Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20.838
Filter
1.
Braz Dent J ; 35: e245461, 2024.
Article in English | MEDLINE | ID: mdl-38775590

ABSTRACT

This study aimed to evaluate the osteogenic potential of hydroxyapatite (HA), Alginate (Alg), and Gelatine (Gel) composite in a critical-size defect model in rats. Twenty-four male rats were divided into three groups: a negative control with no treatment (Control group), a positive control treated with deproteinized bovine bone mineral (DBBM group), and the experimental group treated with the new HA-Alg-Gel composite (HA-Alg-Gel group). A critical size defect (8.5mm) was made in the rat's calvaria, and the bone formation was evaluated by in vivo microcomputed tomography analysis (µCT) after 1, 15, 45, and 90 days. After 90 days, the animals were euthanized and histological and histomorphometric analyses were performed. A higher proportion of mineralized tissue/biomaterial was observed in the DBBM group when compared to the HA-Alg-Gel and Control groups in the µCT analysis during all analysis periods. However, no differences were observed in the mineralized tissue/biomaterial proportion observed on day 1 (immediate postoperative) in comparison to later periods of analysis in all groups. In the histomorphometric analysis, the HA-Alg-Gel and Control groups showed higher bone formation than the DBBM group. Moreover, in histological analysis, five samples of the HA-Alg-Gal group exhibited formed bone spicules adjacent to the graft granules against only two of eight samples in the DBBM group. Both graft materials ensured the maintenance of defect bone thickness, while a tissue thickness reduction was observed in the control group. In conclusion, this study demonstrated the osteoconductive potential of HA-Alg-Gel bone graft by supporting new bone formation around its particles.


Subject(s)
Alginates , Bone Regeneration , Durapatite , Gelatin , Skull , X-Ray Microtomography , Animals , Bone Regeneration/drug effects , Durapatite/pharmacology , Skull/surgery , Skull/diagnostic imaging , Rats , Male , Biocompatible Materials , Glucuronic Acid , Rats, Wistar , Hexuronic Acids , Osteogenesis/drug effects , Bone Substitutes
2.
Stem Cell Res Ther ; 15(1): 135, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38715130

ABSTRACT

BACKGROUND: Biomaterials used in bone tissue engineering must fulfill the requirements of osteoconduction, osteoinduction, and osseointegration. However, biomaterials with good osteoconductive properties face several challenges, including inadequate vascularization, limited osteoinduction and barrier ability, as well as the potential to trigger immune and inflammatory responses. Therefore, there is an urgent need to develop guided bone regeneration membranes as a crucial component of tissue engineering strategies for repairing bone defects. METHODS: The mZIF-8/PLA membrane was prepared using electrospinning technology and simulated body fluid external mineralization method. Its ability to induce biomimetic mineralization was evaluated through TEM, EDS, XRD, FT-IR, zeta potential, and wettability techniques. The biocompatibility, osteoinduction properties, and osteo-immunomodulatory effects of the mZIF-8/PLA membrane were comprehensively evaluated by examining cell behaviors of surface-seeded BMSCs and macrophages, as well as the regulation of cellular genes and protein levels using PCR and WB. In vivo, the mZIF-8/PLA membrane's potential to promote bone regeneration and angiogenesis was assessed through Micro-CT and immunohistochemical staining. RESULTS: The mineralized deposition enhances hydrophilicity and cell compatibility of mZIF-8/PLA membrane. mZIF-8/PLA membrane promotes up-regulation of osteogenesis and angiogenesis related factors in BMSCs. Moreover, it induces the polarization of macrophages towards the M2 phenotype and modulates the local immune microenvironment. After 4-weeks of implantation, the mZIF-8/PLA membrane successfully bridges critical bone defects and almost completely repairs the defect area after 12-weeks, while significantly improving the strength and vascularization of new bone. CONCLUSIONS: The mZIF-8/PLA membrane with dual osteoconductive and immunomodulatory abilities could pave new research paths for bone tissue engineering.


