Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22.225
Filter
1.
J Transl Med ; 22(1): 629, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38970118

ABSTRACT

BACKGROUND: Magnesium is a micronutrient and an intracellular cation responsible for different biochemical reactions involved in energy production and storage, control of neuronal and vasomotor activity, cardiac excitability, and muscle contraction. Magnesium deficiency may result in impaired physical performance. Moreover, magnesium plays an important role on delayed onset muscle soreness after training. Thus, physically active individuals and sport specialists have to pay attention to magnesium supplementation (MgS). However, the type, timing and dosage of magnesium intake are not well elucidated yet. Hence, we aimed to systematically review the literature regarding the effects of MgS on muscle soreness in physically active individuals. We focused exclusively on MgS, excluding those studies in which magnesium was administered together with other substances. METHODS: Three electronic databases and literature sources (PUBMED, SCOPUS and Web of Sciences-Core Collection) were searched, in accordance with PRISMA guidelines. After the database search, 1254 articles were identified, and after excluding duplicates, 960 articles remained. Among these, 955 were excluded following the title and abstract screening. The remaining 5 articles were screened in full text and 4 study met the eligibility criteria. RESULTS: These studies showed that MgS reduced muscle soreness, improved performance, recovery and induced a protective effect on muscle damage. CONCLUSION: To reach these positive effects, individuals engaged in intense exercise should have a Mg requirement 10-20% higher than sedentary people, to be taken in capsules and 2 h before training. Moreover, it is suggested to maintain magnesium levels in the recommended range during the off-season. SYSTEMATIC REVIEW REGISTRATION: PROSPERO registration number: CRD42024501822.


Subject(s)
Dietary Supplements , Exercise , Magnesium , Myalgia , Humans , Male , Magnesium/administration & dosage , Magnesium/pharmacology , Myalgia/drug therapy
2.
Sci Rep ; 14(1): 14693, 2024 06 26.
Article in English | MEDLINE | ID: mdl-38926545

ABSTRACT

Our research aimed to elucidate the mechanism by which aurintricarboxylic acid (ATA) inhibits plasma membrane Ca2+-ATPase (PMCA), a crucial enzyme responsible for calcium transport. Given the pivotal role of PMCA in cellular calcium homeostasis, understanding how it is inhibited by ATA holds significant implications for potentially regulating physiopathological cellular processes in which this pump is involved. Our experimental findings revealed that ATA employs multiple modes of action to inhibit PMCA activity, which are influenced by ATP but also by the presence of calcium and magnesium ions. Specifically, magnesium appears to enhance this inhibitory effect. Our experimental and in-silico results suggest that, unlike those reported in other proteins, ATA complexed with magnesium (ATA·Mg) is the molecule that inhibits PMCA. In summary, our study presents a novel perspective and establishes a solid foundation for future research efforts aimed at the development of new pharmacological molecules both for PMCA and other proteins.


Subject(s)
Aurintricarboxylic Acid , Calcium , Magnesium , Plasma Membrane Calcium-Transporting ATPases , Magnesium/metabolism , Magnesium/pharmacology , Aurintricarboxylic Acid/pharmacology , Plasma Membrane Calcium-Transporting ATPases/metabolism , Plasma Membrane Calcium-Transporting ATPases/antagonists & inhibitors , Calcium/metabolism , Adenosine Triphosphate/metabolism , Cell Membrane/metabolism , Cell Membrane/drug effects , Animals , Humans
3.
J Nanobiotechnology ; 22(1): 368, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38918787

ABSTRACT

Active artificial bone substitutes are crucial in bone repair and reconstruction. Calcium phosphate bone cement (CPC) is known for its biocompatibility, degradability, and ability to fill various shaped bone defects. However, its low osteoinductive capacity limits bone regeneration applications. Effectively integrating osteoinductive magnesium ions with CPC remains a challenge. Herein, we developed magnesium malate-modified CPC (MCPC). Incorporating 5% magnesium malate significantly enhances the compressive strength of CPC to (6.18 ± 0.49) MPa, reduces setting time and improves disintegration resistance. In vitro, MCPC steadily releases magnesium ions, promoting the proliferation of MC3T3-E1 cells without causing significant apoptosis, proving its biocompatibility. Molecularly, magnesium malate prompts macrophages to release prostaglandin E2 (PGE2) and synergistically stimulates dorsal root ganglion (DRG) neurons to synthesize and release calcitonin gene-related peptide (CGRP). The CGRP released by DRG neurons enhances the expression of the key osteogenic transcription factor Runt-related transcription factor-2 (RUNX2) in MC3T3-E1 cells, promoting osteogenesis. In vivo experiments using minipig vertebral bone defect model showed MCPC significantly increases the bone volume fraction, bone density, new bone formation, and proportion of mature bone in the defect area compared to CPC. Additionally, MCPC group exhibited significantly higher levels of osteogenesis and angiogenesis markers compared to CPC group, with no inflammation or necrosis observed in the hearts, livers, or kidneys, indicating its good biocompatibility. In conclusion, MCPC participates in the repair of bone defects in the complex post-fracture microenvironment through interactions among macrophages, DRG neurons, and osteoblasts. This demonstrates its significant potential for clinical application in bone defect repair.


