Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.369
Filter
1.
Int J Implant Dent ; 10(1): 24, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38722448

ABSTRACT

PURPOSE: The objective of the present study was to ascertain the effect of immediate occlusal loading after implant placement on osseointegration and the micro/nanostructure of the surrounding bone. METHODS: After extraction of a rat maxillary right second molar, an implant was placed immediately with initial fixation (2 N< ). The implants were placed to avoid occlusal loading due to mastication, and in the loaded group, a superstructure was fabricated and subjected to occlusal loading. Bone morphometry, collagen fiber anisotropy, and biological apatite (BAp) crystallite alignment were quantitatively evaluated in both groups after extraction and fixation of the jaw bone at Days 7 and 21 after surgery. RESULTS: Osseointegration was observed in both groups. Bone morphometry showed significant differences in bone volume, trabecular number, trabecular thickness and bone mineral density (BMD) at Days 21 postoperatively (P < 0.05). A significant difference was also found in the trabecular separation at Days 7 postoperatively (P < 0.05). In the evaluation of collagen fiber anisotropy, collagen fiber bundles running differently from the existing bone were observed in both groups. In terms of BAp crystallite alignment, a specific structure was observed in the reconstructed new bone after implantation, and preferential orientation of BAp crystallite alignment was observed in the longitudinal direction of the implants in the Day 21 postoperative loaded group. CONCLUSION: When sufficient initial fixation is achieved at the time of dental implant placement, then the applied masticatory load may contribute to rapidly achieving not only bone volume, but also adequate bone quality after implant placement.


Subject(s)
Immediate Dental Implant Loading , Osseointegration , Animals , Rats , Osseointegration/drug effects , Male , Bone Density/physiology , Dental Implants , Rats, Wistar , Maxilla/surgery , Collagen/metabolism , X-Ray Microtomography
2.
Biomed Mater ; 19(4)2024 May 22.
Article in English | MEDLINE | ID: mdl-38740037

ABSTRACT

The purpose of this study was to construct a rutin-controlled release system on the surface of Ti substrates and investigate its effects on osteogenesis and osseointegration on the surface of implants. The base layer, polyethylenimine (PEI), was immobilised on a titanium substrate. Then, hyaluronic acid (HA)/chitosan (CS)-rutin (RT) multilayer films were assembled on the PEI using layer-by-layer (LBL) assembly technology. We used scanning electron microscopy (SEM), Fourier transform infrared (FTIR) spectroscopy and contact angle measurements to examine all Ti samples. The drug release test of rutin was also carried out to detect the slow-release performance. The osteogenic abilities of the samples were evaluated by experiments on an osteoporosis rat model and MC3T3-E1 cells. The results (SEM, FTIR and contact angle measurements) all confirmed that the PEI substrate layer and HA/CS-RT multilayer film were effectively immobilised on titanium. The drug release test revealed that a rutin controlled release mechanism had been successfully established. Furthermore, thein vitrodata revealed that osteoblasts on the coated titanium matrix had greater adhesion, proliferation, and differentiation capacity than the osteoblasts on the pure titanium surface. When MC3T3-E1 cells were exposed to H2O2-induced oxidative stressin vitro, cell-based tests revealed great tolerance and increased osteogenic potential on HA/CS-RT substrates. We also found that the HA/CS-RT coating significantly increased the new bone mass around the implant. The LBL-deposited HA/CS-RT multilayer coating on the titanium base surface established an excellent rutin-controlled release system, which significantly improved osseointegration and promoted osteogenesis under oxidative stress conditions, suggesting a new implant therapy strategy for patients with osteoporosis.


Subject(s)
Coated Materials, Biocompatible , Hyaluronic Acid , Osseointegration , Osteoblasts , Osteogenesis , Osteoporosis , Prostheses and Implants , Rutin , Surface Properties , Titanium , Animals , Titanium/chemistry , Rutin/chemistry , Rutin/pharmacology , Osteogenesis/drug effects , Rats , Osteoporosis/drug therapy , Mice , Osteoblasts/drug effects , Osteoblasts/cytology , Osteoblasts/metabolism , Osseointegration/drug effects , Hyaluronic Acid/chemistry , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Oxidation-Reduction , Chitosan/chemistry , Female , Rats, Sprague-Dawley , Cell Adhesion/drug effects , Spectroscopy, Fourier Transform Infrared , Cell Differentiation/drug effects , Microscopy, Electron, Scanning , Cell Proliferation/drug effects , Polyethyleneimine/chemistry , 3T3 Cells , Oxidative Stress/drug effects , Layer-by-Layer Nanoparticles
3.
Iran Biomed J ; 28(2&3): 82-9, 2024 05 01.
Article in English | MEDLINE | ID: mdl-38770885

