Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 7.720
1.
BMC Oral Health ; 24(1): 501, 2024 May 09.
Article En | MEDLINE | ID: mdl-38725023

BACKGROUND: Releasing of metal ions might implicate in allergic reaction as a negative subsequent of the corrosion of Stainless Steel (SS304) orthodontic wires. The aim of this study was to evaluate the corrosion resistance of zinc-coated (Zn-coated) SS orthodontic wires. METHODS: Zinc coating was applied on SS wires by PVD method. Electrochemical impedance spectroscopy (EIS), Potentiodynamic polarization tests and Tafel analysis methods were used to predict the corrosion behavior of Zn-coated and uncoated SS wires in both neutral and acidic environments. RESULTS: The values of Ecorr ,icorr and Rct ,which were the electrochemical corrosion characteristics, reported better corrosion behavior of Zn-coated SS wires against uncoated ones in both artificial saliva and fluoride-containing environments. Experimental results of the Tafel plot analyses were consistent with that of electrochemical impedance spectroscopy analyses for both biological solutions. CONCLUSION: Applying Zn coating on bare SS orthodontic wire by PVD method might increase the corrosion resistance of the underlying stainless-steel substrate.


Dielectric Spectroscopy , Materials Testing , Orthodontic Wires , Saliva, Artificial , Stainless Steel , Zinc , Corrosion , Stainless Steel/chemistry , Zinc/chemistry , Saliva, Artificial/chemistry , Dental Alloys/chemistry , Coated Materials, Biocompatible/chemistry , Fluorides/chemistry , Hydrogen-Ion Concentration , Humans , Surface Properties , Potentiometry
2.
Cell Mol Biol (Noisy-le-grand) ; 70(3): 67-77, 2024 Mar 31.
Article En | MEDLINE | ID: mdl-38650153

Osteoinduction, and/or osteoconduction, and antibacterial characteristics are prerequisites for achieving successful bone grafting. This study aimed to coat bone allografts with silver nanoparticles and assess their antibacterial activity and biocompatibility compared to uncoated bone allografts. In this study, the bone allografts were coated with varying concentrations of silver nanoparticles (5 mg/l, 10 mg/l, and 50 mg/l) through a simple adsorption technique. Subsequently, the coated samples underwent characterization using SEM, FTIR, EDS, and XRD. The Minimal Inhibitory Concentration (MIC) of the silver nanoparticles was determined against Staphylococcus aureus and Streptococcus mutans. Bacterial growth inhibition was evaluated by measuring turbidity and performing a disk diffusion test. Moreover, qualitative investigation of biofilm formation on the coated bone allograft was conducted using SEM. Following this, MG-63 cell lines, resembling osteoblasts, were cultured on the bone allografts coated with 5 mg/l of silver nanoparticles, as well as on uncoated bone allografts, to assess biocompatibility. The MIC results demonstrated that silver nanoparticles exhibited antimicrobial effects on both microorganisms, inhibiting the growth of isolates at concentrations of 0.78 mg/L for Staphylococcus aureus and 0.39 mg/L for Streptococcus mutans. The bone allografts coated with varying concentrations of silver nanoparticles exhibited significant antibacterial activity against the tested bacteria, completely eradicating bacterial growth and preventing biofilm formation. The osteoblast-like MG-63 cells thrived on the bone allografts coated with 5 mg/l of silver nanoparticles, displaying no significant differences compared to both the uncoated bone allografts and the control group.  Within the limit of this study, it can be concluded that silver nanoparticles have a positive role in controlling graft infection. In addition, simple adsorption technique showed an effective method of coating without overwhelming the healing of the graft.


Allografts , Anti-Bacterial Agents , Biofilms , Bone Substitutes , Metal Nanoparticles , Microbial Sensitivity Tests , Silver , Staphylococcus aureus , Streptococcus mutans , Silver/pharmacology , Silver/chemistry , Metal Nanoparticles/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Streptococcus mutans/drug effects , Staphylococcus aureus/drug effects , Humans , Biofilms/drug effects , Bone Substitutes/chemistry , Bone Substitutes/pharmacology , Allografts/drug effects , Coated Materials, Biocompatible/pharmacology , Coated Materials, Biocompatible/chemistry , Bone Transplantation/methods , Materials Testing , Cell Line
3.
Int J Oral Maxillofac Implants ; 39(2): 235-242, 2024 Apr 24.
Article En | MEDLINE | ID: mdl-38657216

