Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Funct Biomater ; 15(7)2024 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-39057313

RESUMO

Magnesium alloys are some of the most convenient biodegradable materials for bone fracture treatment due to their tailorable degradation rate, biocompatibility, and mechanical properties resembling those of bone. Despite the fact that magnesium-based implants and ZX00 (Mg-0.45Zn-0.45Ca in wt.%), in particular, have been shown to have suitable degradation rates and good osseointegration, knowledge gaps remain in our understanding of the impact of their degradation properties on the bone's ultrastructure. Bone is a hierarchically structured material, where not only the microstructure but also the ultrastructure are important as properties like the local mechanical response are determined by it. This study presents the first comparative analysis of bone ultrastructure parameters with high spatial resolution around ZX00 and Ti implants after 6, 12, and 24 weeks of healing. The mineralization was investigated, revealing a significant decrease in the lattice spacing of the (002) Bragg's peak closer to the ZX00 implant in comparison to Ti, while no significant difference in the crystallite size was observed. The hydroxyapatite platelet thickness and osteon density demonstrated a decrease closer to the ZX00 implant interface. Correlative indentation and strain maps obtained by scanning X-ray diffraction measurements revealed a higher stiffness and faster mechanical adaptation of the bone surrounding Ti implants as compared to the ZX00 ones. Thus, the results suggest the incorporation of Mg2+ ions into the bone ultrastructure, as well as a lower degree of remodeling and stiffness of the bone in the presence of ZX00 implants than Ti.

2.
Bioact Mater ; 39: 456-478, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38873086

RESUMO

Due to matching biomechanical properties and significant biological activity, Mg-based implants present great potential in orthopedic applications. In recent years, the biocompatibility and therapeutic effect of magnesium-based implants have been widely investigated in trauma repair. In contrast, the R&D work of Mg-based implants in spinal fusion is still limited. This review firstly introduced the general background for Mg-based implants. Secondly, the mechanical properties and degradation behaviors of Mg and its traditional and novel alloys were reviewed. Then, different surface modification techniques of Mg-based implants were described. Thirdly, this review comprehensively summarized the biological pathways of Mg degradation to promote bone formation in neuro-musculoskeletal circuit, angiogenesis with H-type vessel formation, osteogenesis with osteoblasts activation and chondrocyte ossification as an integrated system. Fourthly, this review followed the translation process of Mg-based implants via updating the preclinical studies in fracture fixation, sports trauma repair and reconstruction, and bone distraction for large bone defect. Furthermore, the pilot clinical studies were involved to demonstrate the reliable clinical safety and satisfactory bioactive effects of Mg-based implants in bone formation. Finally, this review introduced the background of spine fusion surgeryand the challenges of biological matching cage development. At last, this review prospected the translation potential of a hybrid Mg-PEEK spine fusion cage design.

3.
Bioact Mater ; 38: 374-383, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38770429

RESUMO

Implant-related osteomyelitis is a formidable hurdle in the clinical setting and is characterized by inflammation, infection, and consequential bone destruction. Therefore, effective reactive oxygen species (ROS) scavenging, bacterial killing, and subsequent bone tissue repair are urgently needed for the treatment of difficult-to-heal osteomyelitis. Herein, we utilized the eddy-thermal effect of magnesium (Mg) implants under an alternating magnetic field (AMF) for the controlled release of H2 gas and ions (OH- and Mg2+) for the treatment of osteomyelitis. H2 released by Mg rods under AMFs effectively scavenged cytotoxic ROS, exhibiting anti-inflammatory effects and consequently disrupting the environment of bacterial infections. In addition, the OH- hindered the energy metabolism of bacteria by effectively neutralizing protons within the microenvironment. Moreover, H2 impaired the permeability of bacterial membranes and expedited the damage induced by OH-. This synergistic AMF-induced H2 and proton depletion treatment approach not only killed both gram-negative and gram-positive bacteria but also effectively treated bacterial infections (abscesses and osteomyelitis). Moreover, Mg2+ released from the Mg rods enhanced and accelerated the process of bone osteogenesis. Overall, our work cleverly exploited the eddy-thermal effect and chemical activity of Mg implants under AMFs, aiming to eliminate the inflammatory environment and combat bacterial infections by the simultaneous release of H2, OH-, and Mg2+, thereby facilitating tissue regeneration. This therapeutic strategy achieved multiple benefits in one, thus presenting a promising avenue for clinical application.