ABSTRACT

Background: Osteogenic, antioxidant and anti-inflammatory effects of Whey protein and M. oleifera gel prompted us to evaluate their role alone or in combination on osseointegration in rabbits. Methods: In this study, 24 titanium implants were inserted in the femurs of six rabbits. One implant was placed without treatment, and another one was coated with a mixture of whey protein and M. oleifera gel for each side. The animals were divided into two groups of 2- and 6-week intervals and evaluated using histopathological and immunohistochemical techniques. Results: Histological evaluation revealed a significant difference between the experimental and the control groups after two weeks in osteoblast and osteocyte counts. The experimental group had mature bone development after six weeks of implantation, while the control group had a woven bone. Immunohistochemical results showed that the experimental group, compared to the control group, exhibited early positive expression of osteoblast cells at two weeks after the experiment. Based on histopathological observations, the experimental group showed a tiny area of collagenous fiber in 6th week after the implantation. Conclusion: A mixture of whey protein and M. oleifera could accelerate osseointegration and healing processes.


Subject(s)
Moringa oleifera , Osseointegration , Plant Extracts , Plant Leaves , Whey Proteins , Animals , Whey Proteins/pharmacology , Rabbits , Osseointegration/drug effects , Moringa oleifera/chemistry , Plant Extracts/pharmacology , Plant Leaves/chemistry , Male , Osteoblasts/drug effects , Femur/drug effects , Osteogenesis/drug effects
4.
ACS Biomater Sci Eng ; 10(5): 3255-3267, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38684056

ABSTRACT

Osteoporosis is a metabolic disease characterized by bone density and trabecular bone loss. Bone loss may affect dental implant osseointegration in patients with osteoporosis. To promote implant osseointegration in osteoporotic patients, we further used a nonthermal atmospheric plasma (NTAP) treatment device previously developed by our research group. After the titanium implant (Ti) is placed into the device, the working gas flow and the electrode switches are turned on, and the treatment is completed in 30 s. Previous studies showed that this NTAP device can remove carbon contamination from the implant surface, increase the hydroxyl groups, and improve its wettability to promote osseointegration in normal conditions. In this study, we demonstrated the tremendous osteogenic enhancement effect of NTAP-Ti in osteoporotic conditions in rats for the first time. Compared to Ti, the proliferative potential of osteoporotic bone marrow mesenchymal stem cells on NTAP-Ti increased by 180% at 1 day (P = 0.004), while their osteogenic differentiation increased by 149% at 14 days (P < 0.001). In addition, the results indicated that NTAP-Ti significantly improved osseointegration in osteoporotic rats in vivo. Compared to the Ti, the bone volume fraction (BV/TV) and trabecular number (Tb.N) values of NTAP-Ti in osteoporotic rats, respectively, increased by 18% (P < 0.001) and 25% (P = 0.007) at 6 weeks and the trabecular separation (Tb.Sp) value decreased by 26% (P = 0.02) at 6 weeks. In conclusion, this study proved a novel NTAP irradiation titanium implant that can significantly promote osseointegration in osteoporotic conditions.


Subject(s)
Mesenchymal Stem Cells , Osseointegration , Osteogenesis , Osteoporosis , Plasma Gases , Rats, Sprague-Dawley , Titanium , Titanium/pharmacology , Animals , Osteogenesis/drug effects , Osteoporosis/pathology , Osteoporosis/therapy , Osteoporosis/drug therapy , Plasma Gases/pharmacology , Plasma Gases/therapeutic use , Osseointegration/drug effects , Female , Rats , Mesenchymal Stem Cells/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Prostheses and Implants
5.
Int J Biol Macromol ; 267(Pt 2): 131636, 2024 May.
Article in English | MEDLINE | ID: mdl-38641287

ABSTRACT

Although bioactive peptides enhancing bone healing have demonstrated effectiveness in treating bone defects, in vivo instability poses a challenge to their clinical application. Currently reported peptide delivery systems do not meet the demands of bone tissue repair regarding stability and peptide release efficacy. Herein, the self-assembling recombinant chimeric protein (Sbp5-2RGD) is developed by genetic engineering with cell adhesion peptide RGD as the targeted peptide and a newly discovered scallop byssal-derived protein Sbp5-2 that can assemble into wet stable films as the structural domain. In vitro studies show that the Sbp5-2RGD film exhibits excellent extensibility and biocompatibility. In vitro and in vivo degradation experiments demonstrate that the film remains stable due to the layer-by-layer degradation mode, resulting in sustained delivery of RGD in situ for up to 4 weeks. Consequently, the film can effectively promote osteogenesis, which accelerates bone defect healing and the implants osseointegration. Cell-level studies further show that the film up-regulates the expression of genes and proteins (ALP, OCN, OSX, OPN, RUNX2, VEGF) associated with osteogenesis and angiogenesis. Overall, this novel protein film represents an intelligent platform for peptide immobilization, protection, and release through its self-assembly, dense structure, and degradation mode, providing a therapeutic strategy for bone repair.


