header advert
Results 1 - 8 of 8
Results per page:
Bone & Joint Research
Vol. 6, Issue 4 | Pages 208 - 215
1 Apr 2017
Decambron A Manassero M Bensidhoum M Lecuelle B Logeart-Avramoglou D Petite H Viateau V

Objectives

To compare the therapeutic potential of tissue-engineered constructs (TECs) combining mesenchymal stem cells (MSCs) and coral granules from either Acropora or Porites to repair large bone defects.

Materials and Methods

Bone marrow-derived, autologous MSCs were seeded on Acropora or Porites coral granules in a perfusion bioreactor. Acropora-TECs (n = 7), Porites-TECs (n = 6) and bone autografts (n = 2) were then implanted into 25 mm long metatarsal diaphyseal defects in sheep. Bimonthly radiographic follow-up was completed until killing four months post-operatively. Explants were subsequently processed for microCT and histology to assess bone formation and coral bioresorption. Statistical analyses comprised Mann-Whitney, t-test and Kruskal–Wallis tests. Data were expressed as mean and standard deviation.


Orthopaedic Proceedings
Vol. 99-B, Issue SUPP_1 | Pages 114 - 114
1 Jan 2017
Decambron A Fournet A Manassero M Bensidhoum M Logeart-Avramoglou D Petite H Viateau V
Full Access

Bone tissue engineering constructs (BTEC) combining natural resorbable osteoconductive scaffolds and mesenchymal stem cells (MSCs) have given promising results to repair critical size bone defect. Yet, results remain inconsistent. Adjonction of an osteoinductive factor to these BTEC, such as rh-BMP-2, to improve bone healing, seems to be a relevant strategy. However, currently supraphysiological dose of this protein are used and can lead to adverse effects such as inflammation, ectopic bone and/or bone cyst formation. Interestingly, in a preliminary study conducted in ectopic site in a murine model, a synergistic effect on bone formation was observed only when a low dose of rh-BMP-2 was associated with MSCs-seeded coral scaffolds but not with a high dose.

The objective of the study was then to evaluate a BTEC combining coral scaffold, MSCs and a low dose of rh-BMP-2 in a large animal model of clinical relevance.

Sixteen sheep were used for this study.

MSCs were isolated from an aspirate of bone marrow harvested from the iliac crest of each sheep receiving BTEC with MSCs, cultivated and seeded on Acroporacoral scaffolds one week before implantation.

Rh-BMP-2, used at two different doses (low dose: 68μg/defect and high dose: 680μg/defect), was diluted and absorbed on Acroporacoral scaffold one day before implantation.

Metatarsal segmental bone defects (25 mm) were made in the left metatarsal bone of the sheep, stabilized by plate fixation, and filled with Acroporacoral scaffolds loaded with either (i) MSCs and a low dose of rh-BMP-2 (Group 1;n=6), (ii) a low dose of rh-BMP-2 (Group 2;n=5), (iii) a high dose of rh-BMP-2 (Group 3;n=5). Standard radiographs were taken after each surgery and each month until sheep sacrifice, 4 months postoperatively. Bone healing and scaffold resorption were assessed by micro-computed-tomography (μCT) and histomorphometry. Results were compared to a historical control group in which coral scaffolds were loaded with MSCs.

Bone volumes (BV) evaluated by μCT and bone surfaces (BS) evaluated by histomorphometry did not differ between groups (BV: 1914±870, 1737±841, 1894±1028 and 1835±1342 mm3; BS: 25,41±14,25, 19,85±8,31, 25,54±16,98 and 26,08±22,52 %; groups 1, 2, 3 and control respectively); however, an higher bone union was observed in group 1 compared to the others (3, 1, 2 and 2 sheep with bone union in groups 1, 2, 3 and control respectively). No histological abnormalities were observed in any group. Coral resorption was almost complete in all specimens. No significant difference in coral volumes and coral surfaces was observed between groups. A trend towards a higher variability in coral resorption was noted in group 1 compared to the others.

There seems to be a benefit to associate low dose of rh-BMP-2 with MSCs-seeded coral scaffolds as this strategy allowed an increase of bone unions in our model. Yet, results remain inconsistent. Although, defective coupling between scaffold resorption and bone formation impaired bone healing in some animals, adjunction of rh-BMP-2 (even at low dose) to CSMs loaded construct is a promising strategy for bone tissue engineering.


Orthopaedic Proceedings
Vol. 96-B, Issue SUPP_11 | Pages 238 - 238
1 Jul 2014
Deschepper M Paquet J Manassero M Logeart-Avramoglou D Bensidhoum M Petite H
Full Access

Summary

In this study, we challenged the current paradigm of human Mesenchymal Stem Cells survival, which assigned a pivotal role to oxygen, by testing the hypothesis that exogenous glucose may be key to their survival.

Introduction

The survival of human mesenchymal stem cells (hMSCs) has elicited a great deal of interest, because it is relevant to the efficacy of engineered tissues. However, to date, hMSCs have not met this promise, in part due to the high death rate of cells upon transplantation. In this study, we challenged the current paradigm of hMSC survival, which assigned a pivotal role to oxygen, by testing the hypothesis that exogenous glucose may be key to hMSC survival.


Orthopaedic Proceedings
Vol. 96-B, Issue SUPP_11 | Pages 191 - 191
1 Jul 2014
Sladkova M Manassero M Myrtil V Savari H Fall M Thomas D Bensidhoum M Logeart-Avramoglou D Petite H
Full Access

Introduction

The use of mesenchymal stem cells (MSCs) loaded on osteoconductive scaffolds has emerged as a potential new treatment of large bone defects but has generated marginally successful results in terms of new bone formation. It is supposed that MSC massive death post implantation is a major obstacle for the exhibition of their osteogenic potential. Yet, the very few studies conducted using primary human MSCs derived from bone marrow (hMSCs), a clinically pertinent cell source, did not demonstrate that cell survival is required for new bone formation. In order to elucidate whether cell survival is needed for hMSC to express their osteogenic potential, the present study examined in an ectopic mouse model the relationship between cell survival and osteogenic potential of hMSCs loaded onto osteoconductive scaffold.

Materials and Methods

hMSCs (106) were seeded on 40-mg calcium carbonate (Biocoral) particles (size: 610–1000 µm), wrapped in fibrin gel (Baxter), and implanted subcutaneously into immunodeficient (nu/nu) mice (n=8/group). The fate of implanted cells was analysed using the bioluminescence and immunohistochemistry. For this, hMSCs were transduced with Luc-GFP (Luciferase-Green fluorescent protein) lentiviral vectors prior to experimentation. Bone formation was analysed 8 weeks post implantation on both non-decalcified and decalcified samples.


Orthopaedic Proceedings
Vol. 96-B, Issue SUPP_11 | Pages 70 - 70
1 Jul 2014
Manassero M Viateau V Decambron A Deschepper M Bensidhoum M Logeart D Petite H
Full Access

Summary

Despite similar, early and massive death, hMSCs promote bone formation which was higher in orthotopic than ectopic site suggesting a trophic effect of hMSCs. Ectopic implantation is suitable to evaluate cell survival, but assessment of bone formation requires orthotopic implantation

Introduction

Tissue constructs containing mesenchymal stem cells (MSCs) are appealing strategies for repairing large segmental bone defects but they do not allow consistent bone healing and early and massive MSCs death was identified as a cause of failure. However, little is known about cell survival in the clinical micro-environment encountered during bone healing process, whereas ectopic evaluation is well documented.

In vivo, luciferase-labelled human MSCs survival, within osteoconductive scaffold, was compared in orthotopic and ectopic locations, and bone formation ability of LF-hMSCs-Acropora constructs was evaluated. Interest and limits of each model were highlighted.


Orthopaedic Proceedings
Vol. 96-B, Issue SUPP_11 | Pages 114 - 114
1 Jul 2014
Viateau V Manassero M Petite H Logeart-Avramoglou D Sladkova M Oudina K Bensidhoum M
Full Access

Short Summary

The present study demonstrated the feasibility of culturing a large number of standardised granular MSC-containing constructs in a packed bed/column bioreactor that can produce sheep MSC-containing constructs to repair critical-size bone defects in sheep model.

Introduction

Endogenous tissue regeneration mechanisms do not suffice to repair large segmental long-bone defects. Although autologous bone graft remains the gold standard for bone repair, the pertinent surgical technique is limited. Tissue constructs composed of MSCs seeded onto biocompatible scaffolds have been proposed for repairing bone defects and have been established in clinically-relevant animal models. Producing tissue constructs for healing bone defects of clinically-relevant volume requires a large number of cells to heal an approximately 3 cm segmental bone defect. For this reason, a major challenge is to expand cells from a bone marrow aspirate to a much larger, and sufficient, number of MSCs. In this respect, bioreactor systems which provide a reproducible and well-controlled three-dimensional (3D) environment suitable for either production of multiple or large size tissue constructs are attractive approaches to expand MSCs and obtain MSC-containing constructs of clinical grade. In these bioreactor systems, MSCs loaded onto scaffolds are exposed to fluid flow, a condition that provides both enhanced access to oxygen and nutrients as well as fluid-flow-driven mechanical stimulation to cells. The present study was to evaluate bioreactor containing autologous MSCs loaded on coral scaffolds to repair critical-size bone defects in sheep model.


Orthopaedic Proceedings
Vol. 96-B, Issue SUPP_11 | Pages 90 - 90
1 Jul 2014
Logeart-Avramoglou D Guillot R Becquart P Gilde F Sailhan F Lapeyre A Picart C
Full Access

Summary

Coating of titanium implants with BMP-2-loaded polyelectrolyte multilayer films conferred the implant surface with osteoinductive properties which are fully preserved upon both air-dried storage and γ-sterilization.

Although BMP-2 is recognised as an important molecule for bone regeneration, its supraphysiological doses currently used in clinical practice has raised serious concerns about cost-effectiveness and safety issues. Thus, there is a strong motivation to engineer new delivery systems or to provide already approved materials with new functionalities. Immobilizing the growth factor onto the surface of implants would reduce protein diffusion and increase residence time at the implantation site. To date, modifying the surfaces of metal materials, such as titanium or titanium alloys, at the nanometer scale for achieving dependable, consistent and long-term osseointegration remains a challenging approach.

In this context, we have developed an osteoinductive coating of a porous titanium implant using biomimetic polyelectrolyte multilayer (PEM) films used as carriers of BMP-2. The PEM films were prepared by alternate deposition of 24 layer pairs of poly(L-lysine) (PLL) and hyaluronic acid (HA) layers (∼3.5 µm in thickness); such films were then cross-linked by means of a water-soluble carbodiimide (EDC) at different degrees. The amount of BMP-2 loaded in these films was tuned (ranging from 1.4 to 14.3 µg/cm2) depending on the cross-linking extent of the film and of the BMP-2 initial concentration. Because packaging, and storage of the devices are important issues that may limit a wide application of biologically functionalised materials, we assessed in the present study the osteoinductive performance of the BMP-2 loaded PEM coatings onto custom-made 3D porous scaffolds made of Ti-6Al-4V in vitro and in vivo pertinent to long-term storage in a dry state and to sterilization by gamma irradiation.

Analysis of PEM films by infrared spectroscopy evidenced that the air-dried films were stable for at least one year of storage at 4°C and they resisted exposure to γ-irradiation at clinically approved doses. The preservation of the growth factor bioactivity was evaluated both in vitro (using C2C12 cell model) and in vivo (in a rat ectopic model). In vitro, BMP-2 loaded in dried PEM films exhibited shelf-life stability at 4°C over a one-year period. However, its bioactivity decreased from 50 to 80% after γ-irradiation at 25 and 50 kGy, respectively. Remarkably, the in vivo studies showed that the amount of new bone tissue formation induced by BMP-2 contained in PEM-coated Ti implants was not affected after air-drying of the implants and sterilization at 25 kGy indicating the full preservation of the growth factor bioactivity.

Altogether, our results provided evidence of the remarkable property of PEM film coatings that both sequester BMP-2 and preserve its full in vivo osteoinductive potential upon both storage and γ-sterilization. The protective effects of PEM films on the growth factor bioactivity may be attributed to both the high water content in (PLL/HA) films (∼90%) and to their porosity, which may provide a “protein-friendly” environment similar to the natural extracellular matrix. This novel “off-the-shelf” technology of functionalised implants opens promising applications in prosthetic and tissue engineering fields.


Orthopaedic Proceedings
Vol. 96-B, Issue SUPP_11 | Pages 118 - 118
1 Jul 2014
Logeart-Avramoglou D Monfoulet L Becquart P Pacard E Vandame K Bourguignon M Marchat D Petite H
Full Access

Summary

45S5 bioactive glass combined with hMSC did not permit de novo ectopic bone formation. Such absence of osteogenicity was most likely due to the alkalinization of the 45S5 microenvironment that affects adversely the osteogenic differentiation of stem/precursor cells.

Bone marrow stromal cells (BMSCs) are capable of bone formation and can promote the repair of osseous defects when implanted in appropriate scaffolds. The most promising biomaterials for application in bone tissue engineering (TE) are hydroxyapatite (HA), tricalcium phosphate (TCP), calcium carbonate (coral) ceramics or bioactive glasses (BG) because of their osteoconductive properties and ability to enhance bone formation. However, information regarding the osteogenic potential of hBMSCs in combination with BG scaffolds is strikingly lacking in the TE field. The present study focused on evaluating the osteogenicity of bone constructs prepared from particles of 45S5 BG combined with hBMSCs in comparison with biphasic HA/TCP or coral particles, in a mouse ectopic model.

The in vivo osteogenicity was then correlated with various aspects of the effects of the scaffold materials tested on hBMSCs functions pertinent to bone tissue formation. Particular attention was given to the pH in the microenvironment where the cells reside in TE constructs and its effect on the osteoblastic differentiation of hBMSCs. In vivo experiments evidenced that 45S5 BG constructs with hBMSCs failed to form ectopic bone. In contrast, the cell constructs prepared with either HA/TCP or coral ceramics displayed great and consistent capacity for the ectopic bone formation. The cytocompatibility of hBMSCs on BG material was addressed and no differences were evidenced between HA/TCP and coral substrates related to the adhesion of hBMSCs and their proliferation in vitro. The hBMSCs viability was even higher within the 45S5 BG-containing constructs compared to the other two types of material constructs tested both in vitro and in vivo. These findings indicated that the absence of de novo bone formation in the hBMSCs-containing 45S5 BG constructs was not the result of cytotoxic effects of the BG material.

The potential of osteogenic differentiation of hBMSCs cultured on material substrates was next addressed and the ALP activity of hBMSCs was significantly diminished when these cells were cultured on 45S5 BG as compared to either HA/TCP or coral substrates. Because BG materials are well-known for causing external alkalinisation, the pH was specifically measured in TE constructs. The pH inside the cell-containing BG constructs, measured ex vivo, was 8.0 (i.e. 0.4–0.5 units more alkaline than that measured in the coral- or HA/TCP-constructs). The impact of such external alkalinisation on the osteogenic differentiation of hBMSCs was assessed by culturing the cells over a wide range of alkaline pH. The hBMSCs expression of osteogenic markers, ALP activity and mineralization were not significantly affected at moderate external alkaline pH (≤ 7.90) but were dramatically inhibited at higher pH.

Altogether, these findings provided evidence that despite 45S5 BG are reported to be good osteoconductive materials, they are not necessarily good scaffolds for TE, most likely due to the alkalinization of the 45S5 microenvironment that affects adversely the osteogenic differentiation of precursor cells. Controlling the shifts of pH in the local engineered extracellular environment is a critical issue for the development of bioactive TE scaffolds.