PURPOSE: To analyze the effectiveness of coating of abutments with antimicrobial agents and their influence on the physicochemical and biologic properties of the coated materials. MATERIALS AND METHODS: This work was registered in Open Science Framework (osf.io/6tkcp) and followed the PRISMA protocols. A search of two independent reviewers of articles published up to October 29, 2021, was performed in the Embase, PubMed, Science Direct, and Scopus databases. RESULTS: The databases found a total of 1,474 references. After excluding the duplicates, 1,050 remained. After reading the titles and abstracts and applying the inclusion criteria, 13 articles remained and were read in full. A total of 8 articles were included in this systematic review. Different antimicrobial agents have been used to coat abutments, including graphene oxide, polydopamine, titanium and zirconium nitride, lactoferrin, tetracycline, silver, and doxycycline with varied release times. Titanium-coated silver showed a better antimicrobial agent release time of up to 28 days. Chemical analysis confirmed the presence of antimicrobials on the surface after coating. Different pathogenic microorganisms, such as Streptococcus sanguinis, Streptococcus oralis, and Staphylococcus aureus, were inhibited when in contact with the coated surface. CONCLUSIONS: This review showed that there is still no consensus on which is the better antimicrobial agent and which coated materials have the better performance. However, the association of surface coating of abutments with antimicrobials is feasible and can benefit many patients, which can support their clinical use to favor the healing process and prevent infections that can lead to treatment failure with dental implants.


Anti-Infective Agents , Dental Abutments , Humans , Anti-Infective Agents/pharmacology , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Dental Abutments/microbiology , Surface Properties , Titanium/chemistry
4.
Biomed Mater ; 19(3)2024 Apr 26.
Article En | MEDLINE | ID: mdl-38626780

Wool derived keratin, due to its demonstrated ability to promote bone formation, has been suggested as a potential bioactive material for implant surfaces. The aim of this study was to assess the effects of keratin-coated titanium on osteoblast functionin vitroand bone healingin vivo. Keratin-coated titanium surfaces were fabricated via solvent casting and molecular grafting. The effect of these surfaces on the attachment, osteogenic gene, and osteogenic protein expression of MG-63 osteoblast-like cells were quantifiedin vitro. The effect of these keratin-modified surfaces on bone healing over three weeks using an intraosseous calvaria defect was assessed in rodents. Keratin coating did not affect MG-63 proliferation or viability, but enhanced osteopontin, osteocalcin and bone morphogenetic expressionin vitro. Histological analysis of recovered calvaria specimens showed osseous defects covered with keratin-coated titanium had a higher percentage of new bone area two weeks after implantation compared to that in defects covered with titanium alone. The keratin-coated surfaces were biocompatible and stimulated osteogenic expression in adherent MG-63 osteoblasts. Furthermore, a pilot preclinical study in rodents suggested keratin may stimulate earlier intraosseous calvaria bone healing.


Bone Regeneration , Cell Proliferation , Coated Materials, Biocompatible , Keratins , Osteoblasts , Osteogenesis , Skull , Titanium , Titanium/chemistry , Osteoblasts/drug effects , Osteoblasts/cytology , Osteoblasts/metabolism , Bone Regeneration/drug effects , Animals , Keratins/chemistry , Keratins/metabolism , Humans , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Cell Proliferation/drug effects , Skull/drug effects , Skull/injuries , Osteogenesis/drug effects , Rats , Surface Properties , Male , Cell Line , Cell Adhesion/drug effects , Materials Testing , Cell Survival/drug effects , Rats, Sprague-Dawley
5.
Shanghai Kou Qiang Yi Xue ; 33(1): 6-12, 2024 Feb.
Article Zh | MEDLINE | ID: mdl-38583018

PURPOSE: Bioactive magnesium ions were successfully incorporated into the nanoporous titanium base coating by micro-arc oxidation(MAO), and its physical properties and osteogenic effects were explored. METHODS: Non-magnesium-containing and magnesium-containing titanium porous titanium coatings(MAO, MAO-mg) were prepared by changing the composition of MAO electrolyte and controlling the doping of magnesium in porous titanium coatings. The samples were characterized by scanning electron microscope (SEM), roughness, contact angle and energy dispersive X-ray spectrometer (EDS). Mg2+ release ability of magnesium-doped nanoporous titanium coatings was determined by inductively coupled plasma/optical emission spectrometer(ICP-OES). The structure of the cytoskeleton was determined by live/dead double staining, CCK-8 detection of material proliferation-toxicity, and staining of ß-actin using FITC-phalloidin. The effects of the coating on osteogenic differentiation in vitro were determined by alizarin red (ARS), alkaline phosphatase (ALP) staining and real-time polymerase chain reaction (qRT-PCR). SPSS 25.0 software package was used for statistical analysis. RESULTS: The MAO electrolyte with magnesium ions did not change the surface characteristics of the porous titanium coating. Each group prepared by MAO had similar microporous structure(P>0.05). There was no significant difference in surface roughness and contact angle between MAO treatment group (MAO, MAO-mg)(P>0.05), but significantly higher than that of Ti group (P<0.05). With the passage of cell culture time, MAO-mg group promoted cell proliferation (P<0.05). MAO-mg group was significantly higher than other groups in ALP and ARS staining. The expression of Runx2 mRNA (P<0.05), ALP(P<0.05) and osteocalcin OCN(P<0.05) in MAO-mg group was significantly higher than that in Ti and MAO groups. CONCLUSIONS: MAO successfully prepared magnesium-containing nanoporous titanium coating, and showed a significant role in promoting osteogenic differentiation.