Subject(s)
Genetic Engineering , Oligopeptides , Animals , Humans , Mice , Drug Delivery Systems , Genetic Engineering/methods , Oligopeptides/chemistry , Oligopeptides/pharmacology , Osseointegration/drug effects , Osteogenesis/drug effects , Pectinidae , Rats, Sprague-Dawley , Male , Rats
6.
ACS Biomater Sci Eng ; 10(5): 3454-3469, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38590081

ABSTRACT

Massive unmelted Ti6Al4 V (Ti64) particles presented across all surfaces of additively manufactured Ti64 scaffolds significantly impacted the designed surface topography, mechanical properties, and permeability, reducing the osseointegration of the scaffolds. In this study, the proposed flowing acid etching (FAE) method presented high efficiency in eliminating Ti64 particles and enhancing the surface modification capacity across all surfaces of Ti64 scaffolds. The Ti64 particles across all surfaces of the scaffolds were completely removed effectively and evenly. The surface topography of the scaffolds closely resembled the design after the 75 s FAE treatment. The actual elastic modulus of the treated scaffolds (3.206 ± 0.040 GPa) was closer to the designed value (3.110 GPa), and a micrometer-scale structure was constructed on the inner and outer surfaces of the scaffolds after the 90 s FAE treatment. However, the yield strength of scaffolds was reduced to 89.743 ± 0.893 MPa from 118.251 ± 0.982 MPa after the 90 s FAE treatment. The FAE method also showed higher efficiency in decreasing the roughness and enhancing the hydrophilicity and surface energy of all of the surfaces. The FAE treatment improved the permeability of scaffolds efficiently, and the permeability of scaffolds increased to 11.93 ± 0.21 × 10-10 mm2 from 8.57 ± 0.021 × 10-10 mm2 after the 90 s FAE treatment. The treated Ti64 scaffolds after the 90 s FAE treatment exhibited optimized osseointegration effects in vitro and in vivo. In conclusion, the FAE method was an efficient way to eliminate unmelted Ti64 particles and obtain ideal surface topography, mechanical properties, and permeability to promote osseointegration in additively manufactured Ti64 scaffolds.


Subject(s)
Alloys , Osseointegration , Surface Properties , Tissue Scaffolds , Titanium , Titanium/chemistry , Alloys/chemistry , Osseointegration/drug effects , Animals , Tissue Scaffolds/chemistry , Elastic Modulus , Materials Testing
7.
Biomater Sci ; 12(10): 2648-2659, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38573023

ABSTRACT

Titanium (Ti) and its alloys have been widely employed in the treatment of orthopedics and other hard tissue diseases. However, Ti-based implants are bioinert and suffer from bacterial infections and poor osseointegration in clinical applications. Herein, we successfully modified Ti with a porous N-halaminated spermidine-containing polymeric coating (Ti-SPD-Cl) through alkali-heat treatment, surface grafting and chlorination, and it has both excellent antibacterial and osteogenic abilities to significantly enhance osseointegration. The as-obtained Ti-SPD-Cl contains abundant N-Cl groups and demonstrates effective antibacterial ability against S. aureus and E. coli. Meanwhile, due to the presence of the spermidine component and construction of a porous hydrophilic surface, Ti-SPD-Cl is also beneficial for maintaining cell membrane homeostasis and promoting cell adhesion, exhibiting good biocompatibility and osteogenic ability. The rat osteomyelitis model demonstrates that Ti-SPD-Cl can effectively suppress bacterial infection and enhance bone-implant integration. Thus, Ti-SPD-Cl shows promising clinical applicability in the prevention of orthopedic implant infections and poor osseointegration.


Subject(s)
Anti-Bacterial Agents , Coated Materials, Biocompatible , Escherichia coli , Osseointegration , Rats, Sprague-Dawley , Spermidine , Staphylococcus aureus , Titanium , Titanium/chemistry , Titanium/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Osseointegration/drug effects , Animals , Staphylococcus aureus/drug effects , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Spermidine/pharmacology , Spermidine/chemistry , Escherichia coli/drug effects , Rats , Polymers/chemistry , Polymers/pharmacology , Osteogenesis/drug effects , Mice , Surface Properties , Microbial Sensitivity Tests , Male
8.
Int J Mol Sci ; 25(8)2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38673834

ABSTRACT

In this paper, the in vivo behavior of orthopedic implants covered with thin films obtained by matrix-assisted pulsed laser evaporation and containing bioactive glass, a polymer, and natural plant extract was evaluated. In vivo testing was performed by carrying out a study on guinea pigs who had coated metallic screws inserted in them and also controls, following the regulations of European laws regarding the use of animals in scientific studies. After 26 weeks from implantation, the guinea pigs were subjected to X-ray analyses to observe the evolution of osteointegration over time; the guinea pigs' blood was collected for the detection of enzymatic activity and to measure values for urea, creatinine, blood glucose, alkaline phosphatase, pancreatic amylase, total protein, and glutamate pyruvate transaminase to see the extent to which the body was affected by the introduction of the implant. Moreover, a histopathological assessment of the following vital organs was carried out: heart, brain, liver, and spleen. We also assessed implanted bone with adjacent tissue. Our studies did not find significant variations in biochemical and histological results compared to the control group or significant adverse effects caused by the implant coating in terms of tissue compatibility, inflammatory reactions, and systemic effects.


