Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.428
Filter
1.
Molecules ; 29(15)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39124887

ABSTRACT

This article presents a new method for preparing multifunctional composite biomaterials with applications in advanced biomedical fields. The biomaterials consist of dicalcium phosphate (DCPD) and bioactive silicate glasses (SiO2/Na2O and SiO2/K2O), containing the antibiotic streptomycin sulfate. Materials were deeply characterized by X-ray diffraction and attenuated total reflectance Fourier transform infrared spectroscopy, and zeta potential analysis, UV-visible spectrophotometry, and ion-exchange measurement were applied in a simulating body fluid (SBF) solution. The main results include an in situ chemical transformation of dicalcium phosphate into an apatitic phase under the influence of silicate solutions and the incorporation of the antibiotic. The zeta potential showed a decrease in surface charge from ζ = -24.6 mV to ζ = -16.5 mV. In addition, a controlled and prolonged release of antibiotics was observed over a period of 37 days, with a released concentration of up to 755 ppm. Toxicity tests in mice demonstrated good tolerance of the biomaterials, with no significant adverse effects. Moreover, these biomaterials have shown potent antibacterial activity against various bacterial strains, including Listeria monocytogenes, Staphylococcus aureus, Escherichia coli, and Pseudomonas aeruginosa, suggesting their potential use in tissue engineering, drug delivery, and orthopedic and dental implants. By integrating the antibiotic into the biomaterial composites, we achieved controlled release and prolonged antibacterial efficacy. This research contributes to advancing biomaterials by exploring innovative synthetic routes and showcasing their promise in regenerative medicine and controlled drug delivery.


Subject(s)
Anti-Bacterial Agents , Biocompatible Materials , Regenerative Medicine , Biocompatible Materials/chemistry , Biocompatible Materials/chemical synthesis , Regenerative Medicine/methods , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Animals , Mice , Drug Delivery Systems , X-Ray Diffraction , Microbial Sensitivity Tests , Delayed-Action Preparations/pharmacology , Spectroscopy, Fourier Transform Infrared , Calcium Phosphates/chemistry , Calcium Phosphates/chemical synthesis , Drug Liberation , Streptomycin/pharmacology , Silicon Dioxide/chemistry
2.
Biotechnol J ; 19(7): e2300751, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38987220

ABSTRACT

The compatibility of bone graft substitutes (BGS) with mesenchymal stem cells (MSCs) is an important parameter to consider for their use in repairing bone defects as it eventually affects the clinical outcome. In the present study, a few commercially available BGS - ß-tricalcium phosphate (ß-TCP), calcium sulfate, gelatin sponge, and different forms of hydroxyapatite (HAP) were screened for their interactions with MSCs from adipose tissue (ADSCs). It was demonstrated that HAP block favorably supported ADSC viability, morphology, migration, and differentiation compared to other scaffolds. The results strongly suggest the importance of preclinical evaluation of bone scaffolds for their cellular compatibility. Furthermore, the bone regenerative potential of HAP block with ADSCs was evaluated in an ex vivo bone defect model developed using patient derived trabecular bone explants. The explants were cultured for 45 days in vitro and bone formation was assessed by expression of osteogenic genes, ALP secretion, and high resolution computed tomography. Our findings confirmed active bone repair process in ex vivo settings. Addition of ADSCs significantly accelerated the repair process and improved bone microarchitecture. This ex vivo bone defect model can emerge as a viable alternative to animal experimentation and also as a potent tool to evaluate patient specific bone therapeutics under controlled conditions.


Subject(s)
Adipose Tissue , Bone Regeneration , Cell Differentiation , Mesenchymal Stem Cells , Tissue Engineering , Tissue Scaffolds , Humans , Adipose Tissue/cytology , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Mesenchymal Stem Cells/cytology , Femur Head , Osteogenesis , Cells, Cultured , Bone Substitutes/chemistry , Durapatite/chemistry , Calcium Phosphates/chemistry
3.
Biomed Mater ; 19(5)2024 Jul 24.
Article in English | MEDLINE | ID: mdl-38986475

ABSTRACT

Bioactive and biodegradable scaffolds that mimic the natural extracellular matrix of bone serve as temporary structures to guide new bone tissue growth. In this study, 3D-printed scaffolds composed of poly (lactic acid) (PLA)-tricalcium phosphate (TCP) (90-10 wt.%) were modified with 1%, 5%, and 10 wt.% of ZnO to enhance bone tissue regeneration. A commercial chain extender named Joncryl was incorporated alongside ZnO to ensure the printability of the composites. Filaments were manufactured using a twin-screw extruder and subsequently used to print 3D scaffolds via fused filament fabrication (FFF). The scaffolds exhibited a homogeneous distribution of ZnO and TCP particles, a reproducible structure with 300 µm pores, and mechanical properties suitable for bone tissue engineering, with an elastic modulus around 100 MPa. The addition of ZnO resulted in enhanced surface roughness on the scaffolds, particularly for ZnO microparticles, achieving values up to 241 nm. This rougher topography was responsible for enhancing protein adsorption on the scaffolds, with an increase of up to 85% compared to the PLA-TCP matrix. Biological analyses demonstrated that the presence of ZnO promotes mesenchymal stem cell (MSC) proliferation and differentiation into osteoblasts. Alkaline phosphatase (ALP) activity, an important indicator of early osteogenic differentiation, increased up to 29%. The PLA-TCP composite containing 5% ZnO microparticles exhibited an optimized degradation rate and enhanced bioactivity, indicating its promising potential for bone repair applications.