4.
ACS Biomater Sci Eng ; 10(2): 1062-1076, 2024 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-38245905

RESUMO

Magnesium is a revolutionary biomaterial for orthopedic implants, owing to its eminent mechanical properties and biocompatibility. However, its uncontrolled degradation rate remains a severe challenge for its potential applications. In this study, we developed a self-healing micro arc oxidation (MAO) and dicalcium phosphate dihydrate (DCPD) double-passivated coating on a magnesium membrane (Mg-MAO/DCPD) and investigated its potential for bone-defect healing. The Mg-MAO/DCPD membrane possessed a feasible self-repairing ability and good cytocompatibility. In vitro degradation experiments showed that the Mg contents on the coating surface were 0.3, 3.8, 4.1, 6.1, and 7.9% when the degradation times were 0, 1, 2, 3, and 4 weeks, respectively, exhibiting available corrosion resistance. The slow and sustained release of Mg2+ during the degradation process activated extracellular matrix proteins for bone regeneration, accelerating osteogenic differentiation of human bone marrow mesenchymal stem cells (hBMSCs). The extract solutions of Mg-MAO/DCPD considerably promoted the activation of the Wnt and PI3K/AKT signaling pathways. Furthermore, the evaluation of the rat skull defect model manifested the outstanding bone-healing efficiency of the Mg-MAO/DCPD membrane. Taken together, the Mg-MAO/DCPD membrane demonstrates an optimized degradation rate and excellent bioactivity and is believed to have great application prospects in bone tissue engineering.


Assuntos
Fosfatos de Cálcio , Materiais Revestidos Biocompatíveis , Magnésio , Ratos , Humanos , Animais , Magnésio/farmacologia , Materiais Revestidos Biocompatíveis/farmacologia , Osteogênese , Fosfatidilinositol 3-Quinases
5.
Bioact Mater ; 30: 154-168, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37575877

RESUMO

The utilization of biodegradable magnesium (Mg)-based implants for restoration of bone function following trauma represents a transformative approach in orthopaedic application. One such alloy, magnesium-10 weight percent gadolinium (Mg-10Gd), has been specifically developed to address the rapid degradation of Mg while enhancing its mechanical properties to promote bone healing. Previous studies have demonstrated that Mg-10Gd exhibits favorable osseointegration; however, it exhibits distinct ultrastructural adaptation in comparison to conventional implants like titanium (Ti). A crucial aspect that remains unexplored is the impact of Mg-10Gd degradation on the bone microarchitecture. To address this, we employed hierarchical three-dimensional imaging using synchrotron radiation in conjunction with image-based finite element modelling. By using the methods outlined, the vascular porosity, lacunar porosity and the lacunar-canaliculi network (LCN) morphology of bone around Mg-10Gd in comparison to Ti in a rat model from 4 weeks to 20 weeks post-implantation was investigated. Our investigation revealed that within our observation period, the degradation of Mg-10Gd implants was associated with significantly lower (p < 0.05) lacunar density in the surrounding bone, compared to Ti. Remarkably, the LCN morphology and the fluid flow analysis did not significantly differ for both implant types. In summary, a more pronounced lower lacunae distribution rather than their morphological changes was detected in the surrounding bone upon the degradation of Mg-10Gd implants. This implies potential disparities in bone remodelling rates when compared to Ti implants. Our findings shed light on the intricate relationship between Mg-10Gd degradation and bone microarchitecture, contributing to a deeper understanding of the implications for successful osseointegration.