Subject(s)
Plant Extracts , Animals , Guinea Pigs , Plant Extracts/chemistry , Plant Extracts/pharmacology , Nanostructures/chemistry , Biocompatible Materials/chemistry , Materials Testing , Glass/chemistry , Prostheses and Implants , Male , Osseointegration/drug effects
9.
Biomed Mater ; 19(4)2024 May 09.
Article in English | MEDLINE | ID: mdl-38653261

ABSTRACT

Artificial bone graft with osteoconductivity, angiogenesis, and immunomodulation is promising clinical therapeutics for the reluctant healing process of bone defects. Among various osteogenic substitutes, polymethyl methacrylate (PMMA) bone cement is a quit competitive platform due to its easy deployment to the bone defects with irregular shape and biomimetic mechanical properties. However, the biologically inert essence of PMMA is reliant on the passive osseointegration and cannot provide sufficient biologic cues to induce fast bone repair. Bioactive glass could serve as an efficient platform for the active osteogenesis of PMMA via ionic therapy and construction of alkaline microenvironment. However, the direct of deployment of bioactive glass into PMMA may trigger additional cytotoxicity and hinder cell growth on its surface. Hence we incorporated ionic therapy as osteogenic cue into the PMMA to enhance the biomedical properties. Specifically, we synthesized core-shell microspheres with a strontium-doped bioactive glass (SrBG) core and hydroxyapatite (HA) shell, and then composited them with PMMA to introduce multifunctional effects of HA incorporation, alkaline microenvironment construction, and functional ion release by adding microsphere. We preparedxSrBG@HA/PMMA cements (x= 30, 40, 50) with varied microsphere content and evaluated impacts on mechanical/handling properties, ion release, and investigated the impacts of different composite cements on proliferation, osteogenic differentiation, angiogenic potential, and macrophage polarization. These findings provide new perspectives and methodologies for developing advanced bone biomaterials to promote tissue regeneration.


Subject(s)
Bone Cements , Durapatite , Microspheres , Osteogenesis , Polymethyl Methacrylate , Strontium , Bone Cements/chemistry , Polymethyl Methacrylate/chemistry , Osteogenesis/drug effects , Porosity , Strontium/chemistry , Animals , Mice , Durapatite/chemistry , Biocompatible Materials/chemistry , Materials Testing , Cell Proliferation/drug effects , Osseointegration/drug effects , Cell Differentiation/drug effects , Ceramics/chemistry , Glass/chemistry , Humans , Bone Substitutes/chemistry
10.
Ann Biomed Eng ; 52(6): 1744-1762, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38517621

ABSTRACT

Osseointegration is a complex biological cascade that regulates bone regeneration after implant placement. Implants possessing complex multiscale surface topographies augment this regenerative process through the regulation of bone marrow stromal cells (MSCs) that are in contact with the implant surface. One pathway regulating osteoblastic differentiation is Wnt signaling, and upregulation of non-canonical Wnts increases differentiation of MSCs on these titanium substrates. Wnt16 is a non-canonical Wnt shown to regulate bone morphology in mouse models. This study evaluated the role of Wnt16 during surface-mediated osteoblastic differentiation of MSCs in vitro and osseointegration in vivo. MSCs were cultured on Ti substrates with different surface properties and non-canonical Wnt expression was determined. Subsequently, MSCs were cultured on Ti substrates +/-Wnt16 (100 ng/mL) and anti-Wnt16 antibodies (2 µg/mL). Wnt16 expression was increased in cells grown on microrough surfaces that were processed to be hydrophilic and have nanoscale roughness. However, treatment MSCs on these surfaces with exogenous rhWnt16b increased total DNA content and osteoprotegerin production, but reduced osteoblastic differentiation and production of local factors necessary for osteogenesis. Addition of anti-Wnt16 antibodies blocked the inhibitor effects of Wnt16. The response to Wnt16 was likely independent of other osteogenic pathways like Wnt11-Wnt5a signaling and semaphorin 3a signaling. We used an established rat model of cortical and trabecular femoral bone impairment following botox injections (2 injections of 8 units/leg each, starting and maintenance doses) to assess Wnt16 effects on whole bone morphology and implant osseointegration. Wnt16 injections did not alter whole bone morphology significantly (BV/TV, cortical thickness, restoration of trabecular bone) but were effective at increasing cortical bone-to-implant contact during impaired osseointegration in the botox model. The mechanical quality of the increased bone was not sufficient to rescue the deleterious effects of botox. Clinically, these results are important to understand the interaction of cortical and trabecular bone during implant integration. They suggest a role for Wnt16 in modulating bone remodeling by reducing osteoclastic activity. Targeted strategies to temporally regulate Wnt16 after implant placement could be used to improve osseointegration by increasing the net pool of osteoprogenitor cells.