Subject(s)
Bone Regeneration , Bone Regeneration/drug effects , Animals , Osteogenesis/drug effects , Tissue Engineering/methods , Biocompatible Materials/pharmacology , Biocompatible Materials/chemistry , Mice , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Membranes, Artificial , Guided Tissue Regeneration/methods , Tissue Scaffolds/chemistry , Polyesters/chemistry , Polyesters/pharmacology , Rats
3.
Mol Biol Rep ; 51(1): 632, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38724827

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) play critical roles in the osteogenic differentiation of human bone mesenchymal stem cells (hBMSCs), but the mechanism by which miRNAs indirectly modulate osteogenesis remains unclear. Here, we explored the mechanism by which miRNAs indirectly modulate gene expression through histone demethylases to promote bone regeneration. METHODS AND RESULTS: Bioinformatics analysis was performed on hBMSCs after 7 days of osteogenic induction. The differentially expressed miRNAs were screened, and potential target mRNAs were identified. To determine the bioactivity and stemness of hBMSCs and their potential for bone repair, we performed wound healing, Cell Counting Kit-8 (CCK-8), real-time reverse transcription quantitative polymerase chain reaction (RT‒qPCR), alkaline phosphatase activity, alizarin red S (ARS) staining and radiological and histological analyses on SD rats with calvarial bone defects. Additionally, a dual-luciferase reporter assay was utilized to investigate the interaction between miR-26b-5p and ten-eleven translocation 3 (TET3) in human embryonic kidney 293T cells. The in vitro and in vivo results suggested that miR-26b-5p effectively promoted the migration, proliferation and osteogenic differentiation of hBMSCs, as well as the bone reconstruction of calvarial defects in SD rats. Mechanistically, miR-26b-5p bound to the 3' untranslated region of TET3 mRNA to mediate gene silencing. CONCLUSIONS: MiR-26b-5p downregulated the expression of TET3 to increase the osteogenic differentiation of hBMSCs and bone repair in rat calvarial defects. MiR-26b-5p/TET3 crosstalk might be useful in large-scale critical bone defects.


Subject(s)
Bone Regeneration , Cell Differentiation , Dioxygenases , Mesenchymal Stem Cells , MicroRNAs , Osteogenesis , Rats, Sprague-Dawley , Skull , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Mesenchymal Stem Cells/metabolism , Humans , Osteogenesis/genetics , Cell Differentiation/genetics , Rats , Skull/pathology , Skull/metabolism , Female , Bone Regeneration/genetics , Dioxygenases/genetics , Dioxygenases/metabolism , Cell Proliferation/genetics , HEK293 Cells
4.
Int J Mol Sci ; 25(9)2024 May 02.
Article in English | MEDLINE | ID: mdl-38732198

ABSTRACT

Osteoporotic vertebral compression fractures (OVCFs) significantly increase morbidity and mortality, presenting a formidable challenge in healthcare. Traditional interventions such as vertebroplasty and kyphoplasty, despite their widespread use, are limited in addressing the secondary effects of vertebral fractures in adjacent areas and do not facilitate bone regeneration. This review paper explores the emerging domain of regenerative therapies, spotlighting stem cell therapy's transformative potential in OVCF treatment. It thoroughly describes the therapeutic possibilities and mechanisms of action of mesenchymal stem cells against OVCFs, relying on recent clinical trials and preclinical studies for efficacy assessment. Our findings reveal that stem cell therapy, particularly in combination with scaffolding materials, holds substantial promise for bone regeneration, spinal stability improvement, and pain mitigation. This integration of stem cell-based methods with conventional treatments may herald a new era in OVCF management, potentially improving patient outcomes. This review advocates for accelerated research and collaborative efforts to translate laboratory breakthroughs into clinical practice, emphasizing the revolutionary impact of regenerative therapies on OVCF management. In summary, this paper positions stem cell therapy at the forefront of innovation for OVCF treatment, stressing the importance of ongoing research and cross-disciplinary collaboration to unlock its full clinical potential.


Subject(s)
Fractures, Compression , Osteoporotic Fractures , Regenerative Medicine , Spinal Fractures , Humans , Spinal Fractures/therapy , Fractures, Compression/therapy , Osteoporotic Fractures/therapy , Regenerative Medicine/methods , Bone Regeneration , Animals , Stem Cell Transplantation/methods , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology
5.
Stomatologiia (Mosk) ; 103(2): 41-49, 2024.
Article in Russian | MEDLINE | ID: mdl-38741534

