header advert
Bone & Joint Research Logo

Receive monthly Table of Contents alerts from Bone & Joint Research

Comprehensive article alerts can be set up and managed through your account settings

View my account settings

Visit Bone & Joint Research at:

Loading...

Loading...

Open Access

Research

Stem cell therapy for enhancement of bone consolidation in distraction osteogenesis

A contemporary review of experimental studies



Download PDF

Abstract

Objectives

Distraction osteogenesis (DO) mobilises bone regenerative potential and avoids the complications of other treatments such as bone graft. The major disadvantage of DO is the length of time required for bone consolidation. Mesenchymal stem cells (MSCs) have been used to promote bone formation with some good results.

Methods

We hereby review the published literature on the use of MSCs in promoting bone consolidation during DO.

Results

Studies differed in animal type (mice, rabbit, dog, sheep), bone type (femur, tibia, skull), DO protocols and cell transplantation methods.

Conclusion

The majority of studies reported that the transplantation of MSCs enhanced bone consolidation or formation in DO. Many questions relating to animal model, DO protocol and cell transplantation regime remain to be further investigated. Clinical trials are needed to test and confirm these findings from animal studies.

Cite this article: Y. Yang, S. Lin, B. Wang, W. Gu, G. Li. Stem cell therapy for enhancement of bone consolidation in distraction osteogenesis: A contemporary review of experimental studies. Bone Joint Res 2017;6:385–390. DOI: 10.1302/2046-3758.66.BJR-2017-0023.

Article focus

  • Distraction osteogenesis (DO) harnesses the bone regenerative potential of bone and avoids the complications of other treatments such as bone grafting. The major disadvantage of DO is the length of time required for bone consolidation. Mesenchymal stem cells (MSCs) have been used to promote bone formation in this phase, with some good results. This review focuses on the use of MSCs in promoting bone consolidation during DO. Studies differed in animal type (mice, rabbit, dog, sheep), bone type (femur, tibia, skull), DO protocols and cell transplantation methods. Most studies reported that the transplantation of MSCs enhanced bone consolidation or formation in DO. Many questions relating to the animal model, DO protocol and cell transplantation regime require further investigation. A consensus is required on this before further research is undertaken. Clinical trials are needed to test and confirm these findings once a consensus has been agreed on the protocols involving animal studies.

Key messages

  • The length of time required for bone consolidation remains a problem in distraction osteogenesis (DO);

  • Mesenchymal stem cells can migrate to the sites of injury and stimulate bone regeneration. This may be used to accelerate bone consolidation in DO;

  • Despite the variation in animal model, cell source, cell number, administration method, treatment time-point and outcome assessment, MSCs proved effective in regenerating bone during DO.

Strengths and limitations

  • Strengths: three tables are used to display the animal models, outcomes, DO protocols and characteristics of MSCs which have been reported in the literature.

  • Limitations: with the eligibility criteria, the number of selected publications is only 16. This is too small to come to a convincing conclusion.

Introduction

Bone can repair and regenerate itself following damage, but not in the case of significant/segmental bone loss, when surgery with bone grafting or transplantation is usually required. The goal in bone defect management is to align the bone segments, facilitate union, maintain or obtain equal limb length, and restore the function of the limb. Many factors can influence the outcomes, and many alternative therapies have been suggested.1,2

The distraction osteogenesis (DO) technique is frequently employed to manage large bone defects.3,4 DO is widely applied in the treatment of firearm injuries, deformities, nonunion, lower-limb malignant metaphyseal bone tumours or tumour resection.5-9 Large defects or growth deficiencies severely compromise patients’ limb function and quality of life, and their treatment is a challenge.10 Compared with the conventional techniques for bone reconstruction, DO is a simpler procedure that has a shorter operation time, less blood loss, a shorter duration of hospitalisation, lower cost and risk of complications.11-13 The DO procedure involves three sequential phases: latency; active distraction or lengthening; and consolidation.14,15 It stimulates new bone formation through the controlled separation of two osteogenic fronts and also promotes the regeneration of the surrounding soft tissue.16,17 DO resembles an in vivo form of tissue engineering.18,19 It has been shown that DO induces migration of MSCs from the bone marrow to the site.20,21