6.
J Funct Biomater ; 14(5)2023 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-37233351

RESUMO

In the present work, the surface of a biodegradable Mg alloy was modified to create porous diatomite biocoatings using the method of micro-arc oxidation. The coatings were applied at process voltages in the range of 350-500 V. We have studied the influence of the addition of ZrO2 microparticles on the structure and properties of diatomite-based protective coatings for Mg implants. The structure and properties of the resulting coatings were examined using a number of research methods. It was found that the coatings have a porous structure and contain ZrO2 particles. The coatings were mostly characterized by pores less than 1 µm in size. However, as the voltage of the MAO process increases, the number of larger pores (5-10 µm in size) also increases. However, the porosity of the coatings varied insignificantly and amounted to 5 ± 1%. It has been revealed that the incorporation of ZrO2 particles substantially affects the properties of diatomite-based coatings. The adhesive strength of the coatings has increased by approximately 30%, and the corrosion resistance has increased by two orders of magnitude compared to the coatings without zirconia particles.

7.
ACS Appl Mater Interfaces ; 14(1): 104-122, 2022 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-34958199

RESUMO

In orthopedic surgery, metals are preferred to support or treat damaged bones due to their high mechanical strength. However, the necessity for a second surgery for implant removal after healing creates problems. Therefore, biodegradable metals, especially magnesium (Mg), gained importance, although their extreme susceptibility to galvanic corrosion limits their applications. The focus of this study was to control the corrosion of Mg and enhance its biocompatibility. For this purpose, surfaces of magnesium-calcium (MgCa1) alloys were modified with calcium phosphate (CaP) or CaP doped with zinc (Zn) or gallium (Ga) via microarc oxidation. The effects of surface modifications on physical, chemical, and mechanical properties and corrosion resistance of the alloys were studied using surface profilometry, goniometry, scanning electron microscopy (SEM), energy-dispersive X-ray spectroscopy (EDS), nanoindentation, and electrochemical impedance spectroscopy (EIS). The coating thickness was about 5-8 µm, with grain sizes of 43.1 nm for CaP coating and 28.2 and 58.1 nm for Zn- and Ga-doped coatings, respectively. According to EIS measurements, the capacitive response (Yc) decreased from 11.29 to 8.72 and 0.15 Ω-1 cm-2 sn upon doping with Zn and Ga, respectively. The Ecorr value, which was -1933 mV for CaP-coated samples, was found significantly electropositive at -275 mV for Ga-doped ones. All samples were cytocompatible according to indirect tests. In vitro culture with Saos-2 cells led to changes in the surface compositions of the alloys. The numbers of cells attached to the Zn-doped (2.6 × 104 cells/cm2) and Ga-doped (6.3 × 104 cells/cm2) coatings were higher than that on the surface of the undoped coating (1.0 × 103 cells/cm2). Decreased corrosivity and enhanced cell affinity of the modified MgCa alloys (CaP coated and Zn and Ga doped, with Ga-doped ones having the greatest positive effect) make them novel and promising candidates as biodegradable metallic implant materials for the treatment of bone damages and other orthopedic applications.


Assuntos
Ligas/química , Fosfatos de Cálcio/química , Materiais Revestidos Biocompatíveis/química , Implantes Absorvíveis , Ligas/toxicidade , Animais , Cálcio/química , Cálcio/toxicidade , Fosfatos de Cálcio/toxicidade , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Materiais Revestidos Biocompatíveis/toxicidade , Corrosão , Módulo de Elasticidade , Gálio/química , Gálio/toxicidade , Humanos , Magnésio/química , Magnésio/toxicidade , Teste de Materiais , Camundongos , Molhabilidade , Zinco/química , Zinco/toxicidade
8.
Bioact Mater ; 8: 140-152, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34541392

