Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.615
Filter
1.
Tissue Eng Regen Med ; 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38955905

ABSTRACT

BACKGROUND: This study aimed to identify glycine analogs conducive to the formation of cell-absorbable nanocomplexes, enhancing collagen synthesis and subsequent osteogenesis in combination with BMP2 for improved bone regeneration. METHODS: Glycine and its derivatives were assessed for their effects on osteogenic differentiation in MC3T3-E1 cells and human bone marrow mesenchymal stem cells (BMSCs) under osteogenic conditions or with BMP2. Osteogenic differentiation was assessed through alkaline phosphatase staining and real-time quantitative polymerase chain reaction (RT-qPCR). Nanocomplex formation was examined via scanning electron microscopy, circular dichroism, and ultraviolet-visible spectroscopy. In vivo osteogenic effects were validated using a mouse calvarial defect model, and bone regeneration was evaluated through micro-computed tomography and histomorphometric analysis. RESULTS: Glycine, glycine methyl ester, and glycinamide significantly enhanced collagen synthesis and ALP activity in conjunction with an osteogenic medium (OSM). GA emerged as the most effective inducer of osteoblast differentiation marker genes. Combining GA with BMP2 synergistically stimulated ALP activity and the expression of osteoblast markers in both cell lines. GA readily formed nanocomplexes, facilitating cellular uptake through strong electrostatic interactions. In an in vivo calvarial defect mouse model, the GA and BMP2 combination demonstrated enhanced bone volume, bone volume/tissue volume ratio, trabecular numbers, and mature bone formation compared to other combinations. CONCLUSION: GA and BMP2 synergistically promoted in vitro osteoblast differentiation and in vivo bone regeneration through nanocomplex formation. This combination holds therapeutic promise for individuals with bone defects, showcasing its potential for clinical intervention.

2.
Stem Cell Res Ther ; 15(1): 194, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38956719

ABSTRACT

BACKGROUND: Repairation of bone defects remains a major clinical problem. Constructing bone tissue engineering containing growth factors, stem cells, and material scaffolds to repair bone defects has recently become a hot research topic. Nerve growth factor (NGF) can promote osteogenesis of bone marrow mesenchymal stem cells (BMSCs), but the low survival rate of the BMSCs during transplantation remains an unresolved issue. In this study, we investigated the therapeutic effect of BMSCs overexpression of NGF on bone defect by inhibiting pyroptosis. METHODS: The relationship between the low survival rate and pyroptosis of BMSCs overexpressing NGF in localized inflammation of fractures was explored by detecting pyroptosis protein levels. Then, the NGF+/BMSCs-NSA-Sca bone tissue engineering was constructed by seeding BMSCs overexpressing NGF on the allograft bone scaffold and adding the pyroptosis inhibitor necrosulfonamide(NSA). The femoral condylar defect model in the Sprague-Dawley (SD) rat was studied by micro-CT, histological, WB and PCR analyses in vitro and in vivo to evaluate the regenerative effect of bone repair. RESULTS: The pyroptosis that occurs in BMSCs overexpressing NGF is associated with the nerve growth factor receptor (P75NTR) during osteogenic differentiation. Furthermore, NSA can block pyroptosis in BMSCs overexpression NGF. Notably, the analyses using the critical-size femoral condylar defect model indicated that the NGF+/BMSCs-NSA-Sca group inhibited pyroptosis significantly and had higher osteogenesis in defects. CONCLUSION: NGF+/BMSCs-NSA had strong osteogenic properties in repairing bone defects. Moreover, NGF+/BMSCs-NSA-Sca mixture developed in this study opens new horizons for developing novel tissue engineering constructs.


Subject(s)
Mesenchymal Stem Cells , Nerve Growth Factor , Osteogenesis , Rats, Sprague-Dawley , Tissue Scaffolds , Animals , Nerve Growth Factor/metabolism , Nerve Growth Factor/genetics , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Rats , Tissue Scaffolds/chemistry , Bone Regeneration , Allografts , Male , Tissue Engineering/methods , Pyroptosis , Sulfonamides/pharmacology , Cell Differentiation , Mesenchymal Stem Cell Transplantation/methods , Bone Transplantation/methods
3.
Heliyon ; 10(11): e32686, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38961957