Subject(s)
Cell Differentiation , Cell Proliferation , Mesenchymal Stem Cells , Osseointegration , Rats, Sprague-Dawley , Wnt Proteins , Animals , Wnt Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Rats , Cell Proliferation/drug effects , Osseointegration/drug effects , Bone Diseases, Metabolic/metabolism , Bone Diseases, Metabolic/pathology , Male , Titanium , Disease Models, Animal , Cells, Cultured
11.
Adv Sci (Weinh) ; 11(18): e2307269, 2024 May.
Article in English | MEDLINE | ID: mdl-38445899

ABSTRACT

Surface modification is an important approach to improve osseointegration of the endosseous implants, however it is still desirable to develop a facile yet efficient coating strategy. Herein, a metal-phenolic network (MPN) is proposed as a multifunctional nanocoating on titanium (Ti) implants for enhanced osseointegration through early immunomodulation. With tannic acid (TA) and Sr2+ self-assembled on Ti substrates, the MPN coatings provided a bioactive interface, which can facilitate the initial adhesion and recruitment of bone marrow mesenchymal stem cells (BMSCs) and polarize macrophage toward M2 phenotype. Furthermore, the TA-Sr coatings accelerated the osteogenic differentiation of BMSCs. In vivo evaluations further confirmed the enhanced osseointegration of TA-Sr modified implants via generating a favorable osteoimmune microenvironment. In general, these results suggest that TA-Sr MPN nanocoating is a promising strategy for achieving better and faster osseointegration of bone implants, which can be easily utilized in future clinical applications.


Subject(s)
Immunomodulation , Mesenchymal Stem Cells , Osseointegration , Titanium , Osseointegration/drug effects , Animals , Titanium/chemistry , Immunomodulation/drug effects , Tannins/pharmacology , Tannins/chemistry , Surface Properties , Prostheses and Implants , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Osteogenesis/drug effects , Cell Differentiation/drug effects , Mice , Strontium/chemistry , Strontium/pharmacology , Models, Animal , Rats
12.
Int J Biol Macromol ; 266(Pt 1): 131169, 2024 May.
Article in English | MEDLINE | ID: mdl-38554899

ABSTRACT

Autogenous bone transplantation is a prevalent clinical method for addressing bone defects. However, the limited availability of donor bone and the morbidity associated with bone harvesting have propelled the search for suitable bone substitutes. Bio-inspired scaffolds, particularly those fabricated using electron beam melting (EBM) deposition technology, have emerged as a significant advancement in this field. These 3D-printed titanium alloy scaffolds are celebrated for their outstanding biocompatibility and favorable elastic modulus. Thermosensitive chitosan hydrogel, which transitions from liquid to solid at body temperature, serves as a popular carrier in bone tissue engineering. Icariin (ICA), known for its efficacy in promoting osteoblast differentiation from bone marrow mesenchymal stem cells (BMSCs), plays a crucial role in this context. We developed a system combining a 3D-printed titanium alloy with a thermosensitive chitosan hydrogel, capable of local bone regeneration and integration through ICA delivery. Our in vitro findings reveal that this system can gradually release ICA, demonstrating excellent biocompatibility while fostering BMSC proliferation and osteogenic differentiation. Immunohistochemistry and Micro-CT analyses further confirm the effectiveness of the system in accelerating in vivo bone regeneration and enhancing osseointegration. This composite system lays a significant theoretical foundation for advancing local bone regeneration and integration.


Subject(s)
Alloys , Cell Differentiation , Chitosan , Flavonoids , Hydrogels , Mesenchymal Stem Cells , Osseointegration , Osteogenesis , Printing, Three-Dimensional , Tissue Scaffolds , Titanium , Chitosan/chemistry , Chitosan/pharmacology , Titanium/chemistry , Osseointegration/drug effects , Alloys/chemistry , Alloys/pharmacology , Tissue Scaffolds/chemistry , Animals , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Osteogenesis/drug effects , Hydrogels/chemistry , Hydrogels/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Bone Regeneration/drug effects , Tissue Engineering/methods
13.
J Biomed Mater Res A ; 112(7): 1083-1092, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38411355

ABSTRACT

Porous titanium exhibits low elastic modulus and porous structure is thought to be a promising implant in bone defect repair. However, the bioinert and low mechanical strength of porous titanium have limited its clinical application, especially in load-bearing bone defect repair. Our previous study has reported an infiltration casting and acid corrosion (IC-AC) method to fabricate a novel porous titanium (pTi) with 40% porosity and 0.4 mm pore diameter, which exerts mechanical property matching with cortical bone and interconnected channels. In this study, we introduced a nanoporous coating and incorporated an osteogenic element strontium (Sr) on the surface of porous titanium (named as Sr-micro arch oxidation [MAO]) to improve the osteogenic ability of the pTi by MAO. Better biocompatibility of Sr-MAO was verified by cell adhesion experiment and cell counting kit-8 (CCK-8) test. The in vitro osteogenic-related tests such as immunofluorescence staining, alkaline phosphatase staining and real-time polymerase chain reaction (RT-PCR) demonstrated better osteogenic ability of Sr-MAO. Femoral bone defect repair model was employed to evaluate the osseointegration of samples in vivo. Results of micro-CT scanning, sequential fluorochrome labeling and Van Gieson staining suggested that Sr-MAO showed better in vivo osteogenic ability than other groups. Taking results of both in vitro and in vivo experiment together, this study indicated the Sr-MAO porous titanium could be a promising implant load-bearing bone defect.