RESUMO

Magnesium (Mg) alloys that have both antibacterial and osteogenic properties are suitable candidates for orthopedic implants. However, the fabrication of ideal Mg implants suitable for bone repair remains challenging because it requires implants with interconnected pore structures and personalized geometric shapes. In this study, we fabricated a porous 3D-printed Mg-Nd-Zn-Zr (denoted as JDBM) implant with suitable mechanical properties using selective laser melting technology. The 3D-printed JDBM implant exhibited cytocompatibility in MC3T3-E1 and RAW267.4 cells and excellent osteoinductivity in vitro. Furthermore, the implant demonstrated excellent antibacterial ratios of 90.0% and 92.1% for methicillin-resistant S. aureus (MRSA) and Escherichia coli, respectively. The 3D-printed JDBM implant prevented MRSA-induced implant-related infection in a rabbit model and showed good in vivo biocompatibility based on the results of histological evaluation, blood tests, and Mg2+ deposition detection. In addition, enhanced inflammatory response and TNF-α secretion were observed at the bone-implant interface of the 3D-printed JDBM implants during the early implantation stage. The high Mg2+ environment produced by the degradation of 3D-printed JDBM implants could promote M1 phenotype of macrophages (Tnf, iNOS, Ccl3, Ccl4, Ccl5, Cxcl10, and Cxcl2), and enhance the phagocytic ability of macrophages. The enhanced immunoregulatory effect generated by relatively fast Mg2+ release and implant degradation during the early implantation stage is a potential antibacterial mechanism of Mg-based implant. Our findings indicate that 3D-printed porous JDBM implants, having both antibacterial property and osteoinductivity, hold potential for future orthopedic applications.

9.
J Biomed Mater Res B Appl Biomater ; 110(4): 898-909, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34846806

RESUMO

Implants of different material classes have been used for the reconstruction of damaged hard and soft tissue for decades. The aim is to increase and subsequently maintain the patient's quality of life through implantation. In service, most implants are subjected to cyclic loading, which must be taken particularly into consideration, since the fatigue strength is far below the yield and tensile strength. Inaccurate estimation of the structural strength of implants due to the consideration of yield or tensile strength leads to a miscalculation of the implant's fatigue strength and lifetime, and therefore, to its unexpected early fatigue failure. Thus, fatigue failure of an implant based on overestimated performance capability represents acute danger to human health. The determination of fatigue strength by corresponding tests investigating various stress amplitudes is time-consuming and cost-intensive. This study summarizes four investigation series on the fatigue behavior of different implant materials and components, following a standard and an in vitro short-time testing procedure, which evaluates the material reaction in one enhanced test set-up. The test set-up and the applied characterization methods were adapted to the respective application of the implant with the aim to simulate the surrounding of the human body with laboratory in vitro tests only. It could be shown that by using the short-time testing method the number of tests required to determine the fatigue strength can be drastically reduced. In future, therefore it will be possible to exclude unsuitable implant materials or components before further clinical investigations by using a time-efficient and application-oriented testing method.


Assuntos
Implantes Dentários , Qualidade de Vida , Análise do Estresse Dentário , Humanos , Técnicas In Vitro , Teste de Materiais , Estresse Mecânico , Resistência à Tração , Titânio
10.
Acta Biomater ; 134: 804-817, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34333163

