Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 20.936
1.
Nat Commun ; 15(1): 4575, 2024 Jun 04.
Article En | MEDLINE | ID: mdl-38834586

Bone regeneration requires a well-orchestrated cellular and molecular response including robust vascularization and recruitment of mesenchymal and osteogenic cells. In femoral fractures, angiogenesis and osteogenesis are closely coupled during the complex healing process. Here, we show with advanced longitudinal intravital multiphoton microscopy that early vascular sprouting is not directly coupled to osteoprogenitor invasion during calvarial bone regeneration. Early osteoprogenitors emerging from the periosteum give rise to bone-forming osteoblasts at the injured calvarial bone edge. Microvessels growing inside the lesions are not associated with osteoprogenitors. Subsequently, osteogenic cells collectively invade the vascularized and perfused lesion as a multicellular layer, thereby advancing regenerative ossification. Vascular sprouting and remodeling result in dynamic blood flow alterations to accommodate the growing bone. Single cell profiling of injured calvarial bones demonstrates mesenchymal stromal cell heterogeneity comparable to femoral fractures with increase in cell types promoting bone regeneration. Expression of angiogenesis and hypoxia-related genes are slightly elevated reflecting ossification of a vascularized lesion site. Endothelial Notch and VEGF signaling alter vascular growth in calvarial bone repair without affecting the ossification progress. Our findings may have clinical implications for bone regeneration and bioengineering approaches.


Bone Regeneration , Mesenchymal Stem Cells , Neovascularization, Physiologic , Osteogenesis , Skull , Animals , Bone Regeneration/physiology , Mice , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Vascular Endothelial Growth Factor A/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Male , Receptors, Notch/metabolism , Receptors, Notch/genetics , Mice, Inbred C57BL , Signal Transduction , Female , Angiogenesis
2.
BMC Musculoskelet Disord ; 25(1): 441, 2024 Jun 05.
Article En | MEDLINE | ID: mdl-38840163

BACKGROUND: An ideal synthetic spacer for medial opening wedge high tibial osteotomy (MOWHTO) has not yet been developed. The authors have developed a new ß-tricalcium phosphate (ß-TCP) spacer with 60% porosity (N-CP60) by modifying the micro- and macro-pore structures of a conventional ß-TCP spacer (CP60) that is widely used in clinical practice. The purpose of this study was to compare the absorbability, osteoconductivity, and in vivo strength of the N-CP60 spacer with those of the CP60 spacer, when used in MOWHTO. METHODS: First, the porosity, diameter distribution of macro- and micropores, and compressive strength of each ß-TCP block were examined using methodology of biomaterial science. Secondly, a clinical study was performed using a total of 106 patients (106 knees) with MOWHTO, who were followed up for 18 months after surgery. In these knees, the N-CP60 and CP-60 spacers were implanted into 49 tibias and 57 tibias, respectively. The absorbability and osteoconductivity were radiologically evaluated by measuring the area of the implanted spacer remaining unabsorbed and assessing with the Hemert's score, respectively. The incidence of cracking in the implanted spacers was determined using computed radiography. Statistical comparisons were made with non-parametric tests. The significance level was set at p = 0.05. RESULTS: The N-CP60 and CP60 blocks had almost the same porosity (mean, 61.0% and 58.7%, respectively). The diameter of macropores was significantly larger (p < 0.0001) in the N-CP60 block than in the CP60 block, while the diameter of micropores was significantly smaller (p = 0.019) in the N-CP60 block. The ultimate strength of the N-CP60 block (median, 36.8 MPa) was significantly greater (p < 0.01) than that of the CP60 block (31.6 MPa). As for the clinical evaluations, the absorption rate of the N-CP60 spacer at 18 months after implantation (mean, 48.0%) was significantly greater (p < 0.001) than that of the CP60 spacer (29.0%). The osteoconductivity of the N-CP60 spacer was slightly but significantly higher (p = 0.0408) than that of the CP60 spacer only in zone 1. The incidence of in vivo cracking of the posteriorly located N-CP60 spacer at one month (mean, 75.5%) was significantly lower (p = 0.0035) than that of the CP60 spacer (91.2%). CONCLUSIONS: The absorbability, osteoconductivity, and compressive strength of the new N-CP60 spacer were significantly improved by modifying the macro- and micro-pore structures, compared with the conventional CP60 spacer. The N-CP60 spacer is more clinically useful than the CP60 spacer. TRIAL REGISTRATION NUMBER: H29-0002.


Calcium Phosphates , Osteotomy , Tibia , Calcium Phosphates/therapeutic use , Humans , Female , Tibia/surgery , Tibia/diagnostic imaging , Osteotomy/methods , Osteotomy/instrumentation , Middle Aged , Male , Aged , Porosity , Adult , Bone Regeneration , Treatment Outcome , Absorbable Implants , Osteoarthritis, Knee/surgery , Osteoarthritis, Knee/diagnostic imaging , Follow-Up Studies
3.
Clin Oral Investig ; 28(6): 351, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38822921

OBJECTIVES: This study aimed to assess membrane use with a bone substitute graft for guided bone regeneration (GBR) in experimental dehiscence defects. MATERIALS AND METHODS: Maxillary second incisors (I2) in 9 dogs were extracted. Six weeks later, implants were inserted and experimental dehiscence defects (5 × 3 mm) created on the buccal aspect. The defects and surrounding bone were grafted with deproteinized bovine bone mineral. One side (test) was covered with a resorbable collagen membrane whereas the contralateral side (control) was not. After 6 weeks, histomorphometrical analysis was performed to evaluate: (a) first bone-to-implant contact (fBIC), (b) buccal bone thickness at 1 mm increments from implant shoulder, (c) regenerated area (RA), (d) area and percentages of new bone (B), bone substitute (BS) and mineralized tissue (MT). RESULTS: The histological appearance was similar between test and control sites. At central and lateral sections, there were no differences between groups for fBIC, buccal bone thickness, RA, BS, B, %B, MT and %MT. At central sections, membrane use favoured more %BS and %MT (p = 0.052). There was significantly more B, %B and MT at lateral compared to central sections. CONCLUSIONS: Membrane use tended to retain more bone substitute, but had no effect on new bone ingrowth. Lateral sections showed significantly more bone ingrowth and mineralized tissue compared to central sections, confirming that new bone ingrowth takes place mainly from the lateral walls of the defect. CLINICAL RELEVANCE: Preclinical research to clarify the dynamics of bone regeneration in GBR procedures is relevant in clinical practice.


Bone Substitutes , Membranes, Artificial , Animals , Cattle , Dogs , Bone Substitutes/pharmacology , Bone Regeneration , Incisor , Guided Tissue Regeneration, Periodontal/methods , Maxilla/surgery , Dental Implants , Collagen , Surgical Wound Dehiscence , Minerals
4.
Mol Biol Rep ; 51(1): 702, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38822942

BACKGROUND: The development of cost-effective, simple, environment-friendly biographene is an area of interest. To accomplish environmentally safe, benign culturing that has advantages over other methods to reduce the graphene oxide (GO), extracellular metabolites from actinobacteria associated with mushrooms were used for the first time. METHODS: Bactericidal effect of GO against methicillin-resistant Staphylococcus aureus, antioxidant activity, and hydroxyapatite-like bone layer formation, gene expression analysis and appropriate biodegradation of the microbe-mediated synthesis of graphene was studied. RESULTS: Isolated extracellular contents Streptomyces achromogenes sub sp rubradiris reduced nano-GO to graphene (rGO), which was further examined by spectrometry and suggested an efficient conversion and significant reduction in the intensity of all oxygen-containing moieties and shifted crystalline peaks. Electron microscopic results also suggested the reduction of GO layer. In addition, absence of significant toxicity in MG-63 cell line, intentional free radical scavenging prowess, liver and kidney histopathology, and Wistar rat bone regeneration through modulation of OPG/RANKL/RUNX2/ALP pathways show the feasibility of the prepared nano GO. CONCLUSIONS: The study demonstrates the successful synthesis of biographene from actinobacterial extracellular metabolites, its potential biomedical applications, and its promising role in addressing health and environmental concerns.


Bone Regeneration , Graphite , Osteoprotegerin , RANK Ligand , Rats, Wistar , Graphite/pharmacology , Animals , Bone Regeneration/drug effects , Rats , RANK Ligand/metabolism , Osteoprotegerin/metabolism , Humans , Biocompatible Materials/pharmacology , Core Binding Factor Alpha 1 Subunit/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Actinobacteria/metabolism , Anti-Bacterial Agents/pharmacology , Antioxidants/metabolism , Antioxidants/pharmacology , Signal Transduction/drug effects
5.
J Nanobiotechnology ; 22(1): 320, 2024 Jun 08.
Article En | MEDLINE | ID: mdl-38849820

Simultaneously modulating the inflammatory microenvironment and promoting local bone regeneration is one of the main challenges in treating bone defects. In recent years, osteoimmunology has revealed that the immune system plays an essential regulatory role in bone regeneration and that macrophages are critical components. In this work, a mussel-inspired immunomodulatory and osteoinductive dual-functional hydroxyapatite nano platform (Gold/hydroxyapatite nanocomposites functionalized with polydopamine - PDA@Au-HA) is developed to accelerate bone tissues regeneration by regulating the immune microenvironment. PDA coating endows nanomaterials with the ability to scavenge reactive oxygen species (ROS) and anti-inflammatory properties, and it also exhibits an immunomodulatory ability to inhibit M1 macrophage polarization and activate M2 macrophage secretion of osteogenesis-related cytokines. Most importantly, this nano platform promotes the polarization of M2 macrophages and regulates the crosstalk between macrophages and pre-osteoblast cells to achieve bone regeneration. Au-HA can synergistically promote vascularized bone regeneration through sustained release of Ca and P particles and gold nanoparticles (NPs). This nano platform has a synergistic effect of good compatibility, scavenging of ROS, and anti-inflammatory and immunomodulatory capability to accelerate the bone repair process. Thus, our research offers a possible therapeutic approach by exploring PDA@Au-HA nanocomposites as a bifunctional platform for tissue regeneration.


Bivalvia , Bone Regeneration , Durapatite , Gold , Indoles , Macrophages , Osteogenesis , Bone Regeneration/drug effects , Durapatite/chemistry , Durapatite/pharmacology , Animals , Mice , Gold/chemistry , Gold/pharmacology , Bivalvia/chemistry , RAW 264.7 Cells , Macrophages/drug effects , Indoles/chemistry , Indoles/pharmacology , Osteogenesis/drug effects , Reactive Oxygen Species/metabolism , Polymers/chemistry , Polymers/pharmacology , Nanocomposites/chemistry , Metal Nanoparticles/chemistry , Osteoblasts/drug effects , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Immunologic Factors/pharmacology , Immunologic Factors/chemistry , Cytokines/metabolism
6.
Carbohydr Polym ; 339: 122232, 2024 Sep 01.
Article En | MEDLINE | ID: mdl-38823905

In this study, new types of hybrid double-network (DN) hydrogels composed of polyvinyl alcohol (PVA), chitosan (CH), and sodium alginate (SA) are introduced, with the hypothesis that this combination and incorporating multi-walled carbon nanotubes (MWCNTs) and graphene nanoplatelets (GNPs) will enhance osteogenetic differentiation and the structural and mechanical properties of scaffolds for bone tissue engineering applications. Initially, the impact of varying mass ratios of the PVA/CH/SA mixture on mechanical properties, swelling ratio, and degradability was examined. Based on this investigation, a mass ratio of 4:6:6 was determined to be optimal. At this ratio, the hydrogel demonstrated a Young's modulus of 47.5 ± 5 kPa, a swelling ratio of 680 ± 6 % after 3 h, and a degradation rate of 46.5 ± 5 % after 40 days. In the next phase, following the determination of the optimal mass ratio, CNTs and GNPs were incorporated into the 4:6:6 composite resulting in a significant enhancement in the electrical conductivity and stiffness of the scaffolds. The introduction of CNTs led to a notable increase of 36 % in the viability of MG63 osteoblast cells. Additionally, the inhibition zone test revealed that GNPs and CNTs increased the diameter of the inhibition zone by 49.6 % and 52.6 %, respectively.


Alginates , Bone Regeneration , Chitosan , Hydrogels , Polyvinyl Alcohol , Tissue Engineering , Tissue Scaffolds , Chitosan/chemistry , Alginates/chemistry , Alginates/pharmacology , Polyvinyl Alcohol/chemistry , Tissue Scaffolds/chemistry , Humans , Bone Regeneration/drug effects , Hydrogels/chemistry , Hydrogels/pharmacology , Tissue Engineering/methods , Nanotubes, Carbon/chemistry , Osteoblasts/drug effects , Osteoblasts/cytology , Graphite/chemistry , Graphite/pharmacology , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Cell Survival/drug effects , Cell Line
7.
Carbohydr Polym ; 339: 122174, 2024 Sep 01.
Article En | MEDLINE | ID: mdl-38823938

Segmental bone defects can arise from trauma, infection, metabolic bone disorders, or tumor removal. Hydrogels have gained attention in the field of bone regeneration due to their unique hydrophilic properties and the ability to customize their physical and chemical characteristics to serve as scaffolds and carriers for growth factors. However, the limited mechanical strength of hydrogels and the rapid release of active substances have hindered their clinical utility and therapeutic effectiveness. With ongoing advancements in material science, the development of injectable and biofunctionalized hydrogels holds great promise for addressing the challenges associated with segmental bone defects. In this study, we incorporated lyophilized platelet-rich fibrin (LPRF), which contains a multitude of growth factors, into a genipin-crosslinked gelatin/hyaluronic acid (GLT/HA-0.5 % GP) hydrogel to create an injectable and biofunctionalized composite material. Our findings demonstrate that this biofunctionalized hydrogel possesses optimal attributes for bone tissue engineering. Furthermore, results obtained from rabbit model with segmental tibial bone defects, indicate that the treatment with this biofunctionalized hydrogel resulted in increased new bone formation, as confirmed by imaging and histological analysis. From a translational perspective, this biofunctionalized hydrogel provides innovative and bioinspired capabilities that have the potential to enhance bone repair and regeneration in future clinical applications.


Bone Regeneration , Freeze Drying , Gelatin , Hyaluronic Acid , Hydrogels , Iridoids , Platelet-Rich Fibrin , Animals , Iridoids/chemistry , Iridoids/pharmacology , Gelatin/chemistry , Rabbits , Hydrogels/chemistry , Hydrogels/pharmacology , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Bone Regeneration/drug effects , Platelet-Rich Fibrin/chemistry , Tissue Engineering/methods , Cross-Linking Reagents/chemistry , Tissue Scaffolds/chemistry , Tibia/drug effects , Tibia/surgery
8.
BMC Musculoskelet Disord ; 25(1): 455, 2024 Jun 08.
Article En | MEDLINE | ID: mdl-38851675

BACKGROUND: Masquelet membrane induction technology is one of the treatment strategies for large bone defect (LBD). However, the angiogenesis ability of induced membrane decreases with time and autologous bone grafting is associated with donor site morbidity. This study investigates if the PRP-FG-nHA/PA66 scaffold can be used as a spacer instead of PMMA to improve the angiogenesis ability of induced membrane and reduce the amount of autologous bone graft. METHODS: Platelet rich plasma (PRP) was prepared and PRP-FG-nHA/PA66 scaffold was synthesized and observed. The sustained release of VEGFA and porosity of the scaffold were analyzed. We established a femur LBD model in male SD rats. 55 rats were randomly divided into four groups depending on the spacer filled in the defect area. "Defect only" group (n = 10), "PMMA" group (n = 15), "PRP-nHA/PA66" group (n = 15) and "PRP-FG-nHA/PA66" group (n = 15 ). At 6 weeks, the spacers were removed and the defects were grafted. The induced membrane and bone were collected and stained. The bone formation was detected by micro-CT and the callus union was scored on a three point system. RESULTS: The PRP-FG-nHA/PA66 scaffold was porosity and could maintain a high concentration of VEGFA after 30 days of preparation. The induced membrane in PRP-FG-nHA/PA66 group was thinner than PMMA, but the vessel density was higher.The weight of autogenous bone grafted in PRP-FG-nHA/PA66 group was significantly smaller than that of PMMA group. In PRP-FG-nHA/PA66 group, the bone defect was morphologically repaired. CONCLUSION: The study showed that PRP-FG-nHA/PA66 scaffold can significantly reduce the amount of autologous bone graft, and can achieve similar bone defect repair effect as PMMA. Our findings provide some reference and theoretical support for the treatment of large segmental bone defects in humans.


Femur , Platelet-Rich Plasma , Rats, Sprague-Dawley , Tissue Scaffolds , Animals , Male , Rats , Femur/surgery , Femur/pathology , Vascular Endothelial Growth Factor A , Bone Regeneration/physiology , Neovascularization, Physiologic , Bone Transplantation/methods , Durapatite/chemistry , Disease Models, Animal , Osteogenesis/physiology
9.
Clin Oral Investig ; 28(7): 375, 2024 Jun 15.
Article En | MEDLINE | ID: mdl-38878120

OBJECTIVE: To investigate the impact of mineralized dentin matrix (MDM) on the prognosis on bone regeneration and migration of retained roots after coronectomy. MATERIALS AND METHODS: Patients were divided into three groups based on the type of bone graft after coronectomy: Group C (n = 20, collagen), Group T (n = 20, tricalcium phosphate (TCP) + collagen), and Group D (n = 20, MDM + collagen). CBCT scans, conducted immediately and 6 months after surgery, were analyzed using digital software. Primary outcomes, including changes in bone defect depth and retained root migration distance, were evaluated 6 months after surgery. RESULTS: After 6 months, both Groups D and T exhibited greater reduction of the bone defect and lesser retained root migration than Group C (p < 0.001). Group D had greater regenerated bone volume in the distal 2 mm (73 mm3 vs. 57 mm3, p = 0.011) and lesser root migration (2.18 mm vs. 2.96 mm, p < 0.001) than Group T. The proportion of completely bone embedded retained roots was also greater in Group D than in Group C (70.0% vs. 42.1%, p = 0.003). CONCLUSIONS: MDM is an appropriate graft material for improving bone defect healing and reducing retained root migration after coronectomy. CLINICAL RELEVANCE: MDM is an autogenous material prepared chairside, which can significantly improve bone healing and reduce the risk of retained root re-eruption. MDM holds promise as a routine bone substitute material after M3M coronectomy.


Bone Regeneration , Calcium Phosphates , Collagen , Cone-Beam Computed Tomography , Dentin , Humans , Male , Female , Calcium Phosphates/therapeutic use , Prognosis , Middle Aged , Collagen/therapeutic use , Bone Regeneration/drug effects , Tooth Root/diagnostic imaging , Tooth Root/surgery , Adult , Tooth Crown/surgery , Treatment Outcome , Bone Transplantation/methods , Bone Substitutes/therapeutic use
10.
Int J Biol Macromol ; 272(Pt 1): 132820, 2024 Jun.
Article En | MEDLINE | ID: mdl-38825286

Bone tissue engineering (BTE) aims to develop implantable bone replacements for severe skeletal abnormalities that do not heal. In the field of BTE, chitosan (CS) has become a leading polysaccharide in the development of bone scaffolds. Although CS has several excellent properties, such as biodegradability, biocompatibility, and antibacterial properties, it has limitations for use in BTE because of its poor mechanical properties, increased degradation, and minimal bioactivity. To address these issues, researchers have explored other biomaterials, such as synthetic polymers, ceramics, and CS coatings on metals, to produce CS-based biocomposite scaffolds for BTE applications. These CS-based biocomposite scaffolds demonstrate superior properties, including mechanical characteristics, such as compressive strength, Young's modulus, and tensile strength. In addition, they are compatible with neighboring tissues, exhibit a controlled rate of degradation, and promote cell adhesion, proliferation, and osteoblast differentiation. This review provides a brief outline of the recent progress in making different CS-based biocomposite scaffolds and how to characterize them so that their mechanical properties can be tuned using crosslinkers for bone regeneration.


Biocompatible Materials , Bone and Bones , Chitosan , Tissue Engineering , Tissue Scaffolds , Chitosan/chemistry , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Humans , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Bone and Bones/drug effects , Bone and Bones/physiology , Animals , Bone Regeneration/drug effects , Mechanical Phenomena
11.
Injury ; 55(7): 111627, 2024 Jul.
Article En | MEDLINE | ID: mdl-38834011

BACKGROUND: As a two-stage surgical procedure, Masquelet's technique has been used to care for critical-size bone defects (CSD). We aimed to determine the effects of modified and altered bone cement with biological or chemical enriching agents on the progression of Masquelet's induced membrane (IM) applied to a rat femur CSD model, and to compare the histopathological, biochemical, and immunohistochemical findings of these cements to enhance IM capacity. METHODS: Thirty-five male rats were included in five groups: plain polymethyl methacrylate (PMMA), estrogen-impregnated PMMA (E+PMMA), bone chip added PMMA (BC+PMMA), hydroxyapatite-coated PMMA (HA) and calcium phosphate cement (CPC). The levels of bone alkaline phosphatase (BALP), osteocalcin (OC), and tumor necrosis factor-alpha (TNF-α) were analyzed in intracardiac blood samples collected at the end of 4 weeks of the right femur CSD intervention. All IMs collected were fixed and prepared for histopathological scoring. The tissue levels of rat-specific Transforming Growth Factor-Beta (TGF-ß), Runt-related Transcription Factor 2 (Runx2), and Vascular Endothelial Growth Factor (VEGF) were analyzed immunohistochemically. RESULTS: Serum levels of BALP and OC were significantly higher in E+PMMA and BC+PMMA groups than those of other groups (P = 0.0061 and 0.0019, respectively). In contrast, TNF-α levels of all groups with alternative bone cement significantly decreased compared to bare PMMA (P = 0.0116). Histopathological scores of E+PMMA, BC+PMMA, and CPC groups were 6.86 ± 1.57, 4.71 ± 0.76, and 6.57 ± 1.51, respectively, which were considerably higher than those of PMMA and HA groups (3.14 ± 0.70 and 1.86 ± 0.69, respectively) (P < 0.0001). Significant increases in TGF-ß and VEGF expressions were observed in E+PMMA and CPC groups (P = 0.0001 and <0.0001, respectively) whereas Runx2 expression significantly increased only in the HA group compared to other groups (P < 0.0001). CONCLUSIONS: The modified PMMA with E and BC, and CPC as an alternative spacer resulted in a well-differentiated IM and increased IM progression by elevating BALP and OC levels in serum and by mediating expressions of TGF-ß and VEGF at the tissue level. Estrogen-supplemented cement spacer has yielded promising findings between modified and alternative bone cement.


Bone Cements , Disease Models, Animal , Femur , Polymethyl Methacrylate , Vascular Endothelial Growth Factor A , Animals , Rats , Male , Vascular Endothelial Growth Factor A/metabolism , Femur/pathology , Femur/drug effects , Femoral Fractures/pathology , Core Binding Factor Alpha 1 Subunit/metabolism , Osteocalcin/metabolism , Alkaline Phosphatase/metabolism , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/metabolism , Rats, Sprague-Dawley , Calcium Phosphates , Fracture Healing/drug effects , Fracture Healing/physiology , Bone Regeneration/drug effects , Durapatite
12.
J Oral Implantol ; 50(3): 141-152, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38839071

After dental extraction, a physiological phenomenon of reabsorption of the dentoalveolar process is triggered, especially if periradicular lesions are present, which can sometimes be associated with oroantral communication in the upper posterior maxilla. To investigate a minimally invasive approach, 19 patients undergoing tooth extraction in the posterosuperior maxilla were recruited. All cases presented an oroantral communication with a diameter of 2-5 mm after tooth extraction and the alveolar process and, in some cases, with a partial defect of 1 or more bony walls. In these cases, a single surgical procedure was used to preserve the alveolar ridge using an open barrier technique with an exposed dense polytetrafluoroethylene membrane. The bottom of the extraction socket was filled with a collagen fleece. The residual bone process was reconstructed using a biomaterial based on carbonate-apatite derived from porcine cancellous bone. After 6 months, all patients were recalled and subjected to radiographic control associated with an implant-prosthetic rehabilitation plan. Data relating to the sinus health status and the average height and thickness of the regenerated bone were collected. Radiographic evaluation verified the integrity of the maxillary sinus floor with new bone formation, detecting a vertical bone dimension between 3.1 mm and 7.4 mm (average 5.13 ± 1.15 mm) and a horizontal thickness between 4.2 mm and 9.6 mm (average 6.86 ± 1.55 mm). The goal of this study was to highlight the advantage of managing an oroantral communication and, simultaneously, obtain the preservation and regeneration of the alveolar bone crest. The open barrier technique appears to be effective for the minimally invasive management of oroantral communication up to 5 mm in diameter in postextraction sites, with a good regeneration of hard and soft tissue.


Membranes, Artificial , Oroantral Fistula , Polytetrafluoroethylene , Tooth Extraction , Humans , Retrospective Studies , Oroantral Fistula/surgery , Middle Aged , Male , Female , Alveolar Process/surgery , Alveolar Process/diagnostic imaging , Tooth Socket/surgery , Aged , Adult , Maxilla/surgery , Bone Regeneration/physiology , Alveolar Ridge Augmentation/methods , Collagen/therapeutic use
13.
Med Sci Monit ; 30: e945471, 2024 Jun 12.
Article En | MEDLINE | ID: mdl-38864115

The Editors of Medical Science Monitor wish to inform you that the above manuscript has been retracted from publication due to concerns with the credibility and originality of the study, the manuscript content, and the Figure images. Reference: Rongfeng Zhang, Jianwei Liu, Shengpeng Yu, Dong Sun, Xiaohua Wang, Jingshu Fu, Jie Shen, Zhao Xie. Osteoprotegerin (OPG) Promotes Recruitment of Endothelial Progenitor Cells (EPCs) via CXCR4 Signaling Pathway to Improve Bone Defect Repair. Med Sci Monit, 2019; 25: 5572-5579. DOI: 10.12659/MSM.916838.


Endothelial Progenitor Cells , Osteoprotegerin , Receptors, CXCR4 , Signal Transduction , Endothelial Progenitor Cells/metabolism , Receptors, CXCR4/metabolism , Osteoprotegerin/metabolism , Animals , Bone Regeneration/drug effects , Humans , Bone and Bones/metabolism , Osteogenesis/drug effects , Male , Mice , Wound Healing/drug effects
14.
Sci Rep ; 14(1): 13764, 2024 06 14.
Article En | MEDLINE | ID: mdl-38877025

Chemobrionic systems have attracted great attention in material science for development of novel biomimetic materials. This study aims to design a new bioactive material by integrating biosilica into chemobrionic structure, which will be called biochemobrionic, and to comparatively investigate the use of both chemobrionic and biochemobrionic materials as bone scaffolds. Biosilica, isolated from Amphora sp. diatom, was integrated into chemobrionic structure, and a comprehensive set of analysis was conducted to evaluate their morphological, chemical, mechanical, thermal, and biodegradation properties. Then, the effects of both scaffolds on cell biocompatibility and osteogenic differentiation capacity were assessed. Cells attached to the scaffolds, spread out, and covered the entire surface, indicating the absence of cytotoxicity. Biochemobrionic scaffold exhibited a higher level of mineralization and bone formation than the chemobrionic structure due to the osteogenic activity of biosilica. These results present a comprehensive and pioneering understanding of the potential of (bio)chemobrionics for bone regeneration.


Bone Regeneration , Cell Differentiation , Osteogenesis , Tissue Engineering , Tissue Scaffolds , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Osteogenesis/drug effects , Bone and Bones/physiology , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Diatoms , Humans , Animals
15.
ACS Nano ; 18(22): 14312-14326, 2024 Jun 04.
Article En | MEDLINE | ID: mdl-38767151

Periodontitis, a prevalent chronic inflammatory disease worldwide, is triggered by periodontopathogenic bacteria, resulting in the progressive destruction of periodontal tissue, particularly the alveolar bone. To effectively address periodontitis, this study proposed a nanoformulation known as CuS@MSN-SCS. This formulation involves coating citrate-grafted copper sulfide (CuS) nanoparticles with mesoporous silica (MSNs), followed by surface modification using amino groups and sulfated chitosan (SCS) through electrostatic interactions. The objective of this formulation is to achieve efficient bacteria removal by inducing ROS signaling pathways mediated by Cu2+ ions. Additionally, it aims to promote alveolar bone regeneration through Cu2+-induced pro-angiogenesis and SCS-mediated bone regeneration. As anticipated, by regulating the surface charges, the negatively charged CuS nanoparticles capped with sodium citrate were successfully coated with MSNs, and the subsequent introduction of amine groups using (3-aminopropyl)triethoxysilane was followed by the incorporation of SCS through electrostatic interactions, resulting in the formation of CuS@MSN-SCS. The developed nanoformulation was verified to not only significantly exacerbate the oxidative stress of Fusobacterium nucleatum, thereby suppressing bacteria growth and biofilm formation in vitro, but also effectively alleviate the inflammatory response and promote alveolar bone regeneration without evident biotoxicity in an in vivo rat periodontitis model. These findings contribute to the therapeutic effect on periodontitis. Overall, this study successfully developed a nanoformulation for combating bacteria and facilitating alveolar bone regeneration, demonstrating the promising potential for clinical treatment of periodontitis.


Anti-Bacterial Agents , Bone Regeneration , Chitosan , Copper , Fusobacterium nucleatum , Nanoparticles , Periodontitis , Chitosan/chemistry , Chitosan/pharmacology , Periodontitis/drug therapy , Periodontitis/microbiology , Periodontitis/therapy , Periodontitis/pathology , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Bone Regeneration/drug effects , Rats , Copper/chemistry , Copper/pharmacology , Fusobacterium nucleatum/drug effects , Nanoparticles/chemistry , Rats, Sprague-Dawley , Male , Sulfates/chemistry , Sulfates/pharmacology , Silicon Dioxide/chemistry , Silicon Dioxide/pharmacology , Microbial Sensitivity Tests
16.
Int J Biol Macromol ; 270(Pt 2): 132419, 2024 Jun.
Article En | MEDLINE | ID: mdl-38759859

Bacterial infection is a serious challenge in the treatment of open bone defects, and reliance on antibiotic therapy may contribute to the emergence of drug-resistant bacteria. To solve this problem, this study developed a mineralized hydrogel (PVA-Ag-PHA) with excellent antibacterial properties and osteogenic capabilities. Silver nanoparticles (CNC/TA@AgNPs) were greenly synthesized using natural macromolecular cellulose nanocrystals (CNC) and plant polyphenolic tannins (TA) as stabilizers and reducing agents respectively, and then introduced into polyvinyl alcohol (PVA) and polydopamine-modified hydroxyapatite (PDA@HAP) hydrogel. The experimental results indicate that the PVA-Ag-PHA hydrogel, benefiting from the excellent antibacterial properties of CNC/TA@AgNPs, can not only eliminate Staphylococcus aureus and Escherichia coli, but also maintain a sustained sterile environment. At the same time, the HAP modified by PDA is uniformly dispersed within the hydrogel, thus releasing and maintaining stable concentrations of Ca2+ and PO43- ions in the local environment. The porous structure of the hydrogel with excellent biocompatibility creates a suitable bioactive environment that facilitates cell adhesion and bone regeneration. The experimental results in the rat critical-sized calvarial defect model indicate that the PVA-Ag-PHA hydrogel can effectively accelerate the bone healing process. Thus, this mussel-inspired hydrogel with antibacterial properties provides a feasible solution for the repair of open bone defects, demonstrating the considerable potential for diverse applications in bone repair.


Bone Regeneration , Cellulose , Hydrogels , Metal Nanoparticles , Silver , Skull , Tannins , Silver/chemistry , Silver/pharmacology , Animals , Bone Regeneration/drug effects , Cellulose/chemistry , Cellulose/pharmacology , Metal Nanoparticles/chemistry , Rats , Hydrogels/chemistry , Hydrogels/pharmacology , Skull/drug effects , Skull/injuries , Tannins/chemistry , Tannins/pharmacology , Bivalvia/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Polyvinyl Alcohol/chemistry , Staphylococcus aureus/drug effects , Durapatite/chemistry , Durapatite/pharmacology , Rats, Sprague-Dawley , Escherichia coli/drug effects
17.
Stem Cell Res Ther ; 15(1): 144, 2024 May 20.
Article En | MEDLINE | ID: mdl-38764077

BACKGROUND: The aim of this study was to evaluate potential synergistic effects of a single, local application of human umbilical cord MSC-derived sEVs in combination with a low dose of recombinant human rhBMP-2 to promote the regeneration of a metaphyseal femoral defect in an osteoporotic rat model. METHODS: 6 weeks after induction of osteoporosis by bilateral ventral ovariectomy and administration of a special diet, a total of 64 rats underwent a distal femoral metaphyseal osteotomy using a manual Gigli wire saw. Defects were stabilized with an adapted Y-shaped mini-locking plate and were subsequently treated with alginate only, or alginate loaded with hUC-MSC-sEVs (2 × 109), rhBMP-2 (1.5 µg), or a combination of sEVs and rhBMP-2 (n = 16 for each group). 6 weeks post-surgery, femora were evaluated by µCT, descriptive histology, and biomechanical testing. RESULTS: Native radiographs and µCT analysis confirmed superior bony union with callus formation after treatment with hUC-MSC-sEVs in combination with a low dose of rhBMP-2. This finding was further substantiated by histology, showing robust defect consolidation 6 weeks after treatment. Torsion testing of the explanted femora revealed increased stiffness after application of both, rhBMP-2 alone, or in combination with sEVs, whereas torque was only significantly increased after treatment with rhBMP-2 together with sEVs. CONCLUSION: The present study demonstrates that the co-application of hUC-MSC-sEVs can improve the efficacy of rhBMP-2 to promote the regeneration of osteoporotic bone defects.


Bone Morphogenetic Protein 2 , Extracellular Vesicles , Femur , Osteoporosis , Recombinant Proteins , Umbilical Cord , Animals , Bone Morphogenetic Protein 2/pharmacology , Bone Morphogenetic Protein 2/genetics , Recombinant Proteins/pharmacology , Recombinant Proteins/genetics , Osteoporosis/pathology , Rats , Female , Humans , Femur/pathology , Femur/drug effects , Femur/diagnostic imaging , Umbilical Cord/cytology , Extracellular Vesicles/metabolism , Bone Regeneration/drug effects , Rats, Sprague-Dawley , Transforming Growth Factor beta/pharmacology , Disease Models, Animal , X-Ray Microtomography , Mesenchymal Stem Cells/metabolism
18.
Sci Rep ; 14(1): 12449, 2024 05 30.
Article En | MEDLINE | ID: mdl-38816454

Bone graft granules implanted in bone defects come into physical contact with the host bone and form interconnected porous structure. However, there exists an accidental displacement of granules to unintended locations and leakage of granules from bone defects. Although covering the defect with a barrier membrane prevents granule emanation, this procedure is troublesome. To resolve these problems, we fabricated bioresorbable mesh cages (BRMc) in this study. Bone graft granules composed of carbonate apatite alone (Gr) and bioresorbable mesh cages (BRMc/Gr) introduced the bone graft granules and were implanted into the bone defect in the rabbit femur. Micro-computed tomography and histological analysis were conducted at 4 and 12 weeks after implantation. Osteoprogenitors in the bloodstream from the host bone passed through the pores of BRMc, penetrated the porous structure of graft granules, and might interact with individual granules. Then bone remodeling could progress actively and new bone was formed. The new bone formation was similar to the host bone at 12 weeks and there were minimal signs of local tissue inflammation. BRMc/Gr could reduce the risk of unwanted new bone formation occurring due to loss of granules from the bone defects compared with Gr because BRMc enclosed granules and prevent granules leakage from bone defects and BRMc could not induce unfavorable effects to forme new bone. Additionally, BRMc/Gr could keep granules assembled in one place, avoid displacement of granules to unintended locations, and carry easily. These results demonstrated that BRMc/Gr was effective in bone regeneration and improved clinical handling.


Bone Transplantation , Femur , X-Ray Microtomography , Animals , Rabbits , Femur/surgery , Femur/diagnostic imaging , Femur/pathology , Bone Transplantation/methods , Absorbable Implants , Bone Regeneration , Osteogenesis/drug effects
19.
Biomater Adv ; 161: 213887, 2024 Jul.
Article En | MEDLINE | ID: mdl-38735199

Critical size bone defects cannot heal without aid and current clinical approaches exhibit some limitations, underling the need for novel solutions. Silk fibroin, derived from silkworms, is widely utilized in tissue engineering and regenerative medicine due to its remarkable properties, making it a promising candidate for bone tissue regeneration in vitro and in vivo. However, the clinical translation of silk-based materials requires refinements in 3D architecture, stability, and biomechanical properties. In earlier research, improved mechanical resistance and stability of chemically crosslinked methacrylate silk fibroin (Sil-Ma) sponges over physically crosslinked counterparts were highlighted. Furthermore, the influence of photo-initiator and surfactant concentrations on silk properties was investigated. However, the characterization of sponges with Sil-Ma solution concentrations above 10 % (w/V) was hindered by production optimization challenges, with only cell viability assessed. This study focuses on the evaluation of methacrylate sponges' suitability as temporal bone tissue regeneration scaffolds. Sil-Ma sponge fabrication at a fixed concentration of 20 % (w/V) was optimized and the impact of photo-initiator (LAP) concentrations and surfactant (Tween 80) presence/absence was studied. Their effects on pore formation, silk secondary structure, mechanical properties, and osteogenic differentiation of hBM-MSCs were investigated. We demonstrated that, by tuning silk sponges' composition, the optimal combination boosted osteogenic gene expression, offering a strategy to tailor biomechanical properties for effective bone regeneration. Utilizing Design of Experiment (DoE), correlations between sponge composition, porosity, and mechanical properties are established, guiding tailored material outcomes. Additionally, correlation matrices elucidate the microstructure's influence on gene expressions, providing insights for personalized approaches in bone tissue regeneration.


Bone Regeneration , Fibroins , Surface-Active Agents , Tissue Engineering , Tissue Scaffolds , Fibroins/chemistry , Tissue Scaffolds/chemistry , Surface-Active Agents/chemistry , Animals , Tissue Engineering/methods , Bone Regeneration/drug effects , Humans , Bone and Bones/drug effects , Cross-Linking Reagents/chemistry , Osteogenesis/drug effects , Mesenchymal Stem Cells/drug effects , Biocompatible Materials/chemistry , Porosity
20.
Biomater Adv ; 161: 213899, 2024 Jul.
Article En | MEDLINE | ID: mdl-38772133

Large bone defects, particularly those exceeding the critical size, present a clinical challenge due to the limited regenerative capacity of bone tissue. Traditional treatments like autografts and allografts are constrained by donor availability, immune rejection, and mechanical performance. This study aimed to develop an effective solution by designing gradient gyroid scaffolds with titania (TiO2) surface modification for the repair of large segmental bone defects. The scaffolds were engineered to balance mechanical strength with the necessary internal space to promote new bone formation and nutrient exchange. A gradient design of the scaffold was optimized through Finite Element Analysis (FEA) and Computational Fluid Dynamics (CFD) simulations to enhance fluid flow and cell adhesion. In vivo studies in rabbits demonstrated that the G@TiO2 scaffold, featuring a gradient structure and TiO2 surface modification, exhibited superior healing capabilities compared to the homogeneous structure and TiO2 surface modification (H@TiO2) and gradient structure (G) scaffolds. At 12 weeks post-operation, in a bone defect representing nearly 30 % of the total length of the radius, the implantation of the G@TiO2 scaffold achieved a 27 % bone volume to tissue volume (BV/TV) ratio, demonstrating excellent osseointegration. The TiO2 surface modification provided photothermal antibacterial effects, enhancing the scaffold's biocompatibility and potential for infection prevention. These findings suggest that the gradient gyroid scaffold with TiO2 surface modification is a promising candidate for treating large segmental bone defects, offering a combination of mechanical strength, bioactivity, and infection resistance.


Alloys , Surface Properties , Tissue Scaffolds , Titanium , Titanium/chemistry , Animals , Rabbits , Tissue Scaffolds/chemistry , Alloys/chemistry , Bone Regeneration/drug effects , Osseointegration/drug effects , Bone and Bones , Tissue Engineering/methods , Finite Element Analysis , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology
...