Subject(s)
Bone Cements , Calcitonin Gene-Related Peptide , Calcium Phosphates , Osteogenesis , Swine, Miniature , Animals , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacology , Bone Cements/pharmacology , Bone Cements/chemistry , Mice , Swine , Calcitonin Gene-Related Peptide/metabolism , Osteogenesis/drug effects , Bone Regeneration/drug effects , Spine/surgery , Ganglia, Spinal/metabolism , Ganglia, Spinal/drug effects , Cell Line , Magnesium/pharmacology , Magnesium/chemistry
4.
ACS Appl Mater Interfaces ; 16(25): 32566-32577, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38867413

ABSTRACT

In this work, the hydroxyapatite (HA) microspheres are utilized as carriers for 8-hydroxyquinoline (8-HQ) inhibitors with a sodium alginate-silver nitrate layer (Ag-SA) added to confer chloride-responsive properties. These 8-HQ@Ag-SA-HA microspheres are subsequently integrated into poly(lactic acid) (PLA) coatings to produce biocompatible coatings. The resulting 8-HQ@Ag-SA-HA microsphere exhibits a spherical structure with a diameter of 3.16 µm. Thermogravimetric analysis indicates that the encapsulated 8-HQ inhibitors are approximately 11.83 wt %. Furthermore, the incorporation of these microspheres fills the micropores within the PLA coating, leading to a denser coating surface, enhanced wettability (contact angle value = 88°), and improved adhesion strength, thereby reinforcing the physical barrier effect. Corrosion tests reveal that the coatings exhibit increased resistance to corrosion in simulated body fluid (SBF) solutions. The released 8-HQ inhibitors in response to chloride ions form a protective layer of Mg(HQ)2, providing the coatings with self-healing properties and ensuring their durability in the SBF environment. Additionally, the cell test demonstrates a significant presence of MG-63 cells, accompanied by a low hemolysis rate of 3.81%, confirming the exceptional biocompatibility of the coatings. These findings offer valuable insights into the development of stimuli-responsive biocompatible coatings for effectively protecting Mg alloys.


Subject(s)
Alloys , Chlorides , Coated Materials, Biocompatible , Magnesium , Alloys/chemistry , Alloys/pharmacology , Humans , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Magnesium/chemistry , Magnesium/pharmacology , Chlorides/chemistry , Durapatite/chemistry , Durapatite/pharmacology , Corrosion , Microspheres , Alginates/chemistry , Polyesters/chemistry
5.
PLoS Biol ; 22(6): e3002694, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38900845

ABSTRACT

Fungi and bacteria coexist in many polymicrobial communities, yet the molecular basis of their interactions remains poorly understood. Here, we show that the fungus Candida albicans sequesters essential magnesium ions from the bacterium Pseudomonas aeruginosa. To counteract fungal Mg2+ sequestration, P. aeruginosa expresses the Mg2+ transporter MgtA when Mg2+ levels are low. Thus, loss of MgtA specifically impairs P. aeruginosa in co-culture with C. albicans, but fitness can be restored by supplementing Mg2+. Using a panel of fungi and bacteria, we show that Mg2+ sequestration is a general mechanism of fungal antagonism against gram-negative bacteria. Mg2+ limitation enhances bacterial resistance to polymyxin antibiotics like colistin, which target gram-negative bacterial membranes. Indeed, experimental evolution reveals that P. aeruginosa evolves C. albicans-dependent colistin resistance via non-canonical means; antifungal treatment renders resistant bacteria colistin-sensitive. Our work suggests that fungal-bacterial competition could profoundly impact polymicrobial infection treatment with antibiotics of last resort.


Subject(s)
Anti-Bacterial Agents , Candida albicans , Colistin , Magnesium , Pseudomonas aeruginosa , Magnesium/pharmacology , Magnesium/metabolism , Pseudomonas aeruginosa/drug effects , Anti-Bacterial Agents/pharmacology , Candida albicans/drug effects , Candida albicans/metabolism , Colistin/pharmacology , Microbial Sensitivity Tests , Polymyxins/pharmacology , Drug Resistance, Bacterial/drug effects , Microbial Interactions/drug effects
6.
Front Cell Infect Microbiol ; 14: 1335189, 2024.
Article in English | MEDLINE | ID: mdl-38895735