RESUMO

The nanostructural adaptation of bone is crucial for its biocompatibility with orthopedic implants. The bone nanostructure also determines its mechanical properties and performance. However, the bone's temporal and spatial nanoadaptation around degrading implants remains largely unknown. Here, we present insights into this important bone adaptation by applying scanning electron microscopy, elemental analysis, and small-angle X-ray scattering tensor tomography (SASTT). We extend the novel SASTT reconstruction method and provide a 3D scattering reciprocal space map per voxel of the sample's volume. From this reconstruction, parameters such as the thickness of the bone mineral particles are quantified, which provide additional information on nanostructural adaptation of bone during healing. We selected a rat femoral bone and a degrading ZX10 magnesium implant as model system, and investigated it over the course of 18 months, using a sham as control. We observe that the bone's nanostructural adaptation starts with an initially fast interfacial bone growth close to the implant, which spreads by a re-orientation of the nanostructure in the bone volume around the implant, and is consolidated in the later degradation stages. These observations reveal the complex bulk bone-implant interactions and enable future research on the related biomechanical bone responses. STATEMENT OF SIGNIFICANCE: Traumatic bone injuries are among the most frequent causes of surgical treatment, and often require the placement of an implant. The ideal implant supports and induces bone formation, while being mechanically and chemically adapted to the bone structure, ensuring a gradual load transfer. While magnesium implants fulfill these requirements, the nanostructural changes during bone healing and implant degradation remain not completely elucidated. Here, we unveil these processes in rat femoral bones with ZX10 magnesium implants and show different stages of bone healing in such a model system.


Assuntos
Magnésio , Próteses e Implantes , Animais , Osso e Ossos , Magnésio/farmacologia , Ratos , Tomografia por Raios X , Raios X
11.
Bioact Mater ; 6(5): 1223-1229, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33210020

RESUMO

Magnesium (Mg) and its alloys are promising biodegradable materials for orthopedic applications. However, one of the major problems is their rapid degradation rate with quick evolution of hydrogen gas. To overcome this problem, calcium phosphate (CaP) coatings have been used to improve the degradation resistance and the biocompatibility of Mg materials. This study focuses on the comparison and correlation of the in vitro and in vivo degradation and biocompatibility behaviors of these materials. A CaP coating consisting of dicalcium phosphate dihydrate (DCPD) was deposited on an AZ60 Mg alloy by the chemical conversion method. Then, the in vitro degradation testing including electrochemical and immersion tests, and in vivo implantation of the CaP coated Mg alloy were conducted to compare the degradation behaviors. Next, the in vitro cell behavior and in vivo bone tissue response were also compared on both uncoated and CaP-coated Mg samples. Data showed that the CaP coating provided the Mg alloy with significantly better biodegradation behavior and biocompatibility. The in vitro and in vivo biocompatibility tests exhibited good consistency while not the case for biodegradation. Results showed that the in vitro electrochemical test could be a quick screening tool for the biodegradation rate, while the in vitro immersion degradation rate was often 2-4 folds faster than the in vivo degradation rate.

12.
Acta Biomater ; 116: 426-437, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32890748

RESUMO

Due to its degradability, magnesium holds potential for the application as a base material for local treatment systems. Particularly for the therapy of severe brain-related diseases, local approaches are advantageous. To confirm the suitability of magnesium as a material for neural implants, information on the interaction of brain cells with magnesium is essential. Initial steps of such an evaluation need to include not only cytocompatibility tests but also the analysis of the in vitro material degradation to predict in vivo material performance. Considering the sensitivity and functional importance of neural tissue, an in-depth understanding of the processes involved is of particular relevance. Here, we investigate the influence of four different brain cell types and fibroblasts on magnesium degradation in direct material contact. Our findings indicate cell type as well as cell density-dependent degradation behavior. Metabolic activity (lactate content) appears to be crucial for degradation promotion. Extracellular matrix composition, distribution, and matrix/cell ratios are analyzed to elucidate the cell-material interactions further. Statement of Significance Thanks to their degradability, magnesium (Mg)-based materials could be promising biomaterials for local ion or even drug delivery strategies for the treatment of severe brain-related diseases. To confirm the suitability of Mg as a neural implant material, information on the interaction of brain cells with Mg is essential. Initial steps of such an evaluation need to include cytocompatibility tests and the analysis of the in vitro material degradation to predict in vivo material performance. The present study provides data on the influence of different brain cell types on Mg degradation in direct material contact. Our findings indicate cell type and cell density-dependent degradation behavior, and elucidate the role of cell metabolites and extracellular matrix molecules in the underlying degradation mechanisms.