Subject(s)
Osteogenesis , Titanium , Weight-Bearing , Titanium/chemistry , Porosity , Animals , Osteogenesis/drug effects , Surface Properties , Rabbits , Osseointegration/drug effects , Strontium/chemistry , Strontium/pharmacology , Male , Femur/pathology , Materials Testing , Mice
14.
Int J Mol Sci ; 23(5)2022 Feb 22.
Article in English | MEDLINE | ID: mdl-35269557

ABSTRACT

Standard zirconia implants used in restoration still present problems related to inertness and long-term stability. Various physicochemical approaches have been used to modify the implant surfaces to improve early and late bone-to-implant integration; however, no ideal surface modification has been reported. This study used pulsed laser deposition to deposit a fluorinated hydroxyapatite (FHA) film on a zirconia implant to create a biologically active surface. The film prepared was uniform, dense, and crack-free, and exhibited granular surface droplets; it also presented excellent mechanical strength and favorable biological behavior. The FHA-coated implant was implanted on the femur of Sprague-Dawley rats, and various tests and analyses were performed. Results show that the in vitro initial cell activity on the FHA-coated samples was enhanced. In addition, higher alkaline phosphatase activity and cell mineralization were detected in cells cultured on the FHA-coated groups. Further, the newly formed bone volume of the FHA-coated group was higher than that of the bare micro-adjusted composite nano-zirconia (NANOZR) group. Therefore, the FHA film facilitated osseointegration and may improve the long-term survival rates of dental implants, and could become part of a new treatment technology for implant surfaces, promoting further optimization of NANOZR implant materials.


Subject(s)
Coated Materials, Biocompatible/administration & dosage , Durapatite/chemistry , Femur/surgery , Fluorine/chemistry , Osseointegration/drug effects , Zirconium/administration & dosage , Alkaline Phosphatase/metabolism , Animals , Cells, Cultured , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Dental Implants , Femur/cytology , Femur/drug effects , Femur/metabolism , Lasers , Male , Materials Testing , Nanostructures , Prostheses and Implants , Rats , Rats, Sprague-Dawley , Surface Properties , Zirconium/chemistry , Zirconium/pharmacology
15.
Drug Deliv ; 29(1): 548-560, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35156499

ABSTRACT

The updating and optimization of drug delivery systems is critical for better in vivo behaviors of drugs, as well as for improving impaired implant osseointegration in diabetes. Numerous studies have reported the benefits of exendin-4 on diabetic bone, with the potential to enhance osseointegration in diabetes. To construct an appropriate sustained-release system of exendin-4 targeting implant osseointegration in diabetes, this study fabricated exendin-4-loaded microspheres using poly(lactic-co-glycolic acid) (PLGA) and chitosan. The morphology, size, encapsulation efficiency, and drug release behavior of microspheres were investigated. The bioactivity of drug-loaded microspheres on cell proliferation and osteogenic differentiation of diabetic BMSCs was investigated to examine the pharmacologic action of exendin-4 loaded into chitosan-PLGA microspheres. Further, the influence of microspheres on osseointegration was evaluated using type 2 diabetes mellitus (T2DM) rat implant model. After 4 weeks, the samples were evaluated by radiological and histological analysis. The results of in vitro experiments showed that the prepared exendin-4-loaded chitosan-PLGA microspheres have good properties as a drug delivery system, and the chitosan could improve the encapsulation efficiency and drug release of PLGA microspheres. In addition, exendin-4-loaded microspheres could enhance the proliferation and osteogenic differentiation of diabetic BMSCs. The results of in vivo experiments showed the exendin-4-loaded microspheres significantly improved the impaired osseointegration and bone formation around implants in T2DM rats without affecting blood glucose levels. Thus, the local application of exendin-4-loaded chitosan-PLGA microspheres might be a promising therapeutic strategy for improving the efficacy of dental implants in T2DM individuals.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Drug Implants/chemistry , Exenatide/pharmacology , Hypoglycemic Agents/pharmacology , Microspheres , Osseointegration/drug effects , Animals , Cell Proliferation/drug effects , Chemistry, Pharmaceutical , Delayed-Action Preparations , Drug Liberation , Exenatide/administration & dosage , Hypoglycemic Agents/administration & dosage , Male , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Random Allocation , Rats , Rats, Sprague-Dawley , Surface Properties
16.
Int J Mol Sci ; 23(3)2022 Jan 18.
Article in English | MEDLINE | ID: mdl-35162963