ABSTRACT

Background: Chikungunya virus (CHIKV), which causes chikungunya fever, is an arbovirus of public health concern with no approved antiviral therapies. A significant proportion of patients develop chronic arthritis after an infection. Zinc and magnesium salts help the immune system respond effectively against viral infections. This study explored the antiviral potential of zinc sulphate, zinc acetate, and magnesium sulphate against CHIKV infection. Methods: The highest non-toxic concentration of the salts (100 µM) was used to assess the prophylactic, virucidal, and therapeutic anti-CHIKV activities. Dose-dependent antiviral effects were investigated to find out the 50% inhibitory concentration of the salts. Entry bypass assay was conducted to find out whether the salts affect virus entry or post entry stages. Virus output in all these experiments was estimated using a focus-forming unit assay, real-time RT-PCR, and immunofluorescence assay. Results: Different time- and temperature-dependent assays revealed the therapeutic antiviral activity of zinc and magnesium salts against CHIKV. A minimum exposure of 4 hours and treatment initiation within 1 to 2 hours of infection are required for inhibition of CHIKV. Entry assays revealed that zinc salt affected virus-entry. Entry bypass assays suggested that both salts affected post-entry stages of CHIKV. In infected C57BL6 mice orally fed with zinc and magnesium salts, a reduction in viral RNA copy number was observed. Conclusion: The study results suggest zinc salts exert anti-CHIKV activity at entry and post entry stages of the virus life cycle, while magnesium salt affect CHIKV at post entry stages. Overall, the study highlights the significant antiviral potential of zinc sulphate, zinc acetate, and magnesium sulphate against CHIKV, which can be exploited in designing potential therapeutic strategies for early treatment of chikungunya patients, thereby reducing the virus-associated persistent arthritis.


Subject(s)
Antiviral Agents , Chikungunya Fever , Chikungunya virus , Zinc Acetate , Zinc Sulfate , Chikungunya virus/drug effects , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Chikungunya Fever/drug therapy , Chikungunya Fever/virology , Zinc Acetate/pharmacology , Zinc Acetate/therapeutic use , Zinc Sulfate/pharmacology , Chlorocebus aethiops , Vero Cells , Virus Internalization/drug effects , Mice , Zinc/pharmacology , Zinc/therapeutic use , Humans , Magnesium Sulfate/pharmacology , Magnesium/pharmacology , Virus Replication/drug effects , Inhibitory Concentration 50 , Salts/pharmacology , Cell Line
7.
ACS Appl Mater Interfaces ; 16(24): 30685-30702, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38859670

ABSTRACT

Macrophages play a pivotal role in the crosstalk between the immune and skeletal systems, while Mg-based biomaterials demonstrate immunomodulatory capabilities in this procedure. However, the mechanism of how Mg2+ promotes osteogenesis through the interplay of bone marrow-derived mesenchymal stem cells (BMSCs) and macrophages remains undescribed. Here, we demonstrated that a Mg-cross-linked alginate hydrogel exerted a dual enhancement of BMSCs osteogenic differentiation through the ligand-receptor pairing of the OSM/miR-370-3p-gp130 axis. On the one hand, Mg2+, released from the Mg-cross-linked hydrogel, stimulates bone marrow-derived macrophages to produce and secrete more OSM. On the other hand, Mg2+ lowers the miR-370-3p level in BMSCs and in turn, reverses its suppression on gp130. Then, the OSM binds to the gp130 heterodimer receptor and activates intracellular osteogenic programs in BMSCs. Taken together, this study reveals a novel cross-talk pattern between the skeletal and immune systems under Mg2+ stimulation. This study not only brings new insights into the immunomodulatory properties of Mg-based biomaterials for orthopedic applications but also enriches the miRNA regulatory network and provides a promising target to facilitate bone regeneration in large bone defects.


Subject(s)
Alginates , Bone Regeneration , Hydrogels , Macrophages , Magnesium , Mesenchymal Stem Cells , MicroRNAs , Osteogenesis , Signal Transduction , Hydrogels/chemistry , Hydrogels/pharmacology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , MicroRNAs/metabolism , MicroRNAs/genetics , Animals , Bone Regeneration/drug effects , Alginates/chemistry , Signal Transduction/drug effects , Macrophages/metabolism , Macrophages/drug effects , Osteogenesis/drug effects , Magnesium/chemistry , Magnesium/pharmacology , Mice , Cytokine Receptor gp130/metabolism , Cytokine Receptor gp130/genetics , Cell Differentiation/drug effects
8.
J Nanobiotechnology ; 22(1): 314, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38840113

ABSTRACT

Osteoporosis is the most common bone metabolic disease that affects the health of middle-aged and elderly people, which is hallmarked by imbalanced bone remodeling and a deteriorating immune microenvironment. Magnesium and calcium are pivotal matrix components that participate in the bone formation process, especially in the immune microenvironment regulation and bone remodeling stages. Nevertheless, how to potently deliver magnesium and calcium to bone tissue remains a challenge. Here, we have constructed a multifunctional nanoplatform composed of calcium-based upconversion nanoparticles and magnesium organic frameworks (CM-NH2-PAA-Ald, denoted as CMPA), which features bone-targeting and pH-responsive properties, effectively regulating the inflammatory microenvironment and promoting the coordination of osteogenic functions for treating osteoporosis. The nanoplatform can efficaciously target bone tissue and gradually degrade in response to the acidic microenvironment of osteoporosis to release magnesium and calcium ions. This study validates that CMPA possessing favorable biocompatibility can suppress inflammation and facilitate osteogenesis to treat osteoporosis. Importantly, high-throughput sequencing results demonstrate that the nanoplatform exerts a good inflammatory regulation effect through inhibition of the nuclear factor kappa-B signaling pathway, thereby normalizing the osteoporotic microenvironment. This collaborative therapeutic strategy that focuses on improving bone microenvironment and promoting osteogenesis provides new insight for the treatment of metabolic diseases such as osteoporosis.


Subject(s)
Calcium , Magnesium , Nanoparticles , Osteogenesis , Osteoporosis , Osteogenesis/drug effects , Osteoporosis/drug therapy , Magnesium/pharmacology , Magnesium/chemistry , Calcium/metabolism , Animals , Nanoparticles/chemistry , Mice , Inflammation/drug therapy , Bone and Bones/drug effects , Bone and Bones/metabolism , Humans , Cellular Microenvironment/drug effects , Female , NF-kappa B/metabolism
9.
Sci Rep ; 14(1): 12877, 2024 06 05.
Article in English | MEDLINE | ID: mdl-38834648

ABSTRACT

This study reports the antibacterial and antibiofilm activities of Magnesium ferrite nanoparticles (MgFe2O4) against gram-positive and gram-negative bacteria. The photocatalytic degradation of Carbol Fuchsin (CF) dye (a class of dyestuffs that are resistant to biodegradation) under the influence of UV-light irradiation is also studied. The crystalline magnesium ferrite (MgFe2O4) nanoparticles were synthesized using the co-precipitation method. The morphology of the resulting nanocomposite was examined using scanning electron microscopy (SEM), while transmission electron microscopy (TEM) was employed for further characterization of particle morphology and size. Fourier transform infrared (FTIR) spectroscopy and X-ray diffraction (XRD) were utilized to analyze the crystalline structure, chemical composition, and surface area, respectively. Optical properties were evaluated using UV-Vis spectroscopy. The UV-assisted photocatalytic performance of MgFe2O4 nanoparticles was assessed by studying the decolorization of Carbol fuchsin (CF) azo dye. The crystallite size of the MgFe2O4 nanoparticles at the (311) plane, the most prominent peak, was determined to be 28.5 nm. The photocatalytic degradation of 10 ppm CF using 15 mg of MgFe2O4 nanoparticles resulted in a significant 96% reduction after 135 min at ambient temperature (25 °C) and a pH value of 9. Additionally, MgFe2O4 nanoparticles exhibited potent antibacterial activity against E. coli and S. aureus in a dose dependent manner with maximum utilized concentration of 30 µg/ml. Specifically, MgFe2O4 nanoparticles demonstrated substantial antibacterial activity via disk diffusion and microbroth dilution tests with zones of inhibition and minimum inhibitory concentrations (MIC) for E. coli (26.0 mm, 1.25 µg/ml) and S. aureus (23.0 mm, 2.5 µg/ml), respectively. Moreover, 10.0 µg/ml of MgFe2O4 nanoparticles elicited marked percent reduction in biofilm formation by E. coli (89%) followed by S. aureus (78.5%) after treatment. In conclusion, MgFe2O4 nanoparticles demonstrated efficient dye removal capabilities along with significant antimicrobial and antibiofilm activity against gram-positive and gram-negative bacterial strains suggesting their potential as promising antimicrobial and detoxifying agents.


Subject(s)
Anti-Bacterial Agents , Biofilms , Ferric Compounds , Magnetite Nanoparticles , Biofilms/drug effects , Ferric Compounds/chemistry , Ferric Compounds/pharmacology , Catalysis , Magnetite Nanoparticles/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Microbial Sensitivity Tests , Escherichia coli/drug effects , Ultraviolet Rays , Staphylococcus aureus/drug effects , Magnesium/chemistry , Magnesium/pharmacology , Spectroscopy, Fourier Transform Infrared
10.
Micron ; 184: 103661, 2024 09.
Article in English | MEDLINE | ID: mdl-38833994

ABSTRACT

The silver/magnesium doped hydroxyapatite (AgMgHAp, Ca10-x-yAgxMgy(PO4)6(OH)2, xAg=0.05 and yMg=0.02) nanocomposites coatings were deposited on Si substrate using the dip coating technique. The resulting coatings were characterized by scanning electron microscopy (SEM), energy-dispersive X-ray spectroscopy (EDS), Fourier transform infrared (FTIR-ATR) spectroscopy, atomic force microscopy (AFM) and X-ray photoelectron spectroscopy (XPS). The EDS analysis highlighted the presence of the constitutive elements of the silver/magnesium doped hydroxyapatite (AgMgHAp) nanocomposites coatings. The surface microtexture of the AgMgHAp was assessed by atomic force microscopy (AFM) technique. The AFM data suggested the obtaining of a uniform deposited layer comprised of equally distributed nanoconglomerates. FT-IR studies highlighted the presence of vibrational modes associated with the phosphate and hydroxyl groups. No bands associated with silver or magnesium were observed. The XPS analysis highlighted the presence of the constituent elements of hydroxyapatite (Ca 2p, P 2 s, O 1 s), as well as dopants (Ag 3d, Mg 1 s and Mg 2p). The antifungal evaluation of AgMgHAp coatings was carried out using the Candida albicans ATCC 10231 fungal strain. The results of the antifungal assay revealed that the AgMgHAp coatings exhibited a strong inhibitory antifungal activity. Furthermore, the data highlighted that the AgMgHAp inhibited the development of biofilm on their surface. The results revealed that the antifungal activity of the coating varied based on the duration of incubation. On the other hand, the data also showed that AgMgHAp nanocomposites coatings inhibited the fungal cell adhesion and development from the early stages of the incubation. In addition to morphological analysis, we additionally take advantage of AFM images to investigate and explore the domain of fractal and multifractal analysis applied to the films under evaluation. Our studies indicates that nanocomposite coatings made from AgMgHAp demonstrate strong antifungal properties. Our studies indicates that nanocomposite coatings made from AgMgHAp demonstrate strong antifungal properties. These results suggest the potential of AgMgHAp nanocomposite coatings as a promising solution for developing innovative antifungal devices in biomedical applications.


Subject(s)
Antifungal Agents , Durapatite , Magnesium , Microscopy, Atomic Force , Nanocomposites , Silver , Durapatite/chemistry , Durapatite/pharmacology , Antifungal Agents/pharmacology , Silver/pharmacology , Silver/chemistry , Nanocomposites/chemistry , Magnesium/chemistry , Magnesium/pharmacology , Spectroscopy, Fourier Transform Infrared , Candida albicans/drug effects , Microscopy, Electron, Scanning , Photoelectron Spectroscopy , Microbial Sensitivity Tests , Spectrometry, X-Ray Emission , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Surface Properties
11.
ACS Biomater Sci Eng ; 10(7): 4452-4462, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38875708

ABSTRACT

Mg-based biodegradable metallic implants are gaining increased attraction for applications in orthopedics and dentistry. However, their current applications are hampered by their high rate of corrosion, degradation, and rapid release of ions and gas bubbles into the physiological medium. The aim of the present study is to investigate the osteogenic and angiogenic potential of coated Mg-based implants in a sheep cranial defect model. Although their osteogenic potential was studied to some extent, their potential to regenerate vascularized bone formation was not studied in detail. We have studied the potential of magnesium-calcium (MgCa)-based alloys modified with zinc (Zn)- or gallium (Ga)-doped calcium phosphate (CaP) coatings as a strategy to control their degradation rate while enhancing bone regeneration capacity. MgCa and its implants with CaP coatings (MgCa/CaP) as undoped or as doped with Zn or Ga (MgCa/CaP + Zn and MgCa/CaP + Ga, respectively) were implanted in bone defects created in the sheep cranium. MgCa implants degraded faster than the others at 4 weeks postop and the weight loss was ca. 50%, while it was ca. 15% for MgCa/CaP and <10% in the presence of Zn and Ga with CaP coating. Scanning electron microscopy (SEM) analysis of the implant surfaces also revealed that the MgCa implants had the largest degree of structural breakdown of all the groups. Radiological evaluation revealed that surface modification with CaP to the MgCa implants induced better bone regeneration within the defects as well as the enhancement of bone-implant surface integration. Bone volume (%) within the defect was ca. 25% in the case of MgCa/CaP + Ga, while it was around 15% for undoped MgCa group upon micro-CT evaluation. This >1.5-fold increase in bone regeneration for MgCa/CaP + Ga implant was also observed in the histopathological examination of the H&E- and Masson's trichrome-stained sections. Immunohistochemical analysis of the bone regeneration (antiosteopontin) and neovascularization (anti-CD31) at the defect sites revealed >2-fold increase in the expression of the markers in both Ga- and Zn-doped, CaP-coated implants. Zn-doped implants further presented low inflammatory reaction, notable bone regeneration, and neovascularization among all the implant groups. These findings indicated that Ga- and Zn-doped CaP coating is an important strategy to control the degradation rate as well as to achieve enhanced bone regeneration capacity of the implants made of Mg-based alloys.


Subject(s)
Alloys , Calcium Phosphates , Coated Materials, Biocompatible , Gallium , Magnesium , Osteogenesis , Skull , Zinc , Animals , Zinc/chemistry , Zinc/pharmacology , Sheep , Skull/drug effects , Skull/pathology , Skull/injuries , Osteogenesis/drug effects , Magnesium/pharmacology , Gallium/chemistry , Gallium/pharmacology , Alloys/chemistry , Alloys/pharmacology , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacology , Bone Regeneration/drug effects , Calcium/metabolism , Absorbable Implants
12.
ACS Biomater Sci Eng ; 10(6): 3514-3527, 2024 06 10.
Article in English | MEDLINE | ID: mdl-38723173

ABSTRACT

The field of bone regeneration has always been a hot and difficult research area, and there is no perfect strategy at present. As a new type of biodegradable material, magnesium alloys have excellent mechanical properties and bone promoting ability. Compared with other inert metals, magnesium alloys have significant advantages and broad application prospects in the field of bone regeneration. By searching the official Web sites and databases of various funds, this paper summarizes the research status of magnesium composites in the field of bone regeneration and introduces the latest scientific research achievements and clinical transformations of scholars in various countries and regions, such as improving the corrosion resistance of magnesium alloys by adding coatings. Finally, this paper points out the current problems and challenges, aiming to provide ideas and help for the development of new strategies for the treatment of bone defects and fractures.


Subject(s)
Bone Regeneration , Magnesium , Bone Regeneration/drug effects , Magnesium/therapeutic use , Magnesium/pharmacology , Magnesium/chemistry , Humans , Alloys/chemistry , Alloys/therapeutic use , Biocompatible Materials/therapeutic use , Biocompatible Materials/pharmacology , Biocompatible Materials/chemistry , Animals , Bone Substitutes/therapeutic use , Bone Substitutes/chemistry , Bone Substitutes/pharmacology , Corrosion
13.
Int J Biol Macromol ; 271(Pt 2): 132487, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38768910

ABSTRACT

Due to its biofunctions similar to NO, the CO gas signaling molecule has gradually shown great potential in cardiovascular biomaterials for regulating the in vivo performances after the implantation and has received increasing attention. To construct a bioactive surface with CO-releasing properties on the surface of magnesium-based alloy to augment the anticorrosion and biocompatibility, graphene oxide (GO) was firstly modified using carboxymethyl chitosan (CS), and then CO-releasing molecules (CORM401) were introduced to synthesize a novel biocompatible nanomaterial (GOCS-CO) that can release CO in the physiological environments. The GOCS-CO was further immobilized on the magnesium alloy surface modified by polydopamine coating with Zn2+ (PDA/Zn) to create a bioactive surface capable of releasing CO in the physiological environment. The outcomes showed that the CO-releasing coating can not only significantly enhance the anticorrosion and abate the corrosion degradation rate of the magnesium alloy in a simulated physiological environment, but also endow it with good hydrophilicity and a certain ability to adsorb albumin selectively. Owing to the significant enhancement of anticorrosion and hydrophilicity, coupled with the bioactivity of GOCS, the modified sample not only showed excellent ability to prevent platelet adhesion and activation and reduce hemolysis rate but also can promote endothelial cell (EC) adhesion, proliferation as well as the expression of nitric oxide (NO) and vascular endothelial growth factor (VEGF). In the case of CO release, the hemocompatibility and EC growth behaviors were further significantly improved, suggesting that CO molecules released from the surface can significantly improve the hemocompatibility and EC growth. Consequently, the present study provides a novel surface modification method that can simultaneously augment the anticorrosion and biocompatibility of magnesium-based alloys, which will strongly promote the research and application of CO-releasing bioactive coatings for surface functionalization of cardiovascular biomaterials and devices.


Subject(s)
Alloys , Chitosan , Coated Materials, Biocompatible , Graphite , Magnesium , Graphite/chemistry , Chitosan/chemistry , Chitosan/analogs & derivatives , Chitosan/pharmacology , Magnesium/chemistry , Magnesium/pharmacology , Alloys/chemistry , Alloys/pharmacology , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Humans , Carbon Monoxide/chemistry , Carbon Monoxide/pharmacology , Stents , Hemolysis/drug effects , Platelet Adhesiveness/drug effects , Corrosion , Cell Adhesion/drug effects , Materials Testing , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cell Proliferation/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Animals , Endothelial Cells/drug effects
14.
ACS Appl Mater Interfaces ; 16(19): 24384-24397, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38709640

ABSTRACT

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


Subject(s)
Bone Regeneration , Deferoxamine , Magnesium , Mesenchymal Stem Cells , Oxides , Tantalum , Deferoxamine/chemistry , Deferoxamine/pharmacology , Bone Regeneration/drug effects , Tantalum/chemistry , Animals , Oxides/chemistry , Oxides/pharmacology , Magnesium/chemistry , Magnesium/pharmacology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteogenesis/drug effects , Neovascularization, Physiologic/drug effects , Rats , Mice , Rats, Sprague-Dawley , Cell Proliferation/drug effects , Angiogenesis
15.
Int J Mol Sci ; 25(9)2024 May 03.
Article in English | MEDLINE | ID: mdl-38732212

ABSTRACT

The skin wound healing process consists of hemostatic, inflammatory, proliferative, and maturation phases, with a complex cellular response by multiple cell types in the epidermis, dermis, and immune system. Magnesium is a mineral essential for life, and although magnesium treatment promotes cutaneous wound healing, the molecular mechanism and timing of action of the healing process are unknown. This study, using human epidermal-derived HaCaT cells and human normal epidermal keratinocyte cells, was performed to investigate the mechanism involved in the effect of magnesium on wound healing. The expression levels of epidermal differentiation-promoting factors were reduced by MgCl2, suggesting an inhibitory effect on epidermal differentiation in the remodeling stage of the late wound healing process. On the other hand, MgCl2 treatment increased the expression of matrix metalloproteinase-7 (MMP7), a cell migration-promoting factor, and enhanced cell migration via the MEK/ERK pathway activation. The enhancement of cell migration by MgCl2 was inhibited by MMP7 knockdown, suggesting that MgCl2 enhances cell migration which is mediated by increased MMP7 expression. Our results revealed that MgCl2 inhibits epidermal differentiation but promotes cell migration, suggesting that applying magnesium to the early wound healing process could be beneficial.


Subject(s)
Cell Differentiation , Cell Movement , Keratinocytes , Magnesium , Matrix Metalloproteinase 7 , Wound Healing , Wound Healing/drug effects , Humans , Cell Movement/drug effects , Keratinocytes/drug effects , Keratinocytes/metabolism , Cell Differentiation/drug effects , Magnesium/pharmacology , Magnesium/metabolism , Matrix Metalloproteinase 7/metabolism , Matrix Metalloproteinase 7/genetics , Skin/metabolism , Skin/drug effects , Skin/injuries , MAP Kinase Signaling System/drug effects , Cell Line , Epidermis/drug effects , Epidermis/metabolism , Magnesium Chloride/pharmacology
16.
Acta Biomater ; 182: 139-155, 2024 07 01.
Article in English | MEDLINE | ID: mdl-38750914

ABSTRACT

Additively manufactured (AM) biodegradable zinc (Zn) alloys have recently emerged as promising porous bone-substituting materials, due to their moderate degradation rates, good biocompatibility, geometrically ordered microarchitectures, and bone-mimicking mechanical properties. While AM Zn alloy porous scaffolds mimicking the mechanical properties of trabecular bone have been previously reported, mimicking the mechanical properties of cortical bone remains a formidable challenge. To overcome this challenge, we developed the AM Zn-3Mg alloy. We used laser powder bed fusion to process Zn-3Mg and compared it with pure Zn. The AM Zn-3Mg alloy exhibited significantly refined grains and a unique microstructure with interlaced α-Zn/Mg2Zn11 phases. The compressive properties of the solid Zn-3Mg specimens greatly exceeded their tensile properties, with a compressive yield strength of up to 601 MPa and an ultimate strain of >60 %. We then designed and fabricated functionally graded porous structures with a solid core and achieved cortical bone-mimicking mechanical properties, including a compressive yield strength of >120 MPa and an elastic modulus of ≈20 GPa. The biodegradation rates of the Zn-3Mg specimens were lower than those of pure Zn and could be adjusted by tuning the AM process parameters. The Zn-3Mg specimens also exhibited improved biocompatibility as compared to pure Zn, including higher metabolic activity and enhanced osteogenic behavior of MC3T3 cells cultured with the extracts from the Zn-3Mg alloy specimens. Altogether, these results marked major progress in developing AM porous biodegradable metallic bone substitutes, which paved the way toward clinical adoption of Zn-based scaffolds for the treatment of load-bearing bony defects. STATEMENT OF SIGNIFICANCE: Our study presents a significant advancement in the realm of biodegradable metallic bone substitutes through the development of an additively manufactured Zn-3Mg alloy. This novel alloy showcases refined grains and a distinctive microstructure, enabling the fabrication of functionally graded porous structures with mechanical properties resembling cortical bone. The achieved compressive yield strength and elastic modulus signify a critical leap toward mimicking the mechanical behavior of load-bearing bone. Moreover, our findings reveal tunable biodegradation rates and enhanced biocompatibility compared to pure Zn, emphasizing the potential clinical utility of Zn-based scaffolds for treating load-bearing bony defects. This breakthrough opens doors for the wider adoption of zinc-based materials in regenerative orthopedics.


Subject(s)
Alloys , Cortical Bone , Zinc , Alloys/chemistry , Alloys/pharmacology , Zinc/chemistry , Zinc/pharmacology , Animals , Mice , Cortical Bone/drug effects , Porosity , Magnesium/chemistry , Magnesium/pharmacology , Materials Testing , Compressive Strength , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Absorbable Implants , Elastic Modulus , Cell Line
17.
Nat Commun ; 15(1): 4160, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755128

ABSTRACT

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


Subject(s)
Biocompatible Materials , Bone Regeneration , Fibroins , Magnesium , Mesenchymal Stem Cells , Osteogenesis , Tissue Scaffolds , Fibroins/chemistry , Fibroins/pharmacology , Bone Regeneration/drug effects , Animals , Tissue Scaffolds/chemistry , Male , Osteogenesis/drug effects , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Rats , Magnesium/chemistry , Magnesium/pharmacology , Biocompatible Materials/chemistry , Osteoblasts/drug effects , Cell Differentiation/drug effects , Rats, Sprague-Dawley , Water/chemistry , Cell Proliferation/drug effects , Tissue Engineering/methods , Skull/drug effects , Cell Adhesion/drug effects , Bombyx
18.
Shanghai Kou Qiang Yi Xue ; 33(1): 6-12, 2024 Feb.
Article in Chinese | MEDLINE | ID: mdl-38583018

ABSTRACT

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.


Subject(s)
Nanopores , Titanium , Titanium/pharmacology , Magnesium/chemistry , Magnesium/pharmacology , Osteogenesis/genetics , Electrolytes/pharmacology , Ions/pharmacology , Surface Properties , Coated Materials, Biocompatible/pharmacology , Coated Materials, Biocompatible/chemistry
19.
ACS Appl Bio Mater ; 7(5): 2762-2780, 2024 05 20.
Article in English | MEDLINE | ID: mdl-38629138

ABSTRACT

In the present study, we have discussed the influence of forging temperature (623 K (FT623), 723 K (FT723) and 823 K (FT823)) on microstructure and texture evolution and its implication on mechanical behavior, in vitro-in vivo biocorrosion, antibacterial response, and cytocompatibility of microalloyed Mg-Zr-Sr-Ce alloy. Phase analysis, SEM, and TEM characterization confirm the presence of Mg12Ce precipitate, and its stability was further validated by performing ab initio molecular dynamic simulation study. FT723 exhibits strengthened basal texture, higher fraction of second phases, and particle-stimulated nucleation-assisted DRX grains compared to other two specimens, resulting in superior strength with comparable ductility. FT723 also exhibits superior corrosion resistance mainly due to the strengthened basal texture and lower dislocation density. All the specimens exhibit excellent antibacterial behavior with Gram-negative E. coli, Gram-positive Staphylococcus aureus, and Pseudomonas aeruginosa bacteria. 100% reduction of bacterial growth is observed within 24 h of culture of the specimens. Cytocompatibility was determined by challenging specimen extracts with the MC3T3-E1 cell lines. FT723 specimen exhibits the highest cell proliferation and alkaline phosphatase activity (ALP) because of its superior corrosion resistance. The ability of the specimens to be used in orthopedic implant application was evaluated by in vivo study in rabbit femur. Neither tissue-related infection nor the detrimental effect surrounding the implant was confirmed from histological analysis. Significant higher bone regeneration surrounding the FT723 specimen was observed in SEM analysis and fluorochrome labeling. After 60 days, the FT723 specimen exhibits the highest bone formation, suggesting it is a suitable candidate for orthopedic implant application.


Subject(s)
Alloys , Anti-Bacterial Agents , Biocompatible Materials , Materials Testing , Osteogenesis , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Alloys/chemistry , Alloys/pharmacology , Osteogenesis/drug effects , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Mice , Zirconium/chemistry , Zirconium/pharmacology , Microbial Sensitivity Tests , Particle Size , Cell Differentiation/drug effects , Rabbits , Magnesium/chemistry , Magnesium/pharmacology , Escherichia coli/drug effects , Pseudomonas aeruginosa/drug effects , Cell Proliferation/drug effects , Strontium/chemistry , Strontium/pharmacology , Molecular Dynamics Simulation , Cell Line , Temperature
20.
Acta Biomater ; 180: 183-196, 2024 05.
Article in English | MEDLINE | ID: mdl-38604465

ABSTRACT

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.


Subject(s)
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
SELECTION OF CITATIONS
SEARCH DETAIL
...