Nanopores , Titanium , Titanium/pharmacology , Magnesium/chemistry , Magnesium/pharmacology , Osteogenesis/genetics , Electrolytes/pharmacology , Ions/pharmacology , Surface Properties , Coated Materials, Biocompatible/pharmacology , Coated Materials, Biocompatible/chemistry
6.
J Biomed Mater Res B Appl Biomater ; 112(5): e35407, 2024 May.
Article En | MEDLINE | ID: mdl-38676956

It would be very beneficial to have a method for joining of ceramics to titanium reliably. Although several techniques have been developed and tested to prevent extensive interfacial chemical reactions in titanium-ceramic systems, the main problem of the inherent brittleness of interfaces was still unsolved. To overcome this problem also in dental applications, we decided to make use of an interlayer material that needs to meet the following requirements: First, it has to be biocompatible, second, it should not melt below the bonding temperatures, and third, it should not react too strongly with titanium, so that its plasticity will be maintained. Considering possible material options only the metals: gold, platinum, palladium, and silver, fulfill the first and second requirements. To find out-without an extensive experimental testing program-which of the four metals fulfills the third requirement best, the combined thermodynamic and reaction kinetic modeling was employed to evaluate how many and how thick reaction layers are formed between the interlayer metals and titanium. With the help of theoretical modeling, it was shown that silver fulfills the last requirement best. However, before starting to test experimentally the effect of the silver layer on the mechanical integrity of dental ceramic/Ag/Ti joints it was decided to make use of mechanical analysis of the three-point bending test, the result of which indicated that the silver layer increases significantly the bond strength of the joints. This result encouraged us to develop a new technique for plating silver on titanium. Subsequently, we executed numerous three-point bending tests, which demonstrated that silver-plated titanium-ceramic joints are much stronger than conventional titanium-ceramic joints. Hence, it can be concluded that the combined thermodynamic, reaction kinetic, and mechanical modeling method can also be a very valuable tool in medical research and development work.


Ceramics , Materials Testing , Silver , Titanium , Titanium/chemistry , Silver/chemistry , Ceramics/chemistry , Coated Materials, Biocompatible/chemistry
7.
Int J Oral Maxillofac Implants ; 39(2): 206-223, 2024 Apr 24.
Article En | MEDLINE | ID: mdl-38657215

PURPOSE: To analyze the available evidence and assess the effect of different implant coatings on healing outcomes. MATERIALS AND METHODS: Using the PICOS strategy, a structured question was formed. A protocol was agreed upon and registered with PROSPERO (no. CRD42022321926). The MEDLINE, Embase, Cochrane Database of Systematic Reviews, Scopus, Web of Science, Pubmed, and ScienceDirect databases were searched using a structured strategy. Study selection was independently carried out in duplicate, first by title and abstract, then by full-text assessment. Quality and risk of bias were independently assessed in duplicate using AMSTAR 2 and ROBIS. Data extraction was independently undertaken in duplicate using a predefined extraction form. RESULTS: The search yielded 11 systematic reviews for inclusion. The most commonly assessed coatings were based on calcium phosphate-including hydroxyapatite (HA), brushite, and bioabsorbable nano-HA-followed by bisphosphonate, then bioactive glass coatings. Included reviews most frequently assessed marginal bone loss (MBL), bone-to-implant contact (BIC), and survival/success rates. There was considerable heterogeneity and small sample sizes. The quality assessment suggested low confidence in the reviews and high risk of bias. CONCLUSIONS: The included reviews provide weak evidence that implant coatings improve osseointegration and reduce MBL following implant placement. There was weak evidence for progressive complications for calcium phosphate coatings. Further research and long-term multicenter controlled clinical trials with improved standardization and control of bias are required to better understand the effects of coating implants.