Subject(s)
Biocompatible Materials , Bone Regeneration , Calcium Phosphates , Cell Differentiation , Cell Proliferation , Mesenchymal Stem Cells , Osteoblasts , Polyesters , Printing, Three-Dimensional , Tissue Engineering , Tissue Scaffolds , Zinc Oxide , Tissue Scaffolds/chemistry , Calcium Phosphates/chemistry , Polyesters/chemistry , Bone Regeneration/drug effects , Tissue Engineering/methods , Mesenchymal Stem Cells/cytology , Zinc Oxide/chemistry , Biocompatible Materials/chemistry , Cell Differentiation/drug effects , Osteoblasts/cytology , Osteogenesis/drug effects , Materials Testing , Bone and Bones , Guided Tissue Regeneration/methods , Humans , Animals , Alkaline Phosphatase/metabolism , Elastic Modulus , Porosity , Surface Properties
4.
ACS Appl Mater Interfaces ; 16(28): 37007-37016, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38953613

ABSTRACT

Osteoblasts and osteoclasts are two of the most important types of cells in bone repair, and their bone-forming and bone-resorbing activities influence the process of bone repair. In this study, we proposed a physicochemical bidirectional regulation strategy via ration by physically utilizing hydroxyapatite nanopatterning to recruit and induce MSCs osteogenic differentiation and by chemically inhibiting osteolysis activity through the loaded zoledronate. The nanorod-like hydroxyapatite coating was fabricated via a modified hydrothermal process while the zoledronic acid was loaded through the chelation within the calcium ions. The fabrication of a hydroxyapatite/zoledronic acid composite biomaterial. This biomaterial promotes bone tissue regeneration by physically utilizing hydroxyapatite nanopatterning to recruit and induce MSCs osteogenic differentiation and by chemically inhibiting osteolysis activity through the loaded zoledronate. The nanorod-like hydroxyapatite coating was fabricated via a modified hydrothermal process while the zoledronic acid was loaded through the chelation within the calcium ions. The in vitro results tested on MSCs and RAW 246.7 indicated that the hydroxyapatite enhanced cells' physical sensing system, therefore enhancing the osteogenesis. At the same time the zoledronic acid inhibited osteolysis by downregulating the RANK-related genes. This research provides a promising strategy for enhancing bone regeneration and contributes to the field of orthopedic implants.


Subject(s)
Bone Regeneration , Calcium Phosphates , Mesenchymal Stem Cells , Osteogenesis , Printing, Three-Dimensional , Zoledronic Acid , Bone Regeneration/drug effects , Animals , Osteogenesis/drug effects , Mice , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Zoledronic Acid/pharmacology , Zoledronic Acid/chemistry , Osteolysis/drug therapy , Durapatite/chemistry , Durapatite/pharmacology , Cell Differentiation/drug effects , RAW 264.7 Cells
5.
ACS Appl Mater Interfaces ; 16(28): 35964-35984, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38968558

ABSTRACT

Developing a neurovascular bone repair scaffold with an appropriate mechanical strength remains a challenge. Calcium phosphate (CaP) is similar to human bone, but its scaffolds are inherently brittle and inactive, which require recombination with active ions and polymers for bioactivity and suitable strength. This work discussed the synthesis of amorphous magnesium-calcium pyrophosphate (AMCP) and the subsequent development of a humidity-responsive AMCP/cassava starch (CS) scaffold. The scaffold demonstrated enhanced mechanical properties by strengthening the intermolecular hydrogen bonds and ionic bonds between AMCP and CS during the gelatinization and freeze-thawing processes. The release of active ions was rapid initially and stabilized into a long-term stable release after 3 days, which is well-matched with new bone growth. The release of pyrophosphate ions endowed the scaffold with antibacterial properties. At the cellular level, the released active ions simultaneously promoted the proliferation and mineralization of osteoblasts, the proliferation and migration of endothelial cells, and the proliferation of Schwann cells. At the animal level, the scaffold was demonstrated to promote vascular growth and peripheral nerve regeneration in a rat skull defect experiment, ultimately resulting in the significant and rapid repair of bone defects. The construction of the AMCP/CS scaffold offers practical suggestions and references for neurovascular bone repair.


