Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 12.943
Filter
2.
Otol Neurotol ; 45(8): 913-918, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39142312

ABSTRACT

INTRODUCTION: This study reviews the feasibility of implanting active osseointegrated bone conduction devices in young children, below the prior age for FDA indication (<12 years), which has recently been reduced to 5 years. Outcomes included differences in adverse event rates and operative time between two groups (<12 and 12 years or older). MATERIALS AND METHODS: This study is a retrospective review of children receiving active osseointegrated bone conduction devices at a tertiary referral center academic hospital. One hundred and twenty-four children received 135 active osseointegrated bone conduction devices (May 2018-March 2024). RESULTS: Of 135 devices, 77 (57%) were in children <12 years (mean age (SD) = 7.9 (2.0) years, range = 4.9-11.9 years) and 58 (43%) were in 12 years or older (mean age (SD) = 15.1 (1.7) years, range = 12-18 years). Adverse events were significantly higher in the older group, occurring in 8 (10%) of 77 devices in children <12 years and 15 (26%) of 58 devices in children 12 years and older (26%) (Fisher's exact test = 0.0217 at p < 0.05). Major adverse events occurred in 5/124 (4%) patients, with 2 in patients <12 years (2/73, 3%) and 3 in children 12 and older (3/51, 6%). The proportion of major events between groups was not significantly different (Fisher's exact test = 0.4, p < 0.05). Mean surgical time was significantly less (t = -2.8799, df = 120.26, p = 0.005) in the children <12 years (mean (SD) = 66.5 (22.4) min) compared to those 12 and over (mean (SD) = 78.32 (23.1) min). CONCLUSIONS: Implantation of active osseointegrated bone conduction devices is feasible in children as young as 5 years and demonstrates low rates of complication. Further miniaturization may allow even earlier safe intervention.


Subject(s)
Bone Conduction , Feasibility Studies , Osseointegration , Humans , Child , Bone Conduction/physiology , Retrospective Studies , Male , Female , Child, Preschool , Osseointegration/physiology , Adolescent , Treatment Outcome , Hearing Loss, Conductive/surgery , Hearing Aids , Bone-Anchored Prosthesis , Prosthesis Implantation/methods
3.
F1000Res ; 13: 281, 2024.
Article in English | MEDLINE | ID: mdl-39149510

ABSTRACT

Introduction: Osseointegration stands as a pivotal concept within the realm of dental implants, signifying the intricate process through which a dental implant integrates with the adjoining bone tissue. Graphene oxide (GO) has been shown to promote osseointegration, the process by which the implant fuses with the surrounding bone. The objective of this study was to assess the osseointegrative and antimicrobial properties of GO nano coated dental implants. Methods: A systematic search was conducted using electronic databases (e.g., PubMed, Scopus, Web of Science) to identify relevant studies published. Inclusion criteria encompassed studies that evaluated the effects of GO nano coating on osseointegrative and antimicrobial characteristics of dental implants. Studies not written in English and published before 2012 were excluded. Results: The initial search yielded a total of 127 potential studies, of which six met the inclusion criteria and five were included in the review. These studies provided data on GO nano coated dental implants and their osseointegrative and antimicrobial properties. All the included studies showed moderate risk of bias. None of the studies provided information related to sample size calculation or sampling technique. Discussion: The findings from the included studies demonstrated that GO nano coating had a positive impact on osseointegrative properties of dental implants. Enhanced bone-implant contact and increased bone density were observed in animals and humans receiving GO nano coated implants. Furthermore, the antimicrobial properties of GO nano coating were found to inhibit bacterial colonization and biofilm formation on the implant surface, reducing the risk of implant-associated infections. Conclusion: The findings indicate that GO nano coating holds promise in enhancing the success rate and longevity of dental implants. However, more studies with larger sample sizes, are needed to further strengthen the evidence and determine the long-term effects of GO nano coated dental implants.