ABSTRACT

OBJECTIVE: The aim of the sthudy. To study the influence of general and local factors on the regenerative potential of bone tissue to optimize augmentation mechanisms in the treatment of jaw atrophy. MATERIALS AND METHODS: The influence of general and local factors on the regenerative potential of the bone during the augmentation operation of the alveolar process (part) of the jaw in 68 patients was studied. The survival rate of dental implants and the incidence of complications after completion of dental rehabilitation in the long-term follow-up period (more than 5 years) were evaluated. 18 indicators were studied, which were evaluated by qualitative and quantitative methods. The study of the effect of indicators on regeneration was carried out using an accurate Fischer test for conjugacy tables. The assessment of the strength of the influence was calculated as the ratio of the chances of developing an unfavorable outcome of bone grafting and dental implantation. RESULTS: He highest chances of developing adverse outcomes were found in smoking patients with diabetes mellitus, high body mass index, low survival of bone walls, severe bone atrophy (5.6 and higher class according to Cawood & Howell), three-dimensional bone defect, in the absence of 3 or more bone walls, the presence of subcompensated and decompensated psychological state the patient. In the course of histological examination, the influence of these factors on the mechanisms of reparative osteogenesis morphologically manifested a lower degree of progress of regenerative processes and a lower degree of maturity of bone tissue. CONCLUSION: To reduce the chances of developing an unfavorable outcome, in the presence of negative general and local factors, it is not advisable to use dental implantation simultaneously with bone augmentation procedures. In order to reduce the pathophysiological impact of negative factors during sinus lifting, increase the proportion of autosteal material for potentiating the metabolic activity of bone tissue, as well as prescribe antioxidant, antihypoxant and membrane stabilizing therapy before surgery and in the early postoperative period.


Subject(s)
Bone Regeneration , Humans , Male , Female , Middle Aged , Adult , Bone Transplantation , Alveolar Ridge Augmentation/methods , Dental Implants , Alveolar Process/surgery , Alveolar Process/pathology , Smoking/adverse effects
6.
Int J Mol Sci ; 25(9)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38731827

ABSTRACT

The reunion and restoration of large segmental bone defects pose significant clinical challenges. Conventional strategies primarily involve the combination of bone scaffolds with seeded cells and/or growth factors to regulate osteogenesis and angiogenesis. However, these therapies face inherent issues related to immunogenicity, tumorigenesis, bioactivity, and off-the-shelf transplantation. The biogenic micro-environment created by implanted bone grafts plays a crucial role in initiating the bone regeneration cascade. To address this, a highly porous bi-phasic ceramic synthetic bone graft, composed of hydroxyapatite (HA) and alumina (Al), was developed. This graft was employed to repair critical segmental defects, involving the creation of a 2 cm segmental defect in a canine tibia. The assessment of bone regeneration within the synthetic bone graft post-healing was conducted using scintigraphy, micro-CT, histology, and dynamic histomorphometry. The technique yielded pore sizes in the range of 230-430 µm as primary pores, 40-70 µm as secondary inner microchannels, and 200-400 nm as tertiary submicron surface holes. These three components are designed to mimic trabecular bone networks and to provide body fluid adsorption, diffusion, a nutritional supply, communication around the cells, and cell anchorage. The overall porosity was measured at 82.61 ± 1.28%. Both micro-CT imaging and histological analysis provided substantial evidence of robust bone formation and the successful reunion of the critical defect. Furthermore, an histology revealed the presence of vascularization within the newly formed bone area, clearly demonstrating trabecular and cortical bone formation at the 8-week mark post-implantation.


Subject(s)
Bone Regeneration , Tibia , Tissue Scaffolds , Animals , Dogs , Tissue Scaffolds/chemistry , Tibia/diagnostic imaging , Pilot Projects , Osteogenesis , Porosity , X-Ray Microtomography , Durapatite , Bone Transplantation/methods , Bone Substitutes
7.
Int J Implant Dent ; 10(1): 21, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38691252

ABSTRACT

BACKGROUND: Beta-tricalcium phosphate (ß-TCP) is a biocompatible ceramic material widely used in the field of oral regeneration. Due to its excellent biological and mechanical properties, it is increasingly utilized for alveolar ridge augmentation or guided bone regeneration (GBR). With recent advances in computer-aided design and manufacturing (CAD/CAM), ß-TCP can now be used in the form of digitally designed patient-specific scaffolds for customized bone regeneration (CBR) of advanced defects in a two-stage implant therapy concept. In this case report following the CARE case report guidelines, we present a novel application of a patient-specific ß-TCP scaffold in pre-implant mandibular alveolar ridge augmentation. CASE PRESENTATION: A 63-year-old female patient with significant horizontal bone loss in the posterior mandible was treated with a custom ß-TCP scaffold in the context of a two-stage backward-planned implant therapy. Cone-beam computed tomography nine months after augmentation showed successful integration of the scaffold into the surrounding bone, allowing implant placement. Follow-up until two years after initial surgery showed excellent oral and peri-implant health. CONCLUSIONS: This case highlights the potential of patient-specific ß-TCP scaffolds for alveolar ridge augmentation and their advantage over traditional techniques, including avoidance of xeno-, allo-, and autografts. The results provide encouraging evidence for their use in clinical practice. Patient-specific ß-TCP scaffolds may be a promising alternative for clinicians seeking to provide their patients with safe, predictable, and effective alveolar ridge augmentation results in customized bone regeneration procedures.


Subject(s)
Alveolar Ridge Augmentation , Calcium Phosphates , Cone-Beam Computed Tomography , Tissue Scaffolds , Humans , Alveolar Ridge Augmentation/methods , Calcium Phosphates/therapeutic use , Female , Middle Aged , Mandible/surgery , Bone Regeneration/drug effects , Dental Implantation, Endosseous/methods , Computer-Aided Design , Alveolar Bone Loss/surgery
8.
Front Immunol ; 15: 1396759, 2024.
Article in English | MEDLINE | ID: mdl-38736888

ABSTRACT

Guided bone regeneration (GBR) is one of the most widely used and thoroughly documented alveolar bone augmentation surgeries. However, implanting GBR membranes inevitably triggers an immune response, which can lead to inflammation and failure of bone augmentation. It has been shown that GBR membranes may significantly improve in vivo outcomes as potent immunomodulators, rather than solely serving as traditional barriers. Macrophages play crucial roles in immune responses and participate in the entire process of bone injury repair. The significant diversity and high plasticity of macrophages complicate our understanding of the immunomodulatory mechanisms underlying GBR. This review provides a comprehensive summary of recent findings on the potential role of macrophages in GBR for bone defects in situ. Specifically, macrophages can promote osteogenesis or fibrous tissue formation in bone defects and degradation or fibrous encapsulation of membranes. Moreover, GBR membranes can influence the recruitment and polarization of macrophages. Therefore, immunomodulating GBR membranes are primarily developed by improving macrophage recruitment and aggregation as well as regulating macrophage polarization. However, certain challenges remain to be addressed in the future. For example, developing more rational and sophisticated sequential delivery systems for macrophage activation reagents; addressing the interference of bone graft materials and dental implants; and understanding the correlations among membrane degradation, macrophage responses, and bone regeneration.


Subject(s)
Bone Regeneration , Macrophages , Humans , Bone Regeneration/immunology , Macrophages/immunology , Animals , Guided Tissue Regeneration/methods , Osteogenesis
9.
J Am Acad Orthop Surg ; 32(10): e476-e481, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38700858

ABSTRACT

With an aging population, and an anticipated increase in overall fracture incidence, a sound understanding of bone healing and how technology can optimize this process is crucial. Concentrated bone marrow aspirate (cBMA) is a technology that capitalizes on skeletal stem and progenitor cells (SSPCs) to enhance the regenerative capacity of bone. This overview highlights the science behind cBMA, discusses the role of SSPCs in bone homeostasis and fracture repair, and briefly details the clinical evidence supporting the use of cBMA in fracture healing. Despite promising early clinical results, a lack of standardization in harvest and processing techniques, coupled with patient variability, presents challenges in optimizing the use of cBMA. However, cBMA remains an emerging technology that may certainly play a crucial role in the future of fracture healing augmentation.


Subject(s)
Fracture Healing , Humans , Fracture Healing/physiology , Bone Marrow Transplantation/methods , Cell- and Tissue-Based Therapy/methods , Bone Regeneration/physiology , Fractures, Bone/therapy , Bone Marrow Cells/cytology
10.
ACS Appl Mater Interfaces ; 16(19): 25317-25332, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38706308

ABSTRACT

This investigation aimed to construct a bilayer scaffold integrating alginate and gelatin with nanobioactive glass (BG), recognized for their efficacy in tissue regeneration and drug delivery. Scaffolds, namely, alginate/gelatin (AG), alginate-/actonel gelatin (AGD), alginate actenol/gelatin-45S5 BG (4AGD), and alginate-actonel/gelatin-59S BG (5AGD), were assembled using a cost-effective freeze-drying method, followed by detailed structural investigation via powder X-ray diffraction as well as morphological characterization using field emission scanning electron microscopy (FESEM). FESEM revealed a honeycomb-like morphology with distinct pore sizes for nutrient, oxygen, and drug transport. The scaffolds evidently exhibited hemocompatibility, high porosity, good swelling capacity, and biodegradability. In vitro studies demonstrated sustained drug release, particularly for scaffolds containing actonel. In vivo tests showed that the bilayer scaffold promoted new bone formation, surpassing the control group in bone area increase. The interaction of the scaffold with collagen and released ions improved the osteoblastic function and bone volume fraction. The findings suggest that this bilayer scaffold could be beneficial for treating critical-sized bone defects, especially in the mandibular and femoral regions.