ABSTRACT

After dental implantation, osteopontin (OPN) is deposited on the hydroxyapatite (HA) blasted implant surface followed by direct osteogenesis, which is significantly disturbed in Opn-knockout (KO) mice. However, whether applying OPN on the implant surface promotes direct osteogenesis remains unclarified. This study analyzed the effects of various OPN modified protein/peptides coatings on the healing patterns of the bone-implant interface after immediately placed implantation in the maxilla of four-week-old Opn-KO and wild-type (WT) mice (n = 96). The decalcified samples were processed for immunohistochemistry for OPN and Ki67 and tartrate-resistant acid phosphatase histochemistry. In the WT mice, the proliferative activity in the HA binding peptide-OPN mimic peptide fusion coated group was significantly higher than that in the control group from day 3 to week 1, and the rates of OPN deposition and direct osteogenesis around the implant surface significantly increased in the recombinant-mouse-OPN (rOPN) group compared to the Gly-Arg-Gly-Asp-Ser peptide group in week 2. The rOPN group achieved the same rates of direct osteogenesis and osseointegration as those in the control group in a half period (week 2). None of the implant surfaces could rescue the direct osteogenesis in the healing process in the Opn-KO mice. These results suggest that the rOPN coated implant enhances direct osteogenesis during osseointegration following implantation.


Subject(s)
Durapatite/chemistry , Osseointegration/drug effects , Osteogenesis/drug effects , Osteopontin/administration & dosage , Acid Phosphatase/metabolism , Animals , Dental Implantation , Dental Implants , Gene Knockout Techniques , Mice , Models, Animal , Osteopontin/chemistry , Osteopontin/genetics , Osteopontin/pharmacology , Recombinant Proteins/administration & dosage , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology
17.
Int J Mol Sci ; 23(3)2022 Feb 03.
Article in English | MEDLINE | ID: mdl-35163682

ABSTRACT

A lack of primary stability and osteointegration in metallic implants may result in implant loosening and failure. Adding porosity to metallic implants reduces the stress shielding effect and improves implant performance, allowing the surrounding bone tissue to grow into the scaffold. However, a bioactive surface is needed to stimulate implant osteointegration and improve mechanical stability. In this study, porous titanium implants were produced via powder sintering to create different porous diameters and open interconnectivity. Two strategies were used to generate a bioactive surface on the metallic foams: (1) an inorganic alkali thermochemical treatment, (2) grafting a cell adhesive tripeptide (RGD). RGD peptides exhibit an affinity for integrins expressed by osteoblasts, and have been reported to improve osteoblast adhesion, whereas the thermochemical treatment is known to improve titanium implant osseointegration upon implantation. Bioactivated scaffolds and control samples were implanted into the tibiae of rabbits to analyze the effect of these two strategies in vivo regarding bone tissue regeneration through interconnected porosity. Histomorphometric evaluation was performed at 4 and 12 weeks after implantation. Bone-to-implant contact (BIC) and bone in-growth and on-growth were evaluated in different regions of interest (ROIs) inside and outside the implant. The results of this study show that after a long-term postoperative period, the RGD-coated samples presented higher quantification values of quantified newly formed bone tissue in the implant's outer area. However, the total analyzed bone in-growth was observed to be slightly greater in the scaffolds treated with alkali thermochemical treatment. These results suggest that both strategies contribute to enhancing porous metallic implant stability and osteointegration, and a combination of both strategies might be worth pursuing.


Subject(s)
Alkalies/pharmacology , Coated Materials, Biocompatible/pharmacology , Metallurgy , Oligopeptides/pharmacology , Osseointegration , Temperature , Tissue Scaffolds/chemistry , Titanium/pharmacology , Animals , Female , Implants, Experimental , Osseointegration/drug effects , Osteogenesis/drug effects , Porosity , Powders , Rabbits
18.
Comput Math Methods Med ; 2022: 6537676, 2022.
Article in English | MEDLINE | ID: mdl-35035523

