Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.416
Filter
1.
Front Endocrinol (Lausanne) ; 15: 1417191, 2024.
Article in English | MEDLINE | ID: mdl-38974581

ABSTRACT

Osteoporosis and osteoarthritis continue to pose significant challenges to the aging population, with limited preventive options and pharmacological treatments often accompanied by side effects. Amidst ongoing efforts to discover new therapeutic agents, tocotrienols (TTs) have emerged as potential candidates. Derived from annatto bean and palm oil, TTs have demonstrated efficacy in improving skeletal and joint health in numerous animal models of bone loss and osteoarthritis. Mechanistic studies suggest that TTs exert their effects through antioxidant, anti-inflammatory, Wnt-suppressive, and mevalonate-modulating mechanisms in bone, as well as through self-repair mechanisms in chondrocytes. However, human clinical trials in this field remain scarce. In conclusion, TTs hold promise as agents for preventing osteoporosis and osteoarthritis, pending further evidence from human clinical trials.


Subject(s)
Osteoarthritis , Osteoporosis , Tocotrienols , Tocotrienols/therapeutic use , Tocotrienols/pharmacology , Humans , Animals , Osteoarthritis/drug therapy , Osteoarthritis/prevention & control , Osteoporosis/drug therapy , Osteoporosis/prevention & control , Bone and Bones/drug effects , Bone and Bones/metabolism
2.
Int J Nanomedicine ; 19: 6359-6376, 2024.
Article in English | MEDLINE | ID: mdl-38946885

ABSTRACT

Background: Bone tissue engineering (BTE) is a promising alternative to autologous bone grafting for the clinical treatment of bone defects, and inorganic/organic composite hydrogels as BTE scaffolds are a hot spot in current research. The construction of nano-hydroxyapatite/gelatin methacrylate/oxidized sodium alginate (nHAP/GelMA/OSA), abbreviated as HGO, composite hydrogels loaded with bone morphogenetic protein 7 (BMP7) will provide a suitable 3D microenvironment to promote cell aggregation, proliferation, and differentiation, thus facilitating bone repair and regeneration. Methods: Dually-crosslinked hydrogels were fabricated by combining GelMA and OSA, while HGO hydrogels were formulated by incorporating varying amounts of nHAP. The hydrogels were physically and chemically characterized followed by the assessment of their biocompatibility. BMP7-HGO (BHGO) hydrogels were fabricated by incorporating suitable concentrations of BMP7 into HGO hydrogels. The osteogenic potential of BHGO hydrogels was then validated through in vitro experiments and using rat femoral defect models. Results: The addition of nHAP significantly improved the physical properties of the hydrogel, and the composite hydrogel with 10% nHAP demonstrated the best overall performance among all groups. The selected concentration of HGO hydrogel served as a carrier for BMP7 loading and was evaluated for its osteogenic potential both in vivo and in vitro. The BHGO hydrogel demonstrated superior in vitro osteogenic induction and in vivo potential for repairing bone tissue compared to the outcomes observed in the blank control, BMP7, and HGO groups. Conclusion: Using hydrogel containing 10% HGO appears promising for bone tissue engineering scaffolds, especially when loaded with BMP7 to boost its osteogenic potential. However, further investigation is needed to optimize the GelMA, OSA, and nHAP ratios, along with the BMP7 concentration, to maximize the osteogenic potential.


Subject(s)
Alginates , Bone Morphogenetic Protein 7 , Bone Regeneration , Durapatite , Gelatin , Hydrogels , Osteogenesis , Tissue Engineering , Tissue Scaffolds , Alginates/chemistry , Alginates/pharmacology , Animals , Bone Morphogenetic Protein 7/chemistry , Bone Morphogenetic Protein 7/pharmacology , Gelatin/chemistry , Tissue Engineering/methods , Hydrogels/chemistry , Hydrogels/pharmacology , Durapatite/chemistry , Durapatite/pharmacology , Osteogenesis/drug effects , Rats , Bone Regeneration/drug effects , Tissue Scaffolds/chemistry , Rats, Sprague-Dawley , Methacrylates/chemistry , Male , Humans , Bone and Bones/drug effects
3.
Rev Fac Cien Med Univ Nac Cordoba ; 81(2): 270-284, 2024 06 28.
Article in English | MEDLINE | ID: mdl-38941224

ABSTRACT

When large amounts of Fluoride are consumed produces insulin resistance, but exercise can reverse insulin resistance in rats, because of a high fluoride uptake by bone tissue. However, bone quality has not been studied in those experiments. Therefore, the aim of this work was to evaluate bone quality in rats treated with fluoride when performing exercise. Sprague-Dawley rats were divided into 3 groups (n=6 per group): Control (drinking water without fluoride), Fluoride (drinking water with fluoride 15 mg/L for 30 days) and Exercise (daily running on a treadmill and drinking water with fluoride 15 mg/L for 30 days).  Then, bone mineral density, mechanical and histological properties and bone fluoride level were measured. No effect of treatment on any bone parameters were observed. These results indicate that exercise normalizes glucose metabolism in insulin-resistant rats by bone fluoride uptake; however, this increase in bone fluoride does not manifest in bone deterioration.


Cuando se consumen grandes cantidades de fluoruro se produce resistencia a la insulina, pero la realización de ejercicio puede revertir dicho efecto en ratas, debido a una alta absorción de fluoruro por el tejido óseo. Sin embargo, la calidad ósea no ha sido estudiada. Por ello, el objetivo de este trabajo fue evaluar la calidad ósea en ratas tratadas con flúor que realizan ejercicio. Se trabajó con ratas Sprague-Dawley que se dividieron en 3 grupos (n=6 por grupo): Control (recibiron agua sin flúor), Flúor (recibieron agua con flúor 15 mg/L durante 30 días) y Ejercicio (realizaron ejercicio diariamente en cinta ergométrica y recibieron agua con fluoruro 15 mg/L por 30 días). Luego, se midieron la densidad mineral ósea, las propiedades biomecánicas e histológicas y el nivel de fluoruro óseo. No se observó ningún efecto del tratamiento sobre ningún parámetro óseo. Estos resultados indican que el ejercicio normaliza el metabolismo de la glucosa en ratas resistentes a la insulina mediante la captación ósea de fluoruro; sin embargo, este aumento del fluoruro óseo no se manifiesta en deterioro óseo.


Subject(s)
Bone Density , Fluorides , Insulin Resistance , Physical Conditioning, Animal , Rats, Sprague-Dawley , Animals , Insulin Resistance/physiology , Bone Density/drug effects , Physical Conditioning, Animal/physiology , Fluorides/pharmacology , Rats , Male , Bone and Bones/metabolism , Bone and Bones/drug effects
4.
Nutrients ; 16(11)2024 May 21.
Article in English | MEDLINE | ID: mdl-38892482

ABSTRACT

Skin problems caused by aging have attracted much attention, and marine collagen peptides have been proved to improve these problems, while mammalian collagen peptides are rarely reported. In this study, fermented deer bone collagen peptide (FCP) and non-fermented deer bone collagen peptide (NCP) were extracted from fermented and non-fermented deer bone, respectively, and their peptide sequences and differential proteins were analyzed using LC-MS/MS technology. After they were applied to aging mice induced with D-gal, the skin hydration ability, antioxidant ability, collagen synthesis, and degradation ability of the mice were studied. The results show that FCP and NCP are mainly peptides that constitute type Ⅰ collagen, and their peptide segments are different. In vivo experiments show that FCP and NCP can improve the richness of collagen fibers in the skin of aging mice; improve the hydration ability of skin; promote the activity of antioxidant-related enzymes; and also show that through the TGF-ß and MAPK pathways, the synthesis and degradation of collagen in skin are regulated. These results show that deer bone collagen peptide can improve skin problems caused by aging, promote skin hydration and antioxidant capacity of aging mice, and regulate collagen synthesis and degradation through the MAPK pathway.


Subject(s)
Aging , Antioxidants , Bone and Bones , Collagen , Deer , Skin , Animals , Antioxidants/pharmacology , Mice , Skin/metabolism , Skin/drug effects , Bone and Bones/drug effects , Bone and Bones/metabolism , Collagen/metabolism , Aging/drug effects , Administration, Oral , Peptides/pharmacology , Skin Aging/drug effects , Male , Fermentation , Collagen Type I/metabolism
5.
In Vivo ; 38(4): 1557-1570, 2024.
Article in English | MEDLINE | ID: mdl-38936927

ABSTRACT

BACKGROUND/AIM: This study examined the effects of tocotrienols (TT) in conjunction with statin on glucose homeostasis, bone microstructure, gut microbiome, and systemic and liver inflammatory markers in obese C57BL/6J mice. MATERIALS AND METHODS: Forty male C57BL/6J mice were fed a high-fat diet (HFD) and assigned into four groups in a 2 (no statin vs. 120 mg statin/kg diet)×2 (no TT vs. 400 mg TT/kg diet) factorial design for 14 weeks. RESULTS: Statin and TT improved glucose tolerance only when each was given alone, and only statin supplementation decreased insulin resistance. Consistently, only statin supplementation decreased serum insulin levels and HOMA-IR. Pancreatic insulin was also increased with statin treatment. Statin and TT, alone or in combination, reduced the levels of serum IL-6, but only TT attenuated the increased serum leptin levels induced by a HFD. Statin supplementation increased bone area/total area and connectivity density at LV-4, while TT supplementation increased bone area/total area and trabecular number, but decreased trabecular separation at the distal femur. Statin supplementation, but not TT, reduced hepatic inflammatory cytokine gene expression. Neither TT supplementation nor statin supplementation statistically altered microbiome species evenness or richness. However, they altered the relative abundance of certain microbiome species. Most notably, both TT and statin supplementation increased the relative abundance of Lachnospiraceae UCG-006. CONCLUSION: TT and statin collectively benefit bone microstructure, glucose homeostasis, and microbial ecology in obese mice. Such changes may be, in part, associated with suppression of inflammation in the host.


Subject(s)
Bone and Bones , Diet, High-Fat , Dietary Supplements , Gastrointestinal Microbiome , Homeostasis , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Obesity , Tocotrienols , Animals , Gastrointestinal Microbiome/drug effects , Tocotrienols/pharmacology , Tocotrienols/administration & dosage , Mice , Homeostasis/drug effects , Obesity/drug therapy , Obesity/metabolism , Male , Bone and Bones/drug effects , Bone and Bones/metabolism , Bone and Bones/pathology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Diet, High-Fat/adverse effects , Bixaceae/chemistry , Mice, Obese , Plant Extracts/pharmacology , Plant Extracts/administration & dosage , Glucose/metabolism , Mice, Inbred C57BL , Insulin Resistance , Blood Glucose , Disease Models, Animal , Liver/drug effects , Liver/metabolism , Liver/pathology , Biomarkers , Carotenoids
6.
Biomacromolecules ; 25(7): 4074-4086, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38838242

ABSTRACT

The presence of oxidative stress in bone defects leads to delayed regeneration, especially in the aged population and patients receiving cancer treatment. This delay is attributed to the increased levels of reactive oxygen species (ROS) in these populations due to the accumulation of senescent cells. Tissue-engineered scaffolds are emerging as an alternative method to treat bone defects. In this study, we engineered tissue scaffolds tailored to modulate the adverse effects of oxidative stress and promote bone regeneration. We used polycaprolactone to fabricate nanofibrous mats by using electrospinning. We exploited the ROS-scavenging properties of cerium oxide nanoparticles to alleviate the high oxidative stress microenvironment caused by the presence of senescent cells. We characterized the nanofibers for their physical and mechanical properties and utilized an ionization-radiation-based model to induce senescence in bone cells. We demonstrate that the presence of ceria can modulate ROS levels, thereby reducing the level of senescence and promoting osteogenesis. Overall, this study demonstrates that ceria-infused nanofibrous scaffolds can be used for augmenting the osteogenic activity of senescent progenitor cells, which has important implications for engineering bone tissue scaffolds for patients with low regeneration capabilities.


Subject(s)
Bone Regeneration , Cellular Senescence , Cerium , Nanofibers , Osteogenesis , Reactive Oxygen Species , Tissue Engineering , Tissue Scaffolds , Cerium/chemistry , Cerium/pharmacology , Bone Regeneration/drug effects , Tissue Scaffolds/chemistry , Cellular Senescence/drug effects , Nanofibers/chemistry , Osteogenesis/drug effects , Humans , Tissue Engineering/methods , Reactive Oxygen Species/metabolism , Oxidative Stress/drug effects , Polyesters/chemistry , Animals , Bone and Bones/drug effects
7.
ACS Biomater Sci Eng ; 10(7): 4463-4479, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38848471

ABSTRACT

Scaffold-free bone microtissues differentiated from mesenchymal stem cell (MSC) spheroids offer great potential for bottom-up bone tissue engineering as a direct supply of cells and osteogenic signals. Many biomaterials or biomolecules have been incorporated into bone microtissues to enhance their osteogenic abilities, but these materials are far from clinical approval. Here, we aimed to incorporate hydroxyapatite (HAP) nanoparticles, an essential component of bone matrix, into MSC spheroids to instruct their osteogenic differentiation into bone microtissues and further self-organization into bone organoids with a trabecular structure. Furthermore, the biological interaction between HAP nanoparticles and MSCs and the potential molecular mechanisms in the bone development of MSC spheroids were investigated by both in vitro and in vivo studies. As a result, improved cell viability and osteogenic abilities were observed for the MSC spheroids incorporated with HAP nanoparticles at a concentration of 30 µg/mL. HAP nanoparticles could promote the sequential expression of osteogenic markers (Runx2, Osterix, Sclerostin), promote the expression of bone matrix proteins (OPN, OCN, and Collagen I), promote the mineralization of the bone matrix, and thus promote the bone development of MSC spheroids. The differentiated bone microtissues could further self-organize into linear, lamellar, and spatial bone organoids with trabecular structures. More importantly, adding FAK or Akt inhibitors could decrease the level of HAP-induced osteogenic differentiation of bone microtissues. Finally, excellent new bone regeneration was achieved after injecting bone microtissues into cranial bone defect models, which could also be eliminated by the Akt inhibitor. In conclusion, HAP nanoparticles could promote the development of bone microtissues by promoting the osteogenic differentiation of MSCs and the formation and mineralization of the bone matrix via the FAK/Akt pathway. The bone microtissues could act as individual ossification centers and self-organize into macroscale bone organoids, and in this meaning, the bone microtissues could be called microscale bone organoids. Furthermore, the bone microtissues revealed excellent clinical perspectives for injectable cellular therapies for bone defects.


Subject(s)
Bone Regeneration , Cell Differentiation , Durapatite , Mesenchymal Stem Cells , Nanoparticles , Osteogenesis , Proto-Oncogene Proteins c-akt , Durapatite/chemistry , Durapatite/pharmacology , Bone Regeneration/drug effects , Nanoparticles/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Animals , Osteogenesis/drug effects , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Cell Differentiation/drug effects , Humans , Signal Transduction/drug effects , Tissue Engineering/methods , Focal Adhesion Kinase 1/metabolism , Bone and Bones/drug effects , Mice , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism
8.
Bone ; 186: 117143, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38866125

ABSTRACT

The effects of gender affirming hormone therapy (GAHT) on bone microarchitecture and fracture risk in adult transgender women is unclear. To investigate the concept that skeletal integrity and strength in trans women may be improved by treatment with a higher dose of GAHT than commonly prescribed, we treated adult male mice with a sustained, high dose of estradiol. Adult male mice at 16 weeks of age were administered ~1.3 mg estradiol by silastic implant, implanted intraperitoneally, for 12 weeks. Controls included vehicle treated intact females and males. High-dose estradiol treatment in males stimulated the endocortical deposition of bone at the femoral mid-diaphysis, increasing cortical thickness and bone area. This led to higher stiffness, maximum force, and the work required to fracture the bone compared to male controls, while post-yield displacement was unaffected. Assessment of the material properties of the bone showed an increase in both elastic modulus and ultimate stress in the estradiol treated males. Treatment of male mice with high dose estradiol was also anabolic for trabecular bone, markedly increasing trabecular bone volume, number and thickness in the distal metaphysis which was accompanied by an increase in the histomorphometric markers of bone remodelling, mineralizing surface/bone surface, bone formation rate and osteoclast number. In conclusion, a high dose of estradiol is anabolic for cortical and trabecular bone in a male to female transgender mouse model, increasing both stiffness and strength. These findings suggest that increasing the current dose of GAHT administered to trans women, while considering other potential adverse effects, may be beneficial to preserving their bone microstructure and strength.


Subject(s)
Estradiol , Animals , Male , Estradiol/pharmacology , Estradiol/blood , Female , Mice , Bone and Bones/drug effects , Bone and Bones/diagnostic imaging , Bone Density/drug effects , Anabolic Agents/pharmacology , Organ Size/drug effects , Mice, Inbred C57BL , Humans , Models, Animal , X-Ray Microtomography
9.
Bone ; 186: 117167, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38876270

ABSTRACT

We evaluated the potential of sclerostin antibody (SclAb) therapy to enhance osseointegration of dental and orthopaedic implants in a mouse model (Brtl/+) mimicking moderate to severe Osteogenesis Imperfecta (OI). To address the challenges in achieving stable implant integration in compromised bone conditions, our aim was to determine the effectiveness of sclerostin antibody (SclAb) at improving bone-to-implant contact and implant fixation strength. Utilizing a combination of micro-computed tomography, mechanical push-in testing, immunohistochemistry, and Western blot analysis, we observed that SclAb treatment significantly enhances bone volume fraction (BV/TV) and bone-implant contact (BIC) in Brtl/+ mice, suggesting a normalization of bone structure toward WT levels. Despite variations in implant survival rates between the maxilla and tibia, SclAb treatment consistently improved implant stability and resistance to mechanical forces, highlighting its potential to overcome the inherent challenges of OI in dental and orthopaedic implant integration. These results suggest that SclAb could be a valuable therapeutic approach for enhancing implant success in compromised bone conditions.


Subject(s)
Adaptor Proteins, Signal Transducing , Antibodies , Collagen Type I , Mutation , Osseointegration , Animals , Osseointegration/drug effects , Mice , Mutation/genetics , Collagen Type I/genetics , Collagen Type I/metabolism , Antibodies/pharmacology , X-Ray Microtomography , Bone and Bones/diagnostic imaging , Bone and Bones/drug effects , Bone and Bones/pathology , Intercellular Signaling Peptides and Proteins , Dental Implants , Tibia/diagnostic imaging , Tibia/pathology , Tibia/drug effects
10.
FASEB J ; 38(11): e23726, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38847773

ABSTRACT

Calcitriol and calcimimetics are used to treat hyperparathyroidism secondary to chronic kidney disease (CKD). Calcitriol administration and the subsequent increase in serum calcium concentration decrease parathyroid hormone (PTH) levels, which should reduce bone remodeling. We have previously reported that, when maintaining a given concentration of PTH, the addition of calcimimetics is associated with an increased bone cell activity. Whether calcitriol administration affects bone cell activity while PTH is maintained constant should be evaluated in an animal model of renal osteodystrophy. The aim of the present study was to compare in CKD PTH-clamped rats the bone effects of calcitriol and calcimimetic administration. The results show that the administration of calcitriol and calcimimetic at doses that induced a similar reduction in PTH secretion produced dissimilar effects on osteoblast activity in 5/6 nephrectomized (Nx) rats with secondary hyperparathyroidism and in Nx rats with clamped PTH. Remarkably, in both rat models, the administration of calcitriol decreased osteoblastic activity, whereas calcimimetic increased bone cell activity. In vitro, calcitriol supplementation inhibited nuclear translocation of ß-catenin and reduced proliferation, osteogenesis, and mineralization in mesenchymal stem cells differentiated into osteoblasts. In conclusion, besides the action of calcitriol and calcimimetics at parathyroid level, these treatments have specific effects on bone cells that are independent of the PTH level.


Subject(s)
Calcimimetic Agents , Calcitriol , Osteoblasts , Parathyroid Hormone , Animals , Calcitriol/pharmacology , Rats , Calcimimetic Agents/pharmacology , Calcimimetic Agents/therapeutic use , Parathyroid Hormone/pharmacology , Male , Osteoblasts/drug effects , Osteoblasts/metabolism , Hyperparathyroidism, Secondary/drug therapy , Hyperparathyroidism, Secondary/etiology , Hyperparathyroidism, Secondary/metabolism , Bone and Bones/metabolism , Bone and Bones/drug effects , Rats, Wistar , Renal Insufficiency/drug therapy , Renal Insufficiency/metabolism , Osteogenesis/drug effects , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/complications , Cell Differentiation/drug effects , Calcium/metabolism
11.
Biol Sex Differ ; 15(1): 51, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38890762

ABSTRACT

BACKGROUND: Prenatal alcohol exposure (PAE) can result in lifelong disabilities known as foetal alcohol spectrum disorder (FASD) and is associated with childhood growth deficiencies and increased bone fracture risk. However, the effects of PAE on the adult skeleton remain unclear and any potential sexual dimorphism is undetermined. Therefore, we utilised a murine model to examine sex differences with PAE on in vitro bone formation, and in the juvenile and adult skeleton. METHODS: Pregnant C57BL/6J female mice received 5% ethanol in their drinking water during gestation. Primary calvarial osteoblasts were isolated from neonatal offspring and mineralised bone nodule formation and gene expression assessed. Skeletal phenotyping of 4- and 12-week-old male and female offspring was conducted by micro-computed tomography (µCT), 3-point bending, growth plate analyses, and histology. RESULTS: Osteoblasts from male and female PAE mice displayed reduced bone formation, compared to control (≤ 30%). Vegfa, Vegfb, Bmp6, Tgfbr1, Flt1 and Ahsg were downregulated in PAE male osteoblasts only, whilst Ahsg was upregulated in PAE females. In 12-week-old mice, µCT analysis revealed a sex and exposure interaction across several trabecular bone parameters. PAE was detrimental to the trabecular compartment in male mice compared to control, yet PAE females were unaffected. Both male and female mice had significant reductions in cortical parameters with PAE. Whilst male mice were negatively affected along the tibial length, females were only distally affected. Posterior cortical porosity was increased in PAE females only. Mechanical testing revealed PAE males had significantly reduced bone stiffness compared to controls; maximum load and yield were reduced in both sexes. PAE had no effect on total body weight or tibial bone length in either sex. However, total growth plate width in male PAE mice compared to control was reduced, whilst female PAE mice were unaffected. 4-week-old mice did not display the altered skeletal phenotype with PAE observed in 12-week-old animals. CONCLUSIONS: Evidence herein suggests, for the first time, that PAE exerts divergent sex effects on the skeleton, possibly influenced by underlying sex-specific transcriptional mechanisms of osteoblasts. Establishing these sex differences will support future policies and clinical management of FASD.


Prenatal alcohol exposure (PAE) can lead to a set of lifelong cognitive, behavioural, and physical disabilities known as foetal alcohol spectrum disorder (FASD). FASD is a significant burden on healthcare, justice and education systems, which is set to worsen with rising alcohol consumption rates. FASD children have an increased risk of long bone fracture and adolescents are smaller in stature. However, sex differences and the long-term effects of PAE on the skeleton have not been investigated and was the aim of this study. Using a mouse model of PAE, we examined the function and gene expression of bone-forming cells (osteoblasts). We then analysed the skeletons of male and female mice at 12-weeks-old (adult) and 4-weeks-old (juvenile). PAE reduced osteoblast bone formation in both sexes, compared to control. Differential gene expression was predominantly observed in PAE males and largely involved genes related to blood vessel formation. High resolution x-ray imaging (micro-CT) revealed PAE had a detrimental effect on the inner trabecular bone component in 12-week-old male mice only. Analysis of the outer cortical bone revealed that whilst both male and female PAE mice were negatively affected, anatomical variations were observed. Mechanical testing also revealed differences in bone strength in PAE mice, compared to control. Interestingly, 4-week-old mice did not possess these sex differences observed in our PAE model at 12 weeks of age. Our data suggest PAE has detrimental and yet sex-dependent effects on the skeleton. Establishing these sex differences will support future policies and clinical management of FASD.


Subject(s)
Ethanol , Mice, Inbred C57BL , Osteoblasts , Prenatal Exposure Delayed Effects , Sex Characteristics , Animals , Female , Male , Pregnancy , Ethanol/toxicity , Ethanol/pharmacology , Osteoblasts/drug effects , Osteogenesis/drug effects , Mice , Bone and Bones/drug effects , X-Ray Microtomography
12.
Int J Mol Sci ; 25(11)2024 May 27.
Article in English | MEDLINE | ID: mdl-38892015

ABSTRACT

Ginsenosides, bioactive compounds from the genus Panax, have potential therapeutic effects on diverse ailments, including diabetes. Emerging evidence suggests their involvement in bone metabolism. The present review summarizes the current understanding of the effects of ginsenosides on osteoporosis, periodontal disease, and osteoarthritis. Their mechanisms of action include effects on osteoblasts, osteoclasts, periodontal ligament fibroblasts (PDLFs), and chondrocytes, which are pivotal in maintaining bone, periodontal tissue, and cartilage homeostasis. Ginsenosides may exert their beneficial effects by enhancing PDLF and osteoblast activity, suppressing osteoclast function, augmenting chondrocyte synthesis in the cartilage matrix, and mitigating connective tissue degradation. Moreover, they possess antioxidant, anti-inflammatory, antimicrobial, and anti-pyroptotic properties. Their efficacy in increasing bone density, ameliorating periodontitis, and alleviating osteoarthritis symptoms has been demonstrated in preclinical studies using animal models. In terms of their mechanism of action, ginsenosides modulate cellular differentiation, activity, and key signaling pathway molecules, such as mitogen-activated protein kinases (MAPKs), while also regulating various mediators. Furthermore, the symptomatic relief observed in animal models lends further credence to their therapeutic utility. However, to translate these preclinical findings into clinical practice, rigorous animal and clinical investigations are imperative to ascertain the safety, efficacy, and optimal dosing regimens in human subjects.


Subject(s)
Ginsenosides , Osteoarthritis , Osteoporosis , Periodontal Diseases , Ginsenosides/pharmacology , Ginsenosides/therapeutic use , Humans , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Animals , Osteoporosis/drug therapy , Osteoporosis/metabolism , Periodontal Diseases/drug therapy , Periodontal Diseases/metabolism , Bone and Bones/metabolism , Bone and Bones/drug effects
13.
Int J Mol Sci ; 25(11)2024 May 28.
Article in English | MEDLINE | ID: mdl-38892062

ABSTRACT

Bone health is the result of a tightly regulated balance between bone modeling and bone remodeling, and alterations of these processes have been observed in several diseases both in adult and pediatric populations. The imbalance in bone remodeling can ultimately lead to osteoporosis, which is most often associated with aging, but contributing factors can already act during the developmental age, when over a third of bone mass is accumulated. The maintenance of an adequate bone mass is influenced by genetic and environmental factors, such as physical activity and diet, and particularly by an adequate intake of calcium and vitamin D. In addition, it has been claimed that the integration of specific nutraceuticals such as resveratrol, anthocyanins, isoflavones, lycopene, curcumin, lutein, and ß-carotene and the intake of bioactive compounds from the diet such as honey, tea, dried plums, blueberry, and olive oil can be efficient strategies for bone loss prevention. Nutraceuticals and functional foods are largely used to provide medical or health benefits, but there is an urge to determine which products have adequate clinical evidence and a strong safety profile. The aim of this review is to explore the scientific and clinical evidence of the positive role of nutraceuticals and functional food in bone health, focusing both on molecular mechanisms and on real-world studies.


Subject(s)
Bone and Bones , Dietary Supplements , Functional Food , Humans , Bone and Bones/metabolism , Bone and Bones/physiology , Bone and Bones/drug effects , Osteoporosis/prevention & control , Animals , Bone Remodeling/drug effects , Bone Density/drug effects
14.
Int J Biol Macromol ; 272(Pt 1): 132820, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38825286

ABSTRACT

Bone tissue engineering (BTE) aims to develop implantable bone replacements for severe skeletal abnormalities that do not heal. In the field of BTE, chitosan (CS) has become a leading polysaccharide in the development of bone scaffolds. Although CS has several excellent properties, such as biodegradability, biocompatibility, and antibacterial properties, it has limitations for use in BTE because of its poor mechanical properties, increased degradation, and minimal bioactivity. To address these issues, researchers have explored other biomaterials, such as synthetic polymers, ceramics, and CS coatings on metals, to produce CS-based biocomposite scaffolds for BTE applications. These CS-based biocomposite scaffolds demonstrate superior properties, including mechanical characteristics, such as compressive strength, Young's modulus, and tensile strength. In addition, they are compatible with neighboring tissues, exhibit a controlled rate of degradation, and promote cell adhesion, proliferation, and osteoblast differentiation. This review provides a brief outline of the recent progress in making different CS-based biocomposite scaffolds and how to characterize them so that their mechanical properties can be tuned using crosslinkers for bone regeneration.


Subject(s)
Biocompatible Materials , Bone and Bones , Chitosan , Tissue Engineering , Tissue Scaffolds , Chitosan/chemistry , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Humans , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Bone and Bones/drug effects , Bone and Bones/physiology , Animals , Bone Regeneration/drug effects , Mechanical Phenomena
15.
Stomatologiia (Mosk) ; 103(3): 5-10, 2024.
Article in Russian | MEDLINE | ID: mdl-38904552

ABSTRACT

THE AIM OF THE STUDY: Was to determine the presence of an amoxicillin-based antibiotic in bone implant biopsies by Raman spectroscopy in an experiment. MATERIALS AND METHODS: Experimental animals (n=10, a miniature pig of the Svetlogorsk breed) were divided into 2 groups of 5 animals. Groups 1 and 2 were injected with amoxicillin 2 ml per 20 kg of body weight 30 minutes before dental implantation surgery, then group 2 was additionally injected with 1 ml per 20 kg of body weight for 5 days. Each animal has 6 implants installed. On the 1st, 3rd, 7th, 14th day, an implant-bone biopsy was removed from each animal, micro-preparations were made and Raman spectroscopy was performed to assess the peak matching of the Raman spectrum. RESULTS: In animals of the 1st and 2nd groups, the main peak of the Raman spectrum, which is closest to the values of the antibiotic spectrum of interest to us, is located closer to 1448 cm-1 and 1446 cm-1, respectively. At the same time, in both observations, the peaks relate to the spectrum of bone tissue, which cannot indicate the content of an antibiotic in the drug. CONCLUSION: No scattering spectra corresponding to the antibiotic molecule were found in any animal from both groups, regardless of the mode of administration and dosage of amoxicillin. The detected peaks corresponded to bone tissue without an antibiotic.


Subject(s)
Amoxicillin , Anti-Bacterial Agents , Dental Implants , Spectrum Analysis, Raman , Spectrum Analysis, Raman/methods , Animals , Amoxicillin/analysis , Amoxicillin/administration & dosage , Swine , Anti-Bacterial Agents/analysis , Anti-Bacterial Agents/administration & dosage , Biopsy , Swine, Miniature , Bone and Bones/chemistry , Bone and Bones/drug effects , Bone and Bones/pathology , Dental Implantation/methods
16.
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
18.
Int J Biol Macromol ; 271(Pt 2): 132675, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38845259

ABSTRACT

Novel hydrogel-based multifunctional systems prepared utilizing photocrosslinking and freeze-drying processes (PhotoCross/Freeze-dried) dedicated for bone tissue regeneration are presented. Fabricated materials, composed of methacrylated gelatin, chitosan, and chondroitin sulfate, possess interesting features including bioactivity, biocompatibility, as well as antibacterial activity. Importantly, their degradation and swellability might be easily tuned by playing with the biopolymeric content in the photocrosllinked systems. To broaden the potential application and deliver the therapeutic features, mesoporous silica particles functionalized with methacrylate moieties decorated with hydroxyapatite and loaded with the antiosteoporotic drug, alendronate, (MSP-MA-HAp-ALN) were dispersed within the biopolymeric sol and photocrosslinked. It was demonstrated that the obtained composites are characterized by a significantly extended degradation time, ensuring optimal conditions for balancing hybrids removal with the deposition of fresh bone. We have shown that attachment of MSP-MA-HAp-ALN to the polymeric matrix minimizes the initial burst effect and provides a prolonged release of ALN (up to 22 days). Moreover, the biological evaluation in vitro suggested the capability of the resulted systems to promote bone remodeling. Developed materials might potentially serve as scaffolds that after implantation will fill up bone defects of various origin (osteoporosis, tumour resection, accidents) providing the favourable conditions for bone regeneration and supporting the infections' treatment.


Subject(s)
Bone Regeneration , Chitosan , Chondroitin Sulfates , Gelatin , Chondroitin Sulfates/chemistry , Chondroitin Sulfates/pharmacology , Chitosan/chemistry , Gelatin/chemistry , Bone Regeneration/drug effects , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Tissue Scaffolds/chemistry , Humans , Cross-Linking Reagents/chemistry , Animals , Bone and Bones/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Hydrogels/chemistry , Hydrogels/pharmacology
19.
Anticancer Res ; 44(7): 2887-2897, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38925839

ABSTRACT

BACKGROUND/AIM: This study aimed to investigate the structure and functions of the membrane formed around liquid nitrogen-treated bones in the osteogenesis and revitalization of frozen bone using a rat model. MATERIALS AND METHODS: Segmental defects were created in femurs of rats, and resected bones treated with liquid nitrogen [frozen bone (FB) group, n=20] or polymethylmethacrylate (PMMA group; n=20) were implanted as spacers. Histological analysis and quantitative real-time reverse transcription-polymerase chain reaction (qRT-PCR) of the membrane around each spacer were performed for bone morphogenetic protein 2 (BMP2), transforming growth factor (TGF)-ß1, and vascular endothelial growth factor (VEGF). Furthermore, in week 2, spacers were removed from both groups (n=5 each), and autologous cancellous bone (ACB) harvested from the ilium was grafted into the defect. Radiological analysis was performed until bone union was observed. RESULTS: In week 2, similar two-layered membrane structures were observed in both groups; these matured into fibrous tissues over time. At each evaluation point, qRT-PCR showed higher expression of all factors in the FB than in the PMMA group. In the ACB graft model, the mean period to bone union and new bone volume were significantly shorter and greater, respectively, in the FB. Chondrocytes invaded the osteotomy site from the membrane in the FB, suggesting that endochondral ossification may occur and be related to osteogenesis. Additionally, fibroblasts and capillaries in the membrane invaded the surface of treated bone in week 2, and osteocytes were observed around them in weeks 6 and 8. CONCLUSION: Fibrous membranous tissue formed around liquid nitrogen-treated bones may be vital for osteogenesis and revitalization of frozen bones.


Subject(s)
Osteogenesis , Vascular Endothelial Growth Factor A , Animals , Osteogenesis/drug effects , Rats , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Nitrogen/metabolism , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 2/genetics , Male , Bone Transplantation/methods , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Polymethyl Methacrylate/pharmacology , Femur/drug effects , Femur/metabolism , Femur/pathology , Bone and Bones/drug effects , Bone and Bones/metabolism , Bone and Bones/pathology , Rats, Sprague-Dawley
20.
Medicina (Kaunas) ; 60(6)2024 May 31.
Article in English | MEDLINE | ID: mdl-38929535

ABSTRACT

Introduction: The ongoing concern of the medical profession regarding chronic medication is related to increasing patient adherence and compliance to treatment and reducing medication side effects. In this respect, drugs represented by fixed-dose combinations of active substances within the same tablet have emerged. Such a principle can be extrapolated by following the potential beneficial effects that a chronic medication can have on chronic pathologies affecting different systems. Materials and Methods: The study included 48 female Albino Wistar rats, aged 16-18 months, which were divided into two groups: ovariectomized and non-ovariectomized rats. One batch of 12 non-ovariectomized rats received no treatment, becoming a control batch (NOVX-M). The ovariectomized (OVX) group was divided into 3 batches of 12 rats each: no treatment, control (OVX-M), fenofibrate-treated (OVX-F) and statin-treated (OVX-S) rats. At 12 weeks after ovariectomy, a femoral fracture occurred in the right hind limb of all animals included in the experiment To reveal the changes, at intervals of 2, 4, 6 and 8 weeks post-fracture, the proximal part of the femur was evaluated by NMR diffusiometry, which allows random motion of proton molecules expressed by self-diffusion coefficients, D, thus allowing analysis of the size and complexity of microscopic order cavities within biological structures, such as pores inside bones. Results: The effects of hypolipidemic medication in the absence of estrogen were evidenced, proving the beneficial effect that fenofibrate can have in preserving healthy tissue exposed to osteoporotic risk during the menopausal period. The effects of lipid-lowering medication are also influenced by the duration of administration. Conclusions: Osteoporosis and heart disease are two chronic pathologies that affect mainly female population in the second half of life, and proving the dual therapeutic potential of lipid-lowering medication may also have positive effects by increasing adherence and compliance to treatment.


Subject(s)
Hypolipidemic Agents , Ovariectomy , Rats, Wistar , Animals , Female , Rats , Hypolipidemic Agents/pharmacology , Hypolipidemic Agents/therapeutic use , Hypolipidemic Agents/administration & dosage , Magnetic Resonance Spectroscopy/methods , Fenofibrate/pharmacology , Fenofibrate/therapeutic use , Disease Models, Animal , Femur/drug effects , Bone and Bones/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...