Subject(s)
Anti-Infective Agents , Coated Materials, Biocompatible , Dental Implants , Graphite , Osseointegration , Graphite/chemistry , Graphite/pharmacology , Dental Implants/microbiology , Osseointegration/drug effects , Humans , Coated Materials, Biocompatible/pharmacology , Coated Materials, Biocompatible/chemistry , Anti-Infective Agents/pharmacology , Animals , Nanostructures
4.
ACS Appl Mater Interfaces ; 16(33): 43227-43243, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39121390

ABSTRACT

Implant-associated infections and delayed osseointegration are major challenges for the clinical success of titanium implants. To enhance antibacterial effects and promote early osseointegration, we developed a synergistic photothermal (PTT)/photodynamic (PDT) therapy strategy based on near-infrared (NIR) responsive biomimetic micro/nano titanate/TiO2-X heterostructure coatings (KMNW and NaMNS) in situ constructed on the surface of titanium implants. Specifically, KMNW and NaMNS significantly enhanced photothermal conversion capabilities, achieving localized high temperatures of 48-51 °C and promoting substantial amounts of reactive oxygen species production under 808 nm irradiation. In vitro antibacterial experiments demonstrated that KMNW achieved the highest antibacterial rates against Staphylococcus aureus and Escherichia coli, at 98.78 and 98.33% respectively. Moreover, by mimicking the three-dimensional fibrous network of the extracellular matrix during bone healing, both KMNW and NaMNS markedly promoted the proliferation and osteogenic differentiation of osteoblasts. In vivo implantation studies further confirmed these findings, with KMNW and NaMNS exhibiting superior antibacterial performance under NIR irradiation─94.45% for KMNW and 92.66% for NaMNS. Moreover, KMNW and NaMNS also significantly promoted new bone formation and improved osseointegration in vivo. This study presents a promising PTT/PDT therapeutic strategy for dentistry and orthopedics by employing NIR-responsive biomimetic coatings to combat implant-associated infection and accelerate osseointegration.


Subject(s)
Anti-Bacterial Agents , Escherichia coli , Infrared Rays , Osseointegration , Staphylococcus aureus , Titanium , Titanium/chemistry , Titanium/pharmacology , Osseointegration/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Escherichia coli/drug effects , Animals , Staphylococcus aureus/drug effects , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Biomimetic Materials/radiation effects , Prostheses and Implants , Photochemotherapy , Mice , Photothermal Therapy , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Reactive Oxygen Species/metabolism , Microbial Sensitivity Tests , Osteoblasts/drug effects , Osteoblasts/cytology , Osteogenesis/drug effects
5.
Benef Microbes ; 15(5): 527-536, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39147374

ABSTRACT

The aim of this animal study was to compare the primary/secondary stability and micro-CT bone and tissue volumes of implants that were immersed in Limosilactobacillus reuteri, cholecalciferol-D3 (vitamin D) and injectable platelet-rich fibrin (i-PRF) suspensions/solutions before placement in bone. 40 implants (10 in each group) were placed in the iliac crest of 5 sheep. The implants were immersed in L. reuteri, vitamin D or i-PRF solutions for five minutes before placement or left unsoaked as controls. Implant stability was determined by ISQ values and bone volume around implants was histomorphometrically analysed by micro-CT evaluation. At 4 weeks, implants in the L. reuteri group showed the highest secondary stability and 2- and 3D BV/TV values. Both L. reuteri and vitamin D immersed implants had higher osseointegration values compared to the implants in the i-PRF group and controls. There were no statistical differences between L. reuteri and vitamin D immersed implants. Within the limits of the study, the results suggest that immersing implants in L. reuteri or vitamin D suspensions/solutions before implant placement in bone may have beneficial effects on osseointegration.


Subject(s)
Dental Implants , Limosilactobacillus reuteri , Osseointegration , Animals , Dental Implants/microbiology , Limosilactobacillus reuteri/physiology , Sheep , Osseointegration/drug effects , Platelet-Rich Fibrin , X-Ray Microtomography , Cholecalciferol/pharmacology , Cholecalciferol/administration & dosage , Wound Healing/drug effects
6.
Biomed Environ Sci ; 37(7): 739-753, 2024 Jul 20.
Article in English | MEDLINE | ID: mdl-39198238

