Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Blood Coagul Fibrinolysis ; 32(6): 411-417, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34397450

ABSTRACT

Leukocyte-platelet rich fibrin (PRF) is an autologous biomaterial formed by platelets, cytokines, growth factors and cells imprisoned on a fibrin mesh, produced according to Choukroun's protocol. The aim of the present article was to report the use of PRF, associated with a bone substitute, on the regenerative treatment of a large bone defect resulting from the enucleation of a paradental cyst involving the posterior mandible. The treatment resulted in the maintenance of the bone volume, and radiographic evaluation showed new bone formation after 40 days, suggesting an osteogenic and osteoinductive effect. Also, the current literature was reviewed.


Subject(s)
Bone Substitutes/therapeutic use , Cysts/therapy , Leukocytes/metabolism , Mandibular Injuries/therapy , Platelet-Rich Fibrin/metabolism , Adolescent , Bone Regeneration , Bone Substitutes/metabolism , Female , Humans , Mandibular Injuries/metabolism , Mandibular Injuries/pathology
2.
J Biomed Mater Res B Appl Biomater ; 108(4): 1304-1310, 2020 05.
Article in English | MEDLINE | ID: mdl-31429195

ABSTRACT

Bone grafting procedures have been widely utilized as the current state-of-the-art for bone regeneration, with autogenous bone graft being the gold-standard bone reconstructive option. However, the use of autografts may be limited by secondary donor-site comorbidities, a finite amount of donor supply, increased operating time, and healthcare cost impact. Synthetic materials, or alloplasts, such as the polymeric material, poly(lactic-co-glycolic acid) (PLGA) has previously been utilized as a transient scaffold to support healing of bone defects with the potential to locally delivery osteogenic additives. In this study a novel procedure was adopted to incorporate both the dissolved contents and mechanical components of leukocyte- and platelet-rich fibrin (L-PRF) into an PLGA scaffold through a two-step method: (a) extraction of the L-PRF membrane transudate with subsequent immersion of the PLGA scaffold in transudate followed by (b) delivering a fibrin gel as a low-viscosity component that subsequently polymerizes into a highly viscous, gel-like biological material within the pores of the PLGA scaffold. Two, ~0.40 cm3 , submandibular defects (n = 24) were created per side using rotary instrumentation under continuous irrigation in six sheep. Each site received a PLGA scaffold (Intra-Lock R&D, Boca Raton, FL), with one positive control (without L-PRF exudate addition [nL-PRF]), and one experimental (augmented with PLGA/L-PRF Blocks [L-PRF]). Animals were euthanized 6 weeks postoperatively and mandibles retrieved, en bloc, for histological analysis. Histomorphometric evaluation for bone regeneration was evaluated as bone area fraction occupancy (BAFO) within the region of interest of the cortical bone (with specific image analysis software) and data presented as mean values with the corresponding 95% confidence interval values. Qualitative evaluation of nondecalcified histologic sections revealed extensive bone formation for both groups, with substantially more bone regeneration for the L-PRF induced group relative nL-PRF group. Quantitative BAFO within the defect as function of the effect of L-PRF exudate on bone regeneration, demonstrated significantly (p = .018) higher values for the L-PRF group (38.26% ± 8.5%) relative to the nL-PRF group (~28% ± 4.0%). This in vivo study indicated that L-PRF exudate has an impact on the regeneration of bone when incorporated with the PLGA scaffold in a large translational model. Further studies are warranted in order to evaluate the L-PRF exudate added, as well as exploring the preparation methods, in order to facilitate bone regeneration.


Subject(s)
Bone Regeneration , Mandible , Mandibular Injuries , Osteogenesis , Platelet-Rich Fibrin/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Tissue Scaffolds/chemistry , Animals , Female , Mandible/metabolism , Mandible/pathology , Mandibular Injuries/metabolism , Mandibular Injuries/pathology , Mandibular Injuries/therapy , Sheep
3.
J Biomed Mater Res B Appl Biomater ; 108(4): 1450-1459, 2020 05.
Article in English | MEDLINE | ID: mdl-31622016

ABSTRACT

Since bone apatite is a carbonate apatite containing carbonate in an apatitic structure, carbonate content may be one of the factors governing the osteoconductivity of apatitic bone substitutes. The aim of this study was to evaluate the effects of carbonate content on the osteoconductivity of apatitic bone substitutes using three commercially available bone substitutes for the reconstruction of alveolar bone defects of a beagle mandible with simultaneous dental implant installation. NEOBONE, Bio-Oss, and Cytrans that contain 0.1, 5.5, and 12.0 mass% of carbonate, respectively, were used in this study. The amount of newly formed bone in the upper portion of the alveolar bone defect of the beagle's mandible was 0.7, 6.6, and 39.4% at 4 weeks after surgery and 4.7, 39.5, and 75.2% at 12 weeks after surgery for NEOBONE, Bio-Oss, and Cytrans, respectively. The results indicate that bone-to-implant contact ratio was the largest for Cytrans. Additionally, the continuity of the alveolar ridge was restored in the case of Cytrans, whereas the continuity of the alveolar ridge was not sufficient when using NEOBONE and Bio-Oss. Both Cytrans and Bio-Oss that have a relatively larger carbonate content in their apatitic structure was resorbed with time. We concluded that carbonate content is one of important factors governing the osteoconductivity of apatitic bone substitutes.