Coated Materials, Biocompatible , Dental Implantation, Endosseous , Dental Implants , Humans , Calcium Phosphates , Coated Materials, Biocompatible/chemistry , Dental Implantation, Endosseous/methods , Durapatite , Osseointegration/physiology , Wound Healing
8.
Int J Mol Sci ; 25(8)2024 Apr 12.
Article En | MEDLINE | ID: mdl-38673856

Immune response to biomaterials, which is intimately related to their surface properties, can produce chronic inflammation and fibrosis, leading to implant failure. This study investigated the development of magnetic nanoparticles coated with silica and incorporating the anti-inflammatory drug naproxen, aimed at multifunctional biomedical applications. The synthesized nanoparticles were characterized using various techniques that confirmed the presence of magnetite and the formation of a silica-rich bioactive glass (BG) layer. In vitro studies demonstrated that the nanoparticles exhibited bioactive properties, forming an apatite surface layer when immersed in simulated body fluid, and biocompatibility with bone cells, with good viability and alkaline phosphatase activity. Naproxen, either free or encapsulated, reduced nitric oxide production, an inflammatory marker, while the BG coating alone did not show anti-inflammatory effects in this study. Overall, the magnetic nanoparticles coated with BG and naproxen showed promise for biomedical applications, especially anti-inflammatory activity in macrophages and in the bone field, due to their biocompatibility, bioactivity, and osteogenic potential.


Coated Materials, Biocompatible , Glass , Magnetite Nanoparticles , Naproxen , Naproxen/pharmacology , Naproxen/chemistry , Glass/chemistry , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Magnetite Nanoparticles/chemistry , Animals , Mice , Humans , Nitric Oxide/metabolism , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Silicon Dioxide/chemistry , Cell Survival/drug effects , RAW 264.7 Cells , Osteogenesis/drug effects
9.
ACS Appl Mater Interfaces ; 16(17): 21672-21688, 2024 May 01.
Article En | MEDLINE | ID: mdl-38637290

Titanium (Ti) and its alloys are widely used as hard tissue substitutes in dentistry and orthopedics, but their low bioactivity leads to undesirable osseointegration defects in the early osteogenic phase. Surface modification is an important approach to overcome these problems. In the present study, novel magnesium phosphate (MgP) coatings with controllable structures were fabricated on the surface of Ti using the phosphate chemical conversion (PCC) method. The effects of the microstructure on the physicochemical and biological properties of the coatings on Ti were researched. The results indicated that accelerators in PCC solution were important factors affecting the microstructure and properties of the MgP coatings. In addition, the coated Ti exhibited excellent hydrophilicity, high bonding strength, and good corrosion resistance. Moreover, the biological results showed that the MgP coatings could improve the spread, proliferation, and osteogenic differentiation of mouse osteoblast cells (MC3T3-E1) and vascular differentiation of human umbilical vein endothelial cells (HUVECs), indicating that the coated Ti samples had a great effect on promoting osteogenesis and angiogenesis. Overall, this study provided a new research idea for the surface modification of conventional Ti to enhance osteogenesis and angiogenesis in different bone types for potential biomedical applications.


Cell Differentiation , Cell Proliferation , Coated Materials, Biocompatible , Human Umbilical Vein Endothelial Cells , Magnesium Compounds , Neovascularization, Physiologic , Osteogenesis , Phosphates , Titanium , Titanium/chemistry , Titanium/pharmacology , Osteogenesis/drug effects , Animals , Mice , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Phosphates/chemistry , Phosphates/pharmacology , Magnesium Compounds/chemistry , Magnesium Compounds/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Neovascularization, Physiologic/drug effects , Osteoblasts/drug effects , Osteoblasts/cytology , Surface Properties , Cell Line , Angiogenesis
10.
Colloids Surf B Biointerfaces ; 238: 113916, 2024 Jun.
Article En | MEDLINE | ID: mdl-38636438

The ureteral stent is an effective treatment for clinical ureteral stricture following urological surgery, and the functional coating of the stent could effectively inhibit bacterial colonization and other complications. The present review provides an analysis and description of the materials used in ureteral stents and their coatings. Emphasis is placed on the technological advancements of functional coatings, taking into consideration the characteristics of these materials and the properties of their active substances. Furthermore, recent advances in enhancing the therapeutic efficacy of functional coatings are also reviewed. It is anticipated that this article will serve as a valuable reference providing insights for future research development on new drug-loaded ureteral stents.


Coated Materials, Biocompatible , Polymers , Stents , Ureter , Humans , Ureter/surgery , Polymers/chemistry , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Animals
11.
Colloids Surf B Biointerfaces ; 238: 113914, 2024 Jun.
Article En | MEDLINE | ID: mdl-38663310

Combining with various antibacterial mechanisms is the preferred strategy to fabricate coatings with effective antibacterial performance. Herein, Cu2O nanoparticles and dimethyloctadecyl [3-(trimethoxysilyl) propyl] ammonium chloride, a kind of quaternary ammonium salt (QAS), were simultaneously incorporated into a moisture-curable acrylic resin in order to achieve both contact-killing and release-killing abilities for antibacterial coatings. The surface morphology, surface composition and basic properties of the coatings were thoroughly characterized. The antibacterial performance of the coatings was determined by in-vitro bacteriostatic test. Under the constant total mass fraction of antibacterial agents, both Cu2O and QAS content possessed the highest value on the coating surface at Cu2O/QAS mass ratio of 1:1, and correspondingly, the coatings reached sterilizing rate above 99 % against both E. coli and S. loihica, indicating the existence of synergistic effect between Cu2O and QAS. The synergistic antibacterial mechanism of the coatings involved two aspects. Firstly, the combination of contact-killing and release-killing biocides resulted in high bactericidal and antibiofilm activity against different bacteria. Further, the grafting of QAS molecules on the surface of Cu2O particles brought about the spontaneous migration of nanoparticles to the coating surface. The interaction between Cu2O and QAS also inhibited the phase separation of QAS and prolonged the release of Cu2+ at the same time. The coatings, therefore, exhibited stable antibacterial performance at varied service conditions.


Anti-Bacterial Agents , Copper , Escherichia coli , Microbial Sensitivity Tests , Quaternary Ammonium Compounds , Surface Properties , Copper/chemistry , Copper/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Quaternary Ammonium Compounds/chemistry , Quaternary Ammonium Compounds/pharmacology , Escherichia coli/drug effects , Particle Size , Nanoparticles/chemistry , Metal Nanoparticles/chemistry , Acrylic Resins/chemistry , Acrylic Resins/pharmacology , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology
12.
Colloids Surf B Biointerfaces ; 238: 113908, 2024 Jun.
Article En | MEDLINE | ID: mdl-38677153

In response to the critical demand for advancements in coronary artery stents, this study addresses the challenges associated with arterial recoil and restenosis post-angioplasty and the imperative to encourage rapid re-endothelialization for minimizing thrombosis risks. We employed an innovative approach inspired by mussel adhesion, incorporating placental anticoagulant protein (AnnexinV) on stent design. The introduction of a post-translationally modified catecholic amino acid L-3,4-dihydroxyphenylalanine (L-Dopa), mimicking mussel characteristics, allowed for effective surface modification of Stainless steel stents through genetic code engineering in AnnexinV (AnxDopa). The efficacy of AnxDopa was analyzed through microscale thermophoresis and flow cytometry, confirming AnxDopa's exceptional binding with phosphatidylserine and activated platelets. AnxDopa coated stainless steel demonstrates remarkable bio-, hemo-, and immuno-compatibility, preventing smooth muscle cell proliferation, platelet adhesion, and fibrin formation. It acts as an interface between the stent and biological fluid, which facilitates the anticoagulation and rapid endothelialization. Surface modification of SS verified through XPS analysis and contact angle measurement attests to the efficacy of AnxDopa mediated surface modification. The hydrophilic nature of the AnxDopa-coated surface enhanced the endothelialization through increased protein absorption. This approach represents a significant stride in developing coronary stents with improved biocompatibility and reduced restenosis risks, offering valuable contributions to scientific and clinical realms alike.


Coated Materials, Biocompatible , Stents , Humans , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Coronary Vessels/drug effects , Platelet Adhesiveness/drug effects , Anticoagulants/pharmacology , Anticoagulants/chemistry , Surface Properties , Cell Proliferation/drug effects , Stainless Steel/chemistry , Blood Platelets/drug effects , Blood Platelets/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/cytology , Animals , Levodopa/chemistry , Levodopa/pharmacology
13.
Acta Biomater ; 180: 358-371, 2024 May.
Article En | MEDLINE | ID: mdl-38604464

Glaucoma valves (GVs) play an essential role in treating glaucoma. However, fibrosis after implantation has limited their long-term success in clinical applications. In this study, we aimed to develop a comprehensive surface-engineering strategy to improve the biocompatibility of GVs by constructing a microenvironment-regulated and dual-hydrophilic antifouling coating on a GV material (silicone rubber, SR). The coating was based on a superhydrophilic polydopamine (SPD) coating with good short-range superhydrophilicity and antifouling abilities. In addition, SPD coatings contain many phenolic hydroxyl groups that can effectively resist oxidative stress and the inflammatory microenvironment. Furthermore, based on its in situ photocatalytic free-radical polymerization properties, the SPD coating polymerized poly 2-methylacryloxyethylphosphocholine, providing an additional long-range hydrophilic and antifouling effect. The in vitro test results showed that the microenvironment-regulated and dual-hydrophilic coatings had anti-protein contamination, anti-oxidation, anti-inflammation, and anti-fiber proliferation capabilities. The in vivo test results indicated that this coating substantially reduced the fiber encapsulation formation of the SR material by inhibiting inflammation and fibrosis. This design strategy for dual hydrophilic coatings with microenvironmental regulation can provide a valuable reference for the surface engineering design of novel medical implantable devices. STATEMENT OF SIGNIFICANCE: Superhydrophilic polydopamine (SPD) coatings were prepared on silicone rubber (SR) by a two-electron oxidation method. Introduction of pMPC to SPD surface using photocatalytic radical polymerization to obtain a dual-hydrophilic coating. The dual-hydrophilic coating effectively modulates the oxidative and inflammatory microenvironment. This coating significantly reduced protein contamination and adhesion of inflammatory cells and fibroblasts in vitro. The coating-modified SR inhibits inflammatory and fibrosis responses in vivo, promising to serve the glaucoma valves.


Coated Materials, Biocompatible , Glaucoma Drainage Implants , Hydrophobic and Hydrophilic Interactions , Polymers , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Animals , Polymers/chemistry , Polymers/pharmacology , Indoles/chemistry , Indoles/pharmacology , Surface Properties , Humans , Glaucoma/pathology
14.
Acta Biomater ; 180: 183-196, 2024 May.
Article En | MEDLINE | ID: mdl-38604465

The utilization of biodegradable magnesium (Mg) alloys in the fabrication of temporary non-vascular stents is an innovative trend in biomedical engineering. However, the heterogeneous degradation profiles of these biomaterials, together with potential bacterial colonization that could precipitate infectious or stenotic complications, are critical obstacles precluding their widespread clinical application. In pursuit of overcoming these limitations, this study applies the principles of biomimicry, particularly the hydrophobic and anti-fouling characteristics of lotus leaves, to pioneer the creation of nanocomposite coatings. These coatings integrate poly-trimethylene carbonate (PTMC) with covalent organic frameworks (COFs), to modify the stent's surface property. The strategic design of the coating's topography, porosity, and self-polishing capabilities collectively aims to decelerate degradation processes and minimize biological adhesion. The protective qualities of the coatings were substantiated through rigorous testing in both in vitro dynamic bile tests and in vivo New Zealand rabbit choledochal models. Empirical findings from these trials confirmed that the implementation of COF-based nanocomposite coatings robustly fortifies Mg implantations, conferring heightened resistance to both biocorrosion and biofouling as well as improved biocompatibility within bodily environments. The outcomes of this research elucidate a comprehensive framework for the multifaceted strategies against stent corrosion and fouling, thereby charting a visionary pathway toward the systematic conception of a new class of reliable COF-derived surface modifications poised to amplify the efficacy of Mg-based stents. STATEMENT OF SIGNIFICANCE: Biodegradable magnesium (Mg) alloys are widely utilized in temporary stents, though their rapid degradation and susceptibility to bacterial infection pose significant challenges. Our research has developed a nanocomposite coating inspired by the lotus, integrating poly-trimethylene carbonate with covalent organic frameworks (COF). The coating achieved self-polishing property and optimal surface energy on the Mg substrate, which decelerates stent degradation and reduces biofilm formation. Comprehensive evaluations utilizing dynamic bile simulations and implantation in New Zealand rabbit choledochal models reveal that the coating improves the durability and longevity of the stent. The implications of these findings suggest the potential COF-based Mg alloy stent surface treatments and a leap forward in advancing stent performance and endurance in clinical applications.


Absorbable Implants , Coated Materials, Biocompatible , Magnesium , Nanocomposites , Stents , Animals , Rabbits , Magnesium/chemistry , Magnesium/pharmacology , Nanocomposites/chemistry , Corrosion , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Biofouling/prevention & control , Dioxanes/chemistry , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Polymers/chemistry , Polymers/pharmacology , Alloys/chemistry , Alloys/pharmacology
15.
Acta Biomater ; 180: 46-60, 2024 May.
Article En | MEDLINE | ID: mdl-38615811

Blood-contacting medical devices often succumb to thrombosis, limiting their durability and safety in clinical applications. Thrombosis is fundamentally initiated by the nonspecific adsorption of proteins to the material surface, which is strongly governed by thermodynamic factors established by the nature of the interaction between the material surface, surrounding water molecules, and the protein itself. Along these lines, different surface materials (such as polymeric, metallic, ceramic, or composite) induce different entropic and enthalpic changes at the surface-protein interface, with material wettability significantly impacting this behavior. Consequently, protein adsorption on medical devices can be modulated by altering their wettability and surface energy. A plethora of polymeric coating modifications have been utilized for this purpose; hydrophobic modifications may promote or inhibit protein adsorption determined by van der Waals forces, while hydrophilic materials achieve this by mainly relying on hydrogen bonding, or unbalanced/balanced electrostatic interactions. This review offers a cohesive understanding of the thermodynamics governing these phenomena, to specifically aid in the design and selection of hemocompatible polymeric coatings for biomedical applications. STATEMENT OF SIGNIFICANCE: Blood-contacting medical devices often succumb to thrombosis, limiting their durability and safety in clinical applications. A plethora of polymeric coating modifications have been utilized for addressing this issue. This review offers a cohesive understanding of the thermodynamics governing these phenomena, to specifically aid in the design and selection of hemocompatible polymeric coatings for biomedical applications.