ABSTRACT

OBJECTIVES: In this study, a new type of dental implant by covering the surface of the titanium (Ti) implant with zinc-magnesium (Zn-Mg) alloy was designed, to study the antibacterial and antioxidant effects of Mg alloy on titanium (Ti) implants in oral implant restoration. METHODS: Human gingival fibroblasts (HGFs), S. sanguinis, and F. nucleatum bacteria were used to detect the bioactivity and antibacterial properties of Mg alloy-coated Ti implants. In addition, B6/J mice implanted with different materials were used to further detect their antibacterial and antioxidant properties. RESULTS: The results showed that Mg alloy could better promote the adhesion and proliferation and improve the alkaline phosphatase (ALP) activity of HGFs, which contributed to better improved stability of implant osseointegration. In addition, Mg alloy could better inhibit the proliferation of S. sanguinis, while no significant difference was found in the proliferation of F. nucleatum between the two implants. In the mouse model, the peripheral inflammatory reaction and oxidative stress of the Mg alloy implant were significantly lower than those of the Ti alloy implant. CONCLUSIONS: Zn-Mg alloy-coated Ti implants could better inhibit the growth of Gram-positive bacteria in the oral cavity, inhibit oxidative stress, and facilitate the proliferation activity of HGFs and the potential of osteoblast differentiation, thus, better increasing the stability of implant osseointegration.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antioxidants/pharmacology , Dental Implants , Magnesium/pharmacology , Titanium , Alloys/chemistry , Alloys/pharmacology , Animals , Anti-Bacterial Agents/chemistry , Antioxidants/chemistry , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Computational Biology , Dental Implants/adverse effects , Dental Implants/microbiology , Dental Prosthesis Design , Gingiva/cytology , Gingiva/drug effects , Gingiva/metabolism , Humans , Magnesium/chemistry , Male , Mice , Mice, Inbred C57BL , Osseointegration/drug effects , Oxidative Stress/drug effects , Surface Properties , Titanium/chemistry , Zinc/pharmacology
19.
ACS Appl Mater Interfaces ; 14(1): 1-19, 2022 Jan 12.
Article in English | MEDLINE | ID: mdl-34939784

ABSTRACT

Compared with traditional internal fixation devices, bone adhesives are expected to exhibit remarkable advantages, such as improved fixation of comminuted fractures and maintained spatial location of fractured scattered bone pieces in treating bone injuries. In this review, different bone adhesives are summarized from the aspects of bone tissue engineering, and the applications of bone adhesives are emphasized. The concepts of "liquid scaffold" and "liquid plate" are proposed to summarize two different research directions of bone adhesives. Furthermore, significant advances of bone adhesives in recent years in mechanical strength, osseointegration, osteoconductivity, and osteoinductivity are discussed. We conclude this topic by providing perspectives on the state-of-the-art research progress and future development trends of bone adhesives. We hope this review will provide a comprehensive summary of bone adhesives and inspire more extensive and in-depth research on this subject.


Subject(s)
Fracture Healing/drug effects , Fractures, Bone/drug therapy , Macromolecular Substances/pharmacology , Tissue Adhesives/pharmacology , Animals , Bone Regeneration/drug effects , Bone and Bones/drug effects , Cell Line, Tumor , Humans , Macromolecular Substances/chemistry , Osseointegration/drug effects , Tissue Adhesives/chemistry , Tissue Engineering , Tissue Scaffolds/chemistry
20.
Front Endocrinol (Lausanne) ; 12: 774257, 2021.
Article in English | MEDLINE | ID: mdl-34956085

ABSTRACT

Treatment of osteomyelitis requires prolonged antibiotic therapy which significantly alters the gut microbiota. While the influences on bone mass and microstructure have been extensively studied, it is poorly understood what impact the changes in gut microbiota may have on the host response to osseointegration around an intramedullary nail implanted. Here, we explored the influence of gut microbiota on the bone osseointegration process around an implant under two conditions: implantation of an intramedullary nail in the bone marrow cavity and chronic osteomyelitis (CO) induced by Staphylococcus aureus infection. Body weight, hepatorenal functions, serum levels of proinflammatory cytokines were monitored. The composition of gut microbiota was assessed via 16S rRNA sequencing, and the bone condition was analyzed via micro-computed tomography, hematoxylin and eosin staining, Safranin O-fast green and Goldner's trichrome staining. Osteoblastogenesis and osteoclastogenesis were assessed by detecting tartrate-resistant acid phosphatase and osterix expression. We found that perturbation of gut microbiota (increase in Proteobacteria and decrease in Bacteroidetes) associated with delayed osseointegration and increased levels of proinflammatory cytokines in the serum (p<0.05), lower bone mass (p<0.05), deficient endochondral ossification and bone formation, reduced osteoblastogenesis (p<0.05) and enhanced osteoclastogenesis (p<0.001). Survival rates (p=0.002) and bacterial loads (p=0.0363) in bone differed significantly between the CO and antibiotic-treated CO mice, but cytokines levels, bone mineral density, and bone formation did not differ, likely because of the severely damaged bone structure. In summary, antibiotic treatment perturbed the gut microbiota and significantly interfered with the bone osseointegration around the nail by increasing proinflammatory cytokine levels in circulation, inhibiting osteoblastogenesis, enhancing osteoclastogenesis, and thus leading to higher pathogen colonization as well as higher mortality postinfection. This report of ours is the first to demonstrate antibiotic-induced alterations in the gut microbiota affect bone osseointegration, helping us understand the role of gut microbiota disorders in osteoblastogenesis and osteoclastogenesis following implant insertion with or without infection.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Gastrointestinal Microbiome/drug effects , Osseointegration/drug effects , Osteomyelitis/drug therapy , Staphylococcal Infections/drug therapy , Animals , Bone Nails , Disease Models, Animal , Male , Mice , Osteomyelitis/microbiology , Staphylococcus aureus
SELECTION OF CITATIONS
SEARCH DETAIL
...