Subject(s)
Bone Regeneration , Starch , Tissue Scaffolds , Animals , Bone Regeneration/drug effects , Tissue Scaffolds/chemistry , Rats , Starch/chemistry , Humidity , Humans , Cell Proliferation/drug effects , Rats, Sprague-Dawley , Diphosphates/chemistry , Diphosphates/pharmacology , Osteoblasts/drug effects , Osteoblasts/cytology , Calcium Pyrophosphate/chemistry , Calcium Pyrophosphate/pharmacology , Schwann Cells/drug effects , Schwann Cells/cytology , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Skull/drug effects
6.
Int J Mol Sci ; 25(14)2024 Jul 20.
Article in English | MEDLINE | ID: mdl-39063181

ABSTRACT

This study aimed to compare the biological properties of newly synthesized cements based on calcium phosphate with a commercially used cement, mineral trioxide aggregate (MTA). Strontium (Sr)-, Copper (Cu)-, and Zinc (Zn)-doped hydroxyapatite (miHAp) powder was obtained through hydrothermal synthesis and characterized by scanning electron microscopy (SEM), X-ray diffraction (XRD), and energy dispersive X-ray spectrometry (EDX). Calcium phosphate cement (CPC) was produced by mixing miHAp powder with a 20 wt.% citric acid solution, followed by the assessment of its compressive strength, setting time, and in vitro bioactivity. Acetylsalicylic acid (ASA) was added to the CPC, resulting in CPCA. Biological tests were conducted on CPC, CPCA, and MTA. The biocompatibility of the cement extracts was evaluated in vitro using human dental pulp stem cells (hDPSCs) and in vivo using a zebrafish model. Antibiofilm and antimicrobial effect (quantified by CFUs/mL) were assessed against Streptococcus mutans and Lactobacillus rhamnosus. None of the tested materials showed toxicity, while CPCA even increased hDPSCs proliferation. CPCA showed a better safety profile than MTA and CPC, and no toxic or immunomodulatory effects on the zebrafish model. CPCA exhibited similar antibiofilm effects against S. mutans and L. rhamnosus to MTA.


Subject(s)
Aspirin , Calcium Phosphates , Copper , Strontium , Zinc , Strontium/chemistry , Strontium/pharmacology , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacology , Humans , Animals , Aspirin/pharmacology , Aspirin/chemistry , Copper/chemistry , Zinc/chemistry , Zinc/pharmacology , Dental Cements/chemistry , Dental Cements/pharmacology , Biofilms/drug effects , Materials Testing , Zebrafish , Dental Pulp/cytology , Dental Pulp/drug effects , Streptococcus mutans/drug effects , Stem Cells/drug effects , X-Ray Diffraction , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cell Proliferation/drug effects
7.
Biomed Mater ; 19(5)2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38955344

ABSTRACT

Artificial bone substitutes for bone repair and reconstruction still face enormous challenges. Previous studies have shown that calcium magnesium phosphate cements (CMPCs) possess an excellent bioactive surface, but its clinical application is restricted due to short setting time. This study aimed to develop new CMPC/carboxymethyl chitosan (CMCS) comg of mixed powders of active MgO, calcined MgO and calcium dihydrogen phosphate monohydrate. With this novel strategy, it can adjust the setting time and improve the compressive strength. The results confirmed that CMPC/CMCS composite bone cements were successfully developed with a controllable setting time (18-70 min) and high compressive strength (87 MPa). In addition, the composite bone cements could gradually degrade in PBS with weight loss up to 32% at 28 d. They also promoted the proliferation of pre-osteoblasts, and induced osteogenic differentiation. The findings indicate that CMPC/CMCS composite bone cements hold great promise as a new type of bone repair material in further and in-depth studies.


Subject(s)
Biocompatible Materials , Bone Cements , Calcium Phosphates , Cell Differentiation , Cell Proliferation , Chitosan , Compressive Strength , Magnesium Compounds , Materials Testing , Osteoblasts , Osteogenesis , Chitosan/chemistry , Chitosan/analogs & derivatives , Bone Cements/chemistry , Bone Cements/pharmacology , Osteogenesis/drug effects , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacology , Cell Differentiation/drug effects , Animals , Cell Proliferation/drug effects , Mice , Osteoblasts/drug effects , Osteoblasts/cytology , Magnesium Compounds/chemistry , Magnesium Compounds/pharmacology , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Bone Substitutes/chemistry , Bone Substitutes/pharmacology , Phosphates
8.
J Nanobiotechnology ; 22(1): 407, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987801

