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

Trauma

Osteogenic, stem cell and molecular characterisation of the human induced membrane from extremity bone defects



Download PDF

Abstract

Objectives

The biomembrane (induced membrane) formed around polymethylmethacrylate (PMMA) spacers has value in clinical applications for bone defect reconstruction. Few studies have evaluated its cellular, molecular or stem cell features. Our objective was to characterise induced membrane morphology, molecular features and osteogenic stem cell characteristics.

Methods

Following Institutional Review Board approval, biomembrane specimens were obtained from 12 patient surgeries for management of segmental bony defects (mean patient age 40.7 years, standard deviation 14.4). Biomembranes from nine tibias and three femurs were processed for morphologic, molecular or stem cell analyses. Gene expression was determined using the Affymetrix GeneChip Operating Software (GCOS). Molecular analyses compared biomembrane gene expression patterns with a mineralising osteoblast culture, and gene expression in specimens with longer spacer duration (> 12 weeks) with specimens with shorter durations. Statistical analyses used the unpaired student t-test (two tailed; p < 0.05 was considered significant).

Results

Average PMMA spacer in vivo time was 11.9 weeks (six to 18). Trabecular bone was present in 33.3% of the biomembrane specimens; bone presence did not correlate with spacer duration. Biomembrane morphology showed high vascularity and collagen content and positive staining for the key bone forming regulators, bone morphogenetic protein 2 (BMP2) and runt-related transcription factor 2 (RUNX2). Positive differentiation of cultured biomembrane cells for osteogenesis was found in cells from patients with PMMA present for six to 17 weeks. Stem cell differentiation showed greater variability in pluripotency for osteogenic potential (70.0%) compared with chondrogenic or adipogenic potentials (100% and 90.0%, respectively). Significant upregulation of BMP2 and 6, numerous collagens, and bone gla protein was present in biomembrane compared with the cultured cell line. Biomembranes with longer resident PMMA spacer duration (vs those with shorter residence) showed significant upregulation of bone-related, stem cell, and vascular-related genes.

Conclusion

The biomembrane technique is gaining favour in the management of complicated bone defects. Novel data on biological mechanisms provide improved understanding of the biomembrane’s osteogenic potential and molecular properties.

Cite this article: Dr H. E. Gruber. Osteogenic, stem cell and molecular characterisation of the human induced membrane from extremity bone defects. Bone Joint Res 2016;5:106–115. DOI: 10.1302/2046-3758.54.2000483.

Article focus

  • The induced membrane technique is gaining importance in current bone defect reconstruction.

  • However, limited information is available on the cell biology of the human biomembrane.

  • Objectives were to evaluate the cell biology and stem cell content of the biomembrane formed during the Masquelet technique applied to treatment of segmental bone loss.

Key messages

  • Positive osteogenic differentiation was found in cells from biomembranes residing in the defect for six to 17 weeks.

  • Biomembrane morphology showed high vascularity and collagen content.

  • Biomembranes with longer maturation times showed upregulation of bone morphogenetic proteins, sonic hedgehog, and vascular and stem cell-related genes.

Strengths and limitations

  • Although this study’s sample size was limited to 13 patients, data presented help to advance the understanding of cellular and stem cell properties of the human biomembrane

  • Greater understanding of the biological properties of the biomembrane will facilitate development of methods to optimise bone defect reconstruction strategies.

  • Future research should focus on optimisation of osteogenic features of the cell population, and on ways to direct inherent stem cells towards the osteogenic lineage. These advances will help develop an optimal bone healing microenvironment.

Introduction

High-energy trauma often produces complex limb injuries and large segmental bone defects. While several techniques have been employed to manage large bone defects, there is controversy regarding the optimal treatment.

One promising approach, the two-stage Masquelet technique (the polymethylmethacrylate (PMMA)-induced biomembrane or ‘induced membrane’), has demonstrated moderate success in small clinical series.1-14 The first stage of the technique involves the development of an induced membrane layer of cells around surgically placed methacrylate spacers placed in a segmental bone defect. In the second stage, the spacers are removed, leaving behind the encasing biomembrane into which autologous cancellous bone grafts or other inductive and/or conductive materials may then be placed.

Few studies have attempted to characterise the biological properties of the human biomembrane,14 and the broad extent of its clinical potential in treatment of segmental bone defects remains to be fully explored. Aho et al14 examined histological properties of the biomembrane in 14 subjects and concluded that it consisted of mature vascularised fibrous tissue with some time-sensitive osteogenic and chondrogenic potential. The purpose of the present work was to evaluate the cell biology and stem cell content of the human biomembrane formed during the Masquelet technique for treatment of segmental bone loss. This study specifically explored the morphologic, stem cell, molecular and gene expression features. Improved understanding of the biological properties of the biomembrane will facilitate development of methods to optimise bone defect reconstruction strategies.

Materials and Methods

This study was performed following Institutional Review Board approval. The need for informed consent was waived by the ethical board as the biomembrane tissue was sampled for tissue culture as part of routine surgical practice. Biomembrane tissue was harvested during the routine surgical removal of PMMA spacers in the second stage of the Masquelet technique performed by one of the senior authors (MJB) and transported to the laboratory in sterile media where it was subdivided for studies described below.

Histology and immunohistochemical analysis

Biomembrane fragments were processed for routine histological studies using haematoxylin and eosin (H&E) and Masson trichrome staining. Trabecular bone was identified by direct visualisation at ×200 magnification, by two reviewers. Immunolocalisation for bone morphogenetic protein (BMP)2 and runt-related transcription factor (RUNX)2 used the anti-BMP2 antibody (Bioworld Technology, Inc., Saint Louis Park, Minnesota) at a 1:50 dilution and the anti-RUNX2 antibody (anti-RUNX2 monoclonal antibody, Abnova Corporation, Taipei, Taiwan) at a 1:100 dilution. Endogenous peroxidase was blocked using 3% H2O2 (Sigma-Aldrich, St Louis, Missouri). Universal Rabbit Negative Control and Mouse IgG (both Dako, Carpinteria, California) were used as negative controls for BMP2 and RUNX2, respectively. The secondary reagent was Vector ImmPRESS Reagent, Rabbit (Vector Laboratories Inc., Burlingame, California) for 30 minutes, followed by diaminobenzidine (DAB) (Dako) for five minutes. Slides were rinsed in water, counterstained with light green, dehydrated, cleared and mounted with resinous mounting media.

Cell differentiation analyses

Cells were cultured in monolayer from biomembrane fragments and used to test for stem cell potency using defined media, which allow differentiation to osteogenic, chondrogenic and adipogenic phenotypes using methods previously reported by our laboratory.15,16 Differentiation of osteogenic cells was performed using the Osteogenesis Kit (Lonza Group AG, Basel, Switzerland)17 and assessed with positive alizarin red (Sigma-Aldrich) staining of mineralised matrix following 21 to 28 days of culture. Biomembrane cells were seeded at a density of 50 000 cells/well in a 24-well tissue culture plate, established in culture for between one and nine days, and then supplemented with the kit’s Osteogenic Differentiation Media. Biomembrane cells were differentiated to adipogenic cells using the Mesenchymal Stem Cell Adipogenic Differentiation medium (Lonza); differentiated cells were stained with oil red O (Sigma-Aldrich) to demonstrate fat droplets.

Demonstration of chondrogenic differentiation was based on micromass in vitro formation by cells cultured for two to 18 days in Chondrogenic Induction Medium (Lonza), supplemented with 5% foetal bovine serum (FBS) and 10 ng/mL transforming growth factor-ß3 (TGF-β3). Cells were seeded at a density of 200 000 cells/well in a 24-well tissue culture plate and supplemented three times a week. Control cultures were supplemented with MSCBM (Mesenchymal Stem Cell Basal Medium, Lonza) basal media only. Control and differentiating cultures were supplemented three times per week. Digital images were used to document cell differentiations in vitro.

Human foetal osteoblast culture

The human osteoblast cell line hFOB 1.19 was obtained from the American Type Culture Collection (ATCC; Manassas, Virginia). Cells were grown in a 24-well tissue culture plate at 33.5° C, 95% humidity, and 5% CO2 with Dulbecco’s Modified Eagle’s Medium (DMEM)/Ham’s F10 medium: 1:1 ratio DMEM/Ham’s F12 with L-glutamine (DMEM/F12; Life Technologies, Carlsbad, California), 10% FBS (Atlas Biologicals, Fort Collins, Colorado), 0.3 mg/ml G418 (Geneticin R; Life Technologies). Once confluent, the growth medium was switched to differentiation media which consisted of DMEM/Ham’s F10 with 0.1 mg/ml L-ascorbic acid, 10−8M menadione, 5 mM ß-glycerol phosphate, and 10−7M 1α,25-Dihydroxyvitamin D3 (Sigma-Aldrich). This differentiation methodology and this cell line have been previously described.18,19 Cells were incubated at 39.5° C for seven days. To confirm osteoblast differentiation, wells were stained for bone mineralisation via alizarin red (data not shown).

Microarray gene expression studies

mRNA was harvested from biomembrane specimens following homogenisation; mRNA from the biomembrane and from cultured osteoblasts using Trizol (Life Technologies). Affymetrix microarray analyses were used to compare gene expression patterns of the biomembrane with cultured osteoblast cells (HG-U133 + PM strips; Affymetrix Inc., Santa Clara, California). Affymetrix ‘.cel’ files were uploaded to GeneSifter web-based software (VizX Labs, Seattle, Washington), normalised, and statistical significance determined (p < 0.05) using the unpaired student t-test (two-tailed). Data were corrected for false discovery rates using the Benjamini-Hochberg test and results are expressed as fold changes (2.0 and greater only). Ontology searches (which allow one to avoid searching gene by gene and also provide a controlled vocabulary of search terms for gene characteristics) were used for the following bone-related ontology groupings: angiogenesis and related vascularity categories; collagen; bone development; bone remodeling; negative regulation of bone remodeling; positive regulation of bone remodeling; regulation of bone remodeling; bone resorption; BMP signaling pathway; ossification; regulation of osteoblast differentiation; positive regulation of osteoblast differentiation; osteoblast differentiation; osteoblast development; negative regulation of osteoclast differentiation; positive regulation of osteoclast differentiation; regulation of osteoclast differentiation; osteoclast differentiation; chondrocyte differentiation; chondrocyte development; cartilage development, and the following ontologies for stem cells: differentiation, division, regulation of stem cell division, maintenance, and canonical WNT receptor signaling pathway involved in mesenchymal stem cell differentiation.

An additional analysis of gene expression patterns was carried out, which used the ontology groups described above to test for differences in major bone-, cartilage- and vascular-related genes in specimens with longer spacer duration periods (> 12 weeks) versus specimens with shorter spacer durations.

Statistical analysis

For non-microarray data analysis, standard statistical methods were used employing InStat (GraphPad Software, Inc., San Diego, California). Means and standard deviation (sd) were calculated, and p < 0.05 was set as the significance level. Spearman’s correlation was used to test for linear relationships between the presence (scored as one) or absence (scored as two) of trabecular bone within the biomembrane histological specimen and the duration of the PMMA spacer in the surgical site.

Results

Subjects

Demographic and clinical features of the study population are presented in Table I. Biomembrane specimens were obtained from 12 surgeries for complex fractures (mean age 40.7 years, sd 14.4; four women, eight men) resulting from six motor vehicle or motorcycle accidents, four falls, and two gunshot wounds. Table I presents data on the surgical site, low- or high-energy causes for the trauma, the presence or absence of infection, the length of the bony defect, the volume of the PMMA spacer, and Arbeitsgemeinschaft für Osteosynthesefragen/American Orthopaedic Trauma Association (AO/OTA) classification20 and the Gustilo classification.21 The mean duration of the PMMA spacers within the patient fracture sites was 11.9 weeks (six to 18). Radiographic measurements (in cm) were performed using AP and lateral radiographs of fractures at the time of fixation during PMMA spacer placement. Lengths, widths and depths were obtained and data used to calculate the approximate PMMA spacer volume (in cm3) (Table I). In total, 50% of the subjects had bone infections at the time of PMMA spacer placement (Table I).

Table I.

Demographic and clinical features of study population.*

Age (yrs)/gender Fracture location Cause of trauma AO/OTA class-ification Gustilo class-ification Fracture fixation during cement spacer Bone infection (Y/N) Bony defect length (cm)* Volume of PMMA spacer (cm3) Duration (weeks) of spacer in site Experimental studies performed
18/F Distal tibia Vehicle crash into tree 43-A3 3A ORIF tibia N 5 23 6 H, O, C, A, G
59/M Distal femur* Low-energy fall (fell while standing; periprosthetic fracture) 33-A3 N/A ORIF distal femur Y 12 293 6 H, O, C, A, G
33/F Distal femur Motorcycle/vehicle accident 33-C3 3A ORIF distal femur N 10 220 7 H, O, C, A, G
28/M Diaphyseal femur GSW 32-C3 3A IM nail femur Y 8 185 9 H, O, C, A, G
48/F Distal tibia MVC 43-C3 3B ORIF distal tibia/IM fixation fibula N 9 80 9 H, O, C, A, G
56/M Distal tibia Low energy fall (fell from stool) 43-A3 2 Ilizarov external fixation tibia Y 6 57 11 H, O, C, A, G
52/M Distal tibia Fell out of vehicle and hit by following vehicle 43-A3 3B IM nail tibia N 6 17 13 H, C, G
27/F Distal tibia MVC 43-C3 3B External fixation and ORIF fibula Y 4 37 15 H, O, C, A, G
42/M Diaphyseal tibia Motorcycle/vehicle accident 42-C2 3B IM nail tibia N 13 141 16 H, G
62/M Distal tibia Crush injury (tree limb fell on leg) 43-A3 3B External fixation tibia Y 7 49 16 H, O, C, A, G
30/M Diaphyseal tibia GSW 42-C3 3A Ilizarov external fixation tibia N 14 112 17 H, O, C, A, G
34/M Distal tibia High-energy fall (jumped from balcony) 43-C2 3B ORIF tibia/fibula Y 7 84 18 H, O, C, A
  1. PMMA, polymethylmethacrylate; M, male; F, female; MVC, motor vehicle collision; GSW, gunshot wound, H, histology and immunohistochemistry; O, osteogenesis determination; C, chondrogenesis; A, adipogenesis determination; G, microarray gene expression analysis; AO/OTA, Arbeitsgemeinschaft für Osteosynthesefragen/American Orthopaedic Trauma Association; ORIF, open reduction and internal fixation; IM, intramedullary

  1. *

    Closed fracture (all other cases were open)

Morphological features of the human biomembrane

Biomembrane tissues examined with routine histology showed the presence of trabecular bone in four out of 12 (33.3%) of the biomembrane specimens (Fig. 1). There was no correlation between the presence of trabecular bone in the specimens and the duration of the PMMA spacer in the surgical site. Morphological analysis showed the presence of high collagen content and extensive vascularity (Fig. 2). Immunohistochemistry showed that the osteoinductive factor BMP2 was present in osteoblasts, osteocytes and in cells within the biomembrane stroma (Fig. 3). RUNX2 (also called CBFA1, a regulator of osteoblast differentiation) was also found to be present within the biomembrane (Fig. 4).

Fig. 1 
            Light microscopic features of the biomembrane tissue. Trabecular bone (arrow) was present in 4/13 (33.3%) of specimens examined for morphological features. (Masson trichrome stain; bar = 50 µm).

Fig. 1

Light microscopic features of the biomembrane tissue. Trabecular bone (arrow) was present in 4/13 (33.3%) of specimens examined for morphological features. (Masson trichrome stain; bar = 50 µm).

Fig. 2 
            Light microscopy showing the highly vascular nature of the biomembrane. Arrows mark vasculature. (Masson trichrome stain; bar = 50 µm).

Fig. 2

Light microscopy showing the highly vascular nature of the biomembrane. Arrows mark vasculature. (Masson trichrome stain; bar = 50 µm).

Fig. 3 
            Immunohistochemical localisation of bone morphogenetic protein 2 in the biomembrane. Insert upper right shows an adjacent section processed as a negative control (Bar = 50 µm).

Fig. 3

Immunohistochemical localisation of bone morphogenetic protein 2 in the biomembrane. Insert upper right shows an adjacent section processed as a negative control (Bar = 50 µm).

Fig. 4 
            Immunohistochemical localisation of runt-related transcription factor 2 within the biomembrane. Insert upper right shows an adjacent section processed as a negative control (Bar = 20 µm).

Fig. 4

Immunohistochemical localisation of runt-related transcription factor 2 within the biomembrane. Insert upper right shows an adjacent section processed as a negative control (Bar = 20 µm).

Stem cell capacity of the biomembrane

Cells cultured from the biomembrane grew well and displayed a spindle-shaped morphology in monolayer culture. Positive osteogenic capacity was demonstrated in seven out of ten (70%) tested cultures (Fig. 5). Chondrogenic capacity was demonstrated by formation of micromasses (Fig. 6) with the strong presence of chondroitin sulfate on immunohistochemistry (Fig. 7). Chondrogenic differentiation was seen in 90.9% of specimens (11 out of 12 tested specimens). Adipogenic differentiation was demonstrated by the presence of fat droplets (Fig. 8). Adipogenic differentiation was seen in 90% of the specimens (nine out of ten tested specimens). Additional data were gained on the stem cell features of the biomembrane by microarray analyses which compared the biomembrane to cultured osteoblasts (Table II). Gene expression was stronger in the biomembrane for secreted frizzled-related protein (SFRP) 2 (upregulated 77 fold), an important factor which interacts with Wnt signaling to enhance mesenchymal stem cell engraftment and myocardial repair.22,23

Fig. 5 
            Biomembrane cells cultured in osteogenic differentiation media showed development of calcified nodules as verified here with alizarin red staining. Insert lower right shows no development of calcified nodules in cells grown in control medium. Original magnification ×200.)

Fig. 5

Biomembrane cells cultured in osteogenic differentiation media showed development of calcified nodules as verified here with alizarin red staining. Insert lower right shows no development of calcified nodules in cells grown in control medium. Original magnification ×200.)

Fig. 6 
            Biomembrane cells cultured in chondrogenesis media showed the formation of compact micromasses in vitro (diameter of the culture plate well shown is 16 mm).

Fig. 6

Biomembrane cells cultured in chondrogenesis media showed the formation of compact micromasses in vitro (diameter of the culture plate well shown is 16 mm).

Fig. 7 
            Micromass specimen embedded in paraffin and processed for immunohistochemical localisation of chondroitin sulfate. Note the abundant presence of this matrix component. Insert upper right shows an adjacent section processed as a negative control (Bar = 20 µm).

Fig. 7

Micromass specimen embedded in paraffin and processed for immunohistochemical localisation of chondroitin sulfate. Note the abundant presence of this matrix component. Insert upper right shows an adjacent section processed as a negative control (Bar = 20 µm).

Fig. 8 
            Biomembrane cells cultured for adipogenesis show the presence of fat droplets (stained red in the micrograph). Insert lower right shows that biomembrane cells cultured in control media showed no presence of fat droplets (original magnification ×200).

Fig. 8

Biomembrane cells cultured for adipogenesis show the presence of fat droplets (stained red in the micrograph). Insert lower right shows that biomembrane cells cultured in control media showed no presence of fat droplets (original magnification ×200).

Table II.

Major bone-, cartilage- and vascular-related gene expression findings in biomembrane specimens vs osteoblast cells.

Gene name Direction Fold change p-value Gene identifier Gen ID
Stem cell-related genes
Fibroblast-like growth factor 2 (basic) Down 45.8 < 0.0001 NM_002006 FGF2
Notch homolog 2 Down 2.6 0.001 AF308601 Notch2
Transforming growth factor, beta 2 Down 27.2 < 0.0001 M19154 TGF-β2
Wingless-type MMTV integration site family, member 3 Down 7.2 < 0.0001 AA463626 WNT3
Wingless-type MMTV integration site family, member 5A Down 40.9 0.0002 NM_003392 WNT5A
Insulin-like growth factor 1 (somatomedin C) Up 30.7 0.0001 AU44912 IGF1
Secreted frizzled-related protein 2 Up 77.7 < 0.0001 AF11912 SFRP2N
Bone or cartilage-related genes
Bone gamma-carboxyglutamate (gla) protein Down 29.1 < 0.0001 NM_000711 BGLAP
Cyclin-dependent kinase 6 Down 4.3 0.04 AW274756 CDK6
Fibrillin 2 Down 21.3 0.0009 NM-001999 FBN2
Interleukin 6 Down 22.5 0.0002 NM_000600 IL-6
SATB homeobox 2 Down 3.5 0.013 AB028957 SATB2
Tumour necrosis factor, alpha-induced protein 3 Down 2.6 0.04 AI738896 TNFAIP3
Acid phosphatase 5, tartrate resistant Up 5.0 0.01 NM_001611 ACP5
Asporin Up 208.6 < 0.0001 NM_017680 ASPN
Biglycan Up 16.9 0.03 AA845258 BGN
Bone morphogenetic protein 2 Up 3.6 0.003 NM_001200 BMP2
Bone morphogenetic protein 6 Up 2.1 0.009 NM_0017_PM_at BMP6
Chemokine (C-C motif) ligand 5 Up 4.2 0.006 NM_002985 CCL5
Collagen, type I, alpha 2 Up 3.7 0.006 AA628535 COL1A2
Collagen, type III, alpha 1 Up 4.5 0.02 AU146808 COL3A1
Collagen, type V, alpha 3 Up 9.9 0.001 AI984221 COL5A3
Collagen, type VI, alpha 2 Up 2.8 0.04 AL531750 COL6A2
Collagen, type VIII, alpha 1 Up 7.0 0.008 AI806793 CO:8A2
Collagen, type XI, alpha 1 Up 11.1 0.003 J04177 COL11A1
Collagen, type XII, alpha 1 Up 11.0 0.006 AU146651 COL12A1
Collagen, type XIV, alpha 1 Up 27.8 0.001 BF449063 COL14A1
Decorin Up 7.7 0.0003 AI281593 DCN
Growth hormone receptor Up 4.9 0.04 NM_000163 GHR
Interleukin 23, alpha subunit p19 Up 4.6 0.005 AL559122 IL23A
Matrix Gla protein Up 158.4 < 0.0001 NM_0009000 MGP
Matrix metallopeptidase 14 Up 2.5 0.004 Z48481 MMP14
Matrix metallopeptidase 9 (type IV collagenase) Up 12.0 0.01 NM_004994 MMP9
Runt-related transcription factor 2 Up 6.2 0.0005 AW469546 RUNX2
Vascular-related genes
Angiopoietin 1 Down 2.6 0.003 U83508 ANGPT1
BMP binding endothelial regulator Down 5.0 < 0.0001 AI423201 BMPER
Bradykinin receptor B2 Down 5.5 0.0003 NM_000623 BDKRB2
Endothelin 1 Down 2.4 0.03 J05008 EDN1
Vascular endothelial growth factor A Down 2.1 0.004 M272781 VEGFA
Vascular endothelial growth factor C Down 8.8 0.005 U58111 VEGFC
Vasohibin 2 Down 15.0 < 0.0001 AI961235 VASH2
Angiogenin, RNase A family, 5 Up 5.6 0.01 AI761728 ANG
Angiopoietin 2 Up 12.7 0.001 AA0835514 ANGPT2
Aquaporin 1 Up 34.2 0.0003 AL518391 AQP1
Endothelial cell-specific chemotaxis regulator Up 5.6 0.01 AI422211 ECSCR
Endothelin receptor type B Up 3.1 0.002 NM_003991 EDNRB

Molecular characterisation of bone-, cartilage- and vasculature-related gene expression: comparison of biomembrane expression patterns with osteoblast cell culture

Table II shows findings regarding the upregulation of genes in the biomembrane (in comparison with expression patterns in cultured osteoblasts) for genes related to bone and cartilage (BMP2, upregulated three-fold; BMP6, upregulated three-fold; matrix gla protein, upregulated 158-fold; and RUNX2, upregulated six-fold), and many collagen genes, including collagen type I, alpha 2. Also present were several genes related to vasculature with strong expression patterns, including angiopoietin 2 (upregulated 12-fold), and endothelial cell-specific chemotaxis regulator (upregulated five-fold).

Comparison of biomembranes with longer versus shorter PMMA spacer residence

The second type of gene analysis performed was a comparison of genes expressed in biomembranes in spacers that resided within the host subject for > 12 weeks compared with those from spacers that had a duration of < 12 weeks (Table III). For spacers with longer maturation times, it was noted that there remained a modest upregulation of several genes with recognised relationships to stem cells (BMP7, MYST histone acetyltransferase 3, fibroblast growth factor (FGF) receptors 1 and 2, FGF 4 and sonic hedgehog). With respect to bone- and cartilage-related genes, upregulation was seen in oestrogen receptor 1 (upregulated 3.1-fold), growth and differentiation factor 5 (upregulated 2.4-fold) and a number of matrix-related genes.

Table III.

Major bone-, cartilage- and vascular-related gene expression findings in human biomembrane specimens with longer spacer duration (> 12 weeks) versus those with shorter duration.

Gene name Direction Fold change p-value Gene identifier Gen ID
Stem cell-related genes
Frizzled homolog 1 (Drosophila) Down 4.54 0.02 NM_003505 FZD1
Frizzled homolog 7 (Drosophila) Down 2.89 0.039 AI333651 FZD7
Insulin-like growth factor 1 (somatomedin C) Down 2.27 0.037 M29644 IGF1
MYST histone acetyltransferase 3 Down 5.46 0.009 AI817830 MYST3
Notch homolog 2 Down 3.36 0.03 AU158495 Notch2
Secreted frizzled-related protein 2 Down 6.32 0.026 AW003584 SFRP2
Wingless-type MMTV integration site family, member 3 Down 7.2 <0.0001 AA463626 WNT3
Bone morphogenetic protein 7 Up 2.03 0.009 M60316 BMP7
Fibroblast growth factor receptor 1 Up 2.14 0.009 NM_023110 FGFR1
Fibroblast growth factor receptor 2 Up 3.23 0.009 AB030073 FGFR2
Fibroblast growth factor 4 Up 3.04 0.01 NM_002007 FGF4
Sonic hedgehog homolog (drosophila) Up 2.42 0.01 AI92528 SHH
Bone or cartilage-related genes
ADAM metallopeptidase domain 9 (meltrin gamma) Down 6.65 0.009 NM_003816 ADAM9
ADAM metallopeptidase with thrombospondin type 1 motif, 3 Down 2.53 0.015 AB002364 ADAMTS3
Bone morphogenetic protein receptor, type IA Down 4.26 0.013 AI678679 BMPR1A
Bone morphogenetic protein receptor, type II Down 6.4 0.009 AL046696 BMPR2
Cartilage-associated protein Down 2.49 0.023 AW024741 CRTAP
Chondroitin sulfate N-acetyl-galactosaminyltransferase 1 Down 10.28 0.009 NM_018371 CSGALNACT1
Chondroitin sulfate N-acetyl galactosaminyltransferase 2 Down 3.36 0.016 NM_018590 CSGALNACT2
Chondroitin sulfate synthase 1 Down 5.21 0.011 NM_014918 CHSY1
Collagen type I, alpha 1 Down 3.33 0.026 AI743621 COL1A1
Collagen, type I, alpha 2 Down 2.38 0.011 AA62835 COL1A2
Collagen, type III, alpha 1 Down 3.69 0.010 AU144167 COL3A1
Collagen, type V, alpha 1 Down 5.34 0.020 AI130969 COL5A1
Collagen, type V, alpha 3 Down 3.09 0.009 AI984221 COL5A3
Collagen, type VI, alpha 1 Down 2.98 0.047 AA292373 COL6A1
Collagen, type VI, alpha 2 Down 2.54 0.009 AL531750 COL6A2
Decorin Down 4.35 0.011 NM_001920 DCN
Dystroglycan 1 Down 4.12 0.012 NM_004393 DAG1
Fibroblast growth factor receptor 1 Down 3.19 0.012 M60485 FGFR1
Growth hormone receptor Down 4.21 0.011 NM_000163 GHR
Heparan sulfate 2-O-sulfotransferase 1 Down 4.31 0.011 NM_012262 HS2ST1
Hypoxia inducible factor 1, alpha subunit Down 3.72 0.012 NM_001530 HIF1A
Insulin-like growth factor 1 (somatomedin C) Down 2.27 0.039 M29644 IGF1
Insulin-like growth factor binding protein 3 Down 5.5 0.015 BF340228 IGFBP3
Insulin-like growth factor binding protein 5 Down 2.83 0.047 AW007532 IGFBP5
Interleukin 6 signal transducer Down 7.07 0.009 AW242916 IL6ST
Matrilin 2 Down 4.71 0.049 NM_002380 MATN2
Matrix Gla protein Down 3.55 0.013 AW512787 MGP
Matrix metallopeptidase 2 Down 2.66 0.039 NM_004530 MMP2
Matrix metallopeptidase 9 Down 4.37 0.030 NM_004994 MMP9
Matrix metallopeptidase 13 (collagenase 3) Down 4.13 0.024 NM_002427 MMP13
Osteoglycin Down 4.07 0.042 NM_014057 OGN
Osteoclast stimulating factor 1 Down 4.46 0.009 NM_012383 OSTF1
Periostin, osteoblast specific factor Down 4.65 0.034 AW137148 POSTN
Runt-related transcription factor 2 Down 4.47 0.012 AL353944 RUNX2
SMAD family member 1 Down 7.26 0.009 U54826 SMAD1
Sulfatase 2 Down 3.69 0.012 AL133001 SULF2
Tenascin C Down 7.85 0.016 NM_002160 TNC
Thyroid hormone receptor, beta Down 3.22 0.019 BG494007 THRB
TIMP metallopeptidase inhibitor 2 Down 3.4 0.01 NM_003255 TIMP2
Transforming growth factor, beta 3 Down 2.17 0.011 J03241 TGF-β3
Tumour necrosis factor, alpha-induced protein 3 Down 3.22 0.009 NM_006290 TNFAIP3
Versican Down 9.07 0.011 D32029 VCAN
Vitamin D receptor Down 3.12 0.009 NM_000376 VDR
ADAM metallopeptidase with thrombospondin type 1 motif, 9 Up 3.3 0.009 AB037733 ADAMTS9
Aggrecan Up 3.9 0.009 BC036445 ACAN
Bone morphogenetic protein 7 Up 2.03 0.009 M60316 BMP7
Bone morphogenetic protein 10 Up 2.04 0.019 NM_014482 BMP10
Epidermal growth factor receptor Up 4.69 0.01 AF277897 EGFR
Estrogen receptor 1 Up 3.13 0.009 AF258450 ESR1
Fibrillin 2 Up 2.63 0.009 AF193046 FBN2
Fibroblast growth factor 4 Up 3.04 0.01 NM_002007 FGF4
Fibroblast growth factor receptor 2 Up 3.23 0.009 AB030073 FGFR2
Growth differentiation factor 5 Up 2.4 0.009 NM_000557 GDF5
G protein-coupled receptor 55 Up 2.48 0.027 NM_005683 GPR55
Heparan sulfate (glucosamine) 3-O-sulfotransferase 5 Up 2.43 0.011 AW449310 HS3ST5
Hyaluronan and proteoglycan link protein 4 Up 2.59 0.009 W63783 HAPLN4
Interleukin 21 Up 2.26 0.012 NM_021803 IL21
Interleukin 23, alpha subunit p19 Up 2.86 0.013 AJ296370 IL23A
Natriuretic peptide receptor C/guanylate cyclase C Up 2.35 0.009 AI628360 NPR3
Retinoic acid receptor, gamma Up 2.32 0.011 M57707 RARG
Sonic hedgehog homolog (Drosophila) Up 2.42 0.01 AI192528 SHH
Tenascin R Up 2.23 0.009 Y13359 TNR
Thrombospondin 1 Up 2.65 0.013 BF084105 THBS1
Vascular-related genes
Angiopoietin 1 Down 3.36 0.012 NM_001146 ANGPT1
Angiopoietin 2 Down 6.28 0.01 NM_001147 ANGPT2
Aquaporin 1 Down 3.7 0.011 AL518391 AQP1
Lysyl oxidase Down 5.01 0.013 NM_002317 LOX
Endothelin 1 Down 2.76 0.01 NM_001955 EDN1
Endothelial cell-specific chemotaxis regulator Down 5.13 0.009 AI422211 ECSCR
Endothelin receptor type A Down 6.09 0.009 AU118882 EDNRA
Vascular endothelial growth factor A Down 7.33 0.017 AF022375 VEGFA
Angiopoietin-like 3 Up 2.21 0.009 AV659209 ANGPTL3
Angiopoietin 4 Up 2.33 0.012 NM_015985 ANGPT4
Endoglin Up 2.08 0.009 AA906156 ENG
Epiregulin Up 2.45 0.009 BC03506 EREG
Vasohibin 2 Up 2.7 0.01 BC028194

Discussion

The ability to reconstruct large bone defects that occur as a result of open fractures and/or infections remains a significant challenge in orthopaedic trauma. The Masquelet technique, which uses an induced biomembrane as a conduit for bone graft, has increasingly been used in treatment of patients with complicated segmental bone loss. The purpose of this study was to investigate the cell biology and stem cell content of the human biomembrane formed during the induced biomembrane technique.

Our results demonstrated that the histological presence of isolated bone islands formed with the biomembrane tissue harvested at the time of spacer removal was consistent with intramembranous ossification. This is an important finding in human biomembrane research and is similar to previous studies from our laboratory that have evaluated the properties of biomembranes formed in a rat segmental defect model.24,25

Stem cell differentiation analyses in the current study used cells isolated from the human biomembrane. These cells showed a greater variability in pluripotency for osteogenic potential (70%) compared with chondrogenic or adipogenic potentials (100% and 90%, respectively). Since the clinical outcome desired with the Masquelet approach is bone formation, we chose an osteoblast cell line rather than fibroblasts for comparison with the biomembrane in our gene expression analyses as osteoblasts are more relevant to expression patterns related to bone formation. Strong expression and high fold changes were found for bone-, cartilage-, stem cell- and angiogenesis-related genes (Table II).22,23 The high expression level of asporin (upregulated 208 times) merits comment. As shown in previous osteoarthritis literature, not only can this extracellular matrix protein inhibit TGF-ß and regulate chondrogenic potential with additional evidence for a role in osteoarthritis,26,27 but also, some research points to elevated asporin expression in osteoblasts in subchondral bone in osteoarthritic patients.28 In the second part of our gene analysis studies, we looked for differences in expression patterns in PMMA spacers with longer versus shorter maturation periods. Upregulation of several stem cell-related genes was identified as well as bone-, cartilage- and vascular-related genes (Table III). Downregulation of several genes was also seen. We would postulate that this is because these genes, including type I collagen, were more actively expressed in the early biomembrane stages during formation of the stroma and islands of trabecular bone.

The biomembrane lies within a highly complex biological milieu in which many cytokine systems are active.29-31 The biomembrane is used clinically as both a receptive bed for bone formation and a source of mesenchymal stem cells which may be recruited and directed to this bed during the healing process. Although several small animal models have been used to evaluate the biomembrane, few studies have analysed the cellular features and stem cell content of biomembranes isolated from humans. A recent paper by Aho et al14 analysed properties of the induced biomembrane from 14 human subjects. They concluded that the biomembrane consisted of mature vascularised fibrous tissue with some osteogenic and chondrogenic time-sensitive potential. However, unlike the current study, their work did not extensively evaluate the difference in gene expression patterns of several bone-, cartilage- and vascular-related genes. The gene expression work reported in the present study may serve as a foundation for future studies that focus on specific gene up- or down-regulation in order to optimise the biomembrane osteogenic potential.

This study has several strengths and limitations. One strength is that data presented herein are shown in relation to well characterised clinical findings, including PMMA spacer duration, energy levels of the trauma source, AO/OTA and Gustilo classifications, fracture fixation descriptions, presence or absence of infection, and measurements of bone defect lengths and volume of the PMMA spacer (Table I). One other important consideration and possible limitation of this study is that the biomembrane analyses presented here are based upon specimens derived from variable sites in the tibia and femur, and the biomembranes formed in response to variable PMMA spacer sizes which resided in the site for different time periods (six to 18 weeks). There was also significant variation in subject ages (18 to 62 years). These factors undoubtedly influenced important features such as vascularisation since the vascularity and healing potential in a younger subject would presumably be greater than that of a middle-aged subject. However, this variability in study population structure reflects the potential age range of trauma patients with severe large bone defects who may be effectively treated with the Masquelet technique. Results from age- and site-controlled small animal studies were previously reported by our group.24,25 Another limitation of this study is that it has a relatively small sample size for the gene expression analyses. Although many of the differences in gene expression reached statistical significance, future studies with larger sample sizes are needed.

Our findings add additional clinical cases to the literature, and advance this research by quantifying the osteogenic, chondrogenic and adipogenic differentiation potential of cells within the biomembrane. This novel work establishes baseline data for the process during which biomembrane cells differentiate into important stem cells. We look forward to such information being used in work designed to optimise the osteogenic or chondrogenic potential of the biomembrane, potentially expediting the timeline for healing segmental bone defects in patients with complex fractures.

In conclusion, the Masquelet technique (PMMA-induced biomembrane) is successfully employed in current bone defect reconstruction treatment. Limited data exist on detailed characterisation of the cell biology of the human biomembrane. We suggest that future research directed towards optimising the biological features of the biomembrane should focus on optimisation of the osteogenic features of the cell population and on ways to direct the stem cells present in the biomembrane into the osteogenic lineage (25% of our biomembrane cell specimens did not differentiate into osteoblasts). This is vital since timely formation of high-quality bone is of paramount importance in the clinical patient population with segmental bone defects. The work presented herein represents an important step forward in the advancement of our understanding of the cellular features and stem cell properties of the human biomembrane and highlights the importance for future research in which biomembranes may be modified to create an optimal bone healing microenvironment.


Dr H. E. Gruber; email:
Author Contribution

H. E. Gruber, Conceived of idea, wrote manuscript, obtained research funding, selected gene expression data, prepared illustrations, reviewed histology/immunohistochemistry, did revisions 1 and 2.

G. Ode, Conceived of idea, assisted with manuscript preparation, reviewed patient charts, observed surgeries, reviewed literature.

G. Hoelscher, Performed ontology searches for gene expression data.

J. Ingram, Performed histology and immunohistochemistry on harvested biomembranes, assisted with methods text.

S. Bethea, Performed cell cultures and mRNA harvesting.

M. J. Bosse, Conceived of idea, performed surgeries on patients, helped with obtaining funding, reviewed and revised manuscript, senior MD on project.


The authors acknowledge funding from The Winkler Orthopaedic Fellowship Fund for partial support of this work. The authors thank Nury Steuerwald, Ph.D. and Judy Parsons in the Molecular Biology Core for expert technical assistance with microarray analysis. We also thank James Kellam, M.D. for his many helpful discussions regards the biomembrane and the Masquelet technique.

Funding Statement

Funding has been received from the Winkler Orthopaedic Fellowship Fund which is related to this article.

ICMJE conflict of interest

None declared.

References

1 Christian EP , BosseMJ, RobbG. Reconstruction of large diaphyseal defects, without free fibular transfer, in Grade-IIIB tibial fractures. J Bone Joint Surg [Am]1989;71-A:994-1004.PubMed Google Scholar

2 Bosse MJ , RobbG. Techniques for the reconstruction of large traumatic bony defects with massive autogenous cancellous bone graft. Tech Orthop1992;7:17-26. Google Scholar

3 Al-Muaid J , FranciolliM, KlaueK. Tubular remodeling of massive cancellous bone graft in the treatment of long bone defects. Eur J Trauma Emerg Surg2007;33:654-658.CrossrefPubMed Google Scholar

4 Masquelet AC . Muscle reconstruction in reconstructive surgery: soft tissue repair and long bone reconstruction. Langenbecks Arch Surg2003;388:344-346.CrossrefPubMed Google Scholar

5 Masquelet AC , BegueT. The concept of induced membrane for reconstruction of long bone defects. Orthop Clin North Am2010;41:27-37.CrossrefPubMed Google Scholar

6 Masquelet AC , FitoussiF, BegueT, MullerGP. Reconstruction des os longs par membrane induite et autogreffe spongieuse. Ann Chir Plast Esthet2000;45:346-353.[[Article in French]] Google Scholar

7 Pelissier P , MartinD, BaudetJ, LepreuxS, MasqueletAC. Behaviour of cancellous bone graft placed in induced membranes. Br J Plast Surg2002;55:596-598.CrossrefPubMed Google Scholar

8 Ristiniemi J , LakovaaraM, FlinkkiläT, JalovaaraP. Staged method using antibiotic beads and subsequent autografting for large traumatic tibial bone loss: 22 of 23 fractures healed after 5-20 months. Acta Orthop2007;78:520-527.CrossrefPubMed Google Scholar

9 Uzel A-P , LemonneF, CasoliV. Tibial segmental bone defect reconstruction by Ilizarov type bone transport in an induced membrane. Orthop Traumatol Surg Res2010;96:194-198.CrossrefPubMed Google Scholar

10 Viateau V , GuilleminG, CalandoY, et al.. Reconstruction de perte de substance osseuse massive par la procédure de Masquelet: modèle expérimental chez la brebis. J Bone Joint Surg [Br]2008;90-B(Suppl II):254. Google Scholar

11 Pelissier P , MasqueletAC, BareilleR, PelissierSM, AmedeeJ. Induced membranes secrete growth factors including vascular and osteoinductive factors and could stimulate bone regeneration. J Orthop Res2004;22:73-79.CrossrefPubMed Google Scholar

12 Chong K-W , WoonCYL, WongM-K. Induced membranes–a staged technique of bone-grafting for segmental bone loss: surgical technique. J Bone Joint Surg [Am]2011;93-A:85-91. Google Scholar

13 Woon CYL , ChongK-W, WongM-K. Induced membranes–a staged technique of bone-grafting for segmental bone loss: a report of two cases and a literature review. J Bone Joint Surg [Am]2010;92-A:196-201. Google Scholar

14 Aho O-M , LehenkariP, RistiniemiJ, et al.. The mechanism of action of induced membranes in bone repair. J Bone Joint Surg [Am]2013;95-A:597-604.CrossrefPubMed Google Scholar

15 Tapp H , DeepeR, IngramJA, et al.. Adipose-derived mesenchymal stem cells from the sand rat: transforming growth factor beta and 3D co-culture with human disc cells stimulate proteoglycan and collagen type I rich extracellular matrix. Arthritis Res Ther2008;10:R89.CrossrefPubMed Google Scholar

16 Gruber HE , DeepeR, HoelscherGL, et al.. Human adipose-derived mesenchymal stem cells: direction to a phenotype sharing similarities with the disc, gene expression profiling, and coculture with human annulus cells. Tissue Eng Part A2010;16:2843-2860.CrossrefPubMed Google Scholar

17 Pittenger MF , MackayAM, BeckSC, et al.. Multilineage potential of adult human mesenchymal stem cells. Science1999;284:143-147.CrossrefPubMed Google Scholar

18 Kapinas K , KesslerC, RicksT, GronowiczG, DelanyAM. miR-29 modulates Wnt signaling in human osteoblasts through a positive feedback loop. J Biol Chem2010;285:25221-25231.CrossrefPubMed Google Scholar

19 Guo D , LiQ, LvQ, et al.. MiR-27a targets sFRP1 in hFOB cells to regulate proliferation, apoptosis and differentiation. PLoS One2014;9:e91354.CrossrefPubMed Google Scholar

20 Marsh JL , SlongoTF, AgelJ, et al.. Fracture and dislocation classification compendium - 2007: orthopaedic Trauma Association classification, database and outcomes committee. J Orthop Trauma2007;21:S1-S133.CrossrefPubMed Google Scholar

21 Gustilo RB , AndersonJT. Prevention of infection in the treatment of one thousand and twenty-five open fractures of long bones: retrospective and prospective analyses. J Bone Joint Surg [Am]1976;58-A:453-458.PubMed Google Scholar

22 Mirotsou M , ZhangZ, DebA, et al.. Secreted frizzled related protein 2 (Sfrp2) is the key Akt-mesenchymal stem cell-released paracrine factor mediating myocardial survival and repair. Proc Natl Acad Sci U S A2007;104:1643-1648.CrossrefPubMed Google Scholar

23 Alfaro MP , PagniM, VincentA, et al.. The Wnt modulator sFRP2 enhances mesenchymal stem cell engraftment, granulation tissue formation and myocardial repair. Proc Natl Acad Sci U S A2008;105:18366-18371.CrossrefPubMed Google Scholar

24 Gruber HE , RileyFE, HoelscherGL, et al.. Osteogenic and chondrogenic potential of biomembrane cells from the PMMA-segmental defect rat model. J Orthop Res2012;30:1198-1212.CrossrefPubMed Google Scholar

25 Gruber HE , GettysFK, MontijoHE, et al.. Genomewide molecular and biologic characterization of biomembrane formation adjacent to a methacrylate spacer in the rat femoral segmental defect model. J Orthop Trauma2013;27:290-297.CrossrefPubMed Google Scholar

26 Ikegawa S . Expression, regulation and function of asporin, a susceptibility gene in common bone and joint diseases. Curr Med Chem2008;15:724-728.CrossrefPubMed Google Scholar

27 Xu L , LiZ, LiuS-Y, XuSY, NiGX. Asporin and osteoarthritis. Osteoarthritis Cartilage2015;23:933-939.CrossrefPubMed Google Scholar

28 Sakao K , TakahashiKA, AraiY, et al.. Asporin and transforming growth factor-beta gene expression in osteoblasts from subchondral bone and osteophytes in osteoarthritis. J Orthop Sci2009;14:738-747.CrossrefPubMed Google Scholar

29 Marsell R , EinhornTA. The biology of fracture healing. Injury2011;42:551-555.CrossrefPubMed Google Scholar

30 Hankenson KD , DishowitzM, GrayC, SchenkerM. Angiogenesis in bone regeneration. Injury2011;42:556-561. Google Scholar

31 Jones E , YangX. Mesenchymal stem cells and bone regeneration: current status. Injury2011;42:562-568.CrossrefPubMed Google Scholar