Subject(s)
Apatites , Bone Substitutes/pharmacology , Carbonates , Implants, Experimental , Mandible/metabolism , Mandibular Injuries , Animals , Apatites/chemistry , Apatites/metabolism , Carbonates/chemistry , Carbonates/metabolism , Dogs , Male , Mandible/pathology , Mandibular Injuries/metabolism , Mandibular Injuries/surgery
4.
Tissue Eng Part C Methods ; 25(12): 721-731, 2019 12.
Article in English | MEDLINE | ID: mdl-31850839

ABSTRACT

Angiogenesis is a critical process essential for optimal bone healing. Several in vitro and in vivo systems have been previously used to elucidate some of the mechanisms involved in the process of angiogenesis, and at the same time, to test potential therapeutic agents and bioactive factors that play important roles in neovascularization. Computed tomography (CT) is a noninvasive imaging technique that has recently allowed investigators to obtain a diverse range of high-resolution, three-dimensional characterization of structures, such as bone formation within bony defects. Unfortunately, to date, angiogenesis evaluation relies primarily on histology, or ex vivo imaging and few studies have utilized CT to qualitatively and quantitatively study the vascular response during bone repair. In the current study a clinical CT-based technique was used to evaluate the effects of rhBMP-2 eluting graft treatment on soft tissue vascular architecture surrounding a large segmental bone defect model in the minipig mandible. The objective of this study was to demonstrate the efficacy of contrast-enhanced, clinical 64-slice CT technology in extracting quantitative metrics of vascular architecture over a 12-week period. The results of this study show that the presence of rhBMP-2 had a positive effect on vessel volume from 4 to 12 weeks, which was explained by a concurrent increase in vessel number, which was also significantly higher at 4 weeks for the rhBMP-2 treatment. More importantly, analysis of vessel architecture showed no changes throughout the duration of the study, indicating therapeutic safety. This study validates CT analysis as a relevant imaging method for quantitative and qualitative analysis of morphological characteristics of vascular tissue around a bone healing site. Also important, the study shows that CT technology can be used in large animal models and potentially be translated into clinical models for the development of improved methods to evaluate tissue healing and vascular adaptation processes over the course of therapy. This methodology has demonstrated sensitivity to tracking spatial and temporal changes in vascularization and has the potential to be applied to studying changes in other high-contrast tissues as well. Impact Statement Tissue engineering solutions depend on the surrounding tissue response to support regeneration. The inflammatory environment and surrounding vascular supply are critical to determining if therapies will survive, engraftment occurs, and native physiology is restored. This study for the first time evaluates the blood vessel network changes in surrounding soft tissue to a bone defect site in a large animal model, using clinically available computed tomography tools and model changes in vessel number, size, and architecture. While this study focuses on rhBMP2 delivery impacting surrounding vasculature, this validated method can be extended to studying the vascular network changes in other tissues as well.


Subject(s)
Bone Morphogenetic Protein 2/pharmacology , Bone Regeneration/drug effects , Mandible , Mandibular Injuries , Animals , Drug Implants/pharmacology , Humans , Mandible/blood supply , Mandible/metabolism , Mandible/pathology , Mandibular Injuries/metabolism , Mandibular Injuries/pathology , Mandibular Injuries/therapy , Recombinant Proteins/pharmacology , Swine , Swine, Miniature
5.
Front Immunol ; 10: 2662, 2019.
Article in English | MEDLINE | ID: mdl-31781128

ABSTRACT

Despite high biocompatibility of titanium and its alloys, this metal causes various side effects in the human body. It is believed that titanium biomaterials may induce an innate/adaptive immune response. However, still little is known about changes caused by titanium mandible implants, particularly with regard to bone healing. The latest studies showed disturbances in the antioxidant barrier, increased oxidative/nitrosative stress, as well as mitochondrial abnormalities in the periosteum covering titanium mandible fixations; nevertheless, the impact of titanium implants on free radical production, inflammation, and mandible apoptosis are still unknown. Because severe inflammation and apoptosis are among the main factors responsible for disturbances in osteointegration as well as implant rejection, this study is the first to evaluate pro-oxidant enzymes, cytokines as well as pro- and anti-apoptotic proteins in the periosteum of patients with a broken jaw, treated with titanium miniplates and miniscrews. The study group consisted of 29 patients with double-sided fracture of the mandible body requiring surgical treatment. We found significantly higher activity of NADPH oxidase and xanthine oxidase as well as enhanced rate of free radical production in the periosteum of patients in the study group compared to the control group. The markers of inflammation [interleukin 1 (IL-1), interleukin 6 (IL-6), tumor necrosis factor α (TNF-α), transforming growth factor ß (TGF-ß) and ß-glucuronidase (GLU)] as well as apoptosis [Bax, Bax/Bcl-2 ratio, caspase-3 (CAS-3) and nitric oxide (NO)] were significantly elevated in periosteum covering titanium fixations compared to the control group. In the study group, we also demonstrated an increased content of titanium on the periosteum surface, which positively correlated with CAS-3 activity. The study led us to the conclusion that titanium mandible implants increase the production of pro-inflammatory cytokines, and enhance free radical generation in the periosteum covering titanium miniplates and miniscrews. Additionally, exposure to Ti6Al4V titanium alloy induces apoptosis in the mandible periosteum. However, no clinical signs of the said phenomena have been observed.


Subject(s)
Fractures, Bone/therapy , Mandibular Injuries/therapy , Mandibular Prosthesis/adverse effects , Titanium/adverse effects , Adult , Apoptosis , Caspase 3/metabolism , Cytokines/metabolism , Female , Fractures, Bone/metabolism , Fractures, Bone/pathology , Free Radicals/metabolism , Humans , Inflammation/metabolism , Male , Mandibular Injuries/metabolism , Mandibular Injuries/pathology , NADPH Oxidases/metabolism , Periosteum/chemistry , Periosteum/metabolism , Periosteum/pathology , Titanium/analysis , Xanthine Oxidase/metabolism , Young Adult
6.
Cell Rep ; 28(11): 2757-2766.e5, 2019 09 10.
Article in English | MEDLINE | ID: mdl-31509739

ABSTRACT

Regenerative paradigms exhibit nerve dependency, including regeneration of the mouse digit tip and salamander limb. Denervation impairs regeneration and produces morphological aberrancy in these contexts, but the direct effect of innervation on the stem and progenitor cells enacting these processes is unknown. We devised a model to examine nerve dependency of the mouse skeletal stem cell (mSSC), the progenitor responsible for skeletal development and repair. We show that after inferior alveolar denervation, mandibular bone repair is compromised because of functional defects in mSSCs. We present mSSC reliance on paracrine factors secreted by Schwann cells as the underlying mechanism, with partial rescue of the denervated phenotype by Schwann cell transplantation and by Schwann-derived growth factors. This work sheds light on the nerve dependency of mSSCs and has implications for clinical treatment of mandibular defects.


Subject(s)
Bone Regeneration/physiology , Mandible/cytology , Mandible/metabolism , Mandibular Injuries/metabolism , Neurons/metabolism , Schwann Cells/metabolism , Stem Cells/metabolism , Animals , Bone Regeneration/drug effects , Denervation , Intercellular Signaling Peptides and Proteins/therapeutic use , Mandible/growth & development , Mandible/pathology , Mandibular Injuries/drug therapy , Mandibular Nerve/pathology , Mice , Mice, Inbred C57BL , Neurons/physiology , Paracrine Communication/physiology , Peripheral Nerve Injuries/metabolism , Platelet-Derived Growth Factor/therapeutic use , Schwann Cells/cytology , Wound Healing/physiology
7.
ACS Appl Mater Interfaces ; 11(32): 28610-28620, 2019 Aug 14.
Article in English | MEDLINE | ID: mdl-31328910

ABSTRACT

Advanced bone healing approaches included a wide range of biomaterials that mainly mimic the composition, structure, and properties of bone extracellular matrix with osteogenic activity. The present study aimed to develop a sandwich-like structure of electrospun nanofibers (NFs) based on polycaprolactone (PCL) and chitosan/polyethylene oxide (CS/PEO) composite to stimulate bone fracture healing. The morphology of the fabricated scaffolds was examined using scanning electron microscopy (SEM). Apatite deposition was evaluated using simulated body fluid (SBF). The physicochemical and mechanical properties of samples were analyzed by Fourier transform infrared, differential scanning calorimetry (DSC), thermogravimetric analysis (TGA), and universal testing machine. SEM images exhibited a porous three-dimensional structure with NF diameters of 514-4745 nm and 68-786 nm for PCL NFs layer and the sandwich-like NFs scaffolds, respectively. Deposition of apatite crystal on scaffolds started at week 2 followed by heavy deposition at week 8. This was confirmed by measuring the consumption of calcium and phosphorous ions from SBF. Thermal stability of scaffolds was confirmed using DSC and TGA. Moreover, the PCL NF layer in the middle of the developed sandwich structure reinforced the scaffolds with bear load up to 12.224 ± 1.12 MPa and Young's modulus of 17.53 ± 3.24 MPa. The scaffolds' porous structure enhanced both cell propagation and proliferation. Besides, the presence of CS in the outer NF layers of the scaffolds increased the hydrophilicity, as evidenced by the reduction of contact angle from 116.6 to 57.6°, which is essential for cell attachment. Cell viability study on mesenchymal stem cells proved the cytocompatibility of the fabricated scaffolds. Finally, in vivo mandibular bone defect rabbit model was used to confirm the regeneration of a new healthy bone within 28 days. In conclusion, the developed scaffolds could be a promising solution to stimulate bone regeneration.


Subject(s)
Bone Regeneration , Mandible/metabolism , Mandibular Injuries/therapy , Nanofibers/chemistry , Tissue Scaffolds/chemistry , Animals , Chitosan/chemistry , Humans , Male , Mandible/pathology , Mandibular Injuries/metabolism , Mandibular Injuries/pathology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Polyesters/chemistry , Polyethylene Glycols/chemistry , Rabbits , Rats
8.
Proc Natl Acad Sci U S A ; 116(14): 6954-6963, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30886100

ABSTRACT

Large mandibular defects are clinically challenging to reconstruct due to the complex anatomy of the jaw and the limited availability of appropriate tissue for repair. We envision leveraging current advances in fabrication and biomaterials to create implantable devices that generate bone within the patients themselves suitable for their own specific anatomical pathology. The in vivo bioreactor strategy facilitates the generation of large autologous vascularized bony tissue of customized geometry without the addition of exogenous growth factors or cells. To translate this technology, we investigated its success in reconstructing a mandibular defect of physiologically relevant size in sheep. We fabricated and implanted 3D-printed in vivo bioreactors against rib periosteum and utilized biomaterial-based space maintenance to preserve the native anatomical mandibular structure in the defect site before reconstruction. Nine weeks after bioreactor implantation, the ovine mandibles were repaired with the autologous bony tissue generated from the in vivo bioreactors. We evaluated tissues generated in bioreactors by radiographic, histological, mechanical, and biomolecular assays and repaired mandibles by radiographic and histological assays. Biomaterial-aided mandibular reconstruction was successful in a large superior marginal defect in five of six (83%) sheep. Given that these studies utilized clinically available biomaterials, such as bone cement and ceramic particles, this strategy is designed for rapid human translation to improve outcomes in patients with large mandibular defects.


Subject(s)
Bone Substitutes , Mandible , Mandibular Injuries , Periosteum , Printing, Three-Dimensional , Tissue Engineering , Animals , Bioreactors , Female , Mandible/metabolism , Mandible/pathology , Mandibular Injuries/metabolism , Mandibular Injuries/pathology , Mandibular Injuries/therapy , Periosteum/metabolism , Periosteum/pathology , Sheep
9.
J Biomed Mater Res B Appl Biomater ; 107(5): 1491-1503, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30265782

ABSTRACT

At least 26% of recent battlefield injuries are to the craniomaxillofacial (CMF) region. Recombinant human bone morphogenetic protein 2 (rhBMP-2) is used to treat CMF open fractures, but several complications have been associated with its use. This study tested the efficacy and safety of a lower (30% recommended) dose of rhBMP-2 to treat mandibular fractures. rhBMP-2 delivered via a polyurethane (PUR) and hydroxyapatite/ß-tricalcium phosphate (Mastergraft®) scaffold was evaluated in a 2 cm segmental mandibular defect in minipigs. Bone regeneration was analyzed at 4, 8, and 12 weeks postsurgery using clinical computed tomography (CT) and rhBMP-2, and inflammatory marker concentrations were analyzed in serum and surgery-site drain effluent. CT scans revealed that pigs treated with PUR-Mastergraft® + rhBMP-2 had complete bone bridging, while the negative control group showed incomplete bone-bridging (n = 6). Volumetric analysis of regenerated bone showed that the PUR-Mastergraft® + rhBMP-2 treatment generated significantly more bone than control by 4 weeks, a trend that continued through 12 weeks. Variations in inflammatory analytes were detected in drain effluent samples and saliva but not in serum, suggesting a localized healing response. Importantly, the rhBMP-2 group did not exhibit an excessive increase in inflammatory analytes compared to control. Treatment with low-dose rhBMP-2 increases bone regeneration capacity in pigs with mandibular continuity defects and restores bone quality. Negative complications from rhBMP-2, such as excessive inflammatory analyte levels, were not observed. Together, these results suggest that treatment with low-dose rhBMP-2 is efficacious and may improve safety when treating CMF open fractures. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B: 1491-1503, 2019.


Subject(s)
Bone Morphogenetic Protein 2 , Bone Regeneration/drug effects , Drug Delivery Systems , Mandible , Mandibular Injuries , Animals , Bone Morphogenetic Protein 2/chemistry , Bone Morphogenetic Protein 2/pharmacology , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacokinetics , Calcium Phosphates/pharmacology , Durapatite/chemistry , Durapatite/pharmacokinetics , Durapatite/pharmacology , Humans , Mandible/diagnostic imaging , Mandible/metabolism , Mandible/pathology , Mandibular Injuries/diagnostic imaging , Mandibular Injuries/drug therapy , Mandibular Injuries/metabolism , Mandibular Injuries/pathology , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Swine , Swine, Miniature , Tomography, X-Ray Computed
10.
J Biomed Mater Res B Appl Biomater ; 107(6): 2071-2079, 2019 08.
Article in English | MEDLINE | ID: mdl-30576059

ABSTRACT

This study aimed to investigate implant osseointegration using a new strategy of biphasic calcium phosphate (BCP) bone substitutes with simultaneous implant placement in mandibular reconstruction. Additionally, the temporal transcriptional profile associated with the early biological processes during osseointegration was determined. BCP and hydroxyapatite (HA) bone substitutes with simultaneous implant placement were grafted into mandibular defects created in mini-swine. Radiographic, histological, and biochemical analyses were applied for evaluation of osseointegration effects at 4 months after the grafting procedure. Bone formation around the implant was assessed by the bone area percentage (BA%) and the bone-implant-contact percentage (BIC%). The biomechanical evaluation was performed by the implant pullout test and the removal torque test. Microarray technology was utilized for gene expression comparison analysis at day 14 postoperatively. Radiographic and histological observation indicated enhanced bone formation in the BCP group compared to the HA group. Histomorphometric analyses of BA% and BIC% as well as biochemical analyses of the maximal pull-out force and the ultimate shear strength were all significantly greater in the BCP group (p < 0.05). Transcriptional analysis at an early stage of osseointegration revealed that genes belonging to biological processes associated with cell proliferation, development, osteogenesis, angiogenesis, and neurogenesis as well as the osteogenesis-related TGF-ß/BMP and WNT signaling pathways were upregulated in the BCP group. In conclusion, the reconstruction of mandibular defects using BCP with simultaneous implant placement resulted in superior osseointegration effects. A number of candidate genes that were differentially expressed may contribute to the superior osseointegration effects. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B: 2071-2079, 2019.


Subject(s)
Bone Substitutes , Hydroxyapatites , Mandible , Mandibular Injuries , Osseointegration/drug effects , Animals , Bone Substitutes/chemistry , Bone Substitutes/pharmacology , Hydroxyapatites/chemistry , Hydroxyapatites/pharmacology , Mandible/metabolism , Mandible/pathology , Mandibular Injuries/metabolism , Mandibular Injuries/pathology , Mandibular Injuries/therapy , Pilot Projects , Swine , Swine, Miniature
11.
Acta Biomater ; 76: 275-282, 2018 08.
Article in English | MEDLINE | ID: mdl-29898419

ABSTRACT

A considerable amount of research has focused on improving regenerative therapy strategies for repairing defects in load-bearing bones. The enhancement of tissue regeneration with microRNAs (miRNAs) is being developed because miRNAs can simultaneously regulate multiple signaling pathways in an endogenous manner. In this study, we developed a miR-210-based bone repair strategy. We identified a miRNA (miR-210-3p) that can simultaneously up-regulate the expression of multiple key osteogenic genes in vitro. This process resulted in enhanced bone formation in a subcutaneous mouse model with a miR-210-3p/poly-l-lactic acid (PLLA)/bone marrow-derived stem cell (BMSC) construct. Furthermore, we constructed a model of critical-sized load-bearing bone defects and implanted a miR-210-3p/ß-tricalcium phosphate (ß-TCP)/bone mesenchymal stem cell (BMSC) construct into the defect. We found that the load-bearing defect was almost fully repaired using the miR-210-3p construct. We also identified a new mechanism by which miR-210-3p regulates Sclerostin protein levels. This miRNA-based strategy may yield novel therapeutic methods for the treatment of regenerative defects in vital load-bearing bones by utilizing miRNA therapy for tissue engineering. STATEMENT OF SIGNIFICANCE: The destroyed maxillofacial bone reconstruction is still a real challenge for maxillofacial surgeon, due to that functional bone reconstruction involved load-bearing. Base on the above problem, this paper developed a novel miR-210-3p/ß-tricalcium phosphate (TCP)/bone marrow-derived stem cell (BMSC) construct (miR-210-3p/ß-TCP/BMSCs), which lead to functional reconstruction of critical-size mandible bone defect. We found that the load-bearing defect was almost fully repaired using the miR-210-3p construct. In addition, we also found the mechanism of how the delivered microRNA activated the signaling pathways of endogenous stem cells, leading to the defect regeneration. This miRNA-based strategy can be used to regenerate defects in vital load-bearing bones, thus addressing a critical challenge in regenerative medicine by utilizing miRNA therapy for tissue engineering.


Subject(s)
Bone Morphogenetic Proteins/biosynthesis , Mandible , Mandibular Injuries , MicroRNAs , Osteogenesis/drug effects , Stem Cell Transplantation , Stem Cells , Animals , Dogs , Mandible/metabolism , Mandible/pathology , Mandibular Injuries/metabolism , Mandibular Injuries/pathology , Mandibular Injuries/therapy , Mice , MicroRNAs/chemistry , MicroRNAs/pharmacokinetics , MicroRNAs/pharmacology , Stem Cells/metabolism , Stem Cells/pathology , Weight-Bearing
12.
J Biomed Mater Res B Appl Biomater ; 106(2): 751-759, 2018 02.
Article in English | MEDLINE | ID: mdl-28334520

ABSTRACT

An injectable hydrogel system with sustained bone morphogenetic protein 2 (BMP-2) release ability was developed for vertical bone regeneration at peri-implant sites and enhanced osseointegration of dental implants. In three young male beagle dogs, a pair of defects was created on both sides of the mandibular bone. Next, two implants were transplanted into each defect. In situ gelling polymer solutions with or without BMP-2 were applied to cover the implants and mandibular defects. The effects of the in situ gelling and sustained BMP-2 releasing (IGSR) hydrogel system on peri-implant bone regeneration were evaluated by radiologic examination, micro-computed tomography, and histomorphometric analysis. Twelve weeks after the treatment, significant bone generation at the peri-implant site occurred following BMP-2/IGSR hydrogel treatment. Bone volume and mineral density were increased by 1.7- and 1.3-fold, respectively (p < 0.01 and 0.05 vs. control, respectively) for the BMP-2/IGSR hydrogel system. And, 0.57-0.31 mm vertical bone generation was observed at the peri-implant site for the BMP-2/IGSR hydrogel system, while rare vertical bone generation occurred in the control group. The BMP-2/IGSR hydrogel system significantly increased bone to implant contact % between induced bone and existing bone (p < 0.05 and 0.01 vs. control). These vertical bone regeneration and higher osseointegration levels demonstrated the effectiveness of the BMP-2/IGSR hydrogel system. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 751-759, 2018.


Subject(s)
Bone Morphogenetic Protein 2 , Bone Regeneration/drug effects , Hydrogels , Mandible/metabolism , Mandibular Injuries/therapy , Organophosphorus Compounds , Polymers , Animals , Bone Morphogenetic Protein 2/chemistry , Bone Morphogenetic Protein 2/pharmacology , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacology , Disease Models, Animal , Dogs , Hydrogels/chemistry , Hydrogels/pharmacology , Male , Mandible/pathology , Mandibular Injuries/metabolism , Mandibular Injuries/pathology , Organophosphorus Compounds/chemistry , Organophosphorus Compounds/pharmacology , Polymers/chemistry , Polymers/pharmacology
13.
J Biomed Mater Res B Appl Biomater ; 106(4): 1546-1557, 2018 05.
Article in English | MEDLINE | ID: mdl-28755493

ABSTRACT

The bone-induction capacity of a porous biphasic calcium phosphate (pBCP) using heterotopic implantation in mouse (mHI-model) and its efficacy as substitute for autograft in mandibular critical-size defect in rabbit (rabMCSD-model) was investigated. In mHI-model, pBCP was implanted into the thigh muscles and bone formation was histomorphometrically and immunohistochemically evaluated. In rabMCSD-model, 13 mm bone defects were treated with pBCP or autograft and bone repair comparatively evaluated by radiographic and histomorphometric methods. In mHI-model, formed bone and immunolabeling for bone morphogenetic protein-2 and osteopontin were observed in 90% of pBCP implanted samples after 12 weeks. In rabMCSD-model neither statistically significant difference was found in newly formed bone between pBCP and autograft groups at 4 weeks (18.8 ± 5.5% vs 27.1 ± 5.6%), 8 weeks (22.3 ± 2.7% vs 26.2 ± 5.1), and 12 weeks (19.6 ± 4.7% vs 19.6 ± 2.3%). At 12 weeks, the stability and contour of the mandible were restored in both treatments. Near tooth remaining, pBCP particles were covered by small amount of mineralized tissue exhibiting perpendicular attachments of collagen fiber bundles with histological characteristic of acellular cementum. Within the limitations of this study, it was concluded that pBCP is osteoinductive and able to stimulate the new formation of bone and cementum-like tissues in rabMCSD-model, suggesting that it may be an alternative to treatment of large bone defect and in periodontal regenerative therapy. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 1546-1557, 2018.


Subject(s)
Bone Substitutes , Ceramics , Hydroxyapatites , Mandible , Mandibular Injuries , Osteogenesis/drug effects , Animals , Bone Morphogenetic Protein 2/chemistry , Bone Morphogenetic Protein 2/pharmacokinetics , Bone Morphogenetic Protein 2/pharmacology , Bone Substitutes/chemistry , Bone Substitutes/pharmacokinetics , Bone Substitutes/pharmacology , Bone Transplantation , Ceramics/chemistry , Ceramics/pharmacokinetics , Ceramics/pharmacology , Disease Models, Animal , Hydroxyapatites/chemistry , Hydroxyapatites/pharmacokinetics , Hydroxyapatites/pharmacology , Male , Mandible/metabolism , Mandible/pathology , Mandibular Injuries/metabolism , Mandibular Injuries/pathology , Mandibular Injuries/therapy , Mice , Mice, Inbred BALB C , Rabbits
14.
Biomed Res Int ; 2017: 7141296, 2017.
Article in English | MEDLINE | ID: mdl-29181402

ABSTRACT

The aim of this study was to evaluate the efficacy of growth factor loaded collagen membranes on new bone formation during horizontal bone augmentation. Mandibular defects (4 × 4 × 4 mm) were surgically prepared in six male beagle dogs, which were then protected with one of three types of membranes: (1) titanium mesh, (2) titanium reinforced collagen, or (3) rhBMP-2 loaded titanium reinforced collagen. Animals were euthanized 8 and 16 weeks after surgery, and nondecalcified specimens were prepared and histomorphologically investigated to determine the degree of osteogenesis. Data were analyzed with Friedman test. With respect to the degree of osteogenesis at earlier stage (8 weeks after surgery), there was significantly higher new bone ratio in rhBMP-2 loaded membrane group (p > 0.05). However, with respect to the long-term results (16 weeks after surgery), there were no significant differences among the three membranes (p > 0.05). Based on histomorphometric analysis, there were no significant differences in horizontal bone gaining ratio (p > 0.05).


Subject(s)
Bone Morphogenetic Protein 2/pharmacology , Collagen/pharmacology , Mandible , Mandibular Injuries , Membranes, Artificial , Titanium/pharmacology , Animals , Dogs , Humans , Mandible/metabolism , Mandible/pathology , Mandibular Injuries/metabolism , Mandibular Injuries/pathology , Mandibular Injuries/surgery , Recombinant Proteins/pharmacology
15.
Sci Rep ; 7(1): 7518, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28790361

ABSTRACT

Although bone morphogenetic protein-2 (BMP2) has demonstrated extraordinary potential in bone formation, its clinical applications require supraphysiological milligram-level doses that increase postoperative inflammation and inappropriate adipogenesis, resulting in well-documented life-threatening cervical swelling and cyst-like bone formation. Recent promising alternative biomolecular strategies are toward promoting pro-osteogenic activity of BMP2 while simultaneously suppressing its adverse effects. Here, we demonstrated that small molecular phenamil synergized osteogenesis and bone formation with BMP2 in a rat critical size mandibular defect model. Moreover, we successfully elicited the BMP2 adverse outcomes (i.e. adipogenesis and inflammation) in the mandibular defect by applying high dose BMP2. Phenamil treatment significantly improves the quality of newly formed bone by inhibiting BMP2 induced fatty cyst-like structure and inflammatory soft-tissue swelling. The observed positive phenamil effects were associated with upregulation of tribbles homolog 3 (Trib3) that suppressed adipogenic differentiation and inflammatory responses by negatively regulating PPARγ and NF-κB transcriptional activities. Thus, use of BMP2 along with phenamil stimulation or Trib3 augmentation may be a promising strategy to improve clinical efficacy and safety of current BMP therapeutics.


Subject(s)
Amiloride/analogs & derivatives , Bone Density Conservation Agents/pharmacology , Bone Morphogenetic Protein 2/pharmacology , Bone Regeneration/drug effects , Mandibular Injuries/drug therapy , Osteogenesis/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Adipogenesis/drug effects , Adipogenesis/genetics , Amiloride/pharmacology , Animals , Bone Regeneration/genetics , Cell Differentiation , Drug Synergism , Drug Therapy, Combination , Enzyme Activation/drug effects , Gene Expression Regulation , Inflammation/prevention & control , Male , Mandible/drug effects , Mandible/metabolism , Mandible/pathology , Mandibular Injuries/genetics , Mandibular Injuries/metabolism , Mandibular Injuries/pathology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B/metabolism , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteogenesis/genetics , PPAR gamma/antagonists & inhibitors , PPAR gamma/genetics , PPAR gamma/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Treatment Outcome
16.
Tissue Eng Part A ; 23(5-6): 195-207, 2017 03.
Article in English | MEDLINE | ID: mdl-27771997

ABSTRACT

Growth factor-based therapeutics using bone morphogenetic protein 2 (BMP-2) presents a promising strategy to reconstruct craniofacial bone defects such as mandible. However, clinical applications require supraphysiological BMP doses that often increase inappropriate adipogenesis, resulting in well-documented, cyst-like bone formation. Here we reported a novel complementary strategy to enhance osteogenesis and mandibular bone repair by using small-molecule phenamil that has been shown to be a strong activator of BMP signaling. Phenamil synergistically induced osteogenic differentiation of human bone marrow mesenchymal stem cells with BMP-2 while suppressing their adipogenic differentiation induced by BMP-2 in vitro. The observed pro-osteogenic and antiadipogenic activity of phenamil was mediated by expression of tribbles homolog 3 (Trb3) that enhanced BMP-smad signaling and inhibited expression of peroxisome proliferator-activated receptor gamma (PPARγ), a master regulator of adipogenesis. The synergistic effect of BMP-2+phenamil on bone regeneration was further confirmed in a critical-sized rat mandibular bone defect by implanting polymer scaffolds designed to slowly release the therapeutic molecules. These findings indicate a new complementary osteoinductive strategy to improve clinical efficacy and safety of current BMP-based therapeutics.


Subject(s)
Amiloride/analogs & derivatives , Bone Morphogenetic Protein 2 , Mandible/metabolism , Mandibular Injuries/drug therapy , Amiloride/pharmacokinetics , Amiloride/pharmacology , Animals , Bone Morphogenetic Protein 2/pharmacokinetics , Bone Morphogenetic Protein 2/pharmacology , Drug Implants/pharmacokinetics , Drug Implants/pharmacology , Humans , Mandible/pathology , Mandibular Injuries/metabolism , Mandibular Injuries/pathology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Rats, Sprague-Dawley
17.
J Biomed Mater Res B Appl Biomater ; 105(4): 723-734, 2017 05.
Article in English | MEDLINE | ID: mdl-26708554

ABSTRACT

Three-dimension (3D) scaffolds for bone tissue regeneration were produced combining three different phases: nanometric hydroxyapatite (HA) was synthesized by precipitation method and the crystals nucleation took place directly within collagen fibrils following a biologically inspired mineralization process; polycaprolactone was employed to give the material a 3D structure. The chemico-physical analysis carried out to test the material's properties and composition revealed a high similarity in composition and morphology with biologically mineralized collagen fibrils and a scaffold degradation pattern suitable for physiological processes. The micro- computerized tomography (micro-CT) showed 53.53% porosity and a 97.86% mean interconnected pores. Computer-aided design and computer-aided manufacturing (CAD-CAM) technology was used for molding the scaffold's volume (design/shape) and for guiding the surgical procedure (cutting guides). The custom made scaffolds were implanted in sheep mandible using prototyped surgical guides and customized bone plates. After three months healing, scanning electron microscopy (SEM) analysis of the explanted scaffold revealed a massive cell seeding of the scaffold, with cell infiltration within the scaffold's interconnected pores. The micro-CT of the explanted construct showed a good match between the scaffold and the adjacent host's bone, to shield the implant primary stability. Histology confirmed cell penetration and widely documented neoangiogenesis within the entire scaffold's volume. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 723-734, 2017.


Subject(s)
Biomimetic Materials/chemistry , Bone Regeneration , Bone Substitutes/chemistry , Durapatite/chemistry , Mandible , Mandibular Injuries , Tissue Scaffolds/chemistry , Animals , Mandible/diagnostic imaging , Mandible/metabolism , Mandibular Injuries/diagnostic imaging , Mandibular Injuries/metabolism , Mandibular Injuries/therapy , Materials Testing , Sheep , X-Ray Microtomography
18.
J Tissue Eng Regen Med ; 11(5): 1641-1647, 2017 05.
Article in English | MEDLINE | ID: mdl-26612731

ABSTRACT

Recently it was reported that the implantation of octacalcium phosphate (OCP) and collagen composite (OCP-collagen) was effective at promoting bone healing in small bone defects after cystectomy in humans. In addition, OCP-collagen promoted bone regeneration in a critical-sized bone defect of a rodent or canine model. In this study, OCP-collagen was implanted into a human mandibular bone defect with a longer axis of approximately 40 mm, which was diagnosed as a residual cyst with apical periodontitis. The amount of OCP-collagen implanted was about five times greater than the amounts implanted in previous clinical cases. Postoperative wound healing was satisfactory and no infection or allergic reactions occurred. The OCP-collagen-treated lesion was gradually filled with radio-opaque figures, and the alveolar region occupied the whole of the bone defect 12 months after implantation. This study suggests that OCP-collagen could be a useful bone substitute material for repairing large bone defects in humans that might not heal spontaneously. Copyright © 2015 John Wiley & Sons, Ltd.


Subject(s)
Bone Regeneration/drug effects , Bone Substitutes/administration & dosage , Calcium Phosphates/administration & dosage , Collagen/administration & dosage , Mandibular Injuries/drug therapy , Female , Humans , Male , Mandibular Injuries/diagnostic imaging , Mandibular Injuries/metabolism , Time Factors
19.
Tissue Eng Part A ; 22(13-14): 985-93, 2016 07.
Article in English | MEDLINE | ID: mdl-27369635

ABSTRACT

Here we present a new model for alveolar jaw bone regeneration, which uses human dental pulp cells (hDPCs) combined with tyrosine-derived polycarbonate polymer scaffolds [E1001(1k)] containing beta-tricalcium phosphate (ß-TCP) [E1001(1k)/ß-TCP]. E1001(1k)/ß-TCP scaffolds (5 mm diameter × 1 mm thickness) were fabricated to fit a 5 mm rat mandibular ramus critical bone defect. Five experimental groups were examined in this study: (1) E1001(1k)/ß-TCP scaffolds seeded with a high density of hDPCs, 5.0 × 10(5) hDPCs/scaffold (CH); (2) E1001(1k)/ß-TCP scaffolds seeded with a lower density of hDPCs, 2.5 × 10(5) hDPCs/scaffold (CL); (3) acellular E1001(1k)/ß-TCP scaffolds (SA); (4) acellular E1001(1k)/ß-TCP scaffolds supplemented with 4 µg recombinant human bone morphogenetic protein-2 (BMP); and (5) empty defects (EDs). Replicate hDPC-seeded and acellular E1001(1k)/ß-TCP scaffolds were cultured in vitro in osteogenic media for 1 week before implantation for 3 and 6 weeks. Live microcomputed tomography (µCT) imaging at 3 and 6 weeks postimplantation revealed robust bone regeneration in the BMP implant group. CH and CL groups exhibited similar uniformly distributed mineralized tissue coverage throughout the defects, but less than the BMP implants. In contrast, SA-treated defects exhibited sparse areas of mineralized tissue regeneration. The ED group exhibited slightly reduced defect size. Histological analyses revealed no indication of an immune response. In addition, robust expression of dentin and bone differentiation marker expression was observed in hDPC-seeded scaffolds, whereas, in contrast, BMP and SA implants exhibited only bone and not dentin differentiation marker expression. hDPCs were detected in 3-week but not in 6-week hDPC-seeded scaffold groups, indicating their survival for at least 3 weeks. Together, these results show that hDPC-seeded E1001(1k)/ß-TCP scaffolds support the rapid regeneration of osteo-dentin-like mineralized jaw tissue, suggesting a promising new therapy for alveolar jaw bone repair and regeneration.


Subject(s)
Bone Regeneration , Dental Pulp/metabolism , Mandible/metabolism , Mandibular Injuries/therapy , Polycarboxylate Cement/chemistry , Stem Cell Transplantation , Stem Cells/metabolism , Tissue Scaffolds/chemistry , Animals , Female , Humans , Mandible/pathology , Mandibular Injuries/metabolism , Mandibular Injuries/pathology , Osteogenesis , Rats , Rats, Nude , Tyrosine/chemistry
20.
Morfologiia ; 149(2): 21-6, 2016.
Article in Russian | MEDLINE | ID: mdl-30136790

ABSTRACT

The reactions of the regional lymph nodes, caused by implantation of the autologous multipotent stromal cells of bone marrow origin (AMSCBMO) to accelerate the healing of mandibular bone defect were studied by fluorescent microscopy in inbred male Wag rats aged 6 months (n=62). After the introduction of polyhydroxyalkanoate transplant containing adsorbed AMSCBMO with a transfected Green Fluorescent Protein (GFP) gene into a damaged bone area, the lymphoid nodules in submandibular lymph nodes demonstrated the appearance of numerous large macrophages containing multiple oval fluorescent inclusions in the cytoplasm. The number of these macrophages increased within 2 weeks after surgery and then began to decline. Apparently, AMSCBMO introduced in this way, were partially absorbed by macrophages. After destruction of the structures formed from AMSCBMO, the debris was also phagocytized by macrophages. In either case, these macrophages appeared in the germinal centers of lymphoid nodules in lymph nodes, where the induction of immune responses against DNA and GFP protein was probable.


Subject(s)
Bone Marrow Cells , Cells, Immobilized , Mandibular Injuries , Multipotent Stem Cells , Polyhydroxyalkanoates/chemistry , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cells, Immobilized/metabolism , Cells, Immobilized/pathology , Cells, Immobilized/transplantation , Male , Mandibular Injuries/metabolism , Mandibular Injuries/pathology , Mandibular Injuries/therapy , Multipotent Stem Cells/metabolism , Multipotent Stem Cells/pathology , Multipotent Stem Cells/transplantation , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...