ABSTRACT

Segmental bone defects, arising from factors such as trauma, tumor resection, and congenital malformations, present significant clinical challenges that often necessitate complex reconstruction strategies. Hydrogels loaded with multiple osteogenesis-promoting components have emerged as promising tools for bone defect repair. While the osteogenic potential of the Piezo1 agonist Yoda1 has been demonstrated previously, its hydrophobic nature poses challenges for effective loading onto hydrogel matrices.In this study, we address this challenge by employing Yoda1-pretreated bone marrow-derived mesenchymal stem cell (BMSCs) exosomes (Exo-Yoda1) alongside exosomes derived from BMSCs (Exo-MSC). Comparatively, Exo-Yoda1-treated BMSCs exhibited enhanced osteogenic capabilities compared to both control groups and Exo-MSC-treated counterparts. Notably, Exo-Yoda1-treated cells demonstrated similar functionality to Yoda1 itself. Transcriptome analysis revealed activation of osteogenesis-associated signaling pathways, indicating the potential transduction of Yoda1-mediated signals such as ErK, a finding validated in this study. Furthermore, we successfully integrated Exo-Yoda1 into gelatin methacryloyl (GelMA)/methacrylated sodium alginate (SAMA)/ß-tricalcium phosphate (ß-TCP) hydrogels. These Exo-Yoda1-loaded hydrogels demonstrated augmented osteogenesis in subcutaneous ectopic osteogenesis nude mice models and in rat skull bone defect model. In conclusion, our study introduces Exo-Yoda1-loaded GELMA/SAMA/ß-TCP hydrogels as a promising approach to promoting osteogenesis. This innovative strategy holds significant promise for future widespread clinical applications in the realm of bone defect reconstruction.


Subject(s)
Exosomes , Hydrogels , Mesenchymal Stem Cells , Osteogenesis , Osteogenesis/drug effects , Animals , Exosomes/metabolism , Mesenchymal Stem Cells/metabolism , Hydrogels/chemistry , Mice , MAP Kinase Signaling System/drug effects , Signal Transduction/drug effects , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacology , Rats , Male , Alginates/chemistry , Gelatin/chemistry , Cell Differentiation/drug effects , Bone Regeneration/drug effects , Cells, Cultured
9.
Sci Rep ; 14(1): 17304, 2024 07 27.
Article in English | MEDLINE | ID: mdl-39068177

ABSTRACT

In contrast to abiotically formed carbonates, biogenetic carbonates have been observed to be nanocomposite, organo-mineral structures, the basic build-blocks of which are particles of quasi-uniform size (10-100 nm) organized into complex higher-order hierarchical structures, typically with highly controlled crystal-axis alignments. Some of these characteristics serve as criteria for inferring a biological origin and the state of preservation of fossil carbonate materials, and to determine whether the biomineralization process was biologically induced or controlled. Here we show that a calcium storage structure formed by the American lobster, a gastrolith initially consisting of amorphous calcium carbonate (ACC) and amorphous calcium phosphate (ACP), post-mortem can crystallize into (thus secondary) calcite with structural properties strongly influenced by the inherited organic matrix. This secondary calcite meets many structural criteria for biominerals (thus called the biomorphic calcite), but differs in trace element distributions (e.g., P and Mg). Such observations refine the capability to determine whether a fossil carbonates can be attributed to biogenic processes, with implications for the record of life on Earth and other terrestrial planets.


Subject(s)
Calcium Carbonate , Crystallization , Fossils , Calcium Carbonate/chemistry , Calcium Carbonate/metabolism , Animals , Calcium Phosphates/chemistry , Calcium Phosphates/metabolism , Nephropidae/metabolism , Nephropidae/chemistry , Biomineralization
10.
BMC Biotechnol ; 24(1): 48, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38982413

ABSTRACT

BACKGROUND: Enamelin is an enamel matrix protein that plays an essential role in the formation of enamel, the most mineralized tissue in the human body. Previous studies using animal models and proteins from natural sources point to a key role of enamelin in promoting mineralization events during enamel formation. However, natural sources of enamelin are scarce and with the current study we therefore aimed to establish a simple microbial production method for recombinant human enamelin to support its use as a mineralization agent. RESULTS: In the study the 32 kDa fragment of human enamelin was successfully expressed in Escherichia coli and could be obtained using immobilized metal ion affinity chromatography purification (IMAC), dialysis, and lyophilization. This workflow resulted in a yield of approximately 10 mg enamelin per liter culture. Optimal conditions for IMAC purification were obtained using Ni2+ as the metal ion, and when including 30 mM imidazole during binding and washing steps. Furthermore, in vitro mineralization assays demonstrated that the recombinant enamelin could promote calcium phosphate mineralization at a concentration of 0.5 mg/ml. CONCLUSIONS: These findings address the scarcity of enamelin by facilitating its accessibility for further investigations into the mechanism of enamel formation and open new avenues for developing enamel-inspired mineralized biomaterials.