Subject(s)
Femur , Glass , Mandible , Tissue Scaffolds , Tissue Scaffolds/chemistry , Animals , Glass/chemistry , Mandible/diagnostic imaging , Mandible/surgery , Mandible/drug effects , Femur/drug effects , Femur/diagnostic imaging , Femur/pathology , Gelatin/chemistry , Bone Regeneration/drug effects , Alginates/chemistry , Porosity , Humans , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Tissue Engineering
11.
ACS Appl Mater Interfaces ; 16(19): 24384-24397, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38709640

ABSTRACT

Vascularization and inflammation management are essential for successful bone regeneration during the healing process of large bone defects assisted by artificial implants/fillers. Therefore, this study is devoted to the optimization of the osteogenic microenvironment for accelerated bone healing through rapid neovascularization and appropriate inflammation inhibition that were achieved by applying a tantalum oxide (TaO)-based nanoplatform carrying functional substances at the bone defect. Specifically, TaO mesoporous nanospheres were first constructed and then modified by functionalized metal ions (Mg2+) with the following deferoxamine (DFO) loading to obtain the final product simplified as DFO-Mg-TaO. Scanning electron microscopy (SEM) and transmission electron microscopy (TEM) revealed that the product was homogeneously dispersed hollow nanospheres with large specific surface areas and mesoporous shells suitable for loading Mg2+ and DFO. The biological assessments indicated that DFO-Mg-TaO could enhance the adhesion, proliferation, and osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). The DFO released from DFO-Mg-TaO promoted angiogenetic activity by upregulating the expressions of hypoxia-inducible factor-1 (HIF-1α) and vascular endothelial growth factor (VEGF). Notably, DFO-Mg-TaO also displayed anti-inflammatory activity by reducing the expressions of pro-inflammatory factors, benefiting from the release of bioactive Mg2+. In vivo experiments demonstrated that DFO-Mg-TaO integrated with vascular regenerative, anti-inflammatory, and osteogenic activities significantly accelerated the reconstruction of bone defects. Our findings suggest that the optimized DFO-Mg-TaO nanospheres are promising as multifunctional fillers to speed up the bone healing process.


Subject(s)
Bone Regeneration , Deferoxamine , Magnesium , Mesenchymal Stem Cells , Oxides , Tantalum , Deferoxamine/chemistry , Deferoxamine/pharmacology , Bone Regeneration/drug effects , Tantalum/chemistry , Animals , Oxides/chemistry , Oxides/pharmacology , Magnesium/chemistry , Magnesium/pharmacology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteogenesis/drug effects , Neovascularization, Physiologic/drug effects , Rats , Mice , Rats, Sprague-Dawley , Cell Proliferation/drug effects , Angiogenesis
12.
Stem Cell Res Ther ; 15(1): 144, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38764077

ABSTRACT

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.


Subject(s)
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
13.
Biomed Mater ; 19(4)2024 May 17.
Article in English | MEDLINE | ID: mdl-38756029

ABSTRACT

Hard tissue engineering scaffolds especially 3D printed scaffolds were considered an excellent strategy for craniomaxillofacial hard tissue regeneration, involving crania and facial bones and teeth. Porcine treated dentin matrix (pTDM) as xenogeneic extracellular matrix has the potential to promote the stem cell differentiation and mineralization as it contains plenty of bioactive factors similar with human-derived dentin tissue. However, its application might be impeded by the foreign body response induced by the damage-associated molecular patterns of pTDM, which would cause strong inflammation and hinder the regeneration. Ceria nanoparticles (CNPs) show a great promise at protecting tissue from oxidative stress and influence the macrophages polarization. Using 3D-bioprinting technology, we fabricated a xenogeneic hard tissue scaffold based on pTDM xenogeneic TDM-polycaprolactone (xTDM/PCL) and we modified the scaffolds by CNPs (xTDM/PCL/CNPs). Through series ofin vitroverification, we found xTDM/PCL/CNPs scaffolds held promise at up-regulating the expression of osteogenesis and odontogenesis related genes including collagen type 1, Runt-related transcription factor 2 (RUNX2), bone morphogenetic protein-2, osteoprotegerin, alkaline phosphatase (ALP) and DMP1 and inducing macrophages to polarize to M2 phenotype. Regeneration of bone tissues was further evaluated in rats by conducting the models of mandibular and skull bone defects. Thein vivoevaluation showed that xTDM/PCL/CNPs scaffolds could promote the bone tissue regeneration by up-regulating the expression of osteogenic genes involving ALP, RUNX2 and bone sialoprotein 2 and macrophage polarization into M2. Regeneration of teeth evaluated on beagles demonstrated that xTDM/PCL/CNPs scaffolds expedited the calcification inside the scaffolds and helped form periodontal ligament-like tissues surrounding the scaffolds.


Subject(s)
Cerium , Extracellular Matrix , Nanoparticles , Osteogenesis , Printing, Three-Dimensional , Tissue Engineering , Tissue Scaffolds , Animals , Tissue Scaffolds/chemistry , Tissue Engineering/methods , Swine , Extracellular Matrix/metabolism , Cerium/chemistry , Nanoparticles/chemistry , Rats , Polyesters/chemistry , Dentin/chemistry , Humans , Bone Regeneration/drug effects , Odontogenesis , Cell Differentiation , Regeneration , Macrophages/metabolism , Skull , Rats, Sprague-Dawley
14.
J Orthop Surg Res ; 19(1): 299, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755635

ABSTRACT

BACKGROUND: This study aims to evaluate the optimal ratio of synthetic bone graft (SBG) material and platelet rich fibrin (PRF) mixed in a metal 3D-printed implant to enhance bone regeneration. METHODS: Specialized titanium hollow implants (5 mm in diameter and 6 mm in height for rabbit; 6 mm in diameter and 5 mm in height for pig) were designed and manufactured using 3D printing technology. The implants were divided into three groups and filled with different bone graft combinations, namely (1) SBG alone; (2) PRF to SBG in 1:1 ratio; (3) PRF to SBG in 2:1 ratio. These three groups were replicated tightly into each bone defect in distal femurs of rabbits (nine implants, n = 3) and femoral shafts of pigs (fifteen implants, n = 5). Animal tissue sections were obtained after euthanasia at the 8th postoperative week. The rabbit specimens were stained with analine blue, while the pig specimens were stained with Masson-Goldner's trichrome stain to perform histologically examination. All titanium hollow implants were well anchored, except in fracture specimens (three in the rabbit and one fracture in the pig). RESULT: Rabbit specimens under analine blue staining showed that collagen tissue increased by about 20% and 40% in the 1:1 ratio group and the 2:1 ratio group, respectively. Masson-Goldner's trichrome stain results showed that new bone growth increased by 32% in the 1:1 ratio PRF to SBG, while - 8% in the 2:1 ratio group. CONCLUSION: This study demonstrated that placing a 1:1 ratio combination of PRF and SBG in a stabilized titanium 3D printed implant resulted in an optimal increase in bone growth.


Subject(s)
Bone Regeneration , Platelet-Rich Fibrin , Printing, Three-Dimensional , Titanium , Animals , Rabbits , Bone Regeneration/drug effects , Bone Regeneration/physiology , Swine , Femur/surgery , Bone Substitutes , Bone Transplantation/methods , Prostheses and Implants
15.
Sci Adv ; 10(20): eadl6343, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38758783

ABSTRACT

Trauma rapidly mobilizes the immune response of surrounding tissues and activates regeneration program. Manipulating immune response to promote tissue regeneration shows a broad application prospect. However, the understanding of bone healing dynamics at cellular level remains limited. Here, we characterize the landscape of immune cells after alveolar bone injury and reveal a pivotal role of infiltrating natural killer T (NKT) cells. We observe a rapid increase in NKT cells after injury, which inhibit osteogenic differentiation of mesenchymal stem cells (MSCs) and impair alveolar bone healing. Cxcl2 is up-regulated in NKT cells after injury. Systemic administration of CXCL2-neutralizing antibody or genetic deletion of Cxcl2 improves the bone healing process. In addition, we fabricate a gelatin-based porous hydrogel to deliver NK1.1 depletion antibody, which successfully promotes alveolar bone healing. In summary, our study highlights the importance of NKT cells in the early stage of bone healing and provides a potential therapeutic strategy for accelerating bone regeneration.


Subject(s)
Bone Regeneration , Chemokine CXCL2 , Natural Killer T-Cells , Osteogenesis , Bone Regeneration/drug effects , Animals , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Mice , Osteogenesis/drug effects , Chemokine CXCL2/metabolism , Chemokine CXCL2/genetics , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Cell Differentiation , Mice, Inbred C57BL
16.
Biomed Mater ; 19(4)2024 May 15.
Article in English | MEDLINE | ID: mdl-38697199

ABSTRACT

Porous tantalum scaffolds offer a high degree of biocompatibility and have a low friction coefficient. In addition, their biomimetic porous structure and mechanical properties, which closely resemble human bone tissue, make them a popular area of research in the field of bone defect repair. With the rapid advancement of additive manufacturing, 3D-printed porous tantalum scaffolds have increasingly emerged in recent years, offering exceptional design flexibility, as well as facilitating the fabrication of intricate geometries and complex pore structures that similar to human anatomy. This review provides a comprehensive description of the techniques, procedures, and specific parameters involved in the 3D printing of porous tantalum scaffolds. Concurrently, the review provides a summary of the mechanical properties, osteogenesis and antibacterial properties of porous tantalum scaffolds. The use of surface modification techniques and the drug carriers can enhance the characteristics of porous tantalum scaffolds. Accordingly, the review discusses the application of these porous tantalum materials in clinical settings. Multiple studies have demonstrated that 3D-printed porous tantalum scaffolds exhibit exceptional corrosion resistance, biocompatibility, and osteogenic properties. As a result, they are considered highly suitable biomaterials for repairing bone defects. Despite the rapid development of 3D-printed porous tantalum scaffolds, they still encounter challenges and issues when used as bone defect implants in clinical applications. Ultimately, a concise overview of the primary challenges faced by 3D-printed porous tantalum scaffolds is offered, and corresponding insights to promote further exploration and advancement in this domain are presented.


Subject(s)
Biocompatible Materials , Bone Substitutes , Bone and Bones , Osteogenesis , Printing, Three-Dimensional , Tantalum , Tissue Engineering , Tissue Scaffolds , Tantalum/chemistry , Tissue Scaffolds/chemistry , Porosity , Humans , Biocompatible Materials/chemistry , Tissue Engineering/methods , Animals , Bone Substitutes/chemistry , Materials Testing , Bone Regeneration
17.
Swiss Dent J ; 134(1): 144-157, 2024 Apr 05.
Article in German | MEDLINE | ID: mdl-38741457

ABSTRACT

The clinical impact of platelet-rich fibrin (PRF) and plasma rich in growth factors (PRGF®) respectively has been studied extensively in the field of regenerative dentistry during the last two decades. Literature supports evidence for additional benefits in regenerative periodontal therapy, alveolar ridge preservation, management of extraction sockets, implantology including guided bone regeneration as well as defect management in oral surgery. Regarding gingival wound healing and soft tissue regeneration, there is sufficient evidence for their positive effects which have been confirmed in several systematic reviews. The effects seem less clear in conjunction with osseous regenerative treatments, where the inter-study heterogenity in terms of different PRF-protocols, indications and application forms might hinder a systematic comparison. Nevertheless there is evidence that PRF might have beneficial effects on hard-tissue or its regeneration respectively.For being able to facilitate conclusions in systematic reviews, precise reporting of the used PRF-protocols is mandatory for future (clinical) research in the field of autologous platelet concentrates.


Subject(s)
Platelet-Rich Fibrin , Platelet-Rich Plasma , Humans , Guided Tissue Regeneration, Periodontal/methods , Blood Platelets/physiology , Bone Regeneration/physiology , Bone Regeneration/drug effects , Wound Healing/physiology , Wound Healing/drug effects , Regenerative Medicine/methods
18.
Nat Commun ; 15(1): 4160, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755128

ABSTRACT

The regeneration of critical-size bone defects, especially those with irregular shapes, remains a clinical challenge. Various biomaterials have been developed to enhance bone regeneration, but the limitations on the shape-adaptive capacity, the complexity of clinical operation, and the unsatisfied osteogenic bioactivity have greatly restricted their clinical application. In this work, we construct a mechanically robust, tailorable and water-responsive shape-memory silk fibroin/magnesium (SF/MgO) composite scaffold, which is able to quickly match irregular defects by simple trimming, thus leading to good interface integration. We demonstrate that the SF/MgO scaffold exhibits excellent mechanical stability and structure retention during the degradative process with the potential for supporting ability in defective areas. This scaffold further promotes the proliferation, adhesion and migration of osteoblasts and the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) in vitro. With suitable MgO content, the scaffold exhibits good histocompatibility, low foreign-body reactions (FBRs), significant ectopic mineralisation and angiogenesis. Skull defect experiments on male rats demonstrate that the cell-free SF/MgO scaffold markedly enhances bone regeneration of cranial defects. Taken together, the mechanically robust, personalised and bioactive scaffold with water-responsive shape-memory may be a promising biomaterial for clinical-size and irregular bone defect regeneration.


Subject(s)
Biocompatible Materials , Bone Regeneration , Fibroins , Magnesium , Mesenchymal Stem Cells , Osteogenesis , Tissue Scaffolds , Fibroins/chemistry , Fibroins/pharmacology , Bone Regeneration/drug effects , Animals , Tissue Scaffolds/chemistry , Male , Osteogenesis/drug effects , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Rats , Magnesium/chemistry , Magnesium/pharmacology , Biocompatible Materials/chemistry , Osteoblasts/drug effects , Cell Differentiation/drug effects , Rats, Sprague-Dawley , Water/chemistry , Cell Proliferation/drug effects , Tissue Engineering/methods , Skull/drug effects , Cell Adhesion/drug effects , Bombyx
19.
Bone Res ; 12(1): 28, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38744863

ABSTRACT

Osteomyelitis is a devastating disease caused by microbial infection in deep bone tissue. Its high recurrence rate and impaired restoration of bone deficiencies are major challenges in treatment. Microbes have evolved numerous mechanisms to effectively evade host intrinsic and adaptive immune attacks to persistently localize in the host, such as drug-resistant bacteria, biofilms, persister cells, intracellular bacteria, and small colony variants (SCVs). Moreover, microbial-mediated dysregulation of the bone immune microenvironment impedes the bone regeneration process, leading to impaired bone defect repair. Despite advances in surgical strategies and drug applications for the treatment of bone infections within the last decade, challenges remain in clinical management. The development and application of tissue engineering materials have provided new strategies for the treatment of bone infections, but a comprehensive review of their research progress is lacking. This review discusses the critical pathogenic mechanisms of microbes in the skeletal system and their immunomodulatory effects on bone regeneration, and highlights the prospects and challenges for the application of tissue engineering technologies in the treatment of bone infections. It will inform the development and translation of antimicrobial and bone repair tissue engineering materials for the management of bone infections.


Subject(s)
Tissue Engineering , Humans , Tissue Engineering/methods , Osteomyelitis/microbiology , Osteomyelitis/therapy , Osteomyelitis/drug therapy , Bone Regeneration , Animals
20.
Int J Implant Dent ; 10(1): 25, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38760582

ABSTRACT

PURPOSE: This retrospective cohort study evaluates the influence of connective tissue grafts (CTG) on bone regeneration at implant sites with total loss of the buccal bone wall treated with flapless immediate implant placement (IIP) and reconstruction with autogenous bone chips (AB) within a follow-up of up to 13 years. METHODS: Sixty implants were inserted in 55 patients in sites with total loss of the buccal bone wall between 2008 and 2021. The implants were inserted and the buccal gaps were grafted by AB. A subgroup of 34 sites was grafted additionally with CTG using tunnel technique. Primary outcome was the vertical bone regeneration in height and thickness. Secondary outcome parameters were interproximal marginal bone level, recession, soft tissue esthetics (PES), width of keratinized mucosa (KMW) and probing depths (PPD). RESULTS: Mean follow-up period was 60.8 months. In 55 sites a complete vertical bone regeneration was documented. The mean buccal bone level increased by 10.6 mm significantly. The thickness of the buccal bone wall ranged between 1.7 and 1.9 mm, and was significantly thicker in sites without CTG. Interproximal marginal bone level was at implant shoulder level. The mean recession improved significantly by 1.2 mm. In sites with CTG, recessions and PES improved significantly more. CONCLUSIONS: Additional CTG in extraction sites with total buccal bone loss followed by IIP with simultaneous AB grafting led to improved PES and recession, but also to a thinner buccal bone wall compared to sites grafted just with AB.


Subject(s)
Connective Tissue , Immediate Dental Implant Loading , Humans , Retrospective Studies , Connective Tissue/transplantation , Female , Male , Middle Aged , Immediate Dental Implant Loading/methods , Adult , Bone Transplantation/methods , Aged , Bone Regeneration/physiology , Alveolar Ridge Augmentation/methods , Alveolar Bone Loss/surgery
SELECTION OF CITATIONS
SEARCH DETAIL
...