Assuntos
Magnésio , Preparações Farmacêuticas , Ligas , Encéfalo , Matriz Extracelular , Magnésio/farmacologia , Teste de Materiais
13.
Materials (Basel) ; 13(6)2020 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-32183231

RESUMO

Magnesium (Mg) alloys are being investigated as a biodegradable metallic biomaterial because of their mechanical property profile, which is similar to the human bone. However, implants based on Mg alloys are corroded quickly in the body before the bone fracture is fully healed. Therefore, we aimed to reduce the corrosion rate of Mg using a double protective layer. We used a magnesium-aluminum-zinc alloy (AZ91) and treated its surface with micro-arc oxidation (MAO) technique to first form an intermediate layer. Next, a bioceramic nanocomposite composed of diopside, bredigite, and fluoridated hydroxyapatite (FHA) was coated on the surface of MAO treated AZ91 using the electrophoretic deposition (EPD) technique. Our in vivo results showed a significant enhancement in the bioactivity of the nanocomposite coated AZ91 implant compared to the uncoated control implant. Implantation of the uncoated AZ91 caused a significant release of hydrogen bubbles around the implant, which was reduced when the nanocomposite coated implants were used. Using histology, this reduction in the corrosion rate of the coated implants resulted in an improved new bone formation and reduced inflammation in the interface of the implants and the surrounding tissue. Hence, our strategy using a MAO/EPD of a bioceramic nanocomposite coating (i.e., diopside-bredigite-FHA) can significantly reduce the corrosion rate and improve the bioactivity of the biodegradable AZ91 Mg implant.

14.
Mater Sci Eng C Mater Biol Appl ; 108: 110389, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31923970

RESUMO

Two miniature pig models to assess safety and performance of degradable osteosynthesis implants are presented. Both models provide multiple implantation sites with human size implants. In the first model, different types of magnesium plates and screws for fracture fixation were used to study local and systemic safety aspects in 14 Göttingen minipigs. Implant degradation, gas release and accumulation of alloying elements in organs were assessed for non-coated and plasmaelectrolytic coated magnesium implants and compared to the titanium reference. The observed implant degradation was mostly uniform and did not seem to depend on the implantation site and implant condition. The coating was effective in delaying initial gas release and degradation. No rare earth alloying elements could be detected in local lymph nodes, kidneys, livers or spleens. In the second model with Göttingen und Yucatan minipigs, full osteotomies were inflicted to four different anatomical sites and treated with magnesium plates and screws to assess fracture healing performance. Two Göttingen pilot minipigs showed promising results including a mandible osteosynthesis which healed within 6 weeks. The subsequent study was compromised by the more massive jaws of the used Yucatan minipigs. Three out of seven animals had to be sacrificed within two months as the stability of magnesium and titanium reference implants in the mandible was surpassed. In conclusion, the resorbable magnesium implants showed promising in vivo properties. For the analysis of human standard sized implants under full chewing load conditions, lighter Göttingen minipigs were more suitable than heavier Yucatan minipigs.


Assuntos
Implantes Absorvíveis , Consolidação da Fratura/fisiologia , Animais , Placas Ósseas , Parafusos Ósseos , Feminino , Fixação Interna de Fraturas/métodos , Humanos , Magnésio , Masculino , Mandíbula/cirurgia , Osteotomia Mandibular , Metacrilatos , Costelas/cirurgia , Suínos , Porco Miniatura , Tionas
15.
Acta Biomater ; 101: 637-645, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31734411