ABSTRACT

Periodontitis is an inflammatory disease characterized by the destruction of periodontal tissues, and the promotion of bone tissue regeneration is the key to curing periodontitis. Psoralen is the main component of Psoralea corylifolia Linn, and has multiple biological effects, including anti-osteoporosis and osteogenesis. We constructed a novel hydrogel loaded with psoralen (PSO) and stromal cell-derived factor-1 (SDF-1) for direct endogenous cell homing. This study aimed to evaluate the synergistic effects of PSO/SDF-1 on periodontal bone regeneration in patients with periodontitis. The results of CCK8, alkaline phosphatase (ALP) activity assay, and Alizarin Red staining showed that PSO/SDF-1 combination treatment promoted cell proliferation, chemotaxis ability, and ALP activity of PDLSCs. qRT-PCR and western blotting showed that the expression levels of alkaline phosphatase (ALP), dwarf-associated transcription factor 2 (RUNX2), and osteocalcin (OCN) gene were upregulated. Rat periodontal models were established to observe the effect of local application of the composite hydrogel on bone regeneration. These results proved that the PSO/SDF-1 combination treatment significantly promoted new bone formation. The immunohistochemical (IHC) results confirmed the elevated expression of ALP, RUNX2, and OCN osteogenic genes. PSO/SDF-1 composite hydrogel can synergistically regulate the biological function and promote periodontal bone formation. Thus, this study provides a novel strategy for periodontal bone regeneration.

4.
Int J Biol Macromol ; 275(Pt 1): 133502, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38960259

ABSTRACT

Bone defects resulting from trauma, illness or congenital abnormalities represent a significant challenge to global health. Conventional treatments such as autographs and allografts have limitations, leading to the exploration of bone tissue engineering (BTE) as an alternative approach. This review aims to provide a comprehensive analysis of bone regeneration mechanisms with a focus on the role of chitosan-based biomaterials and mesenchymal stem cells (MSCs) in BTE. In addition, the physiochemical and biological properties of chitosan, its potential for bone regeneration when combined with other materials and the mechanisms through which MSCs facilitate bone regeneration were investigated. In addition, different methods of scaffold development and the incorporation of MSCs into chitosan-based scaffolds were examined. Chitosan has remarkable biocompatibility, biodegradability and osteoconductivity, making it an attractive choice for BTE. Interactions between transcription factors such as Runx2 and Osterix and signaling pathways such as the BMP and Wnt pathways regulate the differentiation of MSCs and bone regeneration. Various forms of scaffolding, including porous and fibrous injections, have shown promise in BTE. The synergistic combination of chitosan and MSCs in BTE has significant potential for addressing bone defects and promoting bone regeneration, highlighting the promising future of clinical challenges posed by bone defects.

5.
Int J Nanomedicine ; 19: 6519-6546, 2024.
Article in English | MEDLINE | ID: mdl-38957181

ABSTRACT

Background: Salidroside (SAL) is the most effective component of Rhodiola rosea, a traditional Chinese medicine. Cryptotanshinone (CT) is the main fat-soluble extract of Salvia miltiorrhiza, exhibiting considerable potential for application in osteogenesis. Herein, a polycaprolactone/gelatin nanofiber membrane loaded with CT and SAL (PSGC membrane) was successfully fabricated via coaxial electrospinning and characterized. Methods and Results: This membrane capable of sustained and controlled drug release was employed in this study. Co-culturing the membrane with bone marrow mesenchymal stem cells and human umbilical vein endothelial cells revealed excellent biocompatibility and demonstrated osteogenic and angiogenic capabilities. Furthermore, drug release from the PSGC membrane activated the Wnt/ß-catenin signaling pathway and promoted osteogenic differentiation and vascularization. Evaluation of the membrane's vascularization and osteogenic capacities involved transplantation onto a rat's subcutaneous area and assessing rat cranium defects for bone regeneration, respectively. Microcomputed tomography, histological tests, immunohistochemistry, and immunofluorescence staining confirmed the membrane's outstanding angiogenic capacity two weeks post-operation, with a higher incidence of osteogenesis observed in rat cranial defects eight weeks post-surgery. Conclusion: Overall, the SAL- and CT-loaded coaxial electrospun nanofiber membrane synergistically enhances bone repair and regeneration.


Subject(s)
Gelatin , Glucosides , Human Umbilical Vein Endothelial Cells , Mesenchymal Stem Cells , Nanofibers , Neovascularization, Physiologic , Osteogenesis , Phenanthrenes , Phenols , Polyesters , Rats, Sprague-Dawley , Osteogenesis/drug effects , Animals , Nanofibers/chemistry , Gelatin/chemistry , Polyesters/chemistry , Glucosides/chemistry , Glucosides/pharmacology , Phenols/chemistry , Phenols/pharmacology , Phenanthrenes/chemistry , Phenanthrenes/pharmacology , Phenanthrenes/pharmacokinetics , Phenanthrenes/administration & dosage , Humans , Neovascularization, Physiologic/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Rats , Male , Bone Regeneration/drug effects , Membranes, Artificial , Coculture Techniques , Drug Liberation , Cell Differentiation/drug effects
6.
J Orthop Translat ; 47: 1-14, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38957270

ABSTRACT

Background: The deployment of bone grafts (BGs) is critical to the success of scaffold-guided bone regeneration (SGBR) of large bone defects. It is thus critical to provide harvesting devices that maximize osteogenic capacity of the autograft while also minimizing graft damage during collection. As an alternative to the Reamer-Irrigator-Aspirator 2 (RIA 2) system - the gold standard for large-volume graft harvesting used in orthopaedic clinics today - a novel intramedullary BG harvesting concept has been preclinically introduced and referred to as the ARA (aspirator + reaming-aspiration) concept. The ARA concept uses aspiration of the intramedullary content, followed by medullary reaming-aspiration of the endosteal bone. This concept allows greater customization of BG harvesting conditions vis-à-vis the RIA 2 system. Following its successful in vitro validation, we hypothesized that an ARA concept-collected BG would have comparable in vivo osteogenic capacity compared to the RIA 2 system-collected BG. Methods: We used 3D-printed, medical-grade polycaprolactone-hydroxyapatite (mPCL-HA, wt 96 %:4 %) scaffolds with a Voronoi design, loaded with or without different sheep-harvested BGs and tested them in an ectopic bone formation rat model for up to 8 weeks. Results: Active bone regeneration was observed throughout the scaffold-BG constructs, particularly on the surface of the bone chips with endochondral bone formation, and highly vascularized tissue formed within the fully interconnected pore architecture. There were no differences between the BGs derived from the RIA 2 system and the ARA concept in new bone volume formation and in compression tests (Young's modulus, p = 0.74; yield strength, p = 0.50). These results highlight that the osteogenic capacities of the mPCL-HA Voronoi scaffold loaded with BGs from the ARA concept and the RIA 2 system are equivalent. Conclusion: In conclusion, the ARA concept offers a promising alternative to the RIA 2 system for harvesting BGs to be clinically integrated into SGBR strategies. The translational potential of this article: Our results show that biodegradable composite scaffolds loaded with BGs from the novel intramedullary harvesting concept and the RIA 2 system have equivalent osteogenic capacity. Thus, the innovative, highly intuitive intramedullary harvesting concept offers a promising alternative to the RIA 2 system for harvesting bone grafts, which are an important component for the routine translation of SGBR concepts into clinical practice.

7.
J Biomed Mater Res A ; 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38963690

ABSTRACT

Approximately 5%-10% of fractures go on to delayed healing and nonunion, posing significant clinical, economic, and social challenges. Current treatment methods involving open bone harvesting and grafting are associated with considerable pain and potential morbidity at the donor site. Hence, there is growing interest in minimally invasive approaches such as bone marrow aspirate concentrate (BMAC), which contains mesenchymal stromal cells (MSCs), macrophages (Mφ), and T cells. However, the use of cultured or activated cells for treatment is not yet FDA-approved in the United States, necessitating further exploration of optimal cell types and proportions for effective bone formation. As our understanding of osteoimmunology advances, it has become apparent that factors from anti-inflammatory Mφ (M2) promote bone formation by MSCs. Additionally, M2 Mφ promote T helper 2 (Th2) cells and Treg cells, both of which enhance bone formation. In this study, we investigated the interactions among MSCs, Mφ, and T cells in bone formation and explored the potential of subsets of BMAC. Coculture experiments were conducted using primary MSCs, Mφ, and CD4+ T cells at specific ratios. Our results indicate that nonactivated T cells had no direct influence on osteogenesis by MSCs, while coculturing MSCs with Mφ and T cells at a ratio of 1:5:10 positively impacted bone formation. Furthermore, higher numbers of T cells led to increased M2 polarization and a higher proportion of Th2 cells in the early stages of coculture. These findings suggest the potential for enhancing bone formation by adjusting immune and mesenchymal cell ratios in BMAC. By understanding the interactions and effects of immune cells on bone formation, we can develop more effective strategies and protocols for treating bone defects and nonunions. Further studies are needed to investigate these interactions in vivo and explore additional factors influencing MSC-based therapies.

8.
Eur J Oral Sci ; : e13006, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38989803

ABSTRACT

Lymphatics are involved in the resolution of inflammation and wound healing, but their role in the oral wound healing process after tooth extraction has never been investigated. We therefore sought to evaluate the healing process following the extraction of maxillary molars in two transgenic mouse models: K14-VEGFR3-Ig mice, which lack initial mucosal lymphatic vessels, and K14-VEGFC mice, which have hyperplastic mucosal lymphatics. Maxillary molars were extracted from both transgenic mouse types and their corresponding wild-type (WT) controls. Mucosal and alveolar bone healing were evaluated. A delayed epithelialization and bone regeneration were observed in K14-VEGFR3-Ig mice compared with their WT littermates. The hampered wound closure was accompanied by decreased levels of epidermal growth factor (EGF) and persistent inflammation, characterized by infiltrates of immune cells and elevated levels of pro-inflammatory markers in the wounds. Hyperplastic mucosal lymphatics did not enhance the healing process after tooth extraction in K14-VEGFC mice. The findings indicate that initial mucosal lymphatics play a major role in the initial phase of the oral wound healing process.

9.
Cureus ; 16(6): e62182, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38993412

ABSTRACT

Bertolotti's syndrome is a syndrome in which the transverse process of the most caudal lumbar vertebra becomes enlarged and articulates with the sacral alar, causing back pain. Here, we report a case of an adolescent basketball player with Bertolotti's syndrome who was unable to resume playing despite conservative treatment and underwent an endoscopic partial transverse process and sacral alar resection. A 16-year-old male basketball player presented to our hospital with a chief complaint of left low back pain during exercise and prolonged sitting for over one month. No obvious neurological abnormality was found. X-rays and CT showed lumbosacral transitional vertebrae, and the left transverse process of the sixth lumbar vertebra articulated with the sacrum and iliac, which was the Castellvi classification IIA. A block injection into the articulated surface produced improvement in pain, but the effect was not sustained. Since the patient was refractory to conservative treatments, such as medication and physiotherapy, surgery was performed. During surgery, the articulated transverse process and sacral alar were partially resected endoscopically. Because of the proximity of the resection site to the S1 nerve root, intraoperative electromyography (free-run EMG) was used to detect nerve root irritation symptoms in real time. The patient had no postoperative complications, his low back pain improved immediately, and he returned to play basketball three months after surgery. One year after surgery, the bone resection site showed gradual bone regeneration, and two years after surgery, the transverse process and sacral alar showed a bony bridge. The transverse process was enlarged compared to immediately after surgery but remained smaller than that before surgery. The patient continued to play basketball for two years after surgery without back pain, and no symptoms due to bone regeneration appeared. In the present case, a partial resection of the transverse process and sacral alar was performed with good results. Because the bone resection site was close to the S1 nerve root, the use of an endoscope and intraoperative free-run EMG allowed for a safer procedure during the bone resection. In addition, the patient did not present with symptoms that would affect his basketball performance, although the bone regenerated and bridging occurred between the transverse process and sacral alar over a two-year postoperative course.

10.
Theranostics ; 14(10): 3859-3899, 2024.
Article in English | MEDLINE | ID: mdl-38994021

ABSTRACT

Osteoporosis is a systemic skeletal disease caused by an imbalance between bone resorption and formation. Current treatments primarily involve systemic medication and hormone therapy. However, these systemic treatments lack directionality and are often ineffective for locally severe osteoporosis, with the potential for complex adverse reactions. Consequently, treatment strategies using bioactive materials or external interventions have emerged as the most promising approaches. This review proposes twelve microenvironmental treatment targets for osteoporosis-related pathological changes, including local accumulation of inflammatory factors and reactive oxygen species (ROS), imbalance of mitochondrial dynamics, insulin resistance, disruption of bone cell autophagy, imbalance of bone cell apoptosis, changes in neural secretions, aging of bone cells, increased local bone tissue vascular destruction, and decreased regeneration. Additionally, this review examines the current research status of effective or potential biophysical and biochemical stimuli based on these microenvironmental treatment targets and summarizes the advantages and optimal parameters of different bioengineering stimuli to support preclinical and clinical research on osteoporosis treatment and bone regeneration. Finally, the review addresses ongoing challenges and future research prospects.


Subject(s)
Osseointegration , Osteoporosis , Humans , Osteoporosis/therapy , Animals , Reactive Oxygen Species/metabolism , Bone Regeneration , Autophagy , Bone and Bones/metabolism , Apoptosis , Bioengineering/methods
11.
Theranostics ; 14(10): 4014-4057, 2024.
Article in English | MEDLINE | ID: mdl-38994032

ABSTRACT

Background: The comprehensive management of diabetic bone defects remains a substantial clinical challenge due to the hostile regenerative microenvironment characterized by aggravated inflammation, excessive reactive oxygen species (ROS), bacterial infection, impaired angiogenesis, and unbalanced bone homeostasis. Thus, an advanced multifunctional therapeutic platform capable of simultaneously achieving immune regulation, bacterial elimination, and tissue regeneration is urgently designed for augmented bone regeneration under diabetic pathological milieu. Methods and Results: Herein, a photoactivated soft-hard combined scaffold system (PGCZ) was engineered by introducing polydopamine-modified zeolitic imidazolate framework-8-loaded double-network hydrogel (soft matrix component) into 3D-printed poly(ε-caprolactone) (PCL) scaffold (hard matrix component). The versatile PGCZ scaffold based on double-network hydrogel and 3D-printed PCL was thus prepared and features highly extracellular matrix-mimicking microstructure, suitable biodegradability and mechanical properties, and excellent photothermal performance, allowing long-term structural stability and mechanical support for bone regeneration. Under periodic near-infrared (NIR) irradiation, the localized photothermal effect of PGCZ triggers the on-demand release of Zn2+, which, together with repeated mild hyperthermia, collectively accelerates the proliferation and osteogenic differentiation of preosteoblasts and potently inhibits bacterial growth and biofilm formation. Additionally, the photoactivated PGCZ system also presents outstanding immunomodulatory and ROS scavenging capacities, which regulate M2 polarization of macrophages and drive functional cytokine secretion, thus leading to a pro-regenerative microenvironment in situ with enhanced vascularization. In vivo experiments further demonstrated that the PGCZ platform in conjunction with mild photothermal therapeutic activity remarkably attenuated the local inflammatory cascade, initiated endogenous stem cell recruitment and neovascularization, and orchestrated the osteoblast/osteoclast balance, ultimately accelerating diabetic bone regeneration. Conclusions: This work highlights the potential application of a photoactivated soft-hard combined system that provides long-term biophysical (mild photothermal stimulation) and biochemical (on-demand ion delivery) cues for accelerated healing of diabetic bone defects.


Subject(s)
Bone Regeneration , Hydrogels , Photothermal Therapy , Tissue Scaffolds , Animals , Mice , Bone Regeneration/drug effects , Photothermal Therapy/methods , Tissue Scaffolds/chemistry , Hydrogels/chemistry , Indoles/chemistry , Indoles/pharmacology , Neovascularization, Physiologic/drug effects , Printing, Three-Dimensional , Osteogenesis/drug effects , Polyesters/chemistry , Diabetes Mellitus, Experimental/therapy , Male , Rats , Polymers/chemistry , Reactive Oxygen Species/metabolism , Wound Healing/drug effects , Angiogenesis
12.
Article in English | MEDLINE | ID: mdl-39001812

ABSTRACT

The utilization of micronano composite scaffolds has been extensively demonstrated to confer the superior advantages in bone repair compared to single nano- or micron-sized scaffolds. Nevertheless, the enhancement of bioactivities within these composite scaffolds remains challenging. In this study, we propose a novel approach to combine melt electrowriting (MEW) and solution electrospinning (SES) techniques for the fabrication of a composite scaffold incorporating hydroxyapatite (HAP), an osteogenic component, and roxithromycin (ROX), an antibacterial active component. Scanning electron microscopy (SEM) and Fourier-transform infrared spectroscopy (FTIR) confirmed the hierarchical architecture of the nanofiber-microgrid within the scaffold, as well as the successful loading of HAP and ROX. The incorporation of HAP enhanced the water absorption capacity of the composite scaffold, thus promoting cell adhesion and proliferation, as well as osteogenic differentiation. Furthermore, ROX resulted in effective antibacterial capability without any observable cytotoxicity. Finally, the scaffolds were applied to a rat calvarial defect model, and the results demonstrated that the 20% HAP group exhibited superior new bone formation without causing adverse reactions. Therefore, our findings present a promising strategy for designing and fabricating bioactive scaffolds for bone regeneration.

13.
Biomed Tech (Berl) ; 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39001571

ABSTRACT

OBJECTIVES: This study aimed to evaluate the intensity of the subacute local biological effects after implantation and osseoconductive potential of novel hydroxyapatite-based bone substitute coated with poly (lactide-co-glycolide), named ALBO-OS, in comparison to Bio-Oss®. METHODS: Fifteen male Wistar rats, randomly assigned into groups: 10, 20, and 30 days (n꞊5), were subcutaneously implanted with ALBO-OS and Bio-Oss®. Furthermore, artificially made bone defects on both rat's tibias were implanted with experimental materials. Unimplanted defects represented negative control. After the animals' euthanizing, tissue samples were prepared and analyzed histologically and histomorphometrically. RESULTS: Normal healing of the epithelial tissue was observed, with no signs of infection or necrosis. Minimal vascular congestion was noted immediately around the graft, with no signs of tissue oedema, with a minimal capsule thickness. The applied material did not cause an inflammatory response (IR) of significant intensity, and 20 days after implantation, the IR was mainly assessed as minimal. The tibial specimen showed that ALBO-OS has good osseoconductive potential, similar to Bio-Oss®, as well as low levels of acute and subacute inflammation. CONCLUSIONS: The tested material exhibits satisfying biocompatibility, similar to Bio-Oss®.

14.
Int J Nanomedicine ; 19: 6359-6376, 2024.
Article in English | MEDLINE | ID: mdl-38946885

ABSTRACT

Background: Bone tissue engineering (BTE) is a promising alternative to autologous bone grafting for the clinical treatment of bone defects, and inorganic/organic composite hydrogels as BTE scaffolds are a hot spot in current research. The construction of nano-hydroxyapatite/gelatin methacrylate/oxidized sodium alginate (nHAP/GelMA/OSA), abbreviated as HGO, composite hydrogels loaded with bone morphogenetic protein 7 (BMP7) will provide a suitable 3D microenvironment to promote cell aggregation, proliferation, and differentiation, thus facilitating bone repair and regeneration. Methods: Dually-crosslinked hydrogels were fabricated by combining GelMA and OSA, while HGO hydrogels were formulated by incorporating varying amounts of nHAP. The hydrogels were physically and chemically characterized followed by the assessment of their biocompatibility. BMP7-HGO (BHGO) hydrogels were fabricated by incorporating suitable concentrations of BMP7 into HGO hydrogels. The osteogenic potential of BHGO hydrogels was then validated through in vitro experiments and using rat femoral defect models. Results: The addition of nHAP significantly improved the physical properties of the hydrogel, and the composite hydrogel with 10% nHAP demonstrated the best overall performance among all groups. The selected concentration of HGO hydrogel served as a carrier for BMP7 loading and was evaluated for its osteogenic potential both in vivo and in vitro. The BHGO hydrogel demonstrated superior in vitro osteogenic induction and in vivo potential for repairing bone tissue compared to the outcomes observed in the blank control, BMP7, and HGO groups. Conclusion: Using hydrogel containing 10% HGO appears promising for bone tissue engineering scaffolds, especially when loaded with BMP7 to boost its osteogenic potential. However, further investigation is needed to optimize the GelMA, OSA, and nHAP ratios, along with the BMP7 concentration, to maximize the osteogenic potential.


Subject(s)
Alginates , Bone Morphogenetic Protein 7 , Bone Regeneration , Durapatite , Gelatin , Hydrogels , Osteogenesis , Tissue Engineering , Tissue Scaffolds , Alginates/chemistry , Alginates/pharmacology , Animals , Bone Morphogenetic Protein 7/chemistry , Bone Morphogenetic Protein 7/pharmacology , Gelatin/chemistry , Tissue Engineering/methods , Hydrogels/chemistry , Hydrogels/pharmacology , Durapatite/chemistry , Durapatite/pharmacology , Osteogenesis/drug effects , Rats , Bone Regeneration/drug effects , Tissue Scaffolds/chemistry , Rats, Sprague-Dawley , Methacrylates/chemistry , Male , Humans , Bone and Bones/drug effects
15.
Sci Rep ; 14(1): 15749, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38977767

ABSTRACT

Although bone dehiscence may occur during orthodontic tooth movement into the narrow alveolar ridge, a non-invasive prevention method is yet to be fully established. We show for the first time prevention of bone dehiscence associated with orthodontic tooth movement by prophylactic injection of bone anabolic agents in mice. In this study, we established a bone dehiscence mouse model by applying force application and used the granular type of scaffold materials encapsulated with bone morphogenetic protein (BMP)-2 and OP3-4, the receptor activator of NF-κB ligand (RANKL)-binding peptide, for the prophylactic injection to the alveolar bone. In vivo micro-computed tomography revealed bone dehiscence with decreased buccal alveolar bone thickness and height after force application, whereas no bone dehiscence was observed with the prophylactic injection after force application, and alveolar bone thickness and height were kept at similar levels as those in the control group. Bone histomorphometry analyses revealed that both bone formation and resorption parameters were significantly higher in the injection with force application group than in the force application without the prophylactic injection group. These findings suggest that the prophylactic local delivery of bone anabolic reagents can prevent bone dehiscence with increased bone remodelling activity.


Subject(s)
Anabolic Agents , Bone Morphogenetic Protein 2 , Tooth Movement Techniques , X-Ray Microtomography , Animals , Mice , Tooth Movement Techniques/adverse effects , Anabolic Agents/pharmacology , Anabolic Agents/administration & dosage , Male , Osteogenesis/drug effects , Bone Remodeling/drug effects , RANK Ligand/metabolism , Alveolar Process/drug effects , Alveolar Process/diagnostic imaging , Alveolar Process/pathology , Disease Models, Animal
16.
Bioact Mater ; 37: 106-118, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39022616

ABSTRACT

Effective control of post-extraction hemorrhage and alveolar bone resorption is critical for successful extraction socket treatment, which remains an unmet clinical challenge. Herein, an injectable Tetra-PEG hydrogel that possesses rapid gelation, firm tissue adhesion, high mechanical strength, suitable degradability, and excellent biocompatibility is developed as a sutureless and coagulation-independent bioadhesive for the management of extraction sockets. Our results demonstrate that the rapid and robust adhesive sealing of the extraction socket by the Tetra-PEG hydrogel can provide reliable protection for the underlying wound and stabilize blood clots to facilitate tissue healing. In vivo experiments using an anticoagulated rat tooth extraction model show that the hydrogel significantly outperformed clinically used cotton and gelatin sponge in hemostatic efficacy, wound closure, alveolar ridge preservation, and in situ alveolar bone regeneration. Histomorphological evaluations reveal the mechanisms for accelerated bone repair through suppressed long-term inflammation, elevated collagen deposition, higher osteoblast activity, and enhanced angiogenesis. Together, our study highlights the clinical potential of the developed injectable Tetra-PEG hydrogel for treating anticoagulant-related post-extraction hemorrhage and improving socket healing.

17.
Biomed Mater ; 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39025110

ABSTRACT

The entangled assembly of bacterial cellulose (BC) nanofibers does not provide a three-dimensional (3D) macroporous structure for cellular infiltration thus hindering its use as a scaffold for bone tissue engineering. In addition, it is difficult to achieve uniform dispersion of bioactive agents in entangled BC nanofibers. To address this, the BC nanofibers were integrated with MXene, a two-dimensional nanomaterial known for its electrical signaling and mechanical strength, along with sodium alginate to form cryogel. The cryogel was fabricated using a cross-linking to enhance its mechanical properties, pores for cellular infilteration. MXene incorporation not only increased water absorption (852% to 1446%) and retention (692% to 973%) ability but also significantly improved the compressive stress (0.85 MPa to 1.43 MPa) and modulus (0.22 MPa to 1.17 MPa) confirming successful MXene reinforcement in cryogel. Biological evaluation revealed that the optimum concentration of MXene increased the cell proliferation and the osteogenic role of fabricated scaffolds was also confirmed through osteogenic gene expressions. The macropores in reconstructed MXene-BC-based cryogel provided ample space for cellular proliferation. The osteogenic role of the scaffold was examined through various gene expressions. The Quantitative polymerase chain reaction (QTPCR) revealed that MXene-loaded scaffolds especially in low concentration, had an obvious osteogenic effect hence concluding that BC can not only be reconstructed into the desired form but osteogenic property can be induced. These findings can open a new way of reconstructing BC into a more optimal structure to overcome its structural limitations and retain its natural bioactivities.

18.
ACS Biomater Sci Eng ; 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39026391

ABSTRACT

Transformation of a fibrous mat into a three-dimensional (3D) scaffold opens up abundant innovative prospects in biomedical research, particularly for studying both soft as well as hard tissues. Electrospun nanofibers, which mimic the extracellular matrix have attracted significant attention in various studies. This research focuses on rapidly converting a fibrous mat made of polycaprolactone (PCL)/pluronic F-127 (PF-127) with different percentages of monetite calcium phosphate (MCP) into desirable 3D matrix cotton using a unique gas foaming technology. These matrix cottons possess biomimetic properties and have oriented porous structures. Using this innovative technique, various shapes of 3D matrix cotton, such as squares, hollow tubes, and other customizable forms, were successfully produced. Importantly, these 3D matrix cottons showed a consistent distribution of monetite particles with total porosity ranging from 90% to 98%. The structure of the 3D matrix cotton, its water/blood absorption capacity, the potential for causing non-hemolysis, and rapid hemostatic properties were thoroughly investigated. Additionally, periodontal cells were cultured on the 3D matrix cotton to assess their viability and morphology, revealing promising results. Furthermore, a coculture study involving NIH-3T3 and MG-63 cells on the 3D matrix cotton showed spheroidal formation within 24 h. Notably, in vitro assessments indicated that the matrix cotton containing 15% monetite (PCL-MMC15%) exhibited superior absorbent capabilities, excellent cell viability, and rapid hemostatic characteristics. Subsequently, the effectiveness of PCL-MMC15% in promoting mandibular bone regeneration was evaluated through an in vivo study on rabbits using a mandibular injury model. The results demonstrated that PCL-MMC15% facilitated the resolution of defects in the mandibular region by initiating new bone formation. Therefore, the presented 3D matrix cotton (PCL-MMC15%) shows significant promise for applications in both mandibular bone regeneration and hemostasis.

19.
J Pak Med Assoc ; 74(7): 1309-1315, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39028060

ABSTRACT

Bone grafting with simultaneous implant placement using the novel bone ring technique was a procedure introduced with the intention of three-dimensional bone augmentation with simultaneous implant placement in both maxilla and mandible. A ring-shaped bone is placed in the socket, which is secured by an implant placed through the ring. The current narrative review was planned to provide a concise summary of the core concepts surrounding bone augmentation, to provide context for understanding the bone ring technique, and to highlight the basics of bone grafting and the origin of the technique to its advancement and its importance in the light of current literature.


Subject(s)
Alveolar Ridge Augmentation , Bone Transplantation , Humans , Bone Transplantation/methods , Alveolar Ridge Augmentation/methods , Dental Implantation, Endosseous/methods , Dental Implants , Mandible/surgery , Maxilla/surgery
20.
Int J Biol Macromol ; 276(Pt 1): 133860, 2024 Jul 14.
Article in English | MEDLINE | ID: mdl-39009256

ABSTRACT

Road accidents and infection-causing diseases during bone surgery are serious problems in orthopedics, and thus, addressing these pressing challenges is crucial. In the present study, the 70S30C calcium silicate bioactive material (BM) is synthesized by a sustainable approach employing a precipitation method using recycled rice husk and eggshells as a precursor of silica and calcium. Further, 70S30C BM is composited with sodium alginate (SA) and polyvinyl alcohol (PVA), and the films were prepared by solvent casting method. The composite films were prepared without the addition of acid, binder, and crosslinking agents. Further, the films were characterized by BET, XRD, ATR-FTIR, SEM, and EDS mapping. The in vitro bioactivity and biodegradation study is performed in the simulated body fluid (SBF). The in vitro haemolysis study is executed using human blood and the results demonstrate haemocompatibility of the composite films. The ex ovo CAM assay also exhibits good neovascularization. The in vitro and in vivo biocompatibility assay proves its non-toxic nature. Further, the in vivo study reveals that the engineered composite film demonstrates accelerated osteogenesis. This work broadens the orthopedic potential of the composite film and offers bioactivity, haemocompatibility, angiogenesis, non-toxicity, and in vivo osteogenesis which would serve as a potential candidate for bone tissue engineering application.

SELECTION OF CITATIONS
SEARCH DETAIL
...