Coated Materials, Biocompatible , Polymers , Thermodynamics , Adsorption , Humans , Polymers/chemistry , Coated Materials, Biocompatible/chemistry , Surface Properties , Thrombosis/prevention & control , Animals , Blood Proteins/chemistry , Blood Proteins/metabolism
16.
Colloids Surf B Biointerfaces ; 238: 113880, 2024 Jun.
Article En | MEDLINE | ID: mdl-38581836

In the field of orthopedics, it's crucial to effectively slow down the degradation rate of Mg alloys. This study aims to improve the degradation behavior of Mg-Zn-Ca alloys by electrodepositing fluorohydroxyapatite (FHA). We investigated the microstructure and bond strength of the deposition, as well as degradation and cellular reactions. After 15-30 days of degradation in Hanks solution, FHA deposited alloys showed enhanced stability and less pH change. The strong interfacial bond between FHA and the Mg-Zn-Ca substrate was verified through scratch tests (Critical loads: 10.73 ± 0.014 N in Mg-Zn-0.5Ca alloys). Cellular studies demonstrated that FHA-coated alloys exhibited good cytocompatibility and promoted the growth of MC3T3-E1 cells. Further tests showed FHA-coated alloys owed improved early bone mineralization and osteogenic properties, especially in Mg-Zn-0.5Ca. This research highlighted the potential of FHA-coated Mg-Zn-0.5Ca alloys in orthopedics applications.


Alloys , Calcium , Magnesium , Zinc , Alloys/chemistry , Alloys/pharmacology , Corrosion , Animals , Zinc/chemistry , Zinc/pharmacology , Magnesium/chemistry , Mice , Calcium/chemistry , Calcium/metabolism , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Surface Properties , Materials Testing , Cell Proliferation/drug effects , Hydroxyapatites/chemistry , Cell Line , Durapatite/chemistry , Durapatite/pharmacology
17.
Int J Mol Sci ; 25(6)2024 Mar 13.
Article En | MEDLINE | ID: mdl-38542241

As the population ages, the number of patients undergoing total hip arthroplasty (THA) and total knee arthroplasty (TKA) continues to increase. Infections after primary arthroplasty are rare but have high rates of morbidity and mortality, as well as enormous financial implications for healthcare systems. Numerous methods including the use of superhydrophobic coatings, the incorporation of antibacterial agents, and the application of topographical treatments have been developed to reduce bacterial attachment to medical devices. However, most of these methods require complex manufacturing processes. Thus, the main purpose of this study was to apply biocoatings to titanium (Ti) surfaces to increase their infection resistance and osteoconductivity via simple processes, without organic reagents. We modified titanium surfaces with a combination of aminomalononitrile (AMN) and an antibiotic-loaded mesoporous bioactive glass (MBG) and evaluated both the antibacterial effects of the coating layer and its effect on osteoblast proliferation and differentiation. The properties of the modified surface, such as the hydrophilicity, roughness, and surface morphology, were characterized via contact angle measurements, atomic force microscopy, and scanning electron microscopy. The cell proliferation reagent WST-1 assay and the alkaline phosphatase (ALP) assay were used to determine the degrees of adhesion and differentiation, respectively, of the MG-63 osteoblast-like cells on the surface. Antimicrobial activity was evaluated by examining the survival rate and inhibition zone of Escherichia coli (E. coli). The AMN coating layer reduced the water contact angle (WCA) of the titanium surface from 87° ± 2.5° to 53° ± 2.3° and this change was retained even after immersion in deionized water for five weeks, demonstrating the stability of the AMN coating. Compared with nontreated titanium and polydopamine (PDA) coating layers, the AMN surface coating increased MG-63 cell attachment, spreading, and early ALP expression; reduced E. coli adhesion; and increased the percentage of dead bacteria. In addition, the AMN coating served as an adhesion layer for the subsequent deposition of MBG-containing antibiotic nanoparticles. The synergistic effects of the AMN layer and antibiotics released from the MBG resulted in an obvious E. coli inhibition zone that was not observed in the nontreated titanium group.


Escherichia coli , Titanium , Humans , Titanium/pharmacology , Titanium/chemistry , Surface Properties , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Hydrophobic and Hydrophilic Interactions , Bacteria , Coated Materials, Biocompatible/pharmacology , Coated Materials, Biocompatible/chemistry , Osteoblasts
18.
Int J Mol Sci ; 25(6)2024 Mar 16.
Article En | MEDLINE | ID: mdl-38542348

Despite a long period of application of metal implants, carbon-carbon medical composites are also widely used for bone defect prosthesis in surgery, dentistry, and oncology. Such implants might demonstrate excellent mechanical properties, but their biocompatibility and integration efficiency into the host should be improved. As a method of enhancing, the electrophoretic deposition of fine-dispersed hydroxyapatite (HAp) on porous carbon substrates might be recommended. With electron microscopy, energy dispersion X-ray and Raman spectroscopy, and X-ray diffraction, we found that the deposition and subsequent heat post-treatment (up to the temperature of 400 °C for 1 h) did not lead to any significant phase and chemical transformations of raw non-stoichometric HAp. The Ca/P ratio was ≈1.51 in the coatings. Their non-toxicity, cyto- and biocompatibility were confirmed by in vitro and in vivo studies and no adverse reactions and side effects had been detected in the test. The proposed coating and subsequent heat treatment procedures provided improved biological responses in terms of resorption and biocompatibility had been confirmed by histological, magnetic resonance and X-ray tomographic ex vivo studies on the resected implant-containing biopsy samples from the BDF1 mouse model. The obtained results are expected to be useful for modern medical material science and clinical applications.


Carbon , Coated Materials, Biocompatible , Animals , Mice , Carbon/chemistry , Coated Materials, Biocompatible/chemistry , Calcium Phosphates , Durapatite/chemistry , Prostheses and Implants , X-Ray Diffraction
19.
Biotechnol J ; 19(3): e2300464, 2024 Mar.
Article En | MEDLINE | ID: mdl-38509814

The present study evaluates the corrosion behavior of poly[xylitol-(1,12-dodecanedioate)](PXDD)-HA coated porous iron (PXDD140/HA-Fe) and its cell-material interaction aimed for temporary bone scaffold applications. The physicochemical analyses show that the addition of 20 wt.% HA into the PXDD polymers leads to a higher crystallinity and lower surface roughness. The corrosion assessments of the PXDD140/HA-Fe evaluated by electrochemical methods and surface chemistry analysis indicate that HA decelerates Fe corrosion due to a lower hydrolysis rate following lower PXDD content and being more crystalline. The cell viability and cell death mode evaluations of the PXDD140/HA-Fe exhibit favorable biocompatibility as compared to bare Fe and PXDD-Fe scaffolds owing to HA's bioactive properties. Thus, the PXDD140/HA-Fe scaffolds possess the potential to be used as a biodegradable bone implant.


Coated Materials, Biocompatible , Xylitol , Materials Testing , Coated Materials, Biocompatible/chemistry , Corrosion , Porosity , Iron , Durapatite/chemistry
20.
J Biomed Mater Res B Appl Biomater ; 112(4): e35403, 2024 Apr.
Article En | MEDLINE | ID: mdl-38520706

For decades, titanium implants have shown impressive advantages in bone repair. However, the preparation of implants with excellent antimicrobial properties as well as better osseointegration ability remains difficult for clinical application. In this study, black phosphorus nanosheets (BPNSs) were doped into hydroxyapatite (HA) coatings using electrophoretic deposition. The coatings' surface morphology, roughness, water contact angle, photothermal properties, and antibacterial properties were investigated. The BP/HA coating exhibited a surface roughness of 59.1 nm, providing an ideal substrate for cell attachment and growth. The water contact angle on the BP/HA coating was measured to be approximately 8.55°, indicating its hydrophilic nature. The BPNSs demonstrated efficient photothermal conversion, with a temperature increase of 42.2°C under laser irradiation. The BP/HA composite coating exhibited a significant reduction in bacterial growth, with inhibition rates of 95.6% and 96.1% against Staphylococcus aureus and Escherichia coli. In addition, the cytocompatibility of the composite coating was evaluated by cell adhesion, CCK8 and AM/PI staining; the effect of the composite coating in promoting angiogenesis was assessed by scratch assay, transwell assay, and protein blotting; and the osteoinductivity of the composite coating was evaluated by alkaline phosphatase assay, alizarin red staining, and Western blot. The results showed that the BP/HA composite coating exhibited superior performance in promoting biological functions such as cell proliferation and adhesion, antibacterial activity, osteogenic differentiation, and angiogenesis, and had potential applications in vascularized bone regeneration.


Durapatite , Titanium , Durapatite/pharmacology , Durapatite/chemistry , Titanium/pharmacology , Titanium/chemistry , Osseointegration , Osteogenesis , Coated Materials, Biocompatible/pharmacology , Coated Materials, Biocompatible/chemistry , Anti-Bacterial Agents/pharmacology , Water/pharmacology , Surface Properties
...