RESUMO

Magnesium alloys are increasingly researched as temporary biodegradable metal implants in bone applications due to their mechanical properties which are more similar to bone than conventional implant metals and the fact that Magnesium occurs naturally within the body. However, the degradation processes in vivo and in particular the interaction of the bone with the degrading material need to be further investigated. In this study we are presenting the first quantitative comparison of the bone ultrastructure formed at the interface of biodegradable Mg-5Gd and Mg-10Gd implants and titanium and PEEK implants after 4, 8 and 12 weeks healing time using two-dimensional small angle X-ray scattering and X-ray diffraction. Differences in mineralization, orientation and thickness of the hydroxyapatite are assessed. We find statistically significant (p < 0.05) differences for the lattice spacing of the (310)-reflex of hydroxyapatite between titanium and Mg-xGd materials, as well as for the (310) crystal size between titanium and Mg-5Gd, indicating a possible deposition of Mg within the bone matrix. The (310) lattice spacing and crystallite size further differ significantly between implant degradation layer and surrounding bone (p < 0.001 for Mg-10Gd), suggesting apatite formation with significant amounts of Gd and Mg within the degradation layer. STATEMENT OF SIGNIFICANCE: Biodegradable Magnesium-based alloys are emerging as a viable alternative for temporary bone implant applications. However, in order to understand if the degradation of the implant material influences the bone ultrastructure, it is necessary to study the bone structure using high-resolution techniques. We have therefore employed 2D small angle X-ray scattering and X-ray diffraction to study the bone ultrastructure surrounding Magnesium-Gadolinium alloys as well as Titanium and PEEK alloys at three different healing times. This is the first time, that the bone ultrastructure around these materials is directly compared and that a statistical evaluation is performed. We found differences indicating a possible deposition of Mg within the bone matrix as well as a local deposition of Mg and/or Gd at the implant site. DATA AVAILABILITY STATEMENT: The raw/processed data required to reproduce these findings cannot be shared at this time as the data also forms part of an ongoing study.


Assuntos
Implantes Absorvíveis , Osso e Ossos/ultraestrutura , Gadolínio/farmacologia , Magnésio/farmacologia , Difração de Raios X , Animais , Plaquetas/efeitos dos fármacos , Cristalização , Durapatita/farmacologia , Masculino , Ratos Sprague-Dawley , Titânio/farmacologia
16.
J Mech Behav Biomed Mater ; 79: 181-188, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29306081

RESUMO

Magnesium-based biomedical implants offer many advantages versus traditional ones although some challenges are still present. In this context, mathematical modeling and computational simulation may be a useful and complementary tool to evaluate in silico the performance of magnesium biomaterials under different conditions. In this paper, a phenomenologically-based model to simulate magnesium corrosion is developed. The model describes the physico-chemical interactions and evolution of species present in this phenomenon. A set of 7 species is considered in the model, which allows to simulate hydrogen release, pH evolution, corrosion products formation as well as degradation of magnesium. The model is developed under the continuum media theory and is implemented in a finite element framework. In the results section, the effect of model parameters on outcomes is firstly explored. Second, model results are qualitative validated versus two examples of application found in the literature. Two main conclusions are derived from this work: (i) the model captures well the experimental trends and allows to analyze the main variables present in magnesium corrosion, (ii) even though further validation is needed the model may be a useful standard in the design of degradable metal implants.


Assuntos
Implantes Absorvíveis , Materiais Biocompatíveis , Modelos Teóricos , Engenharia Tecidual/métodos , Simulação por Computador , Magnésio , Teste de Materiais
17.
Acta Biomater ; 63: 369-382, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28882757