Although DO is recognised as an effective and safe procedure, there are drawbacks associated with it.22 The major disadvantage relates to the duration of the bone consolidation phase, when the external fixators need to be kept in place for a prolonged period (12 months on average).5,17,23 During this period complications can arise including pin site infection and fibrous union; patients and family members may experience physical inconvenience and psychosocial burden due to the retained external fixators, and there is a higher rate of fracture at the docking site.14,24-26 These limitations hamper any large-scale clinical application of DO. The recommended rate of gradual bone lengthening as described by Ilizarov is 1 mm per day. Higher rates (1.3 mm/day or greater) may lead to tissue damage, and researchers have found that the size of the fibrous zone increased more quickly than that of the new bone zone in these conditions.27 Also, the bone-regenerating capability slows with increasing age.28

To reduce the time required for the DO process, numerous animal models were developed to mimic clinical situations and test the new treatments.29 MSCs have been used in a variety of applications owing to their regenerative potential and migration capacity. One of the most important capabilities of MSCs is their migration capacity in response to signals released from the injured tissues.30,31 The application of MSCs has been shown to improve the quality and quantity of bone healing including fractures or DO.3,32,33

Materials and Methods

This review included studies that have been published in English; PubMed, OVID and Google Scholar search engines were used. The key terms “cells” and “distraction osteogenesis” were searched, and 434 titles identified. The inclusion criteria were as follows: DO with bone lengthening and administration of MSCs with control and/or comparison group(s). The exclusion criteria included: distraction of other areas such as mandibular distraction; use of bone grafts in addition to DO; testing of new distractor device or new surgical technique; and outcomes assessing soft-tissue changes, rather than bony outcomes. Review articles, case reports and non-English language publications were excluded.

Results

A total of 16 studies meeting the eligibility criteria were selected from 423 published articles. Six studies used small animal models (rats or mice) and six studies used rabbits, while the other three studies used large animal models (two used dogs and one a goat) and one study involved a human subject (Table I). Among these studies, there were variations in the experimental design, including distraction time point, cell transplantation time point, administration methods and cell quantity, as well as outcome assessment methods. All studies used unilateral limb DO with or without cell injection.

Table I.

Characteristics of animal models and the main outcomes of the study

First author name and Year Animal Gender (n) Cells Used Control Group Main Results Objectives
Yuji Takamine, 200242 Rat Male (73) BMSCs Collagen gel Cells treated group was significantly better than that of control group Promote new bone formation and shorten the consolidation period.
Kazuhiko Kinoshita, 200837 Rabbit Male (54) BMSC Saline Same as above Promote bone regeneration of DO
Koichiro Sato, 201043 Rabbit Male (8) BMSCs PBS Same as above Promote new bone formation.
Qing-Guo Lai, 201144 Rabbit Male (54) BMSCs Saline Same as above Promote bone formation.
Masahito Fujio, 201115 ICR mice Female (83) BMECs+SDF-1 Saline Same as above Shorten the treatment period of DO.
Jan Gessmann, 201245 Human Male(6) female(2) BMSC N/A Same as above Promote bone regeneration of DO
Ozgur Sunay, 201346 Rabbit Female (21) BMSCs Saline Same as above Promote new bone formation and shorten bone consolidation phase.
Issei Nomura, 201447 Rat N/A(60) ADSC+ Collagen gel Saline Same as above Promote bone regeneration of DO
Yuji Ando, 201448 ICR mice Female (12) BMSCs FBS Same as above Shorten the distraction period.
Yohei Harada, 201520 Rabbit Male (42) BMSCs + PBS PBS Same as above Repair of large bone defects.
J. J. Zeng, 201649 Dog Male (27) BMSCs transfected with hBMP-2 PBS Same as above Promote bone regeneration of DO
Xu jia,201650 Rabbit Male(24) Human fetal MSC secretome PBS Same as above Promote bone regeneration of DO
Mohammad Mehdi Dehghan, 201536 Dog Male (10) MSC+PRP PRP Same as above Promote new bone formation and shortened the consolidation period.
El Hadidi, 201629 Goats Female (12) BMSCs PBS Same as above Improve the quality and quantity of DO.
Alexander R. Zheutlin, 201616 Lewis rats Male (30) BMSCs N/A Same as above N/A
Sung Joo Lee, 201651 Rabbit Male (32) ADSC Fibrin glue Same as above Promote bone regeneration of DO

The DO protocols are summarised in Table II. Various custom-made or commercially available distractors/lengtheners were used. The latency phase ranged between four and fourteen days. The distraction rate ranged from 0.5 mm/day to 2 mm/day, the total distraction gap ranged from 1.5 mm to 60.8 mm, and the consolidation period ranged from four days to ten weeks.

Table II.

Characteristics of distraction osteogenesis protocols

First author, year Latency time (days) Rate of lengthening (mm/day) Total lengthening (mm) Consolidation phase (days) Infection rate
Yuji Takamine, 200242 7 0.5 5.0 14/28/42/56 N/A
Kazuhiko Kinoshita, 200837 5 2.0 8.0 N/A N/A
Koichiro Sato, 200943 7 0.5 10 21 N/A
Qing-Guo Lai, 201044 6 0.8 4.8 42 N/A
Masahito Fujio, 201115 5 0.4 3.2 N/A N/A
Jan Gessmann, 201245 N/A N/A Average 82.4 N/A Six local pin infections
Ozgur Sunay, 201333 7 0.7 10.5 56 N/A
Issei Nomura, 201446 7 0.8 6.4 14/28/42 N/A
Yuji Ando, 201447 3 0.8 3.2 N/A N/A
Yohei Harada, 201538 14 N/A 1.5 28/56/84 N/A
J. J. Zeng, 201548 5 1.0 10 14/28/42/56 No infection
Xu Jia, 201549 5 1.0 10 42 N/A
Mohammad Mehdi Dehghan, 201536 7 1.0 60.8 120 N/A
El Hadidi, 201629 5 1.0 10 30 Most animals
Alexander R. Zheutlin, 201616 4 0.6 5.1 28 N/A
Sung Joo Lee, 201650 5 3.0 10 28/56/84 1

As shown in Table III, different cell sources were used: autologous or allogenic bone marrow MSCs from long bone16,20,37,43,45,48,5052 and iliac crest,30,38,44,46,47 autologous adipose stem cells,48,52 and location not specified.15,49 The number of transplanted cells ranged from 1 to 30×106. The cell injection timing included three different time points: in three studies, MSCs were injected during the distraction phase15,37,49 or during the consolidation phase;30,38,43,44,45,47,48,51 in three studies, the MSCs were injected on the day of the operation;16,20,50 and in one study the time of injection was not stated52. The majority of the studies reported that MSC transplantation at the end of the lengthening phase with MSCs over 1 x 106 enhanced bone consolidation or formation in DO in animal models.

Table III.

Characteristics of transplanted cells

First Author Name, Year Cell Type Cell Source Cell Number Time Passage of MSC
Yuji Takamine, 200242 Allogenic Femurs 0.1M When distraction phase finished 3
Kazuhiko Kinoshita, 200837 Autologous Iliac crest 10M When distraction phase finished 3
Koichiro Sato, 201043 Allogenic Iliac bone 30M When distraction phase finished 3-6
Qing-Guo Lai, 201144 Autologous Tibia 10M When distraction phase finished 3
Masahito Fujio, 201115 N/A N/A N/A Every other day from day 4. 3-6
Jan Gessmann, 201245 Autologous iliac crest 2ml At the end of the distraction phase 3
Ozgur Sunay, 201346 Autologous Inguinal regions 5M When distraction phase finished 3
Issei Nomura, 201447 Autologous Femurs 1M after termination of distraction 3
Yuji Ando, 201448 Human MSCs N/A 0.3M The second day at the distraction phase 3
Yohei Harada, 201520 Autologous Tibia 1M At surgery day 4-6
J. J. Zeng, 201649 Allogenic Tibia 1M At surgery day 3
Xu jia,201650 Allogenic Tibia N/A Every 3 days when distraction phase finished 3
Mohammad Mehdi Dehghan, 201536 N/A Tibia 10M Middle and end of the distraction phase 3
El Hadidi, 201629 Allogenic Iliac crest 15M Day 10 and 20 in the consolidation phase 3
Alexander R. Zheutlin, 201616 Allogenic Femurs and ummers 2M At surgery day 3
Sung Joo Lee, 201651 Autologous Tibia 3M N/A 3
  1. M, number of cells in millions.

All studies showed a positive effect in bone regeneration on the cell-treated side. This was regardless of whether the cell sources, cell number, and time points have different effect.

Discussion

In normal DO, the recruitment, proliferation, and osteogenic differentiation of MSCs are sufficient to achieve bone regeneration in the distraction gap (Fig. 1). However, the MSCs may be compromised under conditions such as poor vascularity, severe trauma and radiotherapy. This may result in fibrous union or nonunion.10,29,34 In order to enhance the quality and quantity, and the time to bone formation in DO, DO animal models were developed to test bone regeneration by MSC therapy approaches.35

Fig. 1 
          Summary of studies on the cell therapy including time and number of transplanted cells used.

Fig. 1

Summary of studies on the cell therapy including time and number of transplanted cells used.

In all studies reported, the MSC-treated group showed improvement in the quality and quantity of new bone formation in the distraction gap. Dehghan et al,36 Lee et al,51 and Kinoshita et al37 used the higher rate of lengthening, and the MSC therapy significantly promoted new bone formation and shortened the consolidation period in their DO models.

The total size of the distraction gap is another important factor to consider; at present, most studies conducted ⩽ 10 mm of distraction, which did not reflect the real clinical scenario where most DO treatment exceeds 5 cm or more. In the large gap, the effect of cell therapy may be more or less apparent. This needs to be tested in a clinical setting.

The timing and number of MSCs for cell injection also varied among studies. MSC isolation and cultivation are similar, though they differ from the source. All of them used MSCs at passages 2 to 6, but only one was characterised by CD33, CD45 and CD90. Most authors did the cell injection at the end of the lengthening period and used a dose of 1 x 105 to 3 x 107 MSCs, based on previous reported studies (Fig. 2). No clear explanation was given as to why a certain time was chosen for cell delivery. As shown in Tables II and III, the larger the distraction gap, the more stem cells may be needed.

Fig. 2 
          The role of MSCs in bone defect. MSCs are maintained in quiescence and transiently activated by damage. The use of MSCs to treat damage is attractive as it would implement a reparative process that should be in place and time. MSCs improved the damage healing affecting the callus. Regeneration is followed by two steps, increased migration and proliferation. MSC migration at the defect site is time-and dose-dependent. They must self-renew to produce more stem cells, maintaining tissue homeostasis. MSCs possess the capability to differentiate directly into subsequently bone-forming osteoblasts.

Fig. 2

The role of MSCs in bone defect. MSCs are maintained in quiescence and transiently activated by damage. The use of MSCs to treat damage is attractive as it would implement a reparative process that should be in place and time. MSCs improved the damage healing affecting the callus. Regeneration is followed by two steps, increased migration and proliferation. MSC migration at the defect site is time-and dose-dependent. They must self-renew to produce more stem cells, maintaining tissue homeostasis. MSCs possess the capability to differentiate directly into subsequently bone-forming osteoblasts.

None of the studies has purposely investigated what the optimal cell numbers and optimal time points are for injection. Nor has any study compared the difference between autologous or allogenic MSCs in DO.

The gender of animals may affect the outcome, however, no study has directly compared the effects of cell therapy on the DO process between male and female animals of any species. We know much more about the role of gender in medicine, such as the fact that women are more sensitive to cardiological medicine38 and women who are lactating may have worsened symptoms. With the increasing demand to eliminate gender bias in animal studies,39 future efforts are warranted to clarify gender-related differences of cell therapy in DO.

Another issue related to animal models is animal age and skeletal maturity. All animals used were skeletally mature, except the goats used by El Hadidi et al29 which were of an age comparable with childhood. Older rats display delayed healing of femoral fractures. As DO may be performed on patients of all ages, it is important clinically to interpret the data obtained from animal studies. This is in specific consideration of its application to humans of different skeletal ages. Future studies are needed to compare age-related differences on MSC therapy in DO. Through extensive research, it has become clear that different animal models may have a different optimal DO protocol. Three aspects should be considered carefully during the DO procedure: site of lengthening; lengthening rate; and length.40,41 The optimal rate of lengthening in small and large animals is commonly thought to be 0.5 mm/day and 1 mm/day, respectively, but there have been attempts using a higher rate of lengthening to create a poor bone formation model.

Adverse events, such as infection, were not carefully or purposefully investigated in any of the 16 studies using MSC therapy in DO. Most studies did not mention the infection risk assessment, and animals were simply excluded from the study due to pin site infections. Complications related to MSC therapy during limb lengthening procedure must be taken into consideration. In clinical situations, the emphasis is on the use of the GMP facility to process and prepare the cells in order to minimise the potential risks of infection.

Questions related to animal models, DO protocols and cell transplantation regimes still need further investigation. A consensus is required for the development of such a model. Clinical trials using MSC therapy for enhancement of bone formation and consolidation in patients with DO treatment are warranted, and it is the only way to obtain a definitive answer in this subject area.


G. Li; email:
Author Contribution

Y. Yang: First author, Literature review, Manuscript writing.

S. Lin: Co- first author, Manuscript revision.

B. Wang: Second author.

W. Gu: Corresponding author.

G. Li: Co-corresponding author.


Funding Statement

The work was partially supported by grants from National Natural Science Foundation of China (81371946 and 81374568); Hong Kong Government Research Grant Council, General Research Fund (470813, 14119115 and 9054014) and grants from China Shenzhen City Science and Technology Bureau (GJHZ20140419120051680 and JCYJ20150630165236960). This study was also supported in part by SMART program, Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong and the research was made possible by resources donated by Lui Che Woo Foundation Limited.

Conflicts of Interest Statement

None.

References

1 Liu L , AronsonJ, HuangS, et al.. Rosiglitazone inhibits bone regeneration and causes significant accumulation of fat at sites of new bone formation. Calcif Tissue Int2012;91:139-148.CrossrefPubMed Google Scholar

2 Morcos MW , Al-JalladH, HamdyR. Comprehensive review of adipose stem cells and their implication in distraction osteogenesis and bone regeneration. Biomed Res Int2015.CrossrefPubMed Google Scholar

3 Spiegl U , PätzoldR, FriederichsJ, et al.. Clinical course, complication rate and outcome of segmental resection and distraction osteogenesis after chronic tibial osteitis. Injury2013;44:1049-1056.CrossrefPubMed Google Scholar

4 Yang Z , JinL, TaoH, YangD. Reconstruction of large tibial bone defects following osteosarcoma resection using bone transport distraction: A report of two cases. Oncol Lett2016;12:1445-1447.CrossrefPubMed Google Scholar

5 He X , ZhangHL, HuYC. Limb salvage by distraction osteogenesis for distal tibial osteosarcoma in a young child: A case report. Orthop Surg2016;8:253-256.CrossrefPubMed Google Scholar

6 Xu J , JiaYC, KangQL, ChaiYM. Management of hypertrophic nonunion with failure of internal fixation by distraction osteogenesis. Injury2015;46:2030-2035.CrossrefPubMed Google Scholar

7 Kaim Khani GM , HumailSM, HafeezK, AhmedN. Pattern of bony injuries among civilian gunshot victims at tertiary care hospital in Karachi, Pakistan. Chin J Traumatol2015;18:161-163.CrossrefPubMed Google Scholar

8 Jeong BO , KimTY, SongWJ. Use of Ilizarov external fixation without soft tissue release to correct severe, rigid equinus deformity. J Foot Ankle Surg2015;54:821-825.CrossrefPubMed Google Scholar

9 Grubor P , MilicevicS, GruborM, MeccarielloL. Treatment of bone defects in war wounds: retrospective study. Med Arch2015;69:260-264.CrossrefPubMed Google Scholar

10 Tee BC , SunZ. Mandibular distraction osteogenesis assisted by cell-based tissue engineering: a systematic review. Orthod Craniofac Res2015;18(suppl 1):39-49.CrossrefPubMed Google Scholar

11 Zandi M , DehghanA, SalehM, Seyed HoseiniSR. Osteodistraction of mandibles with a small bone defect at the planned osteotomy site: a histological pilot study in dogs. J Craniomaxillofac Surg2014;42:e204-209.CrossrefPubMed Google Scholar

12 Yamauchi K , NogamiS, Martinez-de la CruzG, et al.. Timed-release system for periosteal expansion osteogenesis using NiTi mesh and absorbable material in the rabbit calvaria. J Craniomaxillofac Surg2016;44:1366-1372.CrossrefPubMed Google Scholar

13 Hüseyin Arslan M . Segmental bone loss in pediatric lower extremity fractures: indications and results of bone transport. J Pediatr Orthop2015;35:8-12.CrossrefPubMed Google Scholar

14 Hong P , BoydD, BeyeaSD, BezuhlyM. Enhancement of bone consolidation in mandibular distraction osteogenesis: a contemporary review of experimental studies involving adjuvant therapies. J Plast Reconstr Aesthet Surg2013;66:883-895.CrossrefPubMed Google Scholar

15 Fujio M , YamamotoA, AndoY, et al.. Stromal cell-derived factor-1 enhances distraction osteogenesis-mediated skeletal tissue regeneration through the recruitment of endothelial precursors. Bone2011;49:693-700.CrossrefPubMed Google Scholar

16 Zheutlin AR , DeshpandeSS, NelsonNS, et al.. Bone marrow stem cells assuage radiation-induced damage in a murine model of distraction osteogenesis: a histomorphometric evaluation. Cytotherapy2016;18:664-672.CrossrefPubMed Google Scholar

17 Rahimnia A , FitoussiF, PenneçotG, MazdaK. Treatment of segmental loss of the tibia by tibialisation of the fibula: a review of the literature. Trauma Mon2012;16:154-159.CrossrefPubMed Google Scholar

18 Konopnicki S , TroulisMJ. Mandibular tissue engineering: past, present, future. J Oral Maxillofac Surg2015;73(suppl):S136-146.CrossrefPubMed Google Scholar

19 Sella EJ . Prevention and management of complications of the Ilizarov treatment method. Foot Ankle Spec2008;1:105-107.CrossrefPubMed Google Scholar

20 Harada Y , NakasaT, MahmoudEE, et al.. Combination therapy with intra-articular injection of mesenchymal stem cells and articulated joint distraction for repair of a chronic osteochondral defect in the rabbit. J Orthop Res2015;33:1466-1473.CrossrefPubMed Google Scholar

21 Hee HT , IsmailHD, LimCT, GohJC, WongHK. Effects of implantation of bone marrow mesenchymal stem cells, disc distraction and combined therapy on reversing degeneration of the intervertebral disc. J Bone Joint Surg [Br]2010;92-B:726-736.CrossrefPubMed Google Scholar

22 Chun CH , KimJW, KimSH, et al.. Clinical and radiological results of femoral head structural allograft for severe bone defects in revision TKA–a minimum 8-year follow-up. Knee2014;21:420-423. Google Scholar

23 Kempton SJ , McCarthyJE, AfifiAM. A systematic review of distraction osteogenesis in hand surgery: what are the benefits, complication rates, and duration of treatment?Plast Reconstr Surg2014;133:1120-1130.CrossrefPubMed Google Scholar

24 Borzunov DY , ChevardinAY, MitrofanovAI. Management of congenital pseudarthrosis of the tibia with the Ilizarov method in a paediatric population: influence of aetiological factors. Int Orthop2016;40:331-339.CrossrefPubMed Google Scholar

25 Hong P . A clinical narrative review of mandibular distraction osteogenesis in neonates with Pierre Robin sequence. Int J Pediatr Otorhinolaryngol2011;75:985-991.CrossrefPubMed Google Scholar

26 Jiang X , ZouS, YeB, et al.. bFGF-Modified BMMSCs enhance bone regeneration following distraction osteogenesis in rabbits. Bone2010;46:1156-1161.CrossrefPubMed Google Scholar

27 Li G . New developments and insights learned from distraction osteogenesis. Curr Opin Orthop2004;15:325-330. Google Scholar

28 Mehta M , StrubeP, PetersA, et al.. Influences of age and mechanical stability on volume, microstructure, and mineralization of the fracture callus during bone healing: is osteoclast activity the key to age-related impaired healing?Bone2010;47:219-228.CrossrefPubMed Google Scholar

29 El Hadidi YN , El KassabyM, El Fatah AhmedSA, KhamisNS. Effect of mesenchymal stem cell application on the distracted bone microstructure: an experimental study. J Oral Maxillofac Surg2016;74:1463.e1-1463.e11.CrossrefPubMed Google Scholar

30 Zomorodian E , Baghaban EslaminejadM. Mesenchymal stem cells as a potent cell source for bone regeneration. Stem Cells Int2012.CrossrefPubMed Google Scholar

31 Granero-Moltó F , WeisJA, MigaMI, et al.. Regenerative effects of transplanted mesenchymal stem cells in fracture healing. Stem Cells2009;27:1887-1898.CrossrefPubMed Google Scholar

32 Wang X , SchröderHC, GrebenjukV, et al.. The marine sponge-derived inorganic polymers, biosilica and polyphosphate, as morphogenetically active matrices/scaffolds for the differentiation of human multipotent stromal cells: potential application in 3D printing and distraction osteogenesis. Mar Drugs2014;12:1131-1147.CrossrefPubMed Google Scholar

33 Sunay O , CanG, CakirZ, et al.. Autologous rabbit adipose tissue-derived mesenchymal stromal cells for the treatment of bone injuries with distraction osteogenesis. Cytotherapy2013;15:690-702.CrossrefPubMed Google Scholar

34 Yao Y , WangG, WangZ, et al.. Synergistic enhancement of new bone formation by recombinant human bone morphogenetic protein-2 and osteoprotegerin in trans-sutural distraction osteogenesis: a pilot study in dogs. J Oral Maxillofac Surg2011;69:e446-e455.CrossrefPubMed Google Scholar

35 Pearce AI , RichardsRG, MilzS, SchneiderE, PearceSG. Animal models for implant biomaterial research in bone: a review. Eur Cell Mater2007;13:1-10.CrossrefPubMed Google Scholar

36 Dehghan MM , Baghaban EslaminejadM, MotallebizadehN, et al.. Transplantation of autologous bone marrow mesenchymal stem cells with platelet-rich plasma accelerate distraction osteogenesis in a canine model. Cell J2015;17:243-252.CrossrefPubMed Google Scholar

37 Kinoshita K , HibiH, YamadaY, UedaM. Promoted new bone formation in maxillary distraction osteogenesis using a tissue-engineered osteogenic material. J Craniofac Surg2008;19:80-87.CrossrefPubMed Google Scholar

38 Hemmati R , GharipourM, ShemiraniH, KhosraviA, KhosraviE; M HRG. Urine albumin to creatinine ratio and echocardiographic left ventricular structure and function in patients with essential hypertension. Am Heart Hosp J2011;9:90-94.CrossrefPubMed Google Scholar

39 Clayton JA , CollinsFS. Policy: NIH to balance sex in cell and animal studies. Nature2014;509:282-283.CrossrefPubMed Google Scholar

40 Chen J , LiuY, PingF, et al.. Two-step transport-disk distraction osteogenesis in reconstruction of mandibular defect involving body and ramus. Int J Oral Maxillofac Surg2010;39:573-579.CrossrefPubMed Google Scholar

41 Demiralp B , EgeT, KoseO, YurttasY, BasbozkurtM. Reconstruction of intercalary bone defects following bone tumor resection with segmental bone transport using an Ilizarov circular external fixator. J Orthop Sci2014;19:1004-1011.CrossrefPubMed Google Scholar

42 Takamine Y , TsuchiyaH, KitakojiT, et al.. Distraction osteogenesis enhanced by osteoblastlike cells and collagen gel. Clin Orthop2002;399:240-246.CrossrefPubMed Google Scholar

43 Sato K , HaruyamaN, ShimizuY, HaraJ, KawamuraH. Osteogenesis by gradually expanding the interface between bone surface and periosteum enhanced by bone marrow stem cell administration in rabbits. Oral Surg Oral Med Oral Pathol Oral Radiol Endod2010;110:32-40.CrossrefPubMed Google Scholar

44 Lai QG , YuanKF, XuX, et al.. Transcription factor osterix modified bone marrow mesenchymal stem cells enhance callus formation during distraction osteogenesis. Oral Surg Oral Med Oral Pathol Oral Radiol Endod2011;111:412-419.CrossrefPubMed Google Scholar

45 Gessmann J , KöllerM, GodryH, SchildhauerTA, SeyboldD. Regenerate augmentation with bone marrow concentrate after traumatic bone loss. Orthop Rev (Pavia)2012;4:e14.CrossrefPubMed Google Scholar

46 Sunay O , CanG, CakirZ, et al.. Autologous rabbit adipose tissue-derived mesenchymal stromal cells for the treatment of bone injuries with distraction osteogenesis. Cytotherapy2013;15:690-702.CrossrefPubMed Google Scholar

47 Nomura I , WatanabeK, MatsubaraH, et al.. Uncultured autogenous adipose-derived regenerative cells promote bone formation during distraction osteogenesis in rats. Clin Orthop Relat Res2014;472:3798-3806.CrossrefPubMed Google Scholar

48 Ando Y , MatsubaraK, IshikawaJ, et al.. Stem cell-conditioned medium accelerates distraction osteogenesis through multiple regenerative mechanisms. Bone2014;61:82-90.CrossrefPubMed Google Scholar

49 Zeng JJ , GuoP, ZhouN, XieQT, LiaoFC. Treatment of large bone defects with a novel biological transport disc in non-vascular transport distraction osteogenesis. Int J Oral Maxillofac Surg2016;45:670-677.CrossrefPubMed Google Scholar

50 Xu J , WangB, SunY, et al.. Human fetal mesenchymal stem cell secretome enhances bone consolidation in distraction osteogenesis. Stem Cell Res Ther2016;7:134.CrossrefPubMed Google Scholar

51 Lee SJ , KimBJ, KimYI, et al.. Effect of recombinant human bone morphogenetic protein-2 and adipose tissue-derived stem cell on new bone formation in high-speed distraction osteogenesis. Cleft Palate Craniofac J2016;53:84-92.CrossrefPubMed Google Scholar

52 Dehghan MM , Baghaban EslaminejadM, et al.. Transplantation of autologous bone marrow mesenchymal stem cells with platelet-rich plasma accelerate distraction osteogenesis in a canine model. Cell J2015;17:243-252.CrossrefPubMed Google Scholar