ABSTRACT

Objective: A high sodium (HS) diet is believed to affect bone metabolism processes. Clarifying its impact on osseointegration of titanium (Ti) implants holds significant implications for postoperative dietary management of implanted patients. Methods: This investigation probed the impact of sodium ions (Na +) on neovascularization and osteogenesis around Ti implants in vivo, utilizing micro-computed tomography, hematoxylin and eosin staining, and immunohistochemical analyses. Concurrently, in vitro experiments assessed the effects of varied Na + concentrations and exposure durations on human umbilical vein endothelial cells (HUVECs) and MC3T3-E1 cells. Results: In vivo, increased dietary sodium (0.8%-6.0%) led to a substantial decline in CD34 positive HUVECs and new bone formation around Ti implants, alongside an increase in inflammatory cells. In vitro, an increase in Na + concentration (140-150 mmol/L) adversely affected the proliferation, angiogenesis, and migration of HUVECs, especially with prolonged exposure. While MC3T3-E1 cells initially exhibited less susceptibility to high Na + concentrations compared to HUVECs during short-term exposure, prolonged exposure to a HS environment progressively diminished their proliferation, differentiation, and osteogenic capabilities. Conclusion: These findings suggest that HS diet had a negative effect on the early osseointegration of Ti implants by interfering with the process of postoperative vascularized bone regeneration.


Subject(s)
Human Umbilical Vein Endothelial Cells , Osseointegration , Titanium , Animals , Osseointegration/drug effects , Humans , Mice , Neovascularization, Physiologic/drug effects , Male , Sodium/metabolism , Osteogenesis/drug effects , Diet
7.
Int J Mol Sci ; 25(16)2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39201648

ABSTRACT

Bisphosphonates are widely used for the treatment of postmenopausal osteoporosis; however, they cause several long-term side effects, necessitating the investigation of local ways to improve osseointegration in compromised bone tissue. The purpose of this study was to evaluate peri-implant bone repair using implants functionalized with zoledronic acid alone (OVX ZOL group, n = 11), zoledronic acid + teriparatide (OVX ZOL + TERI group, n = 11), and zoledronic acid + ruterpy (OVX ZOL + TERPY group, n = 11) compared to the control group (OVX CONV, n = 11). Analyses included computer-assisted microtomography, qualitative histologic analysis, and real-time PCR analysis. Histologically, all functionalized surfaces improved peri-implant repair, with the OVX ZOL + TERI group standing out. Similar results were found in computerized microtomography analysis. In real-time PCR analysis, however, the OVX ZOL and OVX ZOL + TERPY groups showed better results for bone formation, with the OVX ZOL + TERPY group standing out, while there were no statistical differences between the OVX CONV and OVX ZOL + TERI groups for the genes studied at 28 postoperative days. Nevertheless, all functionalized groups showed a reduced rate of bone resorption. In short, all surface functionalization groups outperformed the control group, with overall better results for the OVX ZOL + TERI group.


Subject(s)
Osteoporosis , Zoledronic Acid , Animals , Rats , Female , Zoledronic Acid/administration & dosage , Zoledronic Acid/pharmacology , Osteoporosis/drug therapy , X-Ray Microtomography , Osseointegration/drug effects , Bone Density Conservation Agents/administration & dosage , Bone Density Conservation Agents/pharmacology , Bone Density Conservation Agents/therapeutic use , Drug Delivery Systems/methods , Diphosphonates/administration & dosage , Osteogenesis/drug effects
8.
Int Immunopharmacol ; 140: 112854, 2024 Oct 25.
Article in English | MEDLINE | ID: mdl-39116494

ABSTRACT

The inflammation causes the destroyed osseointegration at the implant-bone interface, significantly increasing the probability of implant loosening in osteoporotic patients. Currently, inhibiting the differentiation of M1 macrophages and the inflammatory response could be a solution to stabilize the microenvironment of implants. Interestingly, some natural products have anti-inflammatory and anti-polarization effects, which could be a promising candidate for stabilizing the implants' microenvironment in osteoporotic patients. This research aims to explore the inhibitory effect of Urolithin B(UB) on macrophage M1 polarization, which ameliorates inflammation, thus alleviating implant instability. We established an osteoporosis mouse model of implant loosening. The mouse tissues were taken out for morphological analysis, staining analysis, and bone metabolic index analysis. In in vitro experiments, RAW264.7 cells were polarized to M1 macrophages using lipopolysaccharide (LPS) and analyzed by immunofluorescence (IF) staining, Western blot (WB), and flow cytometry. The CSP100 plus chip experiments were used to explore the potential mechanisms behind the inhibiting effects of UB. Through observation of these experiments, UB can improve the osseointegration between the implants and femurs in osteoporotic mice and enhance the stability of implants. The UB can inhibit the differentiation of M1 macrophages and local inflammation via inhibiting the phosphorylation of VEGFR2, which can be further proved by the weakened inhibited effects of UB in macrophages with lentivirus-induced overexpression of VEGFR2. Overall, UB can specifically inhibit the activation of VEGFR2, alleviate local inflammation, and improve the stability of implants in osteoporotic mice.


Subject(s)
Cell Differentiation , Coumarins , Macrophages , Osteoporosis , Vascular Endothelial Growth Factor Receptor-2 , Animals , Mice , Coumarins/pharmacology , Coumarins/therapeutic use , Macrophages/immunology , Macrophages/drug effects , Macrophages/metabolism , Cell Differentiation/drug effects , RAW 264.7 Cells , Phosphorylation/drug effects , Osteoporosis/drug therapy , Osteoporosis/immunology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Female , Mice, Inbred C57BL , Down-Regulation/drug effects , Osseointegration/drug effects , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Humans , Inflammation/drug therapy , Disease Models, Animal , Prostheses and Implants
9.
ACS Appl Mater Interfaces ; 16(34): 44451-44466, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39141574

ABSTRACT

Bone and tooth defects can considerably affect the quality of life and health of patients, and orthopedic implants remain the primary method of addressing such defects. However, implant materials cannot coordinate with the immune microenvironment because of their biological inertness, which may lead to implant loosening or failure. Motivated by the microstructure of nacre, we engineered a biomimetic micro/nanoscale topography on a tantalum surface using a straightforward method. This comprised an organized array of tantalum nanotubes arranged in a brick wall structure, with epigallocatechin gallate acting as "mortar." The coating improved the corrosion resistance, biocompatibility, and antioxidant properties. In vitro and in vivo evaluations further confirmed that coatings can create a favorable bone immune microenvironment through the synergistic effects of mechanochemistry and enhance bone integration. This research offers a new viewpoint on the creation of sophisticated functional implants, possessing vast potential for use in the regeneration and repair of bone tissue.


Subject(s)
Osseointegration , Tantalum , Tantalum/chemistry , Osseointegration/drug effects , Animals , Mice , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Catechin/chemistry , Catechin/analogs & derivatives , Catechin/pharmacology , Nanotubes/chemistry , Immunomodulation/drug effects , Surface Properties , Antioxidants/chemistry , Antioxidants/pharmacology , Humans
10.
J Appl Biomater Funct Mater ; 22: 22808000241266665, 2024.
Article in English | MEDLINE | ID: mdl-39129373

ABSTRACT

The pro-inflammatory/anti-inflammatory polarized phenotypes of macrophages (M1/M2) can be used to predict the success of implant integration. Hence, activating and inducing the transformation of immunocytes that promote tissue repair appears to be a highly promising strategy for facilitating osteo-anagenesis. In a previous study, titanium implants were coated with a graphene oxide-hydroxyapatite (GO-HA) nanocomposite via electrophoretic deposition, and the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) was found to be significantly enhanced when the GO content was 2wt%. However, the effectiveness of the GO-HA nanocomposite coating in modifying the in vivo immune microenvironment still remains unclear. In this study, the effects of GO-HA coatings on osteogenesis were investigated based on the GO-HA-mediated immune regulation of macrophages. The HA-2wt%GO nanocomposite coatings exhibited good biocompatibility and favored M2 macrophage polarization. Meanwhile, they could also significantly upregulate IL-10 (anti-inflammatory factor) expression and downregulate TNF-α (pro-inflammatory factor) expression. Additionally, the microenvironment, which was established by M2 macrophages, favored the osteogenesis of BMSCs both in vivo and in vitro. These findings show that the GO-HA nanocomposite coating is a promising surface-modification material. Hence, this study provides a reference for the development of next-generation osteoimmunomodulatory biomaterials.


Subject(s)
Coated Materials, Biocompatible , Durapatite , Graphite , Macrophages , Mesenchymal Stem Cells , Osseointegration , Osteogenesis , Osseointegration/drug effects , Durapatite/chemistry , Durapatite/pharmacology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Macrophages/cytology , Animals , Graphite/chemistry , Graphite/pharmacology , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Osteogenesis/drug effects , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Prostheses and Implants , Immunomodulation/drug effects , Nanocomposites/chemistry , RAW 264.7 Cells , Cell Differentiation/drug effects , Titanium/chemistry , Titanium/pharmacology , Male
11.
J Neuroeng Rehabil ; 21(1): 128, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39085954

ABSTRACT

BACKGROUND: Systems that capture motion under laboratory conditions limit validity in real-world environments. Mobile motion capture solutions such as Inertial Measurement Units (IMUs) can progress our understanding of "real" human movement. IMU data must be validated in each application to interpret with clinical applicability; this is particularly true for diverse populations. Our IMU analysis method builds on the OpenSim IMU Inverse Kinematics toolkit integrating the Versatile Quaternion-based Filter and incorporates realistic constraints to the underlying biomechanical model. We validate our processing method against the reference standard optical motion capture in a case report with participants with transfemoral amputation fitted with a Percutaneous Osseointegrated Implant (POI) and without amputation walking over level ground. We hypothesis that by using this novel pipeline, we can validate IMU motion capture data, to a clinically acceptable degree. RESULTS: Average RMSE (across all joints) between the two systems from the participant with a unilateral transfemoral amputation (TFA) on the amputated and the intact sides were 2.35° (IQR = 1.45°) and 3.59° (IQR = 2.00°) respectively. Equivalent results in the non-amputated participant were 2.26° (IQR = 1.08°). Joint level average RMSE between the two systems from the TFA ranged from 1.66° to 3.82° and from 1.21° to 5.46° in the non-amputated participant. In plane average RMSE between the two systems from the TFA ranged from 2.17° (coronal) to 3.91° (sagittal) and from 1.96° (transverse) to 2.32° (sagittal) in the non-amputated participant. Coefficients of Multiple Correlation (CMC) results between the two systems in the TFA ranged from 0.74 to > 0.99 and from 0.72 to > 0.99 in the non-amputated participant and resulted in 'excellent' similarity in each data set average, in every plane and at all joint levels. Normalized RMSE between the two systems from the TFA ranged from 3.40% (knee level) to 54.54% (pelvis level) and from 2.18% to 36.01% in the non-amputated participant. CONCLUSIONS: We offer a modular processing pipeline that enables the addition of extra layers, facilitates changes to the underlying biomechanical model, and can accept raw IMU data from any vendor. We successfully validate the pipeline using data, for the first time, from a TFA participant using a POI and have proved our hypothesis.


Subject(s)
Amputation, Surgical , Artificial Limbs , Humans , Biomechanical Phenomena , Amputation, Surgical/rehabilitation , Femur/surgery , Osseointegration/physiology , Male , Proof of Concept Study , Amputees/rehabilitation , Walking/physiology , Adult , Bone-Anchored Prosthesis
12.
JBJS Rev ; 12(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38968369

ABSTRACT

¼ The purpose of this article was to review the multidisciplinary, team-based approach necessary for the optimal management of patients with limb loss undergoing osseointegration surgery.¼ In this study, we describe the interdisciplinary process of screening, counseling, and surgical and rehabilitation considerations with an emphasis on principles rather than specific implants or techniques.¼ Integrated perioperative management and long-term surveillance are crucial to ensure the best possible outcomes.¼ We hope this model will service as an implant-agnostic guide to others seeking to development an osseointegration center of excellence.


Subject(s)
Amputation, Surgical , Osseointegration , Humans , Amputation, Surgical/rehabilitation , Patient Care Team , Artificial Limbs
13.
J Orthop Surg Res ; 19(1): 411, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39026349

ABSTRACT

BACKGROUND: This study evaluated the osseointegration potential of functionalised high-performance oxide ceramics (HPOC) in isolation or coated with BMP-2 or RGD peptides in 36 New Zeeland female rabbits using micro-computed tomography (micro CT). The primary outcomes of interest were to assess the amount of ossification evaluating the improvement in the bone volume/ total volume (BV/TV) ratio and trabecular thickness at 6 and 12 weeks. The second outcome of interest was to investigate possible differences in osteointegration between the functionalised silanised HPOC in isolation or coated with Bone Morphogenetic Protein 2 (BMP-2) or RGD peptides. METHODS: 36 adult female New Zealand white rabbits with a minimum weight of three kg were used. One-third of HPOCs were functionalised with silicon suboxide (SiOx), a third with BMP-2 (sHPOC-BMP2), and another third with RGD (sHPOC-RGD). All samples were scanned with a high-resolution micro CT (U-CTHR, MILabs B.V., Houten, The Netherlands) with a reconstructed voxel resolution of 10 µm. MicroCT scans were reconstructed in three planes and processed using Imalytics Preclinical version 2.1 (Gremse-IT GmbH, Aachen, Germany) software. The total volume (TV), bone volume (BV) and ratio BV/TV were calculated within the coating area. RESULTS: BV/TV increased significantly from 6 to 12 weeks in all HPOCs: silanised (P = 0.01), BMP-2 (P < 0.0001), and RGD (P < 0.0001) groups. At 12 weeks, the BMP-2 groups demonstrated greater ossification in the RGD (P < 0.0001) and silanised (P = 0.008) groups. Trabecular thickness increased significantly from 6 to 12 weeks (P < 0.0001). At 12 weeks, BMP-2 promoted greater trabecular thickness compared to the silanised group (P = 0.07), although no difference was found with the RGD (P = 0.1) group. CONCLUSION: Sinalised HPOC in isolation or functionalised with BMP-2 or RGD promotes in vivo osteointegration. The sinalised HOPC functionalised with BMP-2 demonstrated the greatest osseointegration.


Subject(s)
Bone Morphogenetic Protein 2 , Ceramics , Osseointegration , X-Ray Microtomography , Animals , Rabbits , X-Ray Microtomography/methods , Female , Osseointegration/physiology , Oxides , Oligopeptides
14.
Sci Rep ; 14(1): 16441, 2024 07 16.
Article in English | MEDLINE | ID: mdl-39013937

ABSTRACT

Cement mediated peri-implantitis accounts for 1.9-75% of dental implant failures associated with peri-implant diseases. This study evaluated the biological impact of dental cements on osseointegrated implants using Lewis rats. Twenty-two rats were distributed into 6 groups: negative control (NC) soft diet (SD), and hard diet (HD); positive control SD and HD (n = 3); Implant + bio-ceramic Cement (BC) SD and HD which included contralateral Sham sites (n = 5). Titanium implants were placed on either side of the maxillae and allowed to heal for 14 days. Later, both sides of experimental groups underwent a re-entry surgery to simulate clinical cementation. The right side received 0.60 mg of BC. At 14 days post cement application, maxillae were harvested for clinical, microtomographic, and histological evaluations. Clinical and microtomographic evaluations indicated evidence of extensive inflammation and circumferential bone resorption around BC implants in comparison to NC. Histology revealed cement particles surrounded by inflammatory infiltrate in the implant area accompanied by biofilm for SD groups. Both sides of BC indicated intensive bone resorption accompanied by signs of osteolysis when compared to NC. Cemented groups depicted significantly lower bone to implant contact when compared to NC. In conclusion, residual cement extravasation negatively impacted osseointegrated implants after re-entry surgeries.


Subject(s)
Dental Cements , Dental Implants , Peri-Implantitis , X-Ray Microtomography , Animals , Rats , Dental Implants/adverse effects , Peri-Implantitis/pathology , Peri-Implantitis/etiology , Male , Rats, Inbred Lew , Osseointegration , Titanium/adverse effects , Disease Models, Animal , Maxilla/surgery
15.
ACS Appl Mater Interfaces ; 16(30): 39064-39078, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39028896

ABSTRACT

The bacterial infection and poor osseointegration of Ti implants could significantly compromise their applications in bone repair and replacement. Based on the carrier separation ability of the heterojunction and the redox reaction of pseudocapacitive metal oxides, we report an electrically responsive TiO2-SnO2-RuO2 coating with a multilayered heterostructure on a Ti implant. Owing to the band gap structure of the TiO2-SnO2-RuO2 coating, electron carriers are easily enriched at the coating surface, enabling a response to the endogenous electrical stimulation of the bone. With the formation of SnO2-RuO2 pseudocapacitance on the modified surface, the postcharging mode can significantly change the surface chemical state of the coating due to the redox reaction, enhancing the antibacterial ability and osteogenesis-related gene expression of the human bone marrow mesenchymal stem cells. Owing to the attraction for Ca2+, only the negatively postcharged SnO2@RuO2 can promote apatite deposition. The in vivo experiment reveals that the S-SnO2@RuO2-NP could effectively kill the bacteria colonized on the surface and promote osseointegration with the synostosis bonding interface. Thus, negatively charging the electrically responsive coating of TiO2-SnO2-RuO2 is a good strategy to endow modified Ti implants with excellent antibacterial ability and osseointegration.


Subject(s)
Anti-Bacterial Agents , Coated Materials, Biocompatible , Osseointegration , Tin Compounds , Titanium , Titanium/chemistry , Titanium/pharmacology , Osseointegration/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Humans , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Tin Compounds/chemistry , Tin Compounds/pharmacology , Animals , Ruthenium Compounds/chemistry , Ruthenium Compounds/pharmacology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Staphylococcus aureus/drug effects , Surface Properties , Osteogenesis/drug effects
16.
Int J Mol Sci ; 25(13)2024 Jul 03.
Article in English | MEDLINE | ID: mdl-39000425

ABSTRACT

This study investigated the impact of adding hydroxyapatite nanoparticles to implant surfaces treated with zirconia blasting and acid etching (ZiHa), focusing on structural changes and bone healing parameters in low-density bone sites. The topographical characterization of titanium discs with a ZiHa surface and a commercially modified zirconia-blasted and acid-etched surface (Zi) was performed using scanning electron microscopy, profilometry, and surface-free energy. For the in vivo assessment, 22 female rats were ovariectomized and kept for 90 days, after which one implant from each group was randomly placed in each tibial metaphysis of the animals. Histological and immunohistochemical analyses were performed at 14 and 28 days postoperatively (decalcified lab processing), reverse torque testing was performed at 28 days, and histometry from calcified lab processing was performed at 60 days The group ZiHa promoted changes in surface morphology, forming evenly distributed pores. For bone healing, ZiHa showed a greater reverse torque, newly formed bone area, and bone/implant contact values compared to group Zi (p < 0.05; t-test). Qualitative histological and immunohistochemical analyses showed higher features of bone maturation for ZiHa on days 14 and 28. This preclinical study demonstrated that adding hydroxyapatite to zirconia-blasted and acid-etched surfaces enhanced peri-implant bone healing in ovariectomized rats. These findings support the potential for improving osseointegration of dental implants, especially in patients with compromised bone metabolism.


Subject(s)
Durapatite , Nanoparticles , Osseointegration , Surface Properties , Zirconium , Zirconium/chemistry , Animals , Durapatite/chemistry , Durapatite/pharmacology , Female , Rats , Nanoparticles/chemistry , Osseointegration/drug effects , Dental Implants , Titanium/chemistry , Tibia/drug effects , Tibia/surgery , Acid Etching, Dental
17.
Int J Mol Sci ; 25(13)2024 Jul 06.
Article in English | MEDLINE | ID: mdl-39000523

ABSTRACT

The dental implant surface plays a crucial role in osseointegration. The topography and physicochemical properties will affect the cellular functions. In this research, four distinct titanium surfaces have been studied: machined acting (MACH), acid etched (AE), grit blasting (GBLAST), and a combination of grit blasting and subsequent acid etching (GBLAST + AE). Human amniotic mesenchymal (hAMSCs) and epithelial stem cells (hAECs) isolated from the amniotic membrane have attractive stem-cell properties. They were cultured on titanium surfaces to analyze their impact on biological behavior. The surface roughness, microhardness, wettability, and surface energy were analyzed using interferometric microscopy, Vickers indentation, and drop-sessile techniques. The GBLAST and GBLAST + AE surfaces showed higher roughness, reduced hydrophilicity, and lower surface energy with significant differences. Increased microhardness values for GBLAST and GBLAST + AE implants were attributed to surface compression. Cell viability was higher for hAMSCs, particularly on GBLAST and GBLAST + AE surfaces. Alkaline phosphatase activity enhanced in hAMSCs cultured on GBLAST and GBLAST + AE surfaces, while hAECs showed no mineralization signals. Osteogenic gene expression was upregulated in hAMSCs on GBLAST surfaces. Moreover, α2 and ß1 integrin expression enhanced in hAMSCs, suggesting a surface-integrin interaction. Consequently, hAMSCs would tend toward osteoblastic differentiation on grit-blasted surfaces conducive to osseointegration, a phenomenon not observed in hAECs.


Subject(s)
Amnion , Dental Implants , Surface Properties , Titanium , Humans , Titanium/chemistry , Amnion/cytology , Amnion/metabolism , Osteogenesis , Cell Differentiation , Cells, Cultured , Osseointegration , Stem Cells/cytology , Stem Cells/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Cell Survival , Alkaline Phosphatase/metabolism
18.
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
19.
Molecules ; 29(13)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38999015

ABSTRACT

Rehabilitation of fully or partially edentulous patients with dental implants represents one of the most frequently used surgical procedures. The work of Branemark, who observed that a piece of titanium embedded in rabbit bone became firmly attached and difficult to remove, introduced the concept of osseointegration and revolutionized modern dentistry. Since then, an ever-growing need for improved implant materials towards enhanced material-tissue integration has emerged. There is a strong belief that nanoscale materials will produce a superior generation of implants with high efficiency, low cost, and high volume. The aim of this review is to explore the contribution of nanomaterials in implantology. A variety of nanomaterials have been proposed as potential candidates for implant surface customization. They can have inherent antibacterial properties, provide enhanced conditions for osseointegration, or act as reservoirs for biomolecules and drugs. Titania nanotubes alone or in combination with biological agents or drugs are used for enhanced tissue integration in dental implants. Regarding immunomodulation and in order to avoid implant rejection, titania nanotubes, graphene, and biopolymers have successfully been utilized, sometimes loaded with anti-inflammatory agents and extracellular vesicles. Peri-implantitis prevention can be achieved through the inherent antibacterial properties of metal nanoparticles and chitosan or hybrid coatings bearing antibiotic substances. For improved corrosion resistance various materials have been explored. However, even though these modifications have shown promising results, future research is necessary to assess their clinical behavior in humans and proceed to widespread commercialization.


Subject(s)
Dental Implants , Osseointegration , Surface Properties , Titanium , Humans , Animals , Osseointegration/drug effects , Titanium/chemistry , Nanostructures/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry
20.
Int J Implant Dent ; 10(1): 34, 2024 Jul 04.
Article in English | MEDLINE | ID: mdl-38963524

ABSTRACT

Dental implant therapy, established as standard-of-care nearly three decades ago with the advent of microrough titanium surfaces, revolutionized clinical outcomes through enhanced osseointegration. However, despite this pivotal advancement, challenges persist, including prolonged healing times, restricted clinical indications, plateauing success rates, and a notable incidence of peri-implantitis. This review explores the biological merits and constraints of microrough surfaces and evaluates the current landscape of nanofeatured dental implant surfaces, aiming to illuminate strategies for addressing existing impediments in implant therapy. Currently available nanofeatured dental implants incorporated nano-structures onto their predecessor microrough surfaces. While nanofeature integration into microrough surfaces demonstrates potential for enhancing early-stage osseointegration, it falls short of surpassing its predecessors in terms of osseointegration capacity. This discrepancy may be attributed, in part, to the inherent "dichotomy kinetics" of osteoblasts, wherein increased surface roughness by nanofeatures enhances osteoblast differentiation but concomitantly impedes cell attachment and proliferation. We also showcase a controllable, hybrid micro-nano titanium model surface and contrast it with commercially-available nanofeatured surfaces. Unlike the commercial nanofeatured surfaces, the controllable micro-nano hybrid surface exhibits superior potential for enhancing both cell differentiation and proliferation. Hence, present nanofeatured dental implants represent an evolutionary step from conventional microrough implants, yet they presently lack transformative capacity to surmount existing limitations. Further research and development endeavors are imperative to devise optimized surfaces rooted in fundamental science, thereby propelling technological progress in the field.


Subject(s)
Dental Implants , Osseointegration , Surface Properties , Titanium , Humans , Titanium/chemistry , Nanostructures/chemistry , Osteoblasts , Dental Prosthesis Design
SELECTION OF CITATIONS
SEARCH DETAIL