RESUMO

Mg-based alloys, as the potential orthopaedic implant, can self-degrade to avoid second operation for its remove, and enable to promote bone repair; however, the underlying molecular mechanisms remain unclear. In the present study, we examined the effect of Mg ions on osteogenesis, chemotaxis and anti-alkaline stress in hFOB1.19 human osteoblast cells to simulate bone-repairing effect of a biodegradable Mg-based alloy implant in vitro, and explored the regulatory role of the transient receptor potential melastatin 7 (TRPM7)/phosphoinositide 3-kinase (PI3K) signalling pathway in the process of Mg ion-induced bone repair by knockdown of TRPM7 and antagonizing PI3K activity. Results indicate that Mg ions up-regulated the expression of Runx2 and alkaline phosphatase (ALP) through TRPM7/PI3K signalling pathway, which could significantly enhance the osteogenic activity of human osteoblasts. Furthermore, the expression levels of MMP2, MMP9 and vascular endothelial growth factor (VEGF) were increased by TRPM7/PI3K signalling pathway, which recruits osteoblasts from low- to high-Mg ion environments by inducing cell migration. Although an alkaline environment has antibacterial effects, alkaline stress can cause cytotoxicity and induce cell death. Finally, we found that Mg ions could activate PI3K phosphorylation to promote cell growth and survival, protecting cells against the alkaline-stress-induced cytotoxicity caused by the degradation of Mg-based alloy implants. Our study not only revealed the molecular mechanism of Mg in promoting bone repair but also explained the protective effects of Mg ions on osteoblasts in an alkaline environment, which provides a theoretical basis and new directions for the application of Mg-based alloy implant material in orthopaedics fixations and osteosarcoma treatment. STATEMENTS OF SIGNIFICANCE: As a potential biomaterial for orthopaedic implant, biodegradable magnesium has several advantages including self-degradation and bone repair promotion; however, the underlying mechanisms and effective concentration by which molecular regulates the bone repair remain unclear. The present study revealed that Mg ion and its effective concentration for activating PI3K phosphorylation via TRPM7, which causes three processes affecting bone repair, namely, osteoblast recruitment, osteogenesis and resistance to alkaline stress in human osteoblast. Therefore, our results have provided insight into the underlying molecular biological basis, and guidance for manipulating degradation rate, such as surface modification, of orthopaedic Mg-based implants.


Assuntos
Ligas/farmacologia , Magnésio/farmacologia , Osseointegração/efeitos dos fármacos , Osteoblastos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Próteses e Implantes , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Canais de Cátion TRPM/metabolismo , Fosfatase Alcalina/metabolismo , Calcificação Fisiológica/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Concentração de Íons de Hidrogênio , Íons , Modelos Biológicos , Osteoblastos/efeitos dos fármacos , Osteoblastos/enzimologia , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/efeitos dos fármacos , Estresse Fisiológico
18.
ACS Biomater Sci Eng ; 3(6): 936-950, 2017 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-33429566

RESUMO

Magnesium (Mg)-based bioresorbable cardiovascular scaffold (BCS) is a promising alternative to conventional permanent cardiovascular stents, but it faces the challenges of rapid degradation and poor endothelium recovery after device degradation. To address these challenges, we investigated poly(l-lactic acid) (PLLA), poly(lactic-co-glycolic acid) (PLGA) (90:10), PLGA (50:50), and polycaprolactone (PCL) coatings on Mg, respectively, and evaluated their surface and biological properties. Intact polymer coatings with complete coverage on Mg substrate were achieved. The biological performance of the materials was evaluated by culturing with human umbilical vein endothelial cells (HUVECs) in vitro using the direct culture method. The pH of the culture media and Mg2+ and Ca2+ ion concentrations in the media were measured after culture to characterize the degradation rate of the materials in vitro. The results showed that the PLGA (50:50) coating improved the adhesion and spreading of HUVECs the most among the four polymer coatings. Moreover, we found three possible factors that promoted HUVECs directly attached on the surface of PLGA (50:50)-coated Mg: (1) the higher concentration of Mg2+ ions released into culture media with a concentration range of 9-15 mM; (2) the lower Ca2+ ion concentration in culture media at 1.3-1.6 mM; and (3) the favorable surface conditions of PLGA (50:50), when compared with the other sample groups. This in vitro study provided the first evidence that the PLGA (50:50) is a promising coating material for Mg-based biodegradable metals toward potential cardiovascular or neurovascular applications.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA