Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.381
Filter
2.
Int J Artif Organs ; 47(5): 338-346, 2024 May.
Article in English | MEDLINE | ID: mdl-38693724

ABSTRACT

In the present study, porous silk fibroin sponges (SFS) were prepared using silk fibroin (SF), fish bone collagen (FBC), and olive oil (OO). The study investigates the potential use of using this sponge as skin tissue regeneration. The sponge was characterized for its physicochemical, mechanical, antimicrobial, and drug release properties. An in vitro study was carried out using human keratinocyte cell line (HaCaT). Biodegradation study using enzymatic method was carried out. The results showed that the mechanical properties such as tensile strength (23.40 ± 0.05 MPa), elongation at break (14.25 ± 0.02%), and water absorption (30.23 ± 0.01%) of the SFS were excellent, indicating promising performance. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays proved the biocompatible nature of the SFS. The SFS exhibited outstanding antibacterial properties against E. coli (4.72 ± 0.05 mm) and S. aureus (4.98 ± 0.07 mm). The developed SFS promote a promising solution for skin tissue regeneration and wound dressing.


Subject(s)
Anti-Bacterial Agents , Collagen , Fibroins , Regeneration , Skin , Staphylococcus aureus , Tissue Scaffolds , Wound Healing , Fibroins/chemistry , Fibroins/pharmacology , Wound Healing/drug effects , Humans , Collagen/metabolism , Animals , Regeneration/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/administration & dosage , Skin/drug effects , Skin/metabolism , Staphylococcus aureus/drug effects , HaCaT Cells , Escherichia coli/drug effects , Keratinocytes/drug effects , Olive Oil , Bone and Bones/drug effects , Bone and Bones/metabolism , Fishes , Tensile Strength , Porosity , Biocompatible Materials , Cell Line
3.
Int J Biol Macromol ; 269(Pt 2): 132124, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38723802

ABSTRACT

Bacterial cellulose (BC) hydrogel is renowned in the field of tissue engineering for its high biocompatibility, excellent mechanical strength, and eco-friendliness. Herein, we present a biomimetic mineralization method for preparing BC/hydroxyapatite (HAP) composite hydrogel scaffolds with different mineralization time and ion concentration of the mineralized solution. Spherical HAP reinforcement enhanced bone mineralization, thereby imparting increased bioactivity to BC matrix materials. Subsequently, platelet-rich plasma (PRP) was introduced into the scaffold. The PRP-loaded hydrogel enhanced the release of growth factors, which promoted cell adhesion, growth, and bone healing. After 3 weeks of MC3T3-E1 cell-induced osteogenesis, PRP positively affected cell differentiation in BC/HAP@PRP scaffolds. Overall, these scaffolds exhibited excellent biocompatibility, mineralized nodule formation, and controlled release in vitro, demonstrating great potential for application in bone tissue repair.


Subject(s)
Cellulose , Durapatite , Hydrogels , Osteogenesis , Platelet-Rich Plasma , Tissue Engineering , Tissue Scaffolds , Platelet-Rich Plasma/chemistry , Tissue Engineering/methods , Durapatite/chemistry , Durapatite/pharmacology , Cellulose/chemistry , Cellulose/pharmacology , Animals , Mice , Tissue Scaffolds/chemistry , Hydrogels/chemistry , Osteogenesis/drug effects , Bone and Bones/drug effects , Bone and Bones/metabolism , Delayed-Action Preparations/pharmacology , Cell Differentiation/drug effects , Biomimetics/methods , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Cell Line , Bone Regeneration/drug effects
4.
Biomed Mater Eng ; 35(3): 265-278, 2024.
Article in English | MEDLINE | ID: mdl-38728179

ABSTRACT

BACKGROUND: Bone defects arising from diverse causes, such as traffic accidents, contemporary weapon usage, and bone-related disorders, present significant challenges in clinical treatment. Prolonged treatment cycles for bone defects can result in complications, impacting patients' overall quality of life. Efficient and timely repair of bone defects is thus a critical concern in clinical practice. OBJECTIVE: This study aims to assess the scientific progress and achievements of magnesium phosphate bone cement (MPC) as an artificial bone substitute material. Additionally, the research seeks to explore the future development path and clinical potential of MPC bone cement in addressing challenges associated with bone defects. METHODS: The study comprehensively reviews MPC's performance, encompassing e.g. mechanical properties, biocompatibility, porosity, adhesion and injectability. Various modifiers are also considered to broaden MPC's applications in bone tissue engineering, emphasizing drug-loading performance and antibacterial capabilities, which meet clinical diversification requirements. RESULTS: In comparison to alternatives such as autogenous bone transplantation, allograft, polymethyl methacrylate (PMMA), and calcium phosphate cement (CPC), MPC emerges as a promising solution for bone defects. It addresses limitations associated with these alternatives, such as immunological rejection and long-term harm to patients. MPC can control heat release during the curing process, exhibits superior mechanical strength, and has the capacity to stimulate new bone growth. CONCLUSION: MPC stands out as an artificial bone substitute with appropriate mechanical strength, rapid degradation, non-toxicity, and good biocompatibility, facilitating bone repair and regeneration. Modification agents can enhance its clinical versatility. Future research should delve into its mechanical properties and formulations, expanding clinical applications to create higher-performing and more medically valuable alternatives in bone defect repair.


Subject(s)
Bone Cements , Bone Substitutes , Magnesium Compounds , Phosphates , Bone Cements/chemistry , Bone Cements/therapeutic use , Humans , Phosphates/chemistry , Magnesium Compounds/chemistry , Magnesium Compounds/therapeutic use , Bone Substitutes/therapeutic use , Bone Substitutes/chemistry , Animals , Bone Regeneration/drug effects , Porosity , Materials Testing , Bone and Bones/drug effects
5.
Calcif Tissue Int ; 115(1): 85-96, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38733412

ABSTRACT

Autosomal dominant osteopetrosis type 2 (ADO2) is a rare inherited bone disorder characterised by dense but brittle bones. It displays striking phenotypic variability, with the most severe symptoms, including blindness and bone marrow failure. Disease management largely relies on symptomatic treatment since there is no safe and effective treatment. Most ADO2 cases are caused by heterozygous loss-of-function mutations in the CLCN7 gene, which encodes an essential Cl-/H+ antiporter for proper bone resorption by osteoclasts. Thus, siRNA-mediated silencing of the mutant allele is a promising therapeutic approach, but targeting bone for first-in-human translation remains challenging. Here, we demonstrate the utility of silicon-stabilised hybrid lipid nanoparticles (sshLNPs) as a next-generation nucleic acid nanocarrier capable of delivering allele-specific siRNA to bone. Using a Clcn7G213R knock-in mouse model recapitulating one of the most common human ADO2 mutations and based on the 129S genetic background (which produces the most severe disease phenotype amongst current models), we show substantial knockdown of the mutant allele in femur when siRNA targeting the pathogenic variant is delivered by sshLNPs. We observed lower areal bone mineral density in femur and reduced trabecular thickness in femur and tibia, when siRNA-loaded sshLNPs were administered subcutaneously (representing the most relevant administration route for clinical adoption and patient adherence). Importantly, sshLNPs have improved stability over conventional LNPs and enable 'post hoc loading' for point-of-care formulation. The treatment was well tolerated, suggesting that sshLNP-enabled gene therapy might allow successful clinical translation of essential new treatments for ADO2 and potentially other rare genetic bone diseases.


Subject(s)
Alleles , Chloride Channels , Nanoparticles , Osteopetrosis , Phenotype , RNA, Small Interfering , Animals , Chloride Channels/genetics , Osteopetrosis/genetics , Osteopetrosis/therapy , Mice , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Bone and Bones/metabolism , Bone and Bones/drug effects , Disease Models, Animal
6.
Molecules ; 29(10)2024 May 17.
Article in English | MEDLINE | ID: mdl-38792228

ABSTRACT

Vitamin D, an essential micronutrient crucial for skeletal integrity and various non-skeletal physiological functions, exhibits limited bioavailability and stability in vivo. This study is focused on the development of polyethylene glycol (PEG)-grafted phospholipid micellar nanostructures co-encapsulating vitamin D3 and conjugated with alendronic acid, aimed at active bone targeting. Furthermore, these nanostructures are rendered optically traceable in the UV-visible region of the electromagnetic spectrum via the simultaneous encapsulation of vitamin D3 with carbon dots, a newly emerging class of fluorescents, biocompatible nanoparticles characterized by their resistance to photobleaching and environmental friendliness, which hold promise for future in vitro bioimaging studies. A systematic investigation is conducted to optimize experimental parameters for the preparation of micellar nanostructures with an average hydrodynamic diameter below 200 nm, ensuring colloidal stability in physiological media while preserving the optical luminescent properties of the encapsulated carbon dots. Comprehensive chemical-physical characterization of these micellar nanostructures is performed employing optical and morphological techniques. Furthermore, their binding affinity for the principal inorganic constituent of bone tissue is assessed through a binding assay with hydroxyapatite nanoparticles, indicating significant potential for active bone-targeting. These formulated nanostructures hold promise for novel therapeutic interventions to address skeletal-related complications in cancer affected patients in the future.


Subject(s)
Alendronate , Bone and Bones , Cholecalciferol , Micelles , Nanostructures , Cholecalciferol/chemistry , Nanostructures/chemistry , Bone and Bones/drug effects , Bone and Bones/metabolism , Alendronate/chemistry , Polyethylene Glycols/chemistry , Humans , Drug Delivery Systems , Luminescence , Nanoparticles/chemistry , Drug Carriers/chemistry , Quantum Dots/chemistry
7.
Mar Drugs ; 22(5)2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38786592

ABSTRACT

Malnutrition is one of the major factors of bone and cartilage disorders. Pacific cod (Gadus macrocephalus) processing waste is a cheap and highly promising source of bioactive substances, including collagen-derived peptides and amino acids, for bone and cartilage structure stabilization. The addition of these substances to a functional drink is one of the ways to achieve their fast intestinal absorption. Collagen hydrolysate was obtained via enzymatic hydrolysis, ultrafiltration, freeze-drying, and grinding to powder. The lyophilized hydrolysate was a light gray powder with high protein content (>90%), including collagen (about 85% of total protein) and a complete set of essential and non-essential amino acids. The hydrolysate had no observed adverse effect on human mesenchymal stem cell morphology, viability, or proliferation. The hydrolysate was applicable as a protein food supply or a structure-forming food component due to the presence of collagen fiber fragments. An isotonic fitness drink (osmolality 298.1 ± 2.1 mOsm/L) containing hydrolysate and vitamin C as a cofactor in collagen biosynthesis was prepared. The addition of the hydrolysate did not adversely affect its organoleptic parameters. The production of such functional foods and drinks is one of the beneficial ways of fish processing waste utilization.


Subject(s)
Bone and Bones , Cartilage , Collagen , Gadiformes , Protein Hydrolysates , Animals , Collagen/metabolism , Humans , Cartilage/drug effects , Cartilage/metabolism , Bone and Bones/drug effects , Bone and Bones/metabolism , Protein Hydrolysates/pharmacology , Protein Hydrolysates/chemistry , Mesenchymal Stem Cells/drug effects , Beverages , Functional Food , Hydrolysis
8.
Med Sci Monit ; 30: e942485, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38814863

ABSTRACT

BACKGROUND The healing of bone defects is a serious challenge worldwide. One branch of dentistry deals with bone defects. Capsaicin has anti-inflammatory, anti-oxidative, and cholesterol-reducing effects. The aim of this study was to evaluate the effects of systemic capsaicin administered at different doses on bone healing. MATERIAL AND METHODS A total of 32 male wistar rats was used, their weight varying between 250 and 300 g. The rats were randomly divided into 4 groups of 8 rats each. The analyses served to evaluate the effect on healing of different doses of capsaicin and grafts. A significant increase was observed in the number of osteoblasts in the capsaicin-applied groups, compared with the control group. RESULTS The analyses served to evaluate the effect on healing of different doses of capsaicin and grafts. A significant increase was observed in the number of osteoblasts in the capsaicin-applied groups, compared with that of the control group. The inflammation scores showed a significant difference only in the control group and in the group administered with 50 mg/kg capsaicin (P=0.010). The osteoclast counts were significantly different between all groups. CONCLUSIONS As a result of the analyses, positive effects on bone healing were observed when capsaicin 0.25 mg/kg and 0.50 mg/kg was administered intraperitoneally. However, more studies are needed for more accurate information.


Subject(s)
Capsaicin , Osteoblasts , Rats, Wistar , Animals , Capsaicin/pharmacology , Male , Rats , Osteoblasts/drug effects , Osteoblasts/metabolism , Wound Healing/drug effects , Osteoclasts/drug effects , Osteoclasts/metabolism , Bone and Bones/drug effects , Bone Regeneration/drug effects
9.
PLoS One ; 19(5): e0300292, 2024.
Article in English | MEDLINE | ID: mdl-38718051

ABSTRACT

The aim of the study was to investigate the effect of returning to a balanced diet combined with chromium picolinate (CrPic) or chromium nanoparticles (CrNPs) supplementation at a pharmacologically relevant dose of 0.3 mg/kg body weight on the expression level of selected genes and bone turnover markers in the blood and bones of rats fed an obese diet. The results of the study showed that chronic intake of a high-fat obesogenic diet negatively affects bone turnover by impairing processes of both synthesis and degradation of bones. The switch to a healthy diet proved insufficient to regulate bone metabolism disorders induced by an obesogenic diet, even when it was supplemented with chromium, irrespective of its form. Supplementation with CrPic with no change in diet stimulated bone metabolism only at the molecular level, towards increased osteoclastogenesis (bone resorption). In contrast, CrNPs added to the high-fat diet effectively regulated bone turnover by increasing both osteoblastogenesis and osteoclastogenesis, with these changes directed more towards bone formation. The results of the study suggest that unfavourable changes in bone metabolism induced by chronic intake of a high-fat diet can be mitigated by supplementation with CrNPs, whereas a change in eating habits fails to achieve a similar effect.


Subject(s)
Bone Remodeling , Chromium , Diet, High-Fat , Animals , Diet, High-Fat/adverse effects , Rats , Chromium/administration & dosage , Chromium/pharmacology , Male , Bone Remodeling/drug effects , Nanoparticles/chemistry , Dietary Fiber/pharmacology , Picolinic Acids/pharmacology , Picolinic Acids/administration & dosage , Dietary Supplements , Bone and Bones/metabolism , Bone and Bones/drug effects , Rats, Wistar , Metal Nanoparticles/chemistry , Metal Nanoparticles/administration & dosage , Osteogenesis/drug effects
10.
J Transl Med ; 22(1): 437, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38720345

ABSTRACT

BACKGROUND: Biological-derived hydroxyapatite is widely used as a bone substitute for addressing bone defects, but its limited osteoconductive properties necessitate further improvement. The osteo-immunomodulatory properties hold crucial promise in maintaining bone homeostasis, and precise modulation of macrophage polarization is essential in this process. Metabolism serves as a guiding force for immunity, and fluoride modification represents a promising strategy for modulating the osteoimmunological environment by regulating immunometabolism. In this context, we synthesized fluorinated porcine hydroxyapatite (FPHA), and has demonstrated its enhanced biological properties and osteogenic capacity. However, it remains unknown whether and how FPHA affects the immune microenvironment of the bone defects. METHODS: FPHA was synthesized and its composition and structural properties were confirmed. Macrophages were cultured with FPHA extract to investigate the effects of FPHA on their polarization and the related osteo-immune microenvironment. Furthermore, total RNA of these macrophages was extracted, and RNA-seq analysis was performed to explore the underlying mechanisms associated with the observed changes in macrophages. The metabolic states were evaluated with a Seahorse analyzer. Additionally, immunohistochemical staining was performed to evaluate the macrophages response after implantation of the novel bone substitutes in critical size calvarial defects in SD rats. RESULTS: The incorporation of fluoride ions in FPHA was validated. FPHA promoted macrophage proliferation and enhanced the expression of M2 markers while suppressing the expression of M1 markers. Additionally, FPHA inhibited the expression of inflammatory factors and upregulated the expression of osteogenic factors, thereby enhancing the osteogenic differentiation capacity of the rBMSCs. RNA-seq analysis suggested that the polarization-regulating function of FPHA may be related to changes in cellular metabolism. Further experiments confirmed that FPHA enhanced mitochondrial function and promoted the metabolic shift of macrophages from glycolysis to oxidative phosphorylation. Moreover, in vivo experiments validated the above results in the calvarial defect model in SD rats. CONCLUSION: In summary, our study reveals that FPHA induces a metabolic shift in macrophages from glycolysis to oxidative phosphorylation. This shift leads to an increased tendency toward M2 polarization in macrophages, consequently creating a favorable osteo-immune microenvironment. These findings provide valuable insights into the impact of incorporating an appropriate concentration of fluoride on immunometabolism and macrophage mitochondrial function, which have important implications for the development of fluoride-modified immunometabolism-based bone regenerative biomaterials and the clinical application of FPHA or other fluoride-containing materials.


Subject(s)
Durapatite , Glycolysis , Macrophages , Oxidative Phosphorylation , Rats, Sprague-Dawley , Animals , Durapatite/chemistry , Macrophages/metabolism , Macrophages/drug effects , Oxidative Phosphorylation/drug effects , Glycolysis/drug effects , Rats , Swine , Cell Proliferation/drug effects , Male , Osteogenesis/drug effects , Skull/pathology , Skull/drug effects , Mice , Cellular Microenvironment/drug effects , RAW 264.7 Cells , Bone and Bones/metabolism , Bone and Bones/drug effects
11.
Int J Mol Sci ; 25(9)2024 May 06.
Article in English | MEDLINE | ID: mdl-38732267

ABSTRACT

Osteoporosis, characterized by reduced bone density and increased fracture risk, affects over 200 million people worldwide, predominantly older adults and postmenopausal women. The disruption of the balance between bone-forming osteoblasts and bone-resorbing osteoclasts underlies osteoporosis pathophysiology. Standard treatment includes lifestyle modifications, calcium and vitamin D supplementation and specific drugs that either inhibit osteoclasts or stimulate osteoblasts. However, these treatments have limitations, including side effects and compliance issues. Natural products have emerged as potential osteoporosis therapeutics, but their mechanisms of action remain poorly understood. In this study, we investigate the efficacy of natural compounds in modulating molecular targets relevant to osteoporosis, focusing on the Mitogen-Activated Protein Kinase (MAPK) pathway and the gut microbiome's influence on bone homeostasis. Using an in silico and in vitro methodology, we have identified quercetin as a promising candidate in modulating MAPK activity, offering a potential therapeutic perspective for osteoporosis treatment.


Subject(s)
Biological Products , Bone Remodeling , Osteoporosis , Humans , Bone Remodeling/drug effects , Osteoporosis/drug therapy , Osteoporosis/metabolism , Biological Products/pharmacology , Biological Products/therapeutic use , Quercetin/pharmacology , Quercetin/therapeutic use , Osteoblasts/drug effects , Osteoblasts/metabolism , Bone and Bones/metabolism , Bone and Bones/drug effects , MAP Kinase Signaling System/drug effects , Gastrointestinal Microbiome/drug effects , Osteoclasts/metabolism , Osteoclasts/drug effects , Animals
12.
Age Ageing ; 53(5)2024 May 01.
Article in English | MEDLINE | ID: mdl-38770543

ABSTRACT

CONTEXT: Chronic kidney disease (CKD) leads to alterations in fibroblast growth factor 23 (FGF23) and the renal-bone axis. This may be partly driven by altered inflammation and iron status. Vitamin D supplementation may reduce inflammation. OBJECTIVE AND METHODS: Older adults with early CKD (estimated glomerular filtration rate (eGFR) 30-60 ml/min/1.73 m2; CKDG3a/b; n = 35) or normal renal function (eGFR >90 ml/min/1.73 m2; CKDG1; n = 35) received 12,000, 24,000 or 48,000 IU D3/month for 1 year. Markers of the renal-bone axis, inflammation and iron status were investigated pre- and post-supplementation. Predictors of c-terminal and intact FGF23 (cFGF23; iFGF23) were identified by univariate and multivariate regression. RESULTS: Pre-supplementation, comparing CKDG3a/b to CKDG1, plasma cFGF23, iFGF23, PTH, sclerostin and TNFα were significantly higher and Klotho, 1,25-dihydroxyvitamin D and iron were lower. Post-supplementation, only cFGF23, 25(OH)D and IL6 differed between groups. The response to supplementation differed between eGFR groups. Only in the CKDG1 group, phosphate decreased, cFGF23, iFGF23 and procollagen type I N-propeptide increased. In the CKDG3a/b group, TNFα significantly decreased, and iron increased. Plasma 25(OH)D and IL10 increased, and carboxy-terminal collagen crosslinks decreased in both groups. In univariate models cFGF23 and iFGF23 were predicted by eGFR and regulators of calcium and phosphate metabolism at both time points; IL6 predicted cFGF23 (post-supplementation) and iFGF23 (pre-supplementation) in univariate models. Hepcidin predicted post-supplementation cFGF23 in multivariate models with eGFR. CONCLUSION: Alterations in regulators of the renal-bone axis, inflammation and iron status were found in early CKD. The response to vitamin D3 supplementation differed between eGFR groups. Plasma IL6 predicted both cFGF23 and iFGF23 and hepcidin predicted cFGF23.


Subject(s)
Biomarkers , Dietary Supplements , Fibroblast Growth Factor-23 , Fibroblast Growth Factors , Glomerular Filtration Rate , Iron , Kidney , Renal Insufficiency, Chronic , Vitamin D , Humans , Aged , Male , Female , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/physiopathology , Renal Insufficiency, Chronic/drug therapy , Glomerular Filtration Rate/drug effects , Biomarkers/blood , Fibroblast Growth Factors/blood , Iron/blood , Kidney/physiopathology , Kidney/drug effects , Vitamin D/blood , Vitamin D/analogs & derivatives , Aged, 80 and over , Treatment Outcome , Inflammation/blood , Inflammation/drug therapy , Inflammation Mediators/blood , Age Factors , Cholecalciferol/administration & dosage , Cholecalciferol/blood , Time Factors , Bone and Bones/drug effects , Bone and Bones/metabolism
13.
Cell Rep Med ; 5(5): 101574, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38776873

ABSTRACT

The existing suite of therapies for bone diseases largely act to prevent further bone loss but fail to stimulate healthy bone formation and repair. We describe an endogenous osteopeptide (PEPITEM) with anabolic osteogenic activity, regulating bone remodeling in health and disease. PEPITEM acts directly on osteoblasts through NCAM-1 signaling to promote their maturation and formation of new bone, leading to enhanced trabecular bone growth and strength. Simultaneously, PEPITEM stimulates an inhibitory paracrine loop: promoting osteoblast release of the decoy receptor osteoprotegerin, which sequesters RANKL, thereby limiting osteoclast activity and bone resorption. In disease models, PEPITEM therapy halts osteoporosis-induced bone loss and arthritis-induced bone damage in mice and stimulates new bone formation in osteoblasts derived from patient samples. Thus, PEPITEM offers an alternative therapeutic option in the management of diseases with excessive bone loss, promoting an endogenous anabolic pathway to induce bone remodeling and redress the imbalance in bone turnover.


Subject(s)
Bone Resorption , Osteoblasts , Osteogenesis , Animals , Humans , Osteoblasts/metabolism , Osteoblasts/drug effects , Osteogenesis/drug effects , Mice , Bone Resorption/pathology , Bone Resorption/metabolism , Anabolic Agents/pharmacology , Anabolic Agents/therapeutic use , Bone Remodeling/drug effects , Osteoporosis/pathology , Osteoporosis/metabolism , Osteoporosis/drug therapy , RANK Ligand/metabolism , Osteoclasts/metabolism , Osteoclasts/drug effects , Bone Development/drug effects , Osteoprotegerin/metabolism , Female , Signal Transduction/drug effects , Peptides/pharmacology , Male , Mice, Inbred C57BL , Bone and Bones/drug effects , Bone and Bones/metabolism , Bone and Bones/pathology
14.
Sci Rep ; 14(1): 10888, 2024 05 13.
Article in English | MEDLINE | ID: mdl-38740835

ABSTRACT

Ethylenediaminetetraacetic acid (EDTA), a classically used chelating agent of decalcification, maintains good morphological details, but its slow decalcification limits its wider applications. Many procedures have been reported to accelerate EDTA-based decalcification, involving temperature, concentration, sonication, agitation, vacuum, microwave, or combination. However, these procedures, concentrating on purely tissue-outside physical factors to increase the chemical diffusion, do not enable EDTA to exert its full capacity due to tissue intrinsic chemical resistances around the diffusion passage. The resistances, such as tissue inner lipids and electric charges, impede the penetration of EDTA. We hypothesized that delipidation and shielding electric charges would accelerate EDTA-based penetration and the subsequent decalcification. The hypothesis was verified by the observation of speedy penetration of EDTA with additives of detergents and hypertonic saline, testing on tissue-mimicking gels of collagen and adult mouse bones. Using a 26% EDTA mixture with the additives at 45°C, a conventional 7-day decalcification of adult mouse ankle joints could be completed within 24 h while the tissue morphological structure, antigenicity, enzymes, and DNA were well preserved, and mRNA better retained compared to using 15% EDTA at room temperature. The addition of hypertonic saline and detergents to EDTA decalcification is a simple, rapid, and inexpensive method that doesn't disrupt the current histological workflow. This method is equally or even more effective than the currently most used decalcification methods in preserving the morphological details of tissues. It can be highly beneficial for the related community.


Subject(s)
Detergents , Edetic Acid , RNA, Messenger , Animals , Edetic Acid/chemistry , Edetic Acid/pharmacology , Detergents/chemistry , Mice , RNA, Messenger/genetics , Saline Solution, Hypertonic/chemistry , Bone and Bones/metabolism , Bone and Bones/drug effects , Bone and Bones/chemistry , Decalcification Technique/methods
15.
Biomater Adv ; 161: 213887, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38735199

ABSTRACT

Critical size bone defects cannot heal without aid and current clinical approaches exhibit some limitations, underling the need for novel solutions. Silk fibroin, derived from silkworms, is widely utilized in tissue engineering and regenerative medicine due to its remarkable properties, making it a promising candidate for bone tissue regeneration in vitro and in vivo. However, the clinical translation of silk-based materials requires refinements in 3D architecture, stability, and biomechanical properties. In earlier research, improved mechanical resistance and stability of chemically crosslinked methacrylate silk fibroin (Sil-Ma) sponges over physically crosslinked counterparts were highlighted. Furthermore, the influence of photo-initiator and surfactant concentrations on silk properties was investigated. However, the characterization of sponges with Sil-Ma solution concentrations above 10 % (w/V) was hindered by production optimization challenges, with only cell viability assessed. This study focuses on the evaluation of methacrylate sponges' suitability as temporal bone tissue regeneration scaffolds. Sil-Ma sponge fabrication at a fixed concentration of 20 % (w/V) was optimized and the impact of photo-initiator (LAP) concentrations and surfactant (Tween 80) presence/absence was studied. Their effects on pore formation, silk secondary structure, mechanical properties, and osteogenic differentiation of hBM-MSCs were investigated. We demonstrated that, by tuning silk sponges' composition, the optimal combination boosted osteogenic gene expression, offering a strategy to tailor biomechanical properties for effective bone regeneration. Utilizing Design of Experiment (DoE), correlations between sponge composition, porosity, and mechanical properties are established, guiding tailored material outcomes. Additionally, correlation matrices elucidate the microstructure's influence on gene expressions, providing insights for personalized approaches in bone tissue regeneration.


Subject(s)
Bone Regeneration , Fibroins , Surface-Active Agents , Tissue Engineering , Tissue Scaffolds , Fibroins/chemistry , Tissue Scaffolds/chemistry , Surface-Active Agents/chemistry , Animals , Tissue Engineering/methods , Bone Regeneration/drug effects , Humans , Bone and Bones/drug effects , Cross-Linking Reagents/chemistry , Osteogenesis/drug effects , Mesenchymal Stem Cells/drug effects , Biocompatible Materials/chemistry , Porosity
16.
Calcif Tissue Int ; 115(1): 14-22, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38744723

ABSTRACT

Increased ß-adrenergic receptor activity has been hypothesized to cause bone loss in those with dementia. We investigated the effect of long-term ß-blocker use on rate of bone loss in older adults with dementia. We used a linear mixed-effects model to estimate the relationship between long-term ß-blocker use and rate of bone loss in participants from the Health Aging and Body Composition study. Records of 1198 participants were analyzed, 44.7% were men. Among the men, 25.2% had dementia and 20.2% were on ß-blockers, while in the women, 22.5% had dementia and 16.6% received ß-blockers. In the 135 men with dementia, 23 were taking ß-blockers, while 15 of 149 women with dementia were using ß-blockers. In men with dementia, ß-blocker users had 0.00491 g/cm2 less bone mineral density (BMD) loss per year at the femoral neck (i.e., 0.63% less loss per year) than non-users (p < 0.05). No differences were detected in women with or without dementia and men without dementia. ß-blockers may be protective by slowing down bone loss in older men with dementia.


Subject(s)
Adrenergic beta-Antagonists , Bone Density , Dementia , Humans , Male , Female , Adrenergic beta-Antagonists/therapeutic use , Adrenergic beta-Antagonists/pharmacology , Aged , Bone Density/drug effects , Dementia/drug therapy , Aged, 80 and over , Osteoporosis/drug therapy , Bone and Bones/drug effects , Bone and Bones/metabolism
17.
J Trace Elem Med Biol ; 84: 127446, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38615498

ABSTRACT

Osteoporosis is associated with an imbalance in bone formation, with certain drugs used in disease treatment being implicated in its development. Supplementation with trace elements may contribute to bone regeneration, offering an alternative approach by enhancing bone mineral density (BMD) and thereby thwarting the onset of osteoporosis. This review aims to assess the mechanisms through which trace elements such as copper (Cu), iron (Fe), selenium (Se), manganese (Mn), and zinc (Zn) are linked to increased bone mass, thus mitigating the effects of pharmaceuticals. Our findings underscore that the use of drugs such as aromatase inhibitors (AIs), proton pump inhibitors (PPIs), antiretrovirals, glucocorticoids, opioids, or anticonvulsants can result in decreased BMD, a primary contributor to osteoporosis. Research indicates that essential elements like Cu, Fe, Se, Mn, and Zn, through various mechanisms, can bolster BMD and forestall the onset of the disease, owing to their protective effects. Consequently, our study recommends a minimum daily intake of these essential minerals for patients undergoing treatment with the aforementioned drugs, as the diverse mechanisms governing the effects of trace elements Cu, Fe, Mn, Se, and Zn facilitate bone remodeling.


Subject(s)
Osteoporosis , Trace Elements , Humans , Osteoporosis/chemically induced , Osteoporosis/drug therapy , Trace Elements/pharmacology , Bone Regeneration/drug effects , Animals , Bone Density/drug effects , Bone and Bones/drug effects , Bone and Bones/metabolism
18.
Nutrients ; 16(8)2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38674818

ABSTRACT

This study investigated the characteristics of Lactobacillus helveticus-derived whey-calcium chelate (LHWCC) and its effect on the calcium absorption and bone health of rats. Fourier-transform infrared spectroscopy showed that carboxyl oxygen atoms, amino nitrogen atoms, and phosphate ions were the major binding sites with calcium in LHWCC, which has a sustained release effect in simulated in vitro digestion. LHWCC had beneficial effects on serum biochemical parameters, bone biomechanics, and the morphological indexes of the bones of calcium-deficient rats when fed at a dose of 40 mg Ca/kg BW for 7 weeks. In contrast to the inorganic calcium supplement, LHWCC significantly upregulated the gene expression of transient receptor potential cation V5 (TRPV5), TRPV6, PepT1, calcium-binding protein-D9k (Calbindin-D9k), and a calcium pump (plasma membrane Ca-ATPase, PMCA1b), leading to promotion of the calcium absorption rate, whereas Ca3(PO4)2 only upregulated the TRPV6 channel in vivo. These findings illustrate the potential of LHWCC as an organic calcium supplement.


Subject(s)
Bone and Bones , Calcium , Lactobacillus helveticus , Animals , Rats , Calcium/metabolism , Bone and Bones/metabolism , Bone and Bones/drug effects , Male , Rats, Sprague-Dawley , Whey/chemistry , TRPV Cation Channels/metabolism , TRPV Cation Channels/genetics , Calcium, Dietary/pharmacology , Calcium, Dietary/administration & dosage , Dietary Supplements , Calcium Channels/metabolism , Calcium Chelating Agents/pharmacology
19.
Bone ; 184: 117106, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38641232

ABSTRACT

Osteogenesis imperfecta (OI) increases fracture risk due to changes in bone quantity and quality caused by mutations in collagen and its processing proteins. Current therapeutics improve bone quantity, but do not treat the underlying quality deficiencies. Male and female G610C+/- mice, a murine model of OI, were treated with a combination of raloxifene and in vivo axial tibial compressive loading starting at 10 weeks of age and continuing for 6 weeks to improve bone quantity and quality. Bone geometry and mechanical properties were measured to determine whole bone and tissue-level material properties. A colocalized Raman/nanoindentation system was used to measure chemical composition and nanomechanical properties in newly formed bone compared to old bone to determine if bone formed during the treatment regimen differed in quality compared to bone formed prior to treatment. Lastly, lacunar geometry and osteocyte apoptosis were assessed. OI mice were able to build bone in response to the loading, but this response was less robust than in control mice. Raloxifene improved some bone material properties in female but not male OI mice. Raloxifene did not alter nanomechanical properties, but loading did. Lacunar geometry was largely unchanged with raloxifene and loading. However, osteocyte apoptosis was increased with loading in raloxifene treated female mice. Overall, combination treatment with raloxifene and loading resulted in positive but subtle changes to bone quality.


Subject(s)
Disease Models, Animal , Osteogenesis Imperfecta , Raloxifene Hydrochloride , Animals , Raloxifene Hydrochloride/pharmacology , Raloxifene Hydrochloride/therapeutic use , Osteogenesis Imperfecta/drug therapy , Osteogenesis Imperfecta/pathology , Female , Male , Mice , Bone and Bones/drug effects , Bone and Bones/pathology , Biomechanical Phenomena/drug effects , Apoptosis/drug effects , Anabolic Agents/pharmacology , Anabolic Agents/therapeutic use , Weight-Bearing , Osteocytes/drug effects , Osteocytes/metabolism , Osteocytes/pathology
20.
J Mater Chem B ; 12(17): 4172-4183, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38591253

ABSTRACT

Traumatic hemorrhage is one of the main causes of mortality in civilian and military accidents. This study aimed to evaluate the effectiveness of cuttlefish bone (cuttlebone, CB) and CB loaded with cuttlefish ink (CB-CFI) nanoparticles for hemorrhage control. CB and CB-CFI were prepared and characterized using different methods. The hemostasis behavior of constructed biocomposites was investigated in vitro and in vivo using a rat model. Results showed that CFI nanoparticles (NPs) are uniformly dispersed throughout the CB surface. CB-CFI10 (10 mg CFI in 1.0 g of CB) showed the best blood clotting performance in both in vitro and in vivo tests. In vitro findings revealed that the blood clotting time of CB, CFI, and CB-CFI10 was found to be 275.4 ± 12.4 s, 229.9 ± 19.9 s, and 144.0 ± 17.5 s, respectively. The bleeding time in rat liver injury treated with CB, CFI, and CB-CFI10 was 158.1 ± 9.2 s, 114.0 ± 5.7 s, and 46.8 ± 2.7 s, respectively. CB-CFI10 composite resulted in more reduction of aPTT (11.31 ± 1.51 s) in comparison with CB (17.34 ± 2.12 s) and CFI (16.79 ± 1.46 s) (p < 0.05). Furthermore, CB and CB-CFI10 exhibited excellent hemocompatibility. The CB and CB-CFI did not show any cytotoxicity on human foreskin fibroblast (HFF) cells. The CB-CFI has a negative surface charge and may activate coagulation factors through direct contact with their components, including CaCO3, chitin, and CFI-NPs with blood. Thus, the superior hemostatic potential, low cost, abundant, simple, and time-saving preparation process make CB-CFI a very favorable hemostatic material for traumatic bleeding control in clinical applications.


Subject(s)
Decapodiformes , Hemostatics , Ink , Nanoparticles , Animals , Rats , Hemostatics/chemistry , Hemostatics/pharmacology , Nanoparticles/chemistry , Decapodiformes/chemistry , Hemorrhage/drug therapy , Male , Blood Coagulation/drug effects , Rats, Sprague-Dawley , Humans , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Hemostasis/drug effects , Bone and Bones/drug effects , Particle Size
SELECTION OF CITATIONS
SEARCH DETAIL
...