization of stem and progenitor cells in the dental pulp of erupted and unerupted murine molars. Bone 2010; 46(6): 1639–1651
https://doi.org/10.1016/j.bone.2010.02.019 pmid: 20193787
29 B Jing, C Zhang, X Liu, L Zhou, J Liu, Y Yao, J Yu, Y Weng, M Pan, J Liu, Z Wang, Y Sun, YE Sun. Glycosylation of dentin matrix protein 1 is a novel key element for astrocyte maturation and BBB integrity. Protein Cell 2018; 9(3): 298–309
https://doi.org/10.1007/s13238-017-0449-8 pmid: 28822114
30 C Wang, Y Abu-Amer, RJ O’Keefe, J Shen. Loss of Dnmt3b in chondrocytes leads to delayed endochondral ossification and fracture repair. J Bone Miner Res 2018; 33(2): 283–297
https://doi.org/10.1002/jbmr.3305 pmid: 29024060
31 GS Baht, D Silkstone, P Nadesan, H Whetstone, BA Alman. Activation of hedgehog signaling during fracture repair enhances osteoblastic-dependent matrix formation. J Orthop Res 2014; 32(4): 581–586
https://doi.org/10.1002/jor.22562 pmid: 24347536
32 M Majidinia, A Sadeghpour and B Yousefi. The roles of signaling pathways in bone repair and regeneration. J Cell Physiol 2018; 233(4): 2937–2948
https://doi.org/10.1002/jcp.26042 pmid: 28590066
33 MS Ghiasi, J Chen, A Vaziri, EK Rodriguez, A Nazarian. Bone fracture healing in mechanobiological modeling: a review of principles and methods. Bone Rep 2017; 6: 87–100
https://doi.org/10.1016/j.bonr.2017.03.002 pmid: 28377988
34 ME Klontzas, EI Kenanidis, RJ MacFarlane, T Michail, ME Potoupnis, M Heliotis, A Mantalaris, E Tsiridis. Investigational drugs for fracture healing: preclinical & clinical data. Expert Opin Investig Drugs 2016; 25(5): 585–596
https://doi.org/10.1517/13543784.2016.1161757 pmid: 26938853
35 CJ Richards, KW Graf Jr, RP Mashru. The effect of opioids, alcohol, and nonsteroidal anti-inflammatory drugs on fracture union. Orthop Clin North Am 2017; 48(4): 433–443
https://doi.org/10.1016/j.ocl.2017.06.002 pmid: 28870304
36 LC Gerstenfeld, DM Cullinane, GL Barnes, DT Graves, TA Einhorn. Fracture healing as a post-natal developmental process: molecular, spatial, and temporal aspects of its regulation. J Cell Biochem 2003; 88(5): 873–884
https://doi.org/10.1002/jcb.10435 pmid: 12616527
37 E Tsiridis, N Upadhyay, P Giannoudis. Molecular aspects of fracture healing: which are the important molecules? Injury 2007; 38(Suppl 1): S11–S25
https://doi.org/10.1016/j.injury.2007.02.006 pmid: 17383481
38 WK Hsu, PA Anderson. Odontoid fractures: update on management. J Am Acad Orthop Surg 2010; 18(7): 383–394
https://doi.org/10.5435/00124635-201007000-00001 pmid: 20595131
39 DR Williams Jr, AR Presar, AT Richmond, CH Mjaatvedt, S Hoffman, AA Capehart. Limb chondrogenesis is compromised in the versican deficient hdf mouse. Biochem Biophys Res Commun 2005; 334(3): 960–966
https://doi.org/10.1016/j.bbrc.2005.06.189 pmid: 16039617
40 MS Lord, BL Farrugia, J Rnjak-Kovacina, JM Whitelock. Current serological possibilities for the diagnosis of arthritis with special focus on proteins and proteoglycans from the extracellular matrix. Expert Rev Mol Diagn 2015; 15(1): 77–95
https://doi.org/10.1586/14737159.2015.979158 pmid: 25382274
41 D Nikitovic, J Aggelidakis, MF Young, RV Iozzo, NK Karamanos, GN Tzanakakis. The biology of small leucine-rich proteoglycans in bone pathophysiology. J Biol Chem 2012; 287(41): 33926–33933
https://doi.org/10.1074/jbc.R112.379602 pmid: 22879588
42 S Li, J Cao, B Caterson, CE Hughes. Proteoglycan metabolism, cell death and Kashin-Beck disease. Glycoconj J 2012; 29(5-6): 241–248
https://doi.org/10.1007/s10719-012-9421-2 pmid: 22733148
43 KR Kukurba, SB Montgomery. RNA sequencing and analysis. Cold Spring Harb Protoc 2015; 2015(11): 951–969
https://doi.org/10.1101/pdb.top084970 pmid: 25870306
44 T Naka, N Nishimoto, T Kishimoto. The paradigm of IL-6: from basic science to medicine. Arthritis Res 2002; 4(Suppl 3): S233–S242
https://doi.org/10.1186/ar565 pmid: 12110143
45 JJ O’Shea, DM Schwartz, AV Villarino, M Gadina, IB McInnes, A Laurence. The JAK-STAT pathway: impact on human disease and therapeutic intervention. Annu Rev Med 2015; 66(1): 311–328
https://doi.org/10.1146/annurev-med-051113-024537 pmid: 25587654
46 C Liongue, R Sertori, AC Ward. Evolution of cytokine receptor signaling. J Immunol 2016;197(1):11–18
https://doi.org/10.4049/jimmunol.1600372 pmid: 27317733
47 F Beier, RF Loeser. Biology and pathology of Rho GTPase, PI-3 kinase-Akt, and MAP kinase signaling pathways in chondrocytes. J Cell Biochem 2010; 110(3): 573–580
https://doi.org/10.1002/jcb.22604 pmid: 20512918
48 NL Fazzalari. Bone fracture and bone fracture repair. Osteoporos Int 2011; 22(6): 2003–2006
https://doi.org/10.1007/s00198-011-1611-4 pmid: 21523400
49 G Sun, Z Wang, Y Ti, Y Wang, J Wang, J Zhao, H Qian. STAT3 promotes bone fracture healing by enhancing the FOXP3 expression and the suppressive function of regulatory T cells. APMIS 2017; 125(8): 752–760
https://doi.org/10.1111/apm.12706 pmid: 28493494
50 PM Mountziaris, PP Spicer, FK Kasper, AG Mikos. Harnessing and modulating inflammation in strategies for bone regeneration. Tissue Eng Part B Rev 2011; 17(6): 393–402
https://doi.org/10.1089/ten.teb.2011.0182 pmid: 21615330
51 B Osta, G Benedetti, P Miossec. Classical and paradoxical effects of TNF-a on bone homeostasis. Front Immunol 2014; 5: 48
https://doi.org/10.3389/fimmu.2014.00048 pmid: 24592264
52 AC Wu, LJ Raggatt, KA Alexander, AR Pettit. Unraveling macrophage contributions to bone repair. Bonekey Rep 2013; 2: 373
https://doi.org/10.1038/bonekey.2013.107 pmid: 25035807
53 YN Jang, EJ Baik. JAK-STAT pathway and myogenic differentiation. JAK-STAT 2013; 2(2): e23282
https://doi.org/10.4161/jkst.23282 pmid: 24058805
54 J Li. JAK-STAT and bone metabolism. JAK-STAT 2013; 2(3): e23930
https://doi.org/10.4161/jkst.23930 pmid: 24069548
55 M Kondo, K Yamaoka, K Sakata, K Sonomoto, L Lin, K Nakano, Y Tanaka. Contribution of the interleukin-6/STAT-3 signaling pathway to chondrogenic differentiation of human mesenchymal stem cells. Arthritis Rheumatol 2015; 67(5): 1250–1260
https://doi.org/10.1002/art.39036 pmid: 25604648
56 C Pass, VE MacRae, C Huesa, S Faisal Ahmed, C Farquharson. SOCS2 is the critical regulator of GH action in murine growth plate chondrogenesis. J Bone Miner Res 2012; 27(5): 1055–1066
https://doi.org/10.1002/jbmr.1544 pmid: 22228213
57 H Kim, JK Sonn. Rac1 promotes chondrogenesis by regulating STAT3 signaling pathway. Cell Biol Int 2016; 40(9): 976–983
https://doi.org/10.1002/cbin.10635 pmid: 27306109
58 KD Hankenson, M Dishowitz, C Gray, M Schenker. Angiogenesis in bone regeneration. Injury 2011; 42(6): 556–561
https://doi.org/10.1016/j.injury.2011.03.035 pmid: 21489534
59 KD Hankenson, K Gagne, M Shaughnessy. Extracellular signaling molecules to promote fracture healing and bone regeneration. Adv Drug Deliv Rev 2015; 94: 3–12
https://doi.org/10.1016/j.addr.2015.09.008 pmid: 26428617
60 BA Alman. The role of hedgehog signalling in skeletal health and disease. Nat Rev Rheumatol 2015; 11(9): 552–560
https://doi.org/10.1038/nrrheum.2015.84 pmid: 26077918
61 JQ Feng, LM Ward, S Liu, Y Lu, Y Xie, B Yuan, X Yu, F Rauch, SI Davis, S Zhang, H Rios, MK Drezner, LD Quarles, LF Bonewald, KE White. Loss of DMP1 causes rickets and osteomalacia and identifies a role for osteocytes in mineral metabolism. Nat Genet 2006; 38(11): 1310–1315
https://doi.org/10.1038/ng1905 pmid: 17033621
[1] Liting Jiang,Yinyin Xie,Li Wei,Qi Zhou,Ning Li,Xinquan Jiang,Yiming Gao. iTRAQ-based quantitative proteomic analysis on differentially expressed proteins of rat mandibular condylar cartilage induced by reducing dietary loading[J]. Front. Med., 2017, 11(1): 97-109.
[2] Shu Zhang, Yinwei Li, Xianqun Fan. Application of endoscopic techniques in orbital blowout fractures[J]. Front Med, 2013, 7(3): 328-332.
[3] Hengyun SUN, Wei LIU, Guangdong ZHOU, Wenjie ZHANG, Lei CUI, Yilin CAO. Tissue engineering of cartilage, tendon and bone[J]. Front Med, 2011, 5(1): 61-69.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed