1
|
Kaneko Y, Minehara H, Sonobe T, Kameda T, Sekiguchi M, Matsushita T, Konno SI, Matsumoto Y. Differences in macrophage expression in induced membranes by fixation method - Masquelet technique using a mouse's femur critical-sized bone defect model. Injury 2024; 55:111135. [PMID: 37925281 DOI: 10.1016/j.injury.2023.111135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 10/06/2023] [Accepted: 10/14/2023] [Indexed: 11/06/2023]
Abstract
INTRODUCTION Masquelet's induced membrane technique (MIMT) is an emerging method for reconstructing critical-sized bone defects. However, an incomplete understanding of the underlying biological and physical processes hinders further optimization. This study investigated the effect of different bone-defect fixation methods on macrophage expression in an induced membrane using a novel mouse plate-fixed Masquelet model. METHODS Mice were divided into Plate-fixed Masquelet (P-M), Intramedullary-fixed Masquelet (IM-M), Plate-fixed Control (P-C), and Back subfascial (B) groups. In the P-M and IM-M groups, a polymethylmethacrylate (PMMA) spacer was implanted into a 3 mm bone defect, while the defect in the P-C group remained unfilled. In group B, a spacer was inserted under the back fascia to examine membrane formation caused by a simple foreign body reaction. Tissues were collected at 1, 2, and 4 weeks postoperatively. Hematoxylin and eosin (H&E) staining and immunohistochemistry (CD68 and CD163: macrophage markers) were performed to assess macrophage expression within the membrane. qPCR was performed to measure the expression of CD68, CD163, and fibroblast growth factor 2 (FGF2). RESULTS Four weeks post-operation, the P-M group presented with minimal callus growth, whereas the IM-M group exhibited vigorous growth. The P-M and IM-M groups displayed a tri-layered membrane structure, which is consistent with the results of previous studies. The IM-M group had significantly thicker membranes, whereas the P-M group exhibited higher expression levels of CD68, CD163, and FGF2. Group P-C showed no osteogenesis, whereas group B maintained a thin, cell-dense membrane structure. The P-M group consistently showed higher gene expression levels than the P-C and P-B groups. CONCLUSION This study introduced a mouse plate fixation model for MIMT. The induced membranes could be adequately evaluated in this model. Induced membranes are formed by foreign body reactions to PMMA spacers; however, their properties are clearly different from those of simple foreign body reaction capsules and granulation tissues that infiltrate bone defects, suggesting that they are more complex tissues. The characteristics and expression of macrophages within these induced membranes varied according to the bone defect fixation method.
Collapse
Affiliation(s)
- Yota Kaneko
- Department of Orthopaedic Surgery, Fukushima Medical University School of Medicine, Japan
| | - Hiroaki Minehara
- Department of Traumatology, Fukushima Medical University School of Medicine, Japan.
| | - Tatsuru Sonobe
- Department of Orthopaedic Surgery, Fukushima Medical University School of Medicine, Japan
| | - Takuya Kameda
- Department of Orthopaedic Surgery, Fukushima Medical University School of Medicine, Japan
| | - Miho Sekiguchi
- Department of Orthopaedic Surgery, Fukushima Medical University School of Medicine, Japan; Laboratory Animal Research Centor, Fukushima Medical University School of Medicine, Japan
| | - Takashi Matsushita
- Department of Traumatology, Fukushima Medical University School of Medicine, Japan
| | - Shin-Ich Konno
- Department of Orthopaedic Surgery, Fukushima Medical University School of Medicine, Japan
| | - Yoshihiro Matsumoto
- Department of Orthopaedic Surgery, Fukushima Medical University School of Medicine, Japan
| |
Collapse
|
2
|
Wu JH, Bao QW, Wang SK, Zhou PY, Xu SG. Mechanisms of the Masquelet technique to promote bone defect repair and its influencing factors. Chin J Traumatol 2024:S1008-1275(24)00054-3. [PMID: 38734563 DOI: 10.1016/j.cjtee.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 03/21/2024] [Accepted: 04/02/2024] [Indexed: 05/13/2024] Open
Abstract
The Masquelet technique, also known as the induced membrane technique, is a surgical technique for repairing large bone defects based on the use of a membrane generated by a foreign body reaction for bone grafting. This technique is not only simple to perform, with few complications and quick recovery, but also has excellent clinical results. To better understand the mechanisms by which this technique promotes bone defect repair and the factors that require special attention in practice, we examined and summarized the relevant research advances in this technique by searching, reading, and analysing the literature. Literature show that the Masquelet technique may promote the repair of bone defects through the physical septum and molecular barrier, vascular network, enrichment of mesenchymal stem cells, and high expression of bone-related growth factors, and the repair process is affected by the properties of spacers, the timing of bone graft, mechanical environment, intramembrane filling materials, artificial membrane, and pharmaceutical/biological agents/physical stimulation.
Collapse
Affiliation(s)
- Jiang-Hong Wu
- Department of Emergency, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China; Department of Trauma Orthopedics, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Quan-Wei Bao
- Trauma Center, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Shao-Kang Wang
- Department of Trauma Orthopedics, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Pan-Yu Zhou
- Department of Trauma Orthopedics, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Shuo-Gui Xu
- Department of Trauma Orthopedics, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
3
|
Siverino C, Vanvelk N, Nehrbass D, Mischler D, Geoff Richards R, Morgenstern M, Zeiter S, Arens D, Fintan Moriarty T. Comparative bone healing with induced membrane technique (IMT) versus empty defects in septic and aseptic conditions in a novel rabbit humerus model. BMC Musculoskelet Disord 2023; 24:886. [PMID: 37964215 PMCID: PMC10644571 DOI: 10.1186/s12891-023-07031-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 11/09/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND Long bone defects resulting from primary trauma or secondary to debridement of fracture-related infection (FRI) remain a major clinical challenge. One approach often used is the induced membrane technique (IMT). The effectiveness of the IMT in infected versus non-infected settings remains to be definitively established. In this study we present a new rabbit humerus model and compare the IMT approach between animals with prior infection and non-infected equivalents. METHODS A 5 mm defect was created in the humerus of New Zealand White rabbits (n = 53) and fixed with a 2.5 mm stainless steel plate. In the non-infected groups, the defect was either left empty (n = 6) or treated using the IMT procedure (PMMA spacer for 3 weeks, n = 6). Additionally, both approaches were applied in animals that were inoculated with Staphylococcus aureus 4 weeks prior to defect creation (n = 5 and n = 6, respectively). At the first and second revision surgeries, infected and necrotic tissues were debrided and processed for bacteriological quantification. In the IMT groups, the PMMA spacer was removed 3 weeks post implantation and replaced with a beta-tricalcium phosphate scaffold and bone healing observed for a further 10 weeks. Infected groups also received systemic antibiotic therapy. The differences in bone healing between the groups were evaluated radiographically using a modification of the radiographic union score for tibial fractures (RUST) and by semiquantitative histopathology on Giemsa-Eosin-stained sections. RESULTS The presence of S. aureus infection at revision surgery was required for inclusion to the second stage. At the second revision surgery all collected samples were culture negative confirming successful treatment. In the empty defect group, bone healing was increased in the previously infected animals compared with non-infected controls as revealed by radiography with significantly higher RUST values at 6 weeks (p = 0.0281) and at the end of the study (p = 0.0411) and by histopathology with increased cortical bridging (80% and 100% in cis and trans cortical bridging in infected animals compared to 17% and 67% in the non-infected animals). With the IMT approach, both infected and non-infected animals had positive healing assessments. CONCLUSION We successfully developed an in vivo model of bone defect healing with IMT with and without infection. Bone defects can heal after an infection with even better outcomes compared to the non-infected setting, although in both cases, the IMT achieved better healing.
Collapse
Affiliation(s)
- Claudia Siverino
- AO Research Institute Davos, Clavadelerstrasse 1, Davos-Platz, 7270, Switzerland
| | - Niels Vanvelk
- AO Research Institute Davos, Clavadelerstrasse 1, Davos-Platz, 7270, Switzerland
| | - Dirk Nehrbass
- AO Research Institute Davos, Clavadelerstrasse 1, Davos-Platz, 7270, Switzerland
| | - Dominic Mischler
- AO Research Institute Davos, Clavadelerstrasse 1, Davos-Platz, 7270, Switzerland
| | | | - Mario Morgenstern
- Center for Musculoskeletal Infections, Department of Orthopaedic and Trauma Surgery, University Hospital Basel, Basel, Switzerland
| | - Stephan Zeiter
- AO Research Institute Davos, Clavadelerstrasse 1, Davos-Platz, 7270, Switzerland
| | - Daniel Arens
- AO Research Institute Davos, Clavadelerstrasse 1, Davos-Platz, 7270, Switzerland
| | - Thomas Fintan Moriarty
- AO Research Institute Davos, Clavadelerstrasse 1, Davos-Platz, 7270, Switzerland.
- Center for Musculoskeletal Infections, Department of Orthopaedic and Trauma Surgery, University Hospital Basel, Basel, Switzerland.
| |
Collapse
|
4
|
Guimarães JAM, Scorza BJB, Machado JAP, Cavalcanti ADS, Duarte MEL. Characterization of the Masquelet Induced Membrane Technique in a Murine Segmental Bone Defect Model. Rev Bras Ortop 2023; 58:e798-e807. [PMID: 37908532 PMCID: PMC10615599 DOI: 10.1055/s-0043-1771490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 12/16/2022] [Indexed: 11/02/2023] Open
Abstract
Objective To reproduce in an animal model the surgical technique of Masquelet used in the treatment of critical bone defects and to analyze the characteristics of the membrane formed around the bone cement. Methods A 10mm critical defect was created in the femoral shaft of 21 Sprague-Dawley rats. After resection of the central portion of the diaphysis, the defect was stabilized with a Kirschner wire introduced through the medullary canal and with the interposition of a bone cement spacer. After 2, 4, and 6 weeks of the surgical procedure, the animals were euthanized and evaluated on radiographs of the posterior limb regarding the size of the defect, alignment and stability of the osteosynthesis. The membranes formed around the spacer were subjected to histological analysis to assess thickness, connective tissue maturation and vascular density. Results Over time, the membranes initially made up of loose connective tissue were replaced by membranes represented by dense connective tissue, rich in thick collagen fibers. At six weeks, membrane thickness was greater (565 ± 208μm) than at four (186.9 ± 70.21μm, p = 0.0002) and two weeks (252.2 ± 55.1μm, p = 0.001). All membranes from the initial time showed foci of osteogenic differentiation that progressively reduced over time. Conclusion In addition to the structural and protective function of the membrane, its intrinsic biological characteristics can actively contribute to bone regeneration. The biological activity attributed by the presence of foci of osteogenesis confers to the membrane the potential of osteoinduction that favors the local conditions for the integration of the bone graft.
Collapse
Affiliation(s)
| | - Breno Jorge Braga Scorza
- Coordenador de pós-graduação, Instituto Nacional de Traumatologia e Ortopedia, Rio de Janeiro, RJ, Brasil
| | - Jamila Alessandra Perini Machado
- Coordenador de pós-graduação, Instituto Nacional de Traumatologia e Ortopedia, Rio de Janeiro, RJ, Brasil
- Pesquisadora, Laboratório de Pesquisa de Ciências Farmacêuticas, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro, Brasil
| | | | - Maria Eugênia Leite Duarte
- Coordenador de pós-graduação, Instituto Nacional de Traumatologia e Ortopedia, Rio de Janeiro, RJ, Brasil
- Cirurgião ortopédico, Instituto D'Or de Ensino e Pesquisa, IDOR, Rio de Janeiro, RJ, Brasil
| |
Collapse
|
5
|
Chen X, Chen G, Chen Z, Zhang J. Bone defect reconstruction using Masquelet technique for calcaneal chondroblastoma: a case report. J Surg Case Rep 2023; 2023:rjad401. [PMID: 37457866 PMCID: PMC10348867 DOI: 10.1093/jscr/rjad401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 06/24/2023] [Indexed: 07/18/2023] Open
Abstract
Masquelet technique demonstrated superiority in reconstructing long bone defect after trauma or infection. However, reports in foot tumor were rare. A 24-year-old male diagnosed with calcaneal chondroblastoma who had a defect of calcaneal after intralesional curettage. We reconstructed the defect by Masquelet technique. This is the first case as far as we know that reported Masquelet technique for calcaneal tumor. The technique to treat irregular bone defects after operation can be considered in other similar situations.
Collapse
Affiliation(s)
| | | | - Zhifu Chen
- Department of Orthopedics, Cancer Hospital of Yunnan Province, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jing Zhang
- Correspondence address. Department of Orthopedics, Cancer Hospital of Yunnan Province, The Third Affiliated Hospital of Kunming Medical University, Kunming, China. Tel: +86-0871 68189037, Fax: +86-0871 68189037. E-mail:
| |
Collapse
|
6
|
Takase K, Fukui T, Oe K, Sawauchi K, Yoshikawa R, Yamamoto Y, Hayashi S, Matsumoto T, Kuroda R, Niikura T. Effect of low-intensity pulsed ultrasound on osteogenic differentiation of human induced membrane-derived cells in Masquelet technique. Injury 2023:S0020-1383(23)00343-1. [PMID: 37062672 DOI: 10.1016/j.injury.2023.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/02/2023] [Accepted: 04/08/2023] [Indexed: 04/18/2023]
Abstract
INTRODUCTION The Masquelet technique is a relatively new method for large bone defect treatment. In this technique, grafted bone tissue is used, and after the cement is removed, the induced membrane (IM; that form around the cement spacers placed in the bone defect region) is thought to play an important role in promoting bone formation. On the other hand, low-intensity pulsed ultrasound (LIPUS) is known to promote fracture healing and angiogenesis through mechanical stimulation. This study aimed to investigate the in vitro effects of LIPUS on the osteogenic differentiation of human induced membrane-derived cells (IMCs). METHODS Seven patients who had been treated using the Masquelet technique were enrolled. The IM was harvested during the second stage of the technique. IMCs were isolated, cultured in growth medium, and then divided into two groups: (1) control group, IMCs cultured in osteogenic medium without LIPUS, and (2) LIPUS group, IMCs cultured in osteogenic medium with LIPUS treatment. Adherent cells from the IM samples were harvested after the first passage and evaluated for cell surface protein expression using immunostaining. A cell proliferation assay was used to count the number of IMCs using a hemocytometer. Osteogenic differentiation capability was assessed using an alkaline phosphatase (ALP) activity assay, Alizarin Red S staining, and real-time reverse transcription-polymerase chain reaction. RESULTS Cell surface antigen profiling revealed that the IMCs contained cells positive for the mesenchymal stem cell-related markers CD73, CD90, and CD105. No significant difference in cell numbers was found between the control and LIPUS groups. The ALP activity of IMCs in the LIPUS group was significantly higher than that in the control group on days 7 and 14. Alizarin red S staining intensity was significantly higher in the LIPUS group than in the control group on day 21. Runx2 and VEGF expression was significantly upregulated on days 7 and 14, respectively, compared with levels in the control group. CONCLUSION We demonstrated the significant effect of LIPUS on the osteogenic differentiation of human IMCs. This study indicates that LIPUS can be used as an additional tool for the enhancement of the healing process of the Masquelet technique.
Collapse
Affiliation(s)
- Kyohei Takase
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Tomoaki Fukui
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Keisuke Oe
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Kenichi Sawauchi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Ryo Yoshikawa
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Yuya Yamamoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Shinya Hayashi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Tomoyuki Matsumoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Takahiro Niikura
- Department of Orthopaedic Surgery, Hyogo Prefectural Nishinomiya Hospital, 13-9 Rokutanji-cho, Nishinomiya 662-0918, Japan.
| |
Collapse
|
7
|
Li D, Zhao D, Zeng Z, Huang F, Jiang Z, Xiong H, Guan T, Fang B, Li Y. Ternary regulation mechanism of Rhizoma drynariae total flavonoids on induced membrane formation and bone remodeling in Masquelet technique. PLoS One 2022; 17:e0278688. [PMID: 36473008 PMCID: PMC9725127 DOI: 10.1371/journal.pone.0278688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022] Open
Abstract
CONTEXT Rhizoma drynariae total flavonoids (RDTF) are used to treat fractures. CD31hiEmcnhi vessels induced by PDGF-BB secreted by osteoclast precursors, together with osteoblasts and osteoclasts, constitute the ternary regulatory mechanism of bone tissue reconstruction. OBJECTIVE This study aimed to determine whether RDTF can promote bone tissue remodeling and induce membrane growth in the rat Masquelet model and to explore its molecular mechanism based on the ternary regulation theory. METHODS Thirty-six SD rats were randomized to three groups: blank, induced membrane, and RDTF treatment (n = 12/group). The gross morphological characteristics of the new bone tissue were observed after 6 weeks. Sixty SD rats were also randomized to five groups: blank, induction membrane, low-dose RDTF, medium-dose RDTF, and high-dose RDTF (n = 12/group). After 4 weeks, immunohistochemistry and western blot were used to detect the expression of membrane tissue-related proteins. The mRNA expression of key factors of ternary regulation was analyzed by qRT-PCR. RESULTS RDTF positively affected angiogenesis and bone tissue reconstruction in the bone defect area. RDTF could upregulate the expression of key factors (PDGF-BB, CD31, and endomucin), VEGF, and HMGB1 mRNA and proteins in the ternary regulation pathway. DISCUSSION AND CONCLUSION Although the expected CD31hiEmcnhi vessels in the induction membrane were not observed, this study confirmed that RDTF could promote the secretion of angiogenic factors in the induced membrane. The specific mechanisms still need to be further studied.
Collapse
Affiliation(s)
- Ding Li
- Department of Orthopedic, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dun Zhao
- Department of Orthopedic, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhikui Zeng
- Department of Orthopedics, The Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, China
| | - Feng Huang
- Department of Orthopedic, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ziwei Jiang
- Department of Orthopedic, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hao Xiong
- Department of Orthopedic, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tianan Guan
- Department of Orthopedic, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bin Fang
- Department of Orthopedic, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yue Li
- Department of Orthopedic, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
8
|
Effects of PMMA spacer loaded with varying vancomycin concentrations on bone regeneration in the Masquelet technique. Sci Rep 2022; 12:4255. [PMID: 35277575 PMCID: PMC8917238 DOI: 10.1038/s41598-022-08381-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/07/2022] [Indexed: 12/04/2022] Open
Abstract
Whether antibiotics should be included remains greatly debated in Masquelet technique. This study intended to determine the effect of polymethyl methacrylate (PMMA) spacer loaded with different vancomycin concentrations on bone defect repair. Hollow cylindrical spacers consisting of PMMA and varying vancomycin concentrations (0, 1, 2, 4, 6, 8, and 10 g) were prepared. Critical bone defects of rabbits were created at the radial shaft, and spacers were implanted and subsequently intramedullary fixed with retrograde Kirschner’s wires (n = 4 for each vancomycin concentration). After 4 weeks, the induced membranes were opened and cancellous allografts were implanted into the defects. Eight weeks post-operatively, the results of X-ray, histology, and micro-CT revealed that some cortical bone was formed to bridge the gap and the bone marrow cavity was formed over time. Quantitatively, there was more new bone formation in the groups with a relatively lower vancomycin concentration (1–4 g) compared with that in the groups with a higher vancomycin concentration (6–10 g). Our findings suggested that PMMA spacers loaded with relatively lower vancomycin concentrations (1–4 g) did not interfere with new bone formation, whereas spacers loaded with relatively higher vancomycin concentrations (6–10 g) had negative effects on bone formation.
Collapse
|
9
|
The induced membrane technique in animal models: a systematic review. OTA Int 2022; 5:e176. [PMID: 35282388 PMCID: PMC8900461 DOI: 10.1097/oi9.0000000000000176] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 12/15/2021] [Indexed: 01/10/2023]
Abstract
Objectives: Data Sources: Study Selection: Data Extraction: Data Synthesis: Conclusions:
Collapse
|
10
|
Alford AI, Nicolaou D, Hake M, McBride-Gagyi S. Masquelet's induced membrane technique: Review of current concepts and future directions. J Orthop Res 2021; 39:707-718. [PMID: 33382115 PMCID: PMC8005442 DOI: 10.1002/jor.24978] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/16/2020] [Accepted: 12/28/2020] [Indexed: 02/04/2023]
Abstract
Masquelet's induced membrane technique (MIMT) is a relatively new, two-stage surgical procedure to reconstruct segmental bone defects. First performed by Dr. Masquelet in the mid-1980s, MIMT has shown great promise to revolutionize critical-sized bone defect repair and has several advantages over its alternative, distraction osteogenesis (DO). Also, its success in extremely challenging cases (defects > 15 cm) suggests that its study could lead to discovery of novel biological mechanisms that might be at play during segmental defect healing and fracture non-union. MIMT's advantages over DO have led to a world-wide increase in MIMT procedures over the past decades. However, MIMT often needs to be repeated and so the average initial success rate in adults lags significantly behind that of DO (86% vs 95%, respectively). The autologous foreign-body membrane created during the first stage by the immune system's response to a polymethyl methacrylate bone cement spacer is critical to supporting the morselized bone graft implanted in the second stage. However, the biological and/or physical mechanisms by which the membrane supports graft to bone union are unclear. This lack of knowledge makes refining MIMT and improving the success rates through technique improvements and patient selection a significant challenge and hinders wider adoption. In this review, current knowledge from basic, translational, and clinical studies is summarized. The dynamics of both stages under normal conditions as well as with drug or material perturbations is discussed along with perspectives on high-priority future research directions.
Collapse
Affiliation(s)
- Andrea I. Alford
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI
| | - Daemeon Nicolaou
- Department of Orthopaedic Surgery, Saint Louis University, St. Louis, MO
| | - Mark Hake
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI
| | | |
Collapse
|
11
|
Hoit G, Kain MS, Sparkman JW, Norris BL, Conway JD, Watson JT, Tornetta P, Nauth A. The induced membrane technique for bone defects: Basic science, clinical evidence, and technical tips. OTA Int 2021; 4:e106(1-5). [PMID: 37608856 PMCID: PMC10441675 DOI: 10.1097/oi9.0000000000000106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/09/2020] [Accepted: 12/11/2020] [Indexed: 08/24/2023]
Abstract
The clinical management of large bone defects continues to be a difficult clinical problem to manage for treating surgeons. The induced membrane technique is a commonly employed strategy to manage these complex injuries and achieve bone union. Basic science and clinical evidence continue to expand to address questions related to the biology of the membrane and how interventions may impact clinical outcomes. In this review, we discuss the basic science and clinical evidence for the induced membrane technique as well as provide indications for the procedure and technical tips for performing the induced membrane technique.
Collapse
Affiliation(s)
- Graeme Hoit
- Division of Orthopaedic Surgery, University of Toronto, Toronto, ON, Canada
| | - Michael S Kain
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, MA, USA
| | - Jeremy W Sparkman
- Department of Orthopaedic Surgery, Oklahoma State University, Tulsa, OK
| | - Brent L Norris
- Department of Orthopaedic Surgery, Oklahoma State University, Tulsa, OK
| | - Janet D Conway
- Rubin Institute for Advanced Orthopedics, Sinai Hospital of Baltimore, Baltimore, MD
| | - J Tracy Watson
- Department of Orthopaedic Surgery, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - Paul Tornetta
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, MA, USA
| | - Aaron Nauth
- Division of Orthopaedic Surgery, University of Toronto, Toronto, ON, Canada
- Department of Orthopaedic Surgery, St. Michael's Hospital. Toronto ON, Canada
| |
Collapse
|
12
|
Gohel N, Senos R, Goldstein SA, Hankenson KD, Hake ME, Alford AI. Evaluation of global gene expression in regenerate tissues during Masquelet treatment. J Orthop Res 2020; 38:2120-2130. [PMID: 32233004 PMCID: PMC7494657 DOI: 10.1002/jor.24676] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 03/19/2020] [Accepted: 03/25/2020] [Indexed: 02/04/2023]
Abstract
The Masquelet induced-membrane (IM) technique is indicated for large segmental bone defects. Attributes of the IM and local milieu that contribute to graft-to-bone union are unknown. Using a rat model, we compared global gene expression profiles in critically sized femoral osteotomies managed using a cement spacer as per Masquelet to those left empty. At the end of the experiment, IM and bone adjacent to the spacer were collected from the Masquelet side. Nonunion tissue in the defect and bone next to the empty defect were collected from the contralateral side. Tissues were subjected to RNA isolation, sequencing, and differential expression analysis. Cell type enrichment analysis suggested the IM and the bone next to the polymethylmethacrylate (PMMA) spacer were comparatively enriched for osteoblastic genes. The nonunion environment was comparatively enriched for innate and adaptive immune cell markers, but only macrophages were evident in the Masquelet context. iPathwayGuide was utilized to identify cell signaling pathways and protein interaction networks enriched in the Masquelet environment. For IM vs nonunion false-discovery rate correction of P values rendered overall pathway differences nonsignificant, and so only protein interaction networks are presented. For the bone comparison, substantial enrichment of pathways and networks known to contribute to osteogenic mechanisms was revealed. Our results suggest that the PMMA spacer affects the cut bone ends that are in contact with it and at the same time induces the foreign body reaction and formation of the IM. B cells in the empty defect suggest a chronic inflammatory response to a large segmental osteotomy.
Collapse
Affiliation(s)
- Nishant Gohel
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan
| | - Rafael Senos
- Department of Morphology, Universidade Federal Fluminense, Niteroi, Rio de Janeiro, Brazil
| | - Steven A. Goldstein
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan
| | - Kurt D. Hankenson
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan
| | - Mark E. Hake
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan.,Address correspondence to Mark E. Hake: Department of Orthopaedic Surgery, University of Michigan School of Medicine, 1500 E Medical Center Drive, 2912 Taubman Center SPC 5328; Ann Arbor, MI 48109; fax: +1-734-647-3277; telephone: +734-936-9839;
| | - Andrea I. Alford
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan.,Address correspondence to Andrea I. Alford: Department of Orthopaedic Surgery, University of Michigan School of Medicine, A. Alfred Taubman Biomedical Sciences Research Building, Room 2009, Ann Arbor, MI, 48109; fax: +1-734 -647-0003; telephone: +1-734-615-6104;
| |
Collapse
|
13
|
Klein C, Monet M, Barbier V, Vanlaeys A, Masquelet AC, Gouron R, Mentaverri R. The Masquelet technique: Current concepts, animal models, and perspectives. J Tissue Eng Regen Med 2020; 14:1349-1359. [PMID: 32621637 DOI: 10.1002/term.3097] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 06/02/2020] [Accepted: 06/15/2020] [Indexed: 12/20/2022]
Abstract
Bone reconstruction within a critical-sized defect remains a real challenge in orthopedic surgery. The Masquelet technique is an innovative, two-step therapeutic approach for bone reconstruction in which the placement of a poly (methylmethacrylate) spacer into the bone defect induces the neo-formation of a tissue called "induced membrane." This surgical technique has many advantages and is often preferred to a vascularized bone flap or Ilizarov's technique. Although the Masquelet technique has achieved high clinical success rates since its development by Alain-Charles Masquelet in the early 2000s, very little is known about how the process works, and few animal models of membrane induction have been developed. Our successful use of this technique in the clinic and our interest in the mechanisms of tissue regeneration (notably bone regeneration) prompted us to develop a surgical model of the Masquelet technique in rats. Here, we provide a comprehensive review of the literature on animal models of membrane induction, encompassing the defect site, the surgical procedure, and the histologic and osteogenic properties of the induced membrane. We also discuss the advantages and disadvantages of those models to facilitate efforts in characterizing the complex biological mechanisms that underlie membrane induction.
Collapse
Affiliation(s)
- Céline Klein
- Department of Pediatric Orthopedic Surgery, Amiens University Medical Center, Jules Verne University of Picardie, Amiens, France.,MP3CV-EA7517, CURS, miens University Medical Center, Jules Verne University of Picardie, Amiens, France
| | - Michael Monet
- MP3CV-EA7517, CURS, miens University Medical Center, Jules Verne University of Picardie, Amiens, France
| | - Vincent Barbier
- Department of Pediatric Orthopedic Surgery, Amiens University Medical Center, Jules Verne University of Picardie, Amiens, France.,MP3CV-EA7517, CURS, miens University Medical Center, Jules Verne University of Picardie, Amiens, France
| | - Alison Vanlaeys
- MP3CV-EA7517, CURS, miens University Medical Center, Jules Verne University of Picardie, Amiens, France
| | - Alain-Charles Masquelet
- Service de Chirurgie Orthopédique, Traumatologie et Chirurgie de la Main, Saint-Antoine Hospital, Paris, France
| | - Richard Gouron
- Department of Pediatric Orthopedic Surgery, Amiens University Medical Center, Jules Verne University of Picardie, Amiens, France.,MP3CV-EA7517, CURS, miens University Medical Center, Jules Verne University of Picardie, Amiens, France
| | - Romuald Mentaverri
- MP3CV-EA7517, CURS, miens University Medical Center, Jules Verne University of Picardie, Amiens, France.,Department of Biochemistry and Endocrine Biology, Amiens University Medical Center, Jules Verne University of Picardie, Amiens, France
| |
Collapse
|
14
|
Cuthbert RJ, Jones E, Sanjurjo-Rodríguez C, Lotfy A, Ganguly P, Churchman SM, Kastana P, Tan HB, McGonagle D, Papadimitriou E, Giannoudis PV. Regulation of Angiogenesis Discriminates Tissue Resident MSCs from Effective and Defective Osteogenic Environments. J Clin Med 2020; 9:jcm9061628. [PMID: 32481579 PMCID: PMC7355658 DOI: 10.3390/jcm9061628] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 05/20/2020] [Accepted: 05/20/2020] [Indexed: 12/12/2022] Open
Abstract
Background: The biological mechanisms that contribute to atrophic long bone non-union are poorly understood. Multipotential mesenchymal stromal cells (MSCs) are key contributors to bone formation and are recognised as important mediators of blood vessel formation. This study examines the role of MSCs in tissue formation at the site of atrophic non-union. Materials and Methods: Tissue and MSCs from non-union sites (n = 20) and induced periosteal (IP) membrane formed following the Masquelet bone reconstruction technique (n = 15) or bone marrow (n = 8) were compared. MSC content, differentiation, and influence on angiogenesis were measured in vitro. Cell content and vasculature measurements were performed by flow cytometry and histology, and gene expression was measured by quantitative polymerase chain reaction (qPCR). Results: MSCs from non-union sites had comparable differentiation potential to bone marrow MSCs. Compared with induced periosteum, non-union tissue contained similar proportion of colony-forming cells, but a greater proportion of pericytes (p = 0.036), and endothelial cells (p = 0.016) and blood vessels were more numerous (p = 0.001) with smaller luminal diameter (p = 0.046). MSCs showed marked differences in angiogenic transcripts depending on the source, and those from induced periosteum, but not non-union tissue, inhibited early stages of in vitro angiogenesis. Conclusions: In vitro, non-union site derived MSCs have no impairment of differentiation capacity, but they differ from IP-derived MSCs in mediating angiogenesis. Local MSCs may thus be strongly implicated in the formation of the immature vascular network at the non-union site. Attention should be given to their angiogenic support profile when selecting MSCs for regenerative therapy.
Collapse
Affiliation(s)
- R. J. Cuthbert
- Leeds Institute of Rheumatic and Musculoskeletal Disease, University of Leeds, Leeds LS16 7PS, UK; (R.J.C.); (E.J.); (C.S.-R.); (P.G.); (S.M.C.); (H.B.T.); (D.M.)
| | - E. Jones
- Leeds Institute of Rheumatic and Musculoskeletal Disease, University of Leeds, Leeds LS16 7PS, UK; (R.J.C.); (E.J.); (C.S.-R.); (P.G.); (S.M.C.); (H.B.T.); (D.M.)
| | - C. Sanjurjo-Rodríguez
- Leeds Institute of Rheumatic and Musculoskeletal Disease, University of Leeds, Leeds LS16 7PS, UK; (R.J.C.); (E.J.); (C.S.-R.); (P.G.); (S.M.C.); (H.B.T.); (D.M.)
- Department of Biomedical Sciences, Medicine and Physiotherapy, University of A Coruña, CIBER-BBN-Institute of Biomedical Research of A Coruña (INIBIC), A Coruña 15001, Spain
| | - A. Lotfy
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef 62511, Egypt;
| | - P. Ganguly
- Leeds Institute of Rheumatic and Musculoskeletal Disease, University of Leeds, Leeds LS16 7PS, UK; (R.J.C.); (E.J.); (C.S.-R.); (P.G.); (S.M.C.); (H.B.T.); (D.M.)
| | - S. M. Churchman
- Leeds Institute of Rheumatic and Musculoskeletal Disease, University of Leeds, Leeds LS16 7PS, UK; (R.J.C.); (E.J.); (C.S.-R.); (P.G.); (S.M.C.); (H.B.T.); (D.M.)
| | - P. Kastana
- Department of Pharmacy, School of Health Sciences, University of Patras, Patras 265 04, Greece; (P.K.); (E.P.)
| | - H. B. Tan
- Leeds Institute of Rheumatic and Musculoskeletal Disease, University of Leeds, Leeds LS16 7PS, UK; (R.J.C.); (E.J.); (C.S.-R.); (P.G.); (S.M.C.); (H.B.T.); (D.M.)
| | - D. McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Disease, University of Leeds, Leeds LS16 7PS, UK; (R.J.C.); (E.J.); (C.S.-R.); (P.G.); (S.M.C.); (H.B.T.); (D.M.)
| | - E. Papadimitriou
- Department of Pharmacy, School of Health Sciences, University of Patras, Patras 265 04, Greece; (P.K.); (E.P.)
| | - P. V. Giannoudis
- Leeds Institute of Rheumatic and Musculoskeletal Disease, University of Leeds, Leeds LS16 7PS, UK; (R.J.C.); (E.J.); (C.S.-R.); (P.G.); (S.M.C.); (H.B.T.); (D.M.)
- NIHR Leeds Biomedical Research Center, Chapel Allerton Hospital, Leeds LS7 4SA, UK
- Correspondence: ; Tel.: +44-113-392-2750; Fax: +44-113-392-3290
| |
Collapse
|
15
|
DeBaun MR, Stahl AM, Daoud AI, Pan CC, Bishop JA, Gardner MJ, Yang YP. Preclinical induced membrane model to evaluate synthetic implants for healing critical bone defects without autograft. J Orthop Res 2019; 37:60-68. [PMID: 30273977 DOI: 10.1002/jor.24153] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 09/12/2018] [Indexed: 02/04/2023]
Abstract
Critical bone defects pose a formidable orthopaedic problem in patients with bone loss. We developed a preclinical model based on the induced membrane technique using a synthetic graft to replace autograft for healing critical bone defects. Additionally, we used a novel osteoconductive scaffold coupled with a synthetic membrane to evaluate the potential for single-stage bone regeneration. Three experimental conditions were investigated in critical femoral defects in rats. Group A underwent a two-stage procedure with insertion of a polymethylmethacrylate (PMMA) spacer followed by replacement with a 3D printed polycaprolactone(PCL)/β-tricalcium phosphate (β-TCP) osteoconductive scaffold after 4 weeks. Group B received a single-stage PCL/β-TCP scaffold wrapped in a PCL-based microporous polymer film creating a synthetic membrane. Group C received a single-stage bare PCL/β-TCP scaffold. All groups were examined by serial radiographs for callus formation. After 12 weeks, the femurs were explanted and analyzed with micro-CT and histology. Mean callus scores tended to be higher in Group A. Group A showed statistically significant greater bone formation on micro-CT compared with other groups, although bone volume fraction was similar between groups. Histology results suggested extensive bone ingrowth and new bone formation within the macroporous scaffolds in all groups and cell infiltration into the microporous synthetic membrane. This study supports the use of a critical size femoral defect in rats as a suitable model for investigating modifications to the induced membrane technique without autograft harvest. Future investigations should focus on bioactive synthetic membranes coupled with growth factors for single-stage bone healing. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.
Collapse
Affiliation(s)
- Malcolm R DeBaun
- Departiment of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Alexander M Stahl
- Departiment of Orthopaedic Surgery, Stanford University, Stanford, California.,Departiment of Chemistry, Stanford University, Stanford, California
| | - Adam I Daoud
- School of Medicine, Stanford University, Stanford, California
| | - Chi-Chun Pan
- Departiment of Orthopaedic Surgery, Stanford University, Stanford, California.,Departiment of Mechanical Engineering, Stanford University, Stanford, California
| | - Julius A Bishop
- Departiment of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Michael J Gardner
- Departiment of Orthopaedic Surgery, Stanford University, Stanford, California
| | - Yunzhi P Yang
- Departiment of Orthopaedic Surgery, Stanford University, Stanford, California.,Material Science and Engineering, Stanford University, Stanford, California.,Departiment of Bioengineering, Stanford University, Stanford, California
| |
Collapse
|
16
|
Gaio N, Martino A, Toth Z, Watson JT, Nicolaou D, McBride-Gagyi S. Masquelet technique: The effect of altering implant material and topography on membrane matrix composition, mechanical and barrier properties in a rat defect model. J Biomech 2018; 72:53-62. [PMID: 29510858 PMCID: PMC5895482 DOI: 10.1016/j.jbiomech.2018.02.026] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 02/12/2018] [Accepted: 02/18/2018] [Indexed: 12/26/2022]
Abstract
The Masquelet technique is a surgical procedure to regenerate segmental bone defects. The two-phase treatment relies on the production of a vascularized foreign-body membrane to support bone grafts over three times larger than the traditional maximum. Historically, the procedure has always utilized a bone cement spacer to evoke membrane production. However, membrane formation can easily be effected by implant surface properties such as material and topology. This study sought to determine if the membrane's mechanical or barrier properties are affected by changing the spacer material to titanium or roughening the surface finish. Ten-week-old, male Sprague Dawley rats were given an externally stabilized, 6 mm femur defect which was filled with a pre-made spacer of bone cement (PMMA) or titanium (TI) with a smooth (∼1 μm) or roughened (∼8 μm) finish. After 4 weeks of implantation, the membranes were harvested, and the matrix composition, tensile mechanics, shrinkage, and barrier function was assessed. Roughening the spacers resulted in significantly more compliant membranes. TI spacers created membranes that inhibited solute transport more. There were no differences between groups in collagen or elastin distribution. This suggests that different membrane characteristics can be created by altering the spacer surface properties. Surgeons may unknowingly effecting membrane formation via bone cement preparation techniques.
Collapse
Affiliation(s)
- Natalie Gaio
- Department of Orthopaedic Surgery, Saint Louis University School of Medicine, 1402 S. Grand Blvd, Schwitalla Hall M176, St. Louis, MO 63132, USA
| | - Alice Martino
- Department of Orthopaedic Surgery, Saint Louis University School of Medicine, 1402 S. Grand Blvd, Schwitalla Hall M176, St. Louis, MO 63132, USA
| | - Zacharie Toth
- Department of Orthopaedic Surgery, Saint Louis University School of Medicine, 1402 S. Grand Blvd, Schwitalla Hall M176, St. Louis, MO 63132, USA
| | - J Tracy Watson
- Department of Orthopaedic Surgery, Saint Louis University School of Medicine, 1402 S. Grand Blvd, Schwitalla Hall M176, St. Louis, MO 63132, USA
| | - Daemeon Nicolaou
- Department of Orthopaedic Surgery, Saint Louis University School of Medicine, 1402 S. Grand Blvd, Schwitalla Hall M176, St. Louis, MO 63132, USA
| | - Sarah McBride-Gagyi
- Department of Orthopaedic Surgery, Saint Louis University School of Medicine, 1402 S. Grand Blvd, Schwitalla Hall M176, St. Louis, MO 63132, USA.
| |
Collapse
|
17
|
McBride-Gagyi S, Toth Z, Kim D, Ip V, Evans E, Watson JT, Nicolaou D. Altering spacer material affects bone regeneration in the Masquelet technique in a rat femoral defect. J Orthop Res 2018; 36:10.1002/jor.23866. [PMID: 29424019 PMCID: PMC6785358 DOI: 10.1002/jor.23866] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 01/30/2018] [Indexed: 02/04/2023]
Abstract
The Masquelet technique depends on pre-development of a foreign-body membrane to support bone regeneration with grafts over three times larger than the traditional maximum. To date, the procedure has always used spacers made of bone cement, which is the polymer polymethyl methacrylate (PMMA), to induce the foreign-body membrane. This study sought to compare (i) morphology, factor expression, and cellularity in membranes formed by PMMA, titanium, and polyvinyl alcohol sponge (PVA) spacers in the Masquelet milieu and (ii) subsequent bone regeneration in the same groups. Ten-week-old, male Sprague-Dawley rats were given an externally stabilized, 6 mm femur defect, and a pre-made spacer of PMMA, titanium, or PVA was implanted. All animals were given 4 weeks to form a membrane, and those receiving an isograft were given 10 weeks post-implantation to union. All samples were scanned with microCT to measure phase 1 and phase 2 bone formation. Membrane samples were processed for histology to measure membrane morphology, cellularity, and expression of the factors BMP2, TGFβ, VEGF, and IL6. PMMA and titanium spacers created almost identical membranes and phase 1 bone. PVA spacers were uniformly infiltrated with tissue and cells and did not form a distinct membrane. There were no quantitative differences in phase 2 bone formation. However, PMMA induced membranes supported functional union in 6 of 7 samples while a majority of titanium and PVA groups failed to achieve the same. Spacer material can alter the membrane enough to disrupt phase 2 bone formation. The membrane's role in bone regeneration is likely more than just as a physical barrier. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.
Collapse
Affiliation(s)
- Sarah McBride-Gagyi
- Department of Orthopaedic Surgery, Saint Louis University School of Medicine, St. Louis, MO
| | - Zacharie Toth
- Department of Orthopaedic Surgery, Saint Louis University School of Medicine, St. Louis, MO
| | - Daniel Kim
- Department of Orthopaedic Surgery, Saint Louis University School of Medicine, St. Louis, MO
| | - Victoria Ip
- Department of Orthopaedic Surgery, Saint Louis University School of Medicine, St. Louis, MO
| | - Emily Evans
- Department of Orthopaedic Surgery, Saint Louis University School of Medicine, St. Louis, MO
| | - J Tracy Watson
- Department of Orthopaedic Surgery, Saint Louis University School of Medicine, St. Louis, MO
| | - Daemeon Nicolaou
- Department of Orthopaedic Surgery, Saint Louis University School of Medicine, St. Louis, MO
| |
Collapse
|
18
|
Scientific Understanding of the Induced Membrane Technique: Current Status and Future Directions. J Orthop Trauma 2017; 31 Suppl 5:S3-S8. [PMID: 28938383 DOI: 10.1097/bot.0000000000000981] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVES To review the most recent basic science advances made in relation to the induced membrane technique and how those relate to clinical practice, applications, and future research directions. DESIGN Review of the literature. SETTING Any trauma center which might encounter large segmental bone defects. ARTICLES REVIEWED Basic science articles that looked at characteristics of the induced membrane published in the past 30 years. INTERVENTION None.
Collapse
|
19
|
Azi ML, de Almeida Teixeira AA, Cotias RB, Joeris A, Kfuri Junior M. Bone union with an in situ spacer after the first stage of the induced membrane technique. Injury 2017; 48 Suppl 4:S17-S20. [PMID: 29145962 DOI: 10.1016/s0020-1383(17)30770-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
INTRODUCTION We report a case of an infected bone defect in the tibia in which the treatment was stopped in the first stage of the induced membrane technique. The polymethylmethacrylate (PMMA) spacer, retained in the bone defect, was encapsulated by the bone regeneration. CASE REPORT A 37-year-old male patient with a 7-cm infected bone defect in the tibia was submitted to the first stage of the induced membrane technique with debridement and implantation of a PMMA spacer with antibiotics. The patient refused the second stage of the procedure and achieved bone union with the spacer in situ. There was no recurrence of infection at the 6-year follow-up. CONCLUSION his is the first report of a case in which bone union was achieved with the spacer in situ after the first stage of the induced membrane technique. Keeping the spacer in the bone defect could be an option in some exceptional situations.
Collapse
Affiliation(s)
- Matheus Lemos Azi
- Manoel Victorino Hospital, Secretary of Health for the State of Bahia, Conselheiro Almeida Couto square S/N, 40050-410, Salvador, Bahia, Brazil.
| | - Armando Augusto de Almeida Teixeira
- Manoel Victorino Hospital, Secretary of Health for the State of Bahia, Conselheiro Almeida Couto square S/N, 40050-410, Salvador, Bahia, Brazil
| | - Ricardo Britto Cotias
- Manoel Victorino Hospital, Secretary of Health for the State of Bahia, Conselheiro Almeida Couto square S/N, 40050-410, Salvador, Bahia, Brazil
| | - Alexander Joeris
- AO Clinical Investigation and Documentation. Stettbachstrasse 6, 8600, Dübendorf, Switzerland
| | - Mauricio Kfuri Junior
- Department of Biomechanics, Medicine and Rehabilitation of the Locomotor Apparatus - Ribeirao Preto Medical School - University of Sao Paulo (FMRP-USP). Av. Bandeirantes 3900, 14048-900, Ribeirão Preto, São Paulo, Brazil; Department of Orthopaedic Surgery, University of Missouri, 1100 Virginia Avenue, Columbia, Missouri, USA
| |
Collapse
|
20
|
Shah SR, Smith BT, Tatara AM, Molina ER, Lee EJ, Piepergerdes TC, Uhrig BA, Guldberg RE, Bennett GN, Wenke JC, Mikos AG. Effects of Local Antibiotic Delivery from Porous Space Maintainers on Infection Clearance and Induction of an Osteogenic Membrane in an Infected Bone Defect. Tissue Eng Part A 2017; 23:91-100. [PMID: 27998243 PMCID: PMC5312600 DOI: 10.1089/ten.tea.2016.0389] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 12/07/2016] [Indexed: 02/06/2023] Open
Abstract
Reconstruction of large bone defects can be complicated by the presence of both infection and local antibiotic administration. This can be addressed through a two-stage reconstructive approach, called the Masquelet technique, that involves the generation of an induced osteogenic membrane over a temporary poly(methyl methacrylate) (PMMA) space maintainer, followed by definitive reconstruction after the induced membrane is formed. Given that infection and antibiotic delivery each have independent effects on local tissue response, the objective of this study is to evaluate the interaction between local clindamycin release and bacterial contamination with regards to infection prevention and the restoration of pro-osteogenic gene expression in the induced membrane. Porous PMMA space maintainers with or without clindamycin were implanted in an 8 mm rat femoral defect model with or without Staphylococcus aureus inoculation for 28 days in a full-factorial study design (four groups, n = 8/group). Culture results demonstrated that 8/8 animals in the inoculated/no antibiotic group were infected at 4 weeks, which was significantly reduced to 1/8 animals in the inoculated/antibiotic group. Quantitative polymerase chain reaction analysis demonstrated that clindamycin treatment restores inflammatory cytokine and growth factor expression to the same levels as the no inoculation/no antibiotic group, demonstrating that clindamycin can ameliorate the negative effects of bacterial inoculation and does not itself negatively impact the expression of important cytokines. Main effect analysis shows that bacterial inoculation and clindamycin treatment have independent and interacting effects on the gene expression profile of the induced membrane, further highlighting that antibiotics play an important role in the regeneration of infected defects apart from their antimicrobial properties.
Collapse
Affiliation(s)
- Sarita R. Shah
- Department of Bioengineering, Rice University, Houston, Texas
| | | | | | - Eric R. Molina
- Department of Bioengineering, Rice University, Houston, Texas
| | - Esther J. Lee
- Department of Bioengineering, Rice University, Houston, Texas
| | | | - Brent A. Uhrig
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia
| | - Robert E. Guldberg
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia
| | | | - Joseph C. Wenke
- Extremity Trauma & Regenerative Medicine, U.S. Army Institute of Surgical Research, San Antonio, Texas
| | | |
Collapse
|
21
|
Abstract
OBJECTIVE To evaluate the union rate of posttraumatic bone defects treated with the induced membrane technique. DESIGN Single-center retrospective case series. SETTING Level I trauma center. PATIENTS/PARTICIPANTS Thirty-three patients who sustained 34 posttraumatic bone defects (19 tibia, 15 femur). INTERVENTION Staged management using the induced membrane technique described by Masquelet. After extensive debridement at the fracture site, a polymethylmethacrylate (PMMA) spacer was inserted into the resulting void. After soft tissue recovery, the spacer was removed, and the void, now enveloped by an induced membrane, was filled with an autologous iliac crest bone graft. MAIN OUTCOME MEASURES Bone union rate, time to achieve bone union, length of hospital stay, number of surgeries, infection resolution, range of motion, musculoskeletal tumor society system functional score, and limb shortening. RESULTS The mean defect size was 6.7 cm, and infection was present in 23 (68%) of the bone defects. Bone union was evident in 91% of cases (31/34). The average time to union was 8.5 months. In 7 of 23 (30%) of infected cases, the infection recurred, and in 3 of them, the graft was resorbed, resulting in treatment failure. CONCLUSION The induced membrane technique was effective for managing posttraumatic bone defects. A recurrence of infection was associated with treatment failure. LEVEL OF EVIDENCE Therapeutic Level IV. See Instructions for Authors for a complete description of levels of evidence.
Collapse
|
22
|
Gruber HE, Ode G, Hoelscher G, Ingram J, Bethea S, Bosse MJ. Osteogenic, stem cell and molecular characterisation of the human induced membrane from extremity bone defects. Bone Joint Res 2016; 5:106-15. [PMID: 27056768 PMCID: PMC5009235 DOI: 10.1302/2046-3758.54.2000483] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 02/25/2016] [Indexed: 01/08/2023] Open
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.
Collapse
Affiliation(s)
- H E Gruber
- Carolinas Medical Center, Orthopaedic Research Biology, Cannon Building, Room 304, PO Box 32861, Charlotte, NC 28232, USA
| | - G Ode
- Department of Orthopaedic Surgery, Carolinas Medical Center, Morehead Medical Plaza, 1025 Morehead Medical Drive, Suite 300, Charlotte, NC 28204, USA
| | - G Hoelscher
- Carolinas Medical Center, Orthopaedic Research Biology, Cannon Building, Room 304, PO Box 32861, Charlotte, NC 28232 USA
| | - J Ingram
- Carolinas Medical Center, Orthopaedic Research Biology, Cannon Building, Room 304, PO Box 32861, Charlotte, NC 28232, USA
| | - S Bethea
- Carolinas Medical Center, Orthopaedic Research Biology, Cannon Research Center, Room 304, PO Box 32861, Charlotte, NC 28232, USA
| | - M J Bosse
- Department of Orthopaedic Surgery, Morehead Medical Plaza, 1025 Morehead Medical Drive, Suite 300, Charlotte, NC 28204, USA
| |
Collapse
|
23
|
Shah SR, Tatara AM, Lam J, Lu S, Scott DW, Bennett GN, van den Beucken JJJP, Jansen JA, Wong ME, Mikos AG. Polymer-Based Local Antibiotic Delivery for Prevention of Polymicrobial Infection in Contaminated Mandibular Implants. ACS Biomater Sci Eng 2016; 2:558-566. [DOI: 10.1021/acsbiomaterials.5b00545] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Sarita R. Shah
- Department
of Bioengineering, Rice University, Houston, Texas 77030, United States
| | - Alexander M. Tatara
- Department
of Bioengineering, Rice University, Houston, Texas 77030, United States
| | - Johnny Lam
- Department
of Bioengineering, Rice University, Houston, Texas 77030, United States
| | - Steven Lu
- Department
of Bioengineering, Rice University, Houston, Texas 77030, United States
| | - David W. Scott
- Department
of Statistics, Rice University, Houston, Texas 77251, United States
| | - George N. Bennett
- Department
of BioSciences, Rice University, Houston, Texas 77005, United States
| | | | - John A. Jansen
- Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mark E. Wong
- University of Texas Health Science Center at Houston, Houston, Texas 77030, United States
| | - Antonios G. Mikos
- Department
of Bioengineering, Rice University, Houston, Texas 77030, United States
| |
Collapse
|
24
|
Wang X, Wei F, Luo F, Huang K, Xie Z. Induction of granulation tissue for the secretion of growth factors and the promotion of bone defect repair. J Orthop Surg Res 2015; 10:147. [PMID: 26381122 PMCID: PMC4574139 DOI: 10.1186/s13018-015-0287-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Accepted: 09/04/2015] [Indexed: 02/07/2023] Open
Abstract
Background The use of the Masquelet technique in the repair of large bone defects has gained increased acceptance in recent years. The core of this technique is the induction of granulation tissue membrane formation and the implantation of an autologous cancellous bone to reconstruct bone defects in the membrane. In this study, we purpose to explore the structure of induced membrane and the content of growth factors as well to compare between the structure and the effects on osteogenesis of induced membranes and the periosteum in animal models. Methods Bilateral radial bone defects were generated in 32 healthy adult rabbits. The defects were implanted with bone cement. The induced membranes and periosteum were removed after 2, 4, 6, and 8 weeks. Thereafter, hematoxylin-eosin staining (HE) and an enzyme-linked immunosorbent assay (ELISA) were performed to detect vascular endothelial growth factor (VEGF), angiotensin II (ANG-II), bone morphogenetic protein 2 (BMP2), fibroblast growth factor 2 (FGF2), and prostaglandin E2 (PGE2). Proteins isolated from total cell lysates were cultured with mesenchymal stem cells to test the cell proliferation and alkaline phosphatase activity using epimysium as a control. Results The induced membrane and periosteum exhibited similar structures and growth factor levels after 4 and 6 weeks. The highest concentration of BMP-2 and VEGF in the induced membranes occurred in week 6, and FGF-2 and ANG-II concentrations peaked in week 4. The thickness and vascular density of induced membranes gradually decreased with time. Conclusion Induced membrane matured between the 4th and the 6th week and secreted growth factors to promote osteogenesis. The matured induced membrane and periosteum had similar structures and abilities to promote the osteogenesis of mesenchymal stem cells. However, the induced membrane was thicker than the periosteum.
Collapse
Affiliation(s)
- Xiaohua Wang
- National & Regional United Engineering Laboratory of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Third Military Medical University, Chongqing, 400038, The People's Republic of China
| | - Fuda Wei
- National & Regional United Engineering Laboratory of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Third Military Medical University, Chongqing, 400038, The People's Republic of China
| | - Fei Luo
- National & Regional United Engineering Laboratory of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Third Military Medical University, Chongqing, 400038, The People's Republic of China
| | - Ke Huang
- National & Regional United Engineering Laboratory of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Third Military Medical University, Chongqing, 400038, The People's Republic of China
| | - Zhao Xie
- National & Regional United Engineering Laboratory of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, Third Military Medical University, Chongqing, 400038, The People's Republic of China.
| |
Collapse
|
25
|
de Monès E, Schlaubitz S, Oliveira H, d'Elbée JM, Bareille R, Bourget C, Couraud L, Fricain JC. Comparative study of membranes induced by PMMA or silicone in rats, and influence of external radiotherapy. Acta Biomater 2015; 19:119-27. [PMID: 25770925 DOI: 10.1016/j.actbio.2015.03.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 02/27/2015] [Accepted: 03/05/2015] [Indexed: 12/11/2022]
Abstract
The induced membrane technique has been used for long bone defect reconstruction after traumatism. One of the major drawbacks of this method is the difficult removal of the polymethyl methacrylate spacer after membrane formation. We therefore replaced the stiff PMMA spacer with a semi-flexible medical grade silicone spacer. This study aimed to compare subcutaneously formed membranes, induced by PMMA and silicone, in the irradiated or not irradiated areas within 28 rats that received the spacers. Histological analysis was performed to evaluate the composition of the membrane and to quantify the amount of vessels. Histomorphometric measurements were used to evaluate membranes' thickness, while fibrosis and inflammation were scored. The expression of VEGF and BMP-2 in lysates of the crushed membranes was determined by Western blotting. ALP expression was analyzed in HBMSC cultures in contact with the same lysates. Non-irradiated membranes induced by the two spacer types were non-inflammatory, fibrous and organized in layers. Irradiation did not change the macroscopic properties of membranes that were induced by silicone, while PMMA induced membranes were sensitive to the radiotherapy, resulting in thicker, strongly inflammatory membranes. Irradiated membranes showed an overall reduced osteogenic potential. Medical grade silicone is safe for the use in radiotherapy and might therefore be of great advantage for patients in need of cancer treatment.
Collapse
Affiliation(s)
- Erwan de Monès
- INSERM U1026, Tissue Bioengineering, 146 rue Léo-Saignat, F-33000 Bordeaux, France; Univ. Bordeaux, 146 rue Léo-Saignat, F-33000 Bordeaux, France; Department of Otolaryngology - Head and Neck Surgery, Univ. Bordeaux, Place Amélie Raba Léon, F-33000 Bordeaux, France
| | - Silke Schlaubitz
- Univ. Bordeaux, 146 rue Léo-Saignat, F-33000 Bordeaux, France; Bordeaux University Hospital, CIC 1401, PTIB, Xavier Arnozan Hospital, Avenue Haut Lévêque, F-33000 Bordeaux, France
| | - Hugo Oliveira
- INSERM U1026, Tissue Bioengineering, 146 rue Léo-Saignat, F-33000 Bordeaux, France; Univ. Bordeaux, 146 rue Léo-Saignat, F-33000 Bordeaux, France
| | - Jean-Marie d'Elbée
- INSERM U1026, Tissue Bioengineering, 146 rue Léo-Saignat, F-33000 Bordeaux, France; Univ. Bordeaux, 146 rue Léo-Saignat, F-33000 Bordeaux, France
| | - Reine Bareille
- INSERM U1026, Tissue Bioengineering, 146 rue Léo-Saignat, F-33000 Bordeaux, France; Univ. Bordeaux, 146 rue Léo-Saignat, F-33000 Bordeaux, France
| | - Chantal Bourget
- INSERM U1026, Tissue Bioengineering, 146 rue Léo-Saignat, F-33000 Bordeaux, France; Univ. Bordeaux, 146 rue Léo-Saignat, F-33000 Bordeaux, France
| | - Lionel Couraud
- Bordeaux University Hospital, CIC 1401, PTIB, Xavier Arnozan Hospital, Avenue Haut Lévêque, F-33000 Bordeaux, France
| | - Jean-Christophe Fricain
- INSERM U1026, Tissue Bioengineering, 146 rue Léo-Saignat, F-33000 Bordeaux, France; Univ. Bordeaux, 146 rue Léo-Saignat, F-33000 Bordeaux, France; Department of Dentistry, Univ. Bordeaux, Place Amélie Raba Léon, F-33000 Bordeaux, France.
| |
Collapse
|
26
|
Bosemark P, Perdikouri C, Pelkonen M, Isaksson H, Tägil M. The masquelet induced membrane technique with BMP and a synthetic scaffold can heal a rat femoral critical size defect. J Orthop Res 2015; 33:488-95. [PMID: 25639666 DOI: 10.1002/jor.22815] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 12/19/2014] [Indexed: 02/04/2023]
Abstract
Long bone defects can be managed by the induced membrane technique together with autologous bone graft. However, graft harvest is associated with donor site morbidity. This study investigates if a tricalcium phosphate hydroxyapatite scaffold can be used alone or in combination with bone active drugs to improve healing. Sprague Dawley rats (n = 40) were randomized into four groups. (A) scaffold, (B) BMP-7, (C) BMP-7 + scaffold, and (D) BMP-7 + scaffold + systemic bisphosphonate at 2 weeks. Locked femoral nailing was followed by 6 mm segment removal and implantation of an epoxy spacer. At 4 weeks, the spacers were removed and the defects grafted. Eleven weeks later, the bones were explanted for evaluation with radiography, manual assessment, micro-CT, histology, and Fourier Transform Infrared spectroscopy (FTIR). Isolated scaffolds (A) did not heal any defects, whereas the other treatments led to healing in 7/10 (B), 10/10 (C), and 9/10 (D) rats. Group D had greater volume of highly mineralized bone (p < 0.01) and higher bone volume fraction (p < 0.01) compared to all other groups. A synthetic scaffold + BMP-7 combined with a bisphosphonate improved the callus properties in a rat femoral critical size defect, compared to both BMP-7 and scaffold alone or the two combined.
Collapse
Affiliation(s)
- Per Bosemark
- Department of Orthopaedics, Clinical Sciences, Lund University, Lund, Sweden
| | | | | | | | | |
Collapse
|
27
|
Cuthbert RJ, Churchman SM, Tan HB, McGonagle D, Jones E, Giannoudis PV. Induced periosteum a complex cellular scaffold for the treatment of large bone defects. Bone 2013; 57:484-92. [PMID: 23954755 DOI: 10.1016/j.bone.2013.08.009] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 08/02/2013] [Accepted: 08/09/2013] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Surgically induced periosteal membrane holds great potential for the treatment of large bone defects representing a simple alternative to combinations of exogenous stem cells, scaffolds and growth factors. The purpose of this study was to explore the biological basis for this novel regenerative medicine strategy in man. METHODS Eight patients with critical size defects were treated with the induced membrane (IM) technique. After membrane formation 1cm(2) biopsy was taken together with matched, healthy diaphyseal periosteum (P) for comparative analysis. Morphological characteristics, cell composition and growth factor expression were compared. Functional and molecular evaluation of mesenchymal stromal cell (MSC) activity was performed. RESULTS Both tissues shared similar morphology although IM was significantly thicker than P (p=0.032). The frequency of lymphocytes, pericytes (CD45(-)CD34(-)CD146(+)) and cells expressing markers consistent with bone marrow MSCs (CD45(-/low)CD271(bright)) were 31. 3 and 15.5-fold higher respectively in IM (all p=0.043). IM contained 3-fold more cells per gramme of tissue with a similar proportion of endothelial cells (CD45(-)CD31(+)). Expressed bone morphogenic protein 2, vascular endothelial growth factor and stromal derived factor 1 (SDF-1) are key tissue regeneration mediators. Adherent expanded cells from both tissues had molecular profiles similar to bone marrow MSCs but cells from IM expressed greater than 2 fold relative abundance of SDF-1transcript compared to P (p=0.043). CONCLUSION The IM is a thick, vascularised structure that resembles periosteum with a cellular composition and molecular profile facilitating large defect repair and therefore may be described as an "induced-periosteum". This tissue offers a powerful example of in situ tissue engineering.
Collapse
Affiliation(s)
- Richard J Cuthbert
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, LS9 7TF, UK; Leeds NIHR Biomedical Research Unit, Leeds Institute of Molecular Medicine, Beckett Street, Leeds, LS9 7TF, UK
| | | | | | | | | | | |
Collapse
|
28
|
Abstract
Orthopedic injuries are common and a source of much misery and economic stress. Several relevant tissues, such as cartilage, meniscus, and intra-articular ligaments, do not heal. And even bone, which normally regenerates spontaneously, can fail to mend. The regeneration of orthopedic tissues requires 4 key components: cells, morphogenetic signals, scaffolds, and an appropriate mechanical environment. Although differentiated cells from the tissue in question can be used, most cellular research focuses on the use of mesenchymal stem cells. These can be retrieved from many different tissues, and one unresolved question is the degree to which the origin of the cells matters. Embryonic and induced pluripotent stem cells are also under investigation. Morphogenetic signals are most frequently supplied by individual recombinant growth factors or native mixtures provided by, for example, platelet-rich plasma; mesenchymal stem cells are also a rich source of trophic factors. Obstacles to the sustained delivery of individual growth factors can be addressed by gene transfer or smart scaffolds, but we still lack detailed, necessary information on which delivery profiles are needed. Scaffolds may be based on natural products, synthetic materials, or devitalized extracellular matrix. Strategies to combine these components to regenerate tissue can follow traditional tissue engineering practices, but these are costly, cumbersome, and not well suited to treating large numbers of individuals. More expeditious approaches make full use of intrinsic biological processes in vivo to avoid the need for ex vivo expansion of autologous cells and multiple procedures. Clinical translation remains a bottleneck.
Collapse
Affiliation(s)
- Christopher H Evans
- Center for Advanced Orthopaedic Studies, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; Collaborative Research Center, AO Foundation, Davos, Switzerland.
| |
Collapse
|
29
|
Genomewide molecular and biologic characterization of biomembrane formation adjacent to a methacrylate spacer in the rat femoral segmental defect model. J Orthop Trauma 2013; 27:290-7. [PMID: 23609788 DOI: 10.1097/bot.0b013e3182691288] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVES This study focuses upon the morphologic and molecular features of the layer of cells, termed the "biomembrane," which forms around methacrylate spacers in bone segmental defects. The objective of this research was to assess the biomembrane formed in a novel rodent femoral segmental defect model at 4, 8, and 16 weeks with histologic and molecular studies. METHODS Following Institutional Animal Care and Use Committee approval, a segmental defect was created in the rat femur and stabilized with the AO LockingRatNail and analyzed at 4, 8, and 16 weeks postsurgery using digital radiologic imaging, morphological and immunohistochemical studies, and genomewide gene expression studies employing microarray analysis. RESULTS The biomembrane formed around the methacrylate spacer was rich in vasculature, which showed vascular endothelial growth factor immunolocalization. The biomembrane supported development of foci of bone and cartilage within it. Bone morphogenetic protein 2 immunolocalization and gene expression were positive within developing osseous and chondrocyte foci. Microarray analysis showed significant expression of key genes related to bone and cartilage formation and angiogenesis. CONCLUSIONS This rat bone model was effective in creation of the biomembrane. Bone and cartilage foci were formed within the vascularized biomembrane with associated expression of genes critical for bone and cartilage development/formation and vascularization. The polymethyl methacrylate-induced biomembrane offers an exciting potential solution for segmental defects; the biomembrane, may act as a receptive bed and also serve as a source for mesenchymal stem cells, which could be recruited/directed for the healing process.
Collapse
|