Subject(s)
Dental Enamel Proteins , Escherichia coli , Recombinant Proteins , Humans , Escherichia coli/genetics , Escherichia coli/metabolism , Dental Enamel Proteins/metabolism , Dental Enamel Proteins/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Chromatography, Affinity , Calcium Phosphates/metabolism , Calcium Phosphates/chemistry
11.
J Agric Food Chem ; 72(28): 15523-15529, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38963614

ABSTRACT

The eggshell is a composite and highly ordered structure formed by biomineralization. Besides other functions, it has a vital and intricate role in the protection of an embryo from various potentially harsh environmental conditions. Solid-state nuclear magnetic resonance (SSNMR) has been used for detailed structural investigations of the chicken, tinamou, and flamingo eggshell materials. 31P NMR spectra reveal that hydroxyapatite and ß-tricalcium phosphate in the ratio 3:2 represent major constituents of phosphate species in the eggshells. All three eggshells exhibit similar spectra, except for the line widths, which implies different structural order of phosphate species in the chicken, tinamou, and flamingo eggshells. 1H NMR spectra for these materials are comparable, differentiating overlapped peaks in three spectral regions at around 7, 4-5, and 1-2 ppm. These spectral regions have been attributed to protons from NH or CaHCO3, water, and possibly isolated monomeric water molecules or hydroxyl groups in calcium-deficient hydroxyapatite. 1H-13C CP MAS NMR revealed the presence of organic matter in the form of lipids and proteins. Two overlapped resonances in the carbonyl region at around 173 and 169 ppm are assigned to the carbonyls of the peptide bonds and the bicarbonate unit in calcite, respectively. Fourier-transform infrared spectroscopy (FTIR) spectra confirmed the presence of structural units detected in the NMR spectra.


Subject(s)
Chickens , Egg Shell , Magnetic Resonance Spectroscopy , Animals , Egg Shell/chemistry , Magnetic Resonance Spectroscopy/methods , Durapatite/chemistry , Birds , Calcium Phosphates/chemistry
12.
J Nanobiotechnology ; 22(1): 453, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-39080653

ABSTRACT

Bioactive agents have demonstrated regenerative potential for cell-free bone tissue engineering. Nevertheless, certain challenges persist, including ineffective delivery methods and confined therapeutic potency. Here, we demonstrated that the biomimetic calcium phosphate coating system (BioCaP) could effectively uptake and slowly release the incorporated bioactive agents compared to the surface absorption system via osteoclast-mediated degradation of BioCaP coatings. The release kinetics were determined as a function of time. The release rate was stable without remarkable burst release during the first 1 day, followed by a sustained release from day 7 to day 19. Then, we developed the bi-functional BioCaP-coated silk fibroin scaffolds enabling the effective co-delivery of TGF-ß3 and BMP-2 (SFI-T/SFI-B) and the corresponding slow release of TGF-ß3 and BMP-2 exhibited superior potential in promoting chondrogenesis and osteogenesis without impairing cell vitality in vitro. The SFI-T/SFI-B scaffolds could improve cartilage and bone regeneration in 5 × 4 mm rabbit osteochondral (OC) defect. These findings indicate that the biomimetic calcium-phosphate coated silk fibroin scaffolds with slowly co-released TGF-ß3 and BMP-2 effectively promote the repair of OC defects, hence facilitating the future clinical translation of controlled drug delivery in tissue engineering.


Subject(s)
Bone Morphogenetic Protein 2 , Bone Regeneration , Calcium Phosphates , Fibroins , Osteogenesis , Tissue Engineering , Tissue Scaffolds , Transforming Growth Factor beta3 , Fibroins/chemistry , Fibroins/pharmacology , Animals , Bone Morphogenetic Protein 2/pharmacology , Transforming Growth Factor beta3/pharmacology , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacology , Rabbits , Tissue Scaffolds/chemistry , Bone Regeneration/drug effects , Tissue Engineering/methods , Osteogenesis/drug effects , Chondrogenesis/drug effects , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Bombyx , Male
13.
Int J Pharm ; 661: 124467, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39004293

ABSTRACT

Tablet disintegration is crucial for drug release and subsequent systemic absorption. Although factors affecting the disintegrant's functionality have been extensively studied, the impact of wet granulation on the performance of disintegrants in a poorly water-soluble matrix has received much less attention. In this study, the disintegrants, crospovidone (XPVP), croscarmellose sodium (CCS) and sodium starch glycolate (SSG), were wet-granulated with dibasic calcium phosphate dihydrate as the poorly water-soluble matrix and polyvinylpyrrolidone as the binder. The effect of wet granulation was studied by evaluating tablet tensile strength and disintegratability. Comparison between tablets with granulated or ungranulated disintegrants as well those without disintegrants were also made. Different formulations showed different degrees of sensitivity to changes in tablet tensile strength and disintegratability post-wet granulation. Tablet tensile strength decreased for tablets with granulated disintegrant XPVP or CCS, but to a smaller extent for SSG. While tablets with granulated XPVP or CCS had increased disintegration time, the increment was lesser than for SSG, suggesting that wet granulation impacted a swelling disintegrant more. The findings showed that tablets with wet-granulated disintegrant had altered the disintegrant's functionality. These findings could provide better insights into changes in the disintegrant's functionality after wet granulation.


Subject(s)
Calcium Phosphates , Carboxymethylcellulose Sodium , Excipients , Povidone , Solubility , Starch , Tablets , Tensile Strength , Water , Carboxymethylcellulose Sodium/chemistry , Povidone/chemistry , Starch/chemistry , Starch/analogs & derivatives , Excipients/chemistry , Water/chemistry , Calcium Phosphates/chemistry , Drug Compounding/methods , Drug Liberation , Chemistry, Pharmaceutical/methods
14.
J Mater Sci Mater Med ; 35(1): 40, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39073605

ABSTRACT

Bone infections are still a major problem in surgery. To avoid severe side effects of systemically administered antibiotics, local antibiotic therapy is increasingly being considered. Using a pressure-based method developed in our group, microporous ß-TCP ceramics, which had previously been characterized, were loaded with 2% w/v alginate containing 50 mg/mL clindamycin and 10 µg/mL rhBMP-2. Release experiments were then carried out over 28 days with changes of liquid at defined times (1, 2, 3, 6, 9, 14, 21 and 28d). The released concentrations of clindamycin were determined by HPLC and those of rhBMP-2 by ELISA. Continuous release (anomalous transport) of clindamycin and uniform release (Fick's diffusion) of BMP-2 were determined. The composites were biocompatible (live/dead, WST-I and LDH) and the released concentrations were all antimicrobially active against Staph. aureus. The results were very promising and clindamycin was detected in concentrations above the MIC as well as a constant rhBMP-2 release over the entire study period. Biocompatibility was also not impaired by either the antibiotic or the BMP-2. This promising approach can therefore be seen as an alternative to the common treatment with PMMA chains containing gentamycin, as the new composite is completely biodegradable and no second operation is necessary for removal or replacement.


Subject(s)
Anti-Bacterial Agents , Biocompatible Materials , Bone Morphogenetic Protein 2 , Clindamycin , Staphylococcus aureus , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/administration & dosage , Bone Morphogenetic Protein 2/chemistry , Bone Morphogenetic Protein 2/pharmacokinetics , Clindamycin/administration & dosage , Clindamycin/chemistry , Clindamycin/pharmacokinetics , Humans , Biocompatible Materials/chemistry , Staphylococcus aureus/drug effects , Kinetics , Calcium Phosphates/chemistry , Animals , Materials Testing , Recombinant Proteins/chemistry , Ceramics/chemistry , Transforming Growth Factor beta , Alginates/chemistry , Absorbable Implants , Microbial Sensitivity Tests
15.
Biomater Adv ; 163: 213968, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39059113

ABSTRACT

Strontium, cobalt, and manganese ions are present in the composition of bone and useful for bone metabolism, even when combined with calcium phosphate in the composition of biomaterials. Herein we explored the possibility to include these ions in the composition of apatitic materials prepared through the cementitious reaction between ion-substituted calcium phosphate dibasic dihydrate, CaHPO4·2H2O (DCPD) and tetracalcium phosphate, Ca4(PO4)2O (TTCP). The results of the chemical, structural, morphological and mechanical characterization indicate that cobalt and manganese exhibit a greater delaying effect than strontium (about 15 at.%) on the cementitious reaction, even though they are present in smaller amounts within the materials (about 0.8 and 4.5 at.%, respectively). Furthermore, the presence of the foreign ions in the apatitic materials leads to a slight reduction of porosity and to enhancement of compressive strength. The results of biological tests show that the presence of strontium and manganese, as well as calcium, in the apatitic materials cultured in direct contact with human mesenchymal stem cells (hMSCs) stimulates their viability and activity. In contrast, the apatitic material containing cobalt exhibits a lower metabolic activity. All the materials have a positive effect on the expression of Vascular Endothelial Growth Factor (VEGF) and Von Willebrand Factor (vWF). Moreover, the apatitic material containing strontium induces the most significant reduction in the differentiation of preosteoclasts into osteoclasts, demonstrating not only osteogenic and angiogenic properties, but also ability to regulate bone resorption.


Subject(s)
Bone Regeneration , Cobalt , Manganese , Mesenchymal Stem Cells , Osteogenesis , Strontium , Strontium/pharmacology , Strontium/chemistry , Cobalt/chemistry , Humans , Osteogenesis/drug effects , Manganese/chemistry , Manganese/pharmacology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Bone Regeneration/drug effects , Neovascularization, Physiologic/drug effects , Biocompatible Materials/pharmacology , Biocompatible Materials/chemistry , Vascular Endothelial Growth Factor A/metabolism , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacology , Cell Survival/drug effects , Angiogenesis
16.
Int J Nanomedicine ; 19: 6659-6676, 2024.
Article in English | MEDLINE | ID: mdl-38975320

ABSTRACT

Background: Vital pulp therapy (VPT) is considered a conservative treatment for preserving pulp viability in caries and trauma-induced pulpitis. However, Mineral trioxide aggregate (MTA) as the most frequently used repair material, exhibits limited efficacy under inflammatory conditions. This study introduces an innovative nanocomposite hydrogel, tailored to simultaneously target anti-inflammation and dentin mineralization, aiming to efficiently preserve vital pulp tissue. Methods: The L-(CaP-ZnP)/SA nanocomposite hydrogel was designed by combining L-Arginine modified calcium phosphate/zinc phosphate nanoparticles (L-(CaP-ZnP) NPs) with sodium alginate (SA), and was characterized with TEM, SEM, FTIR, EDX, ICP-AES, and Zeta potential. In vitro, we evaluated the cytotoxicity and anti-inflammatory properties. Human dental pulp stem cells (hDPSCs) were cultured with lipopolysaccharide (LPS) to induce an inflammatory response, and the cell odontogenic differentiation was measured and possible signaling pathways were explored by alkaline phosphatase (ALP)/alizarin red S (ARS) staining, qRT-PCR, immunofluorescence staining, and Western blotting, respectively. In vivo, a pulpitis model was utilized to explore the potential of the L-(CaP-ZnP)/SA nanocomposite hydrogel in controlling pulp inflammation and enhancing dentin mineralization by Hematoxylin and eosin (HE) staining and immunohistochemistry staining. Results: In vitro experiments revealed that the nanocomposite hydrogel was synthesized successfully and presented desirable biocompatibility. Under inflammatory conditions, compared to MTA, the L-(CaP-ZnP)/SA nanocomposite hydrogel demonstrated superior anti-inflammatory and pro-odontogenesis effects. Furthermore, the nanocomposite hydrogel significantly augmented p38 phosphorylation, implicating the involvement of the p38 signaling pathway in pulp repair. Significantly, in a rat pulpitis model, the L-(CaP-ZnP)/SA nanocomposite hydrogel downregulated inflammatory markers while upregulating mineralization-related markers, thereby stimulating the formation of robust reparative dentin. Conclusion: The L-(CaP-ZnP)/SA nanocomposite hydrogel with good biocompatibility efficiently promoted inflammation resolution and enhanced dentin mineralization by activating p38 signal pathway, as a pulp-capping material, offering a promising and advanced solution for treatment of pulpitis.


Subject(s)
Alginates , Anti-Inflammatory Agents , Dental Pulp , Hydrogels , Nanocomposites , Dental Pulp/cytology , Dental Pulp/drug effects , Humans , Hydrogels/chemistry , Hydrogels/pharmacology , Nanocomposites/chemistry , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Alginates/chemistry , Alginates/pharmacology , Pulpitis/therapy , Stem Cells/drug effects , Stem Cells/cytology , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacology , Silicates/chemistry , Silicates/pharmacology , Rats , Cell Differentiation/drug effects , Calcium Compounds/chemistry , Calcium Compounds/pharmacology , Cells, Cultured , Aluminum Compounds/chemistry , Aluminum Compounds/pharmacology , Arginine/chemistry , Arginine/pharmacology , Rats, Sprague-Dawley , Drug Combinations , Male , Oxides/chemistry , Oxides/pharmacology
17.
Int J Biol Macromol ; 274(Pt 1): 133267, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38906359

ABSTRACT

While it is known that calcium phosphate (CaP) minerals deposit in elastin-rich medial layers of arteries during medial calcification, their nucleation and growth sites are still debated. Neutral carbonyl groups and carboxylate groups are possible candidates. Also, while it is known that elastin degradation leads to calcification, it is unclear whether this is due to formation of new carboxylate groups or elastin fragmentation. In this work, we disentangle effects of carboxylate groups and particle size on elastin calcification; in doing so, we shed light on CaP mineralization sites on elastin. We find carboxylate groups accelerate calcification only in early stages; they mainly function as Ca2+ ion chelation sites but not calcification sites. Their presence promotes formation (likely on Ca2+ ions adsorbed on nearby carbonyl groups) of CaP minerals with high calcium-to-phosphate ratio as intermediate phases. Larger elastin particles calcify slower but reach similar amounts of CaP minerals in late stages; they promote direct formation of hydroxyapatite and CaP minerals with low calcium-to-phosphate ratio as intermediate phases. This work provides new perspectives on how carboxylate groups and elastin particle size influence calcification; these parameters can be tuned to study the mechanism of medial calcification and design drugs to inhibit the process.


Subject(s)
Calcium Phosphates , Elastin , Particle Size , Elastin/metabolism , Elastin/chemistry , Calcium Phosphates/chemistry , Calcium Phosphates/metabolism , Animals , Carboxylic Acids/chemistry , Vascular Calcification/metabolism , Vascular Calcification/pathology , Calcium/metabolism , Durapatite/chemistry
18.
Biomed Res ; 45(3): 103-113, 2024.
Article in English | MEDLINE | ID: mdl-38839353

ABSTRACT

Kidney stone disease is a serious disease due to the severe pain it causes, high morbidity, and high recurrence rate. Notably, calcium oxalate stones are the most common type of kidney stone. Calcium oxalate appears in two forms in kidney stones: the stable phase, monohydrate (COM), and the metastable phase, dihydrate (COD). Particularly, COM stones with concentric structures are hard and difficult to treat. However, the factor determining the growth of either COM or COD crystals in the urine, which is supersaturated for both phases, remains unclear. This study shows that calcium phosphate ingredients preferentially induce COM crystal nucleation and growth, by observing and analyzing kidney stones containing both COM and COD crystals. The forms of calcium phosphate are not limited to Randall's plaques (1-2 mm size aggregates, which contain calcium phosphate nanoparticles and proteins, and form in the renal papilla). For example, aggregates of strip-shaped calcium phosphate crystals and fields of dispersed calcium phosphate microcrystals (nano to micrometer order) also promote the growth of concentric COM structures. This suggests that patients who excrete urine with a higher quantity of calcium phosphate crystals may be more prone to forming hard and troublesome COM stones.


Subject(s)
Calcium Oxalate , Calcium Phosphates , Crystallization , Kidney Calculi , Calcium Phosphates/metabolism , Calcium Phosphates/chemistry , Calcium Oxalate/chemistry , Calcium Oxalate/metabolism , Calcium Oxalate/urine , Kidney Calculi/chemistry , Kidney Calculi/metabolism , Humans , Animals
19.
Int J Nanomedicine ; 19: 5109-5123, 2024.
Article in English | MEDLINE | ID: mdl-38846643

ABSTRACT

Introduction: Lumbar interbody fusion is widely employed for both acute and chronic spinal diseases interventions. However, large incision created during interbody cage implantation may adversely impair spinal tissue and influence postoperative recovery. The aim of this study was to design a shape memory interbody fusion device suitable for small incision implantation. Methods: In this study, we designed and fabricated an intervertebral fusion cage that utilizes near-infrared (NIR) light-responsive shape memory characteristics. This cage was composed of bisphenol A diglycidyl ether, polyether amine D-230, decylamine and iron oxide nanoparticles. A self-hardening calcium phosphate-starch cement (CSC) was injected internally through the injection channel of the cage for healing outcome improvement. Results: The size of the interbody cage is reduced from 22 mm to 8.8 mm to minimize the incision size. Subsequent NIR light irradiation prompted a swift recovery of the cage shape within 5 min at the lesion site. The biocompatibility of the shape memory composite was validated through in vitro MC3T3-E1 cell (osteoblast-like cells) adhesion and proliferation assays and subcutaneous implantation experiments in rats. CSC was injected into the cage, and the relevant results revealed that CSC is uniformly dispersed within the internal space, along with the cage compressive strength increasing from 12 to 20 MPa. Conclusion: The results from this study thus demonstrated that this integrated approach of using a minimally invasive NIR shape memory spinal fusion cage with CSC has potential for lumbar interbody fusion.


Subject(s)
Spinal Fusion , Spinal Fusion/instrumentation , Spinal Fusion/methods , Animals , Mice , Rats , Calcium Phosphates/chemistry , Minimally Invasive Surgical Procedures/instrumentation , Minimally Invasive Surgical Procedures/methods , Lumbar Vertebrae/surgery , Rats, Sprague-Dawley , Male , Compressive Strength , Cell Proliferation/drug effects , Bone Cements/chemistry , Smart Materials/chemistry , Cell Adhesion/drug effects
20.
J Biomed Mater Res B Appl Biomater ; 112(6): e35434, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38874589

ABSTRACT

Bioactive degradable scaffolds that facilitate bone healing while fighting off initial bacterial infection have the potential to change established strategies of dealing with traumatic bone injuries. To achieve this a composite material made from calcium phosphate graphene (CaPG), and MXene was synthesized. CaPG was created by functionalizing graphene oxide with phosphate groups in the presence of CaBr with a Lewis acid catalyst. Through this transformation, Ca2+ and PO4 3- inducerons are released as the material degrades thereby aiding in the process of osteogenesis. The 2D MXene sheets, which have shown to have antibacterial properties, were made by etching the Al from a layered Ti3AlC2 (MAX phase) using HF. The hot-pressed scaffolds made of these materials were designed to combat the possibility of infection during initial surgery and failure of osteogenesis to occur. These two failure modes account for a large percentage of issues that can arise during the treatment of traumatic bone injuries. These scaffolds were able to retain induceron-eluting properties in various weight percentages and bring about osteogenesis with CaPG alone and 2 wt% MXene scaffolds demonstrating increased osteogenic activity as compared to no treatment. Additionally, added MXene provided antibacterial properties that could be seen at as little as 2 wt%. This CaPG and MXene composite provides a possible avenue for developing osteogenic, antibacterial materials for treating bone injuries.


Subject(s)
Anti-Bacterial Agents , Calcium Phosphates , Graphite , Osteogenesis , Tissue Scaffolds , Titanium , Osteogenesis/drug effects , Graphite/chemistry , Graphite/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Titanium/chemistry , Titanium/pharmacology , Tissue Scaffolds/chemistry , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacology , Animals , Humans , Escherichia coli/drug effects , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL