Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 12.169
Filter
1.
Sci Rep ; 14(1): 12449, 2024 05 30.
Article in English | MEDLINE | ID: mdl-38816454

ABSTRACT

Bone graft granules implanted in bone defects come into physical contact with the host bone and form interconnected porous structure. However, there exists an accidental displacement of granules to unintended locations and leakage of granules from bone defects. Although covering the defect with a barrier membrane prevents granule emanation, this procedure is troublesome. To resolve these problems, we fabricated bioresorbable mesh cages (BRMc) in this study. Bone graft granules composed of carbonate apatite alone (Gr) and bioresorbable mesh cages (BRMc/Gr) introduced the bone graft granules and were implanted into the bone defect in the rabbit femur. Micro-computed tomography and histological analysis were conducted at 4 and 12 weeks after implantation. Osteoprogenitors in the bloodstream from the host bone passed through the pores of BRMc, penetrated the porous structure of graft granules, and might interact with individual granules. Then bone remodeling could progress actively and new bone was formed. The new bone formation was similar to the host bone at 12 weeks and there were minimal signs of local tissue inflammation. BRMc/Gr could reduce the risk of unwanted new bone formation occurring due to loss of granules from the bone defects compared with Gr because BRMc enclosed granules and prevent granules leakage from bone defects and BRMc could not induce unfavorable effects to forme new bone. Additionally, BRMc/Gr could keep granules assembled in one place, avoid displacement of granules to unintended locations, and carry easily. These results demonstrated that BRMc/Gr was effective in bone regeneration and improved clinical handling.


Subject(s)
Bone Transplantation , Femur , X-Ray Microtomography , Animals , Rabbits , Femur/surgery , Femur/diagnostic imaging , Femur/pathology , Bone Transplantation/methods , Absorbable Implants , Bone Regeneration , Osteogenesis/drug effects
2.
Int J Med Sci ; 21(6): 1155-1164, 2024.
Article in English | MEDLINE | ID: mdl-38774749

ABSTRACT

Introduction: Clinical studies have shown that endodontically-treated nonvital teeth exhibit less root resorption during orthodontic tooth movement. The purpose of this study was to explore whether hypoxic dental pulp stem cells (DPSCs) can promote osteoclastogenesis in orthodontically induced inflammatory root resorption (OIIRR). Methods: Succinate in the supernatant of DPSCs under normal and hypoxic conditions was measured by a succinic acid assay kit. The culture supernatant of hypoxia-treated DPSCs was used as conditioned medium (Hypo-CM). Bone marrow-derived macrophages (BMDMs) from succinate receptor 1 (SUCNR1)-knockout or wild-type mice were cultured with conditioned medium (CM), exogenous succinate or a specific inhibitor of SUCNR1 (4c). Tartrate-resistant acid phosphatase (TRAP) staining, Transwell assays, qPCR, Western blotting, and resorption assays were used to evaluate osteoclastogenesis-related changes. Results: The concentration of succinate reached a maximal concentration at 6 h in the supernatant of hypoxia-treated DPSCs. Hypo-CM-treated macrophages were polarized to M1 proinflammatory macrophages. Hypo-CM treatment significantly increased the formation and differentiation of osteoclasts and increased the expression of osteoclastogenesis-related genes, and this effect was inhibited by the specific succinate inhibitor 4c. Succinate promoted chemotaxis and polarization of M1-type macrophages with increased expression of osteoclast generation-related genes. SUCNR1 knockout decreased macrophage migration, M1 macrophage polarization, differentiation and maturation of osteoclasts, as shown by TRAP and NFATc1 expression and cementum resorption. Conclusions: Hypoxic DPSC-derived succinate may promote osteoclast differentiation and root resorption. The regulation of the succinate-SUCNR1 axis may contribute to the reduction in the OIIRR.


Subject(s)
Dental Pulp , Mice, Knockout , Osteoclasts , Osteogenesis , Root Resorption , Stem Cells , Succinic Acid , Animals , Mice , Dental Pulp/cytology , Dental Pulp/drug effects , Dental Pulp/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , Root Resorption/pathology , Root Resorption/metabolism , Humans , Succinic Acid/metabolism , Osteogenesis/drug effects , Stem Cells/metabolism , Stem Cells/drug effects , Cell Differentiation/drug effects , Macrophages/metabolism , Macrophages/drug effects , Cell Hypoxia/drug effects , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Culture Media, Conditioned/pharmacology , Cells, Cultured
3.
Molecules ; 29(9)2024 May 02.
Article in English | MEDLINE | ID: mdl-38731604

ABSTRACT

Edible grey oyster mushroom, Pleurotus sajor-caju, ß (1,3), (1,6) glucan possesses a wide range of biological activities, including anti-inflammation, anti-microorganism and antioxidant. However, its biological activity is limited by low water solubility resulting from its high molecular weight. Our previous study demonstrated that enzymatic hydrolysis of grey oyster mushroom ß-glucan using Hevea ß-1,3-glucanase isozymes obtains a lower molecular weight and higher water solubility, Pleurotus sajor-caju glucanoligosaccharide (Ps-GOS). Additionally, Ps-GOS potentially reduces osteoporosis by enhancing osteoblast-bone formation, whereas its effect on osteoclast-bone resorption remains unknown. Therefore, our study investigated the modulatory activities and underlying mechanism of Ps-GOS on Receptor activator of nuclear factor kappa-Β ligand (RANKL) -induced osteoclastogenesis in pre-osteoclastic RAW 264.7 cells. Cell cytotoxicity of Ps-GOS on RAW 264.7 cells was determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay and its effect on osteoclast differentiation was determined by tartrate-resistant acid phosphatase (TRAP) staining. Additionally, its effect on osteoclast bone-resorptive ability was detected by pit formation assay. The osteoclastogenic-related factors were assessed by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR), Western blot and immunofluorescence. The results revealed that Ps-GOS was non-toxic and significantly suppressed the formation of mature osteoclast multinucleated cells and their resorption activity by reducing the number of TRAP-positive cells and pit formation areas in a dose-dependent manner. Additionally, Ps-GOS attenuated the nuclear factor kappa light chain-enhancer of activated B cells' P65 (NFκB-P65) expression and their subsequent master osteoclast modulators, including nuclear factor of activated T cell c1 (NFATc1) and Fos proto-oncogene (cFOS) via the NF-κB pathway. Furthermore, Ps-GOS markedly inhibited RANK expression, which serves as an initial transmitter of many osteoclastogenesis-related cascades and inhibited proteolytic enzymes, including TRAP, matrix metallopeptidase 9 (MMP-9) and cathepsin K (CTK). These findings indicate that Ps-GOS could potentially be beneficial as an effective natural agent for bone metabolic disease.


Subject(s)
Cell Differentiation , NF-kappa B , NFATC Transcription Factors , Osteoclasts , Pleurotus , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Signal Transduction , Animals , Mice , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoclasts/cytology , RAW 264.7 Cells , RANK Ligand/metabolism , Cell Differentiation/drug effects , Signal Transduction/drug effects , NF-kappa B/metabolism , Pleurotus/chemistry , Receptor Activator of Nuclear Factor-kappa B/metabolism , NFATC Transcription Factors/metabolism , Proto-Oncogene Proteins c-fos/metabolism , beta-Glucans/pharmacology , beta-Glucans/chemistry , Oligosaccharides/pharmacology , Oligosaccharides/chemistry , Osteogenesis/drug effects
4.
J Physiol Pharmacol ; 75(2): 173-183, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38736264

ABSTRACT

Quercetin is widely distributed in plants as a flavonol compound with multiple biological activities. It has been found that quercetin can regulate bone homeostasis through multiple pathways and targets. This study investigated the role and specific molecular mechanisms of quercetin in regulating osteoblast viability, proliferation, migration and osteogenic differentiation. A mouse model of traumatic fracture was established and then 100 mg/kg quercetin corn oil suspension was gavaged at the same time every day for 28 days. miR-6089 and E2F transcription factor 2 (E2F2) expression levels in mice were measured. Fracture healing in mice was observed. MC3T3-E1 cells were transfected with plasmids targeting miR-6089 and E2F2, and cell viability, proliferation, migration, apoptosis, and osteogenic differentiation were determined. The targeting relationship between miR-6089 and E2F2 was verified. In vivo experiments showed that quercetin significantly increased osteocalcin (OCN) expression (P<0.05) and promoted fracture healing in traumatic fracture (TF) mice. miR-6089 expression was down-regulated (P<0.05) and E2F2 expression was up-regulated (P<0.05) in TF mice. Quercetin promoted miR-6089 expression and inhibited E2F2 expression (both P<0.05). In vitro results showed that quercetin promoted miR-6089 expression and inhibited E2F2 expression in a dose-dependent manner (both P<0.05). Quercetin dose-dependently promoted MC3T3-E1 cell viability, proliferation, migration, and osteogenic differentiation, and inhibited MC3T3-E1 cell apoptosis (all P<0.05). Up-regulating miR-6089 further promoted MC3T3-E1 cell viability, proliferation, migration and osteogenic differentiation, and inhibited MC3T3-E1 cell apoptosis (all P<0.05). miR-6089 targeted and regulated E2F2 expression. Up-regulating E2F2 attenuated the promoting effect of up-regulated miR-6089 on MC3T3-E1 cell viability, proliferation, migration, osteogenic differentiation, and inhibition of apoptosis (all P<0.05). We conclude that quercetin enhances osteoblast viability, proliferation, migration, and osteogenic differentiation by modulating the miR-6089/E2F2 axis, thereby promoting fracture healing.


Subject(s)
E2F2 Transcription Factor , Fracture Healing , MicroRNAs , Osteoblasts , Osteogenesis , Quercetin , Animals , Male , Mice , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , E2F2 Transcription Factor/metabolism , E2F2 Transcription Factor/genetics , Fracture Healing/drug effects , MicroRNAs/genetics , MicroRNAs/metabolism , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteogenesis/drug effects , Quercetin/pharmacology
5.
Iran Biomed J ; 28(2&3): 82-9, 2024 05 01.
Article in English | MEDLINE | ID: mdl-38770885

ABSTRACT

Background: Osteogenic, antioxidant and anti-inflammatory effects of Whey protein and M. oleifera gel prompted us to evaluate their role alone or in combination on osseointegration in rabbits. Methods: In this study, 24 titanium implants were inserted in the femurs of six rabbits. One implant was placed without treatment, and another one was coated with a mixture of whey protein and M. oleifera gel for each side. The animals were divided into two groups of 2- and 6-week intervals and evaluated using histopathological and immunohistochemical techniques. Results: Histological evaluation revealed a significant difference between the experimental and the control groups after two weeks in osteoblast and osteocyte counts. The experimental group had mature bone development after six weeks of implantation, while the control group had a woven bone. Immunohistochemical results showed that the experimental group, compared to the control group, exhibited early positive expression of osteoblast cells at two weeks after the experiment. Based on histopathological observations, the experimental group showed a tiny area of collagenous fiber in 6th week after the implantation. Conclusion: A mixture of whey protein and M. oleifera could accelerate osseointegration and healing processes.


Subject(s)
Moringa oleifera , Osseointegration , Plant Extracts , Plant Leaves , Whey Proteins , Animals , Whey Proteins/pharmacology , Rabbits , Osseointegration/drug effects , Moringa oleifera/chemistry , Plant Extracts/pharmacology , Plant Leaves/chemistry , Male , Osteoblasts/drug effects , Femur/drug effects , Osteogenesis/drug effects
6.
Nanoscale ; 16(20): 9861-9874, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38712977

ABSTRACT

A guided bone regeneration (GBR) membrane can act as a barrier to prevent the invasion and interference from foreign soft tissues, promoting infiltration and proliferation of osteoblasts in the bone defect area. Herein, a composite scaffold with dual functions of osteogenesis and antibacterial effects was prepared for GBR. A polycaprolactone (PCL)/nano-hydroxyapatite (n-HA) aerogel produced by electrospinning and freeze-drying techniques was fabricated as the loose layer of the scaffold, while a PCL nanofiber membrane was used as the dense layer. Chitosan (CS) solution served as a middle layer to provide mechanical support and antibacterial effects between the two layers. Morphological results showed that the loose layer had a porous structure with n-HA successfully dispersed in the aerogels, while the dense layer possessed a sufficiently dense structure. In vitro antibacterial experiments illustrated that the CS solution in the middle layer stabilized the scaffold structure and endowed the scaffold with good antibacterial properties. The cytocompatibility results indicated that both fibroblasts and osteoblasts exhibited superior cell activity on the dense and loose layers, respectively. In particular, the dense layer made of nanofibers could work as a barrier layer to inhibit the infiltration of fibroblasts into the loose layer. In vitro osteogenesis analysis suggested that the PCL/n-HA aerogel could enhance the bone induction ability of bone mesenchymal stem cells, which was confirmed by the increased expression of the alkaline phosphatase activity. The loose structure facilitated the infiltration and migration of bone mesenchymal stem cells for better osteogenesis. In summary, such a composite scaffold exhibited excellent osteogenic and antibacterial properties as well as the barrier effect, thus holding promising potential for use as GBR materials.


Subject(s)
Anti-Bacterial Agents , Bone Regeneration , Chitosan , Durapatite , Nanofibers , Osteoblasts , Osteogenesis , Polyesters , Chitosan/chemistry , Chitosan/pharmacology , Durapatite/chemistry , Durapatite/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bone Regeneration/drug effects , Nanofibers/chemistry , Polyesters/chemistry , Polyesters/pharmacology , Animals , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteogenesis/drug effects , Mice , Tissue Scaffolds/chemistry , Gels/chemistry , Staphylococcus aureus/drug effects , Fibroblasts/drug effects , Fibroblasts/cytology
7.
ACS Appl Mater Interfaces ; 16(20): 25799-25812, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38727024

ABSTRACT

The excess production of reactive oxygen species (ROS) will delay tooth extraction socket (TES) healing. In this study, we developed an injectable thermosensitive hydrogel (NBP@BP@CS) used to treat TES healing. The hydrogel formulation incorporated black phosphorus (BP) nanoflakes, recognized for their accelerated alveolar bone regeneration and ROS-scavenging properties, and dl-3-n-butylphthalide (NBP), a vasodilator aimed at enhancing angiogenesis. In vivo investigations strongly demonstrated that NBP@BP@CS improved TES healing due to antioxidation and promotion of alveolar bone regeneration by BP nanoflakes. The sustained release of NBP from the hydrogel promoted neovascularization and vascular remodeling. Our results demonstrated that the designed thermosensitive hydrogel provided great opportunity not only for ROS elimination but also for the promotion of osteogenesis and angiogenesis, reflecting the "three birds with one stone" concept, and has tremendous potential for rapid TES healing.


Subject(s)
Hydrogels , Phosphorus , Tooth Extraction , Wound Healing , Animals , Hydrogels/chemistry , Hydrogels/pharmacology , Wound Healing/drug effects , Phosphorus/chemistry , Tooth Socket/drug effects , Neovascularization, Physiologic/drug effects , Reactive Oxygen Species/metabolism , Osteogenesis/drug effects , Rats , Bone Regeneration/drug effects , Male
8.
Scand Cardiovasc J ; 58(1): 2353070, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38757904

ABSTRACT

Objectives: The role of diabetes mellitus as a risk factor for the development of calcific aortic valve disease has not been fully clarified. Aortic valve interstitial cells (VICs) have been suggested to be crucial for calcification of the valve. Induced calcification in cultured VICs is a good in vitro model for aortic valve calcification. The purpose of this study was to investigate whether increased glucose levels increase experimentally induced calcification in cultured human VICs. Design: VICs were isolated from explanted calcified aortic valves after valve replacement. Osteogenic medium induced calcification of cultured VICs at different glucose levels (5, 15, and 25 mM). Calcium deposits were visualized using Alizarin Red staining and measured spectrophotometrically. Results: The higher the glucose concentration, the lower the level of calcification. High glucose (25 mM) reduced calcification by 52% compared with calcification at a physiological (5 mM) glucose concentration (correlation and regression analysis: r = -0.55, p = .025 with increased concentration of glucose). Conclusions: In vitro hyperglycemia-like conditions attenuated calcification in VICs. High glucose levels may trigger a series of events that secondarily stimulate calcification of VICs in vivo.


Subject(s)
Aortic Valve Stenosis , Aortic Valve , Calcinosis , Glucose , Hyperglycemia , Humans , Aortic Valve/pathology , Aortic Valve/metabolism , Aortic Valve/surgery , Calcinosis/pathology , Calcinosis/metabolism , Cells, Cultured , Glucose/metabolism , Hyperglycemia/metabolism , Aortic Valve Stenosis/pathology , Aortic Valve Stenosis/metabolism , Aortic Valve Stenosis/surgery , Male , Middle Aged , Aged , Female , Dose-Response Relationship, Drug , Osteogenesis/drug effects
9.
Pak J Pharm Sci ; 37(1(Special)): 223-229, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38747273

ABSTRACT

In this study, the anti-osteogenic properties of the volatile oil extracted from Homalomena gigantea rhizome using ethyl acetate (EtOAc) and methanol (MeOH) were examined. Gas chromatography-mass spectrometry (GC-MS) was employed for the identification of volatile components. Following this, bioassays were performed to evaluate their effects on osteogenesis, encompassing parameters like cell viability, osteoblast differentiation, collagen synthesis and mineralization. The GC-MS analysis revealed 19 compounds in the EtOAc extract and 36 compounds in the MeOH extract. In the MeOH extract, major constituents included bis(2-ethylhexyl) terephthalate (13.83%), linalool (9.58%), palmitic acid (6.55%) and stearic acid (4.29%). The EtOAc extract contained bis(2-ethylhexyl) terephthalate (16.64%), palmitic acid (5.60%) and stearic acid (3.11%) as the predominant components. Both the EtOAc and MeOH extracts of H. gigantea exhibited promising potential for further investigation in anti-osteoporosis research. These findings contribute to the exploration of natural compounds with potential anti-osteoporotic properties, expanding our understanding of their therapeutic potential.


Subject(s)
Gas Chromatography-Mass Spectrometry , Oils, Volatile , Osteogenesis , Plant Extracts , Rhizome , Osteogenesis/drug effects , Rhizome/chemistry , Plant Extracts/pharmacology , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Oils, Volatile/pharmacology , Oils, Volatile/chemistry , Oils, Volatile/isolation & purification , Animals , Cell Survival/drug effects , Osteoblasts/drug effects , Cell Differentiation/drug effects , Mice , Palmitic Acid/pharmacology , Acyclic Monoterpenes/pharmacology
10.
Braz Oral Res ; 38: e037, 2024.
Article in English | MEDLINE | ID: mdl-38747824

ABSTRACT

Dental pulp stem cells (DPSCs) and periodontal ligament stem cells (PDLSCs) can differentiate into osteoblasts, indicating that both are potential candidates for bone tissue engineering. Osteogenesis is influenced by many environmental factors, one of which is lipopolysaccharide (LPS). LPS-induced NF-κB activity affects the osteogenic potencies of different types of MSCs differently. This study evaluated the effect of LPS-induced NF-κB activity and its inhibition in DPSCs and PDLSCs. DPSCs and PDLSCs were cultured in an osteogenic medium, pretreated with/without NF-κB inhibitor Bay 11-7082, and treated with/without LPS. Alizarin red staining was performed to assess bone nodule formation, which was observed under an inverted light microscope. NF-κB and alkaline phosphatase (ALP) activities were measured to examine the effect of Bay 11-7082 pretreatment and LPS supplementation on osteogenic differentiation of DPSCs and PDLSCs. LPS significantly induced NF-κB activity (p = 0.000) and reduced ALP activity (p = 0.000), which inhibited bone nodule formation in DPSCs and PDLSCs. Bay 11-7082 inhibited LPS-induced NF-κB activity, and partially maintained ALP activity and osteogenic potency of LPS-supplemented DPSCs and PDLSCs. Thus, inhibition of LPS-induced NF-κB activity can maintain the osteogenic potency of DPSCs and PDLSCs.


Subject(s)
Alkaline Phosphatase , Cell Differentiation , Dental Pulp , Lipopolysaccharides , NF-kappa B , Nitriles , Osteogenesis , Periodontal Ligament , Stem Cells , Humans , Lipopolysaccharides/pharmacology , Periodontal Ligament/cytology , Periodontal Ligament/drug effects , Osteogenesis/drug effects , Osteogenesis/physiology , Dental Pulp/cytology , Dental Pulp/drug effects , NF-kappa B/metabolism , Alkaline Phosphatase/analysis , Cell Differentiation/drug effects , Stem Cells/drug effects , Stem Cells/physiology , Cells, Cultured , Nitriles/pharmacology , Sulfones/pharmacology , Reproducibility of Results , Time Factors , Young Adult , Adolescent
11.
Cells ; 13(9)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38727297

ABSTRACT

Spinal fusion, a common surgery performed for degenerative lumbar conditions, often uses recombinant human bone morphogenetic protein 2 (rhBMP-2) that is associated with adverse effects. Mesenchymal stromal/stem cells (MSCs) and their extracellular vesicles (EVs), particularly exosomes, have demonstrated efficacy in bone and cartilage repair. However, the efficacy of MSC exosomes in spinal fusion remains to be ascertained. This study investigates the fusion efficacy of MSC exosomes delivered via an absorbable collagen sponge packed in a poly Ɛ-caprolactone tricalcium phosphate (PCL-TCP) scaffold in a rat posterolateral spinal fusion model. Herein, it is shown that a single implantation of exosome-supplemented collagen sponge packed in PCL-TCP scaffold enhanced spinal fusion and improved mechanical stability by inducing bone formation and bridging between the transverse processes, as evidenced by significant improvements in fusion score and rate, bone structural parameters, histology, stiffness, and range of motion. This study demonstrates for the first time that MSC exosomes promote bone formation to enhance spinal fusion and mechanical stability in a rat model, supporting its translational potential for application in spinal fusion.


Subject(s)
Exosomes , Mesenchymal Stem Cells , Rats, Sprague-Dawley , Spinal Fusion , Animals , Exosomes/metabolism , Exosomes/transplantation , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Spinal Fusion/methods , Rats , Osteogenesis/drug effects , Calcium Phosphates/pharmacology , Male , Humans , Tissue Scaffolds/chemistry , Bone Morphogenetic Protein 2/metabolism , Mesenchymal Stem Cell Transplantation/methods
12.
ACS Appl Mater Interfaces ; 16(19): 24384-24397, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38709640

ABSTRACT

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


Subject(s)
Bone Regeneration , Deferoxamine , Magnesium , Mesenchymal Stem Cells , Oxides , Tantalum , Deferoxamine/chemistry , Deferoxamine/pharmacology , Bone Regeneration/drug effects , Tantalum/chemistry , Animals , Oxides/chemistry , Oxides/pharmacology , Magnesium/chemistry , Magnesium/pharmacology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteogenesis/drug effects , Neovascularization, Physiologic/drug effects , Rats , Mice , Rats, Sprague-Dawley , Cell Proliferation/drug effects , Angiogenesis
13.
Gut Microbes ; 16(1): 2351532, 2024.
Article in English | MEDLINE | ID: mdl-38727248

ABSTRACT

Emerging evidence indicates that alteration of gut microbiota plays an important role in chronic kidney disease (CKD)-related vascular calcification (VC). We aimed to investigate the specific gut microbiota and the underlying mechanism involved in CKD-VC. We identified an increased abundance of Prevotella copri (P. copri) in the feces of CKD rats (induced by using 5/6 nephrectomy followed by a high calcium and phosphate diet) with aortic calcification via amplicon sequencing of 16S rRNA genes. In patients with CKD, we further confirmed a positive correlation between abundance of P. copri and aortic calcification scores. Moreover, oral administration of live P. copri aggravated CKD-related VC and osteogenic differentiation of vascular smooth muscle cells in vivo, accompanied by intestinal destruction, enhanced expression of Toll-like receptor-4 (TLR4), and elevated lipopolysaccharide (LPS) levels. In vitro and ex vivo experiments consistently demonstrated that P. copri-derived LPS (Pc-LPS) accelerated high phosphate-induced VC and VSMC osteogenic differentiation. Mechanistically, Pc-LPS bound to TLR4, then activated the nuclear factor κB (NF-κB) and nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3 (NLRP3) inflammasome signals during VC. Inhibition of NF-κB reduced NLRP3 inflammasome and attenuated Pc-LPS-induced VSMC calcification. Our study clarifies a novel role of P. copri in CKD-related VC, by the mechanisms involving increased inflammation-regulating metabolites including Pc-LPS, and activation of the NF-κB/NLRP3 signaling pathway. These findings highlight P. copri and its-derived LPS as potential therapeutic targets for VC in CKD.


Subject(s)
Gastrointestinal Microbiome , Lipopolysaccharides , NF-kappa B , Prevotella , Renal Insufficiency, Chronic , Signal Transduction , Toll-Like Receptor 4 , Vascular Calcification , Animals , Vascular Calcification/metabolism , Vascular Calcification/pathology , NF-kappa B/metabolism , Lipopolysaccharides/metabolism , Rats , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/microbiology , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/pathology , Humans , Male , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/genetics , Prevotella/metabolism , Rats, Sprague-Dawley , Myocytes, Smooth Muscle/metabolism , Osteogenesis/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Feces/microbiology , Inflammasomes/metabolism
14.
Stem Cell Res Ther ; 15(1): 135, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38715130

ABSTRACT

BACKGROUND: Biomaterials used in bone tissue engineering must fulfill the requirements of osteoconduction, osteoinduction, and osseointegration. However, biomaterials with good osteoconductive properties face several challenges, including inadequate vascularization, limited osteoinduction and barrier ability, as well as the potential to trigger immune and inflammatory responses. Therefore, there is an urgent need to develop guided bone regeneration membranes as a crucial component of tissue engineering strategies for repairing bone defects. METHODS: The mZIF-8/PLA membrane was prepared using electrospinning technology and simulated body fluid external mineralization method. Its ability to induce biomimetic mineralization was evaluated through TEM, EDS, XRD, FT-IR, zeta potential, and wettability techniques. The biocompatibility, osteoinduction properties, and osteo-immunomodulatory effects of the mZIF-8/PLA membrane were comprehensively evaluated by examining cell behaviors of surface-seeded BMSCs and macrophages, as well as the regulation of cellular genes and protein levels using PCR and WB. In vivo, the mZIF-8/PLA membrane's potential to promote bone regeneration and angiogenesis was assessed through Micro-CT and immunohistochemical staining. RESULTS: The mineralized deposition enhances hydrophilicity and cell compatibility of mZIF-8/PLA membrane. mZIF-8/PLA membrane promotes up-regulation of osteogenesis and angiogenesis related factors in BMSCs. Moreover, it induces the polarization of macrophages towards the M2 phenotype and modulates the local immune microenvironment. After 4-weeks of implantation, the mZIF-8/PLA membrane successfully bridges critical bone defects and almost completely repairs the defect area after 12-weeks, while significantly improving the strength and vascularization of new bone. CONCLUSIONS: The mZIF-8/PLA membrane with dual osteoconductive and immunomodulatory abilities could pave new research paths for bone tissue engineering.


Subject(s)
Bone Regeneration , Bone Regeneration/drug effects , Animals , Osteogenesis/drug effects , Tissue Engineering/methods , Biocompatible Materials/pharmacology , Biocompatible Materials/chemistry , Mice , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Membranes, Artificial , Guided Tissue Regeneration/methods , Tissue Scaffolds/chemistry , Polyesters/chemistry , Polyesters/pharmacology , Rats
15.
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
16.
Mol Med ; 30(1): 57, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38698308

ABSTRACT

BACKGROUND: Ossification of the posterior longitudinal ligament (OPLL), an emerging heterotopic ossification disease, causes spinal cord compression, resulting in motor and sensory dysfunction. The etiology of OPLL remains unclear but may involve integrin αVß3 regulating the process of osteogenesis and angiogenesis. In this study, we focused on the role of integrin αVß3 in OPLL and explored the underlying mechanism by which the c(RGDyk) peptide acts as a potent and selective integrin αVß3 inhibitor to inhibit osteogenesis and angiogenesis in OPLL. METHODS: OPLL or control ligament samples were collected in surgery. For OPLL samples, RNA-sequencing results revealed activation of the integrin family, particularly integrin αVß3. Integrin αVß3 expression was detected by qPCR, Western blotting, and immunohistochemical analysis. Fluorescence microscopy was used to observe the targeted inhibition of integrin αVß3 by the c(RGDyk) peptide on ligaments fibroblasts (LFs) derived from patients with OPLL and endothelial cells (ECs). The effect of c(RGDyk) peptide on the ossification of pathogenic LFs was detected using qPCR, Western blotting. Alkaline phosphatase staining or alizarin red staining were used to test the osteogenic capability. The effect of the c(RGDyk) peptide on angiogenesis was determined by EC migration and tube formation assays. The effects of the c(RGDyk) peptide on heterotopic bone formation were evaluated by micro-CT, histological, immunohistochemical, and immunofluorescence analysis in vivo. RESULTS: The results indicated that after being treated with c(RGDyk), the osteogenic differentiation of LFs was significantly decreased. Moreover, the c(RGDyk) peptide inhibited the migration of ECs and thus prevented the nutritional support required for osteogenesis. Furthermore, the c(RGDyk) peptide inhibited ectopic bone formation in mice. Mechanistic analysis revealed that c(RGDyk) peptide could inhibit osteogenesis and angiogenesis in OPLL by targeting integrin αVß3 and regulating the FAK/ERK pathway. CONCLUSIONS: Therefore, the integrin αVß3 appears to be an emerging therapeutic target for OPLL, and the c(RGDyk) peptide has dual inhibitory effects that may be valuable for the new therapeutic strategy of OPLL.


Subject(s)
Integrin alphaVbeta3 , Ossification of Posterior Longitudinal Ligament , Osteogenesis , Integrin alphaVbeta3/metabolism , Integrin alphaVbeta3/antagonists & inhibitors , Humans , Osteogenesis/drug effects , Animals , Mice , Ossification of Posterior Longitudinal Ligament/metabolism , Ossification of Posterior Longitudinal Ligament/drug therapy , Male , Female , Middle Aged , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Fibroblasts/metabolism , Fibroblasts/drug effects , Neovascularization, Physiologic/drug effects , Cell Movement/drug effects , Disease Models, Animal , Oligopeptides/pharmacology , Oligopeptides/chemistry , Angiogenesis
17.
J Biomed Mater Res B Appl Biomater ; 112(6): e35409, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38786580

ABSTRACT

The challenge of integrating hydroxyapatite nanoparticles (nHAp) with polymers is hindered by the conflict between the hydrophilic and hygroscopic properties of nHAp and the hydrophobic properties of polymers. This conflict particularly affects the materials when calcium phosphates, including nHAp, are used as a filler in composites in thermal processing applications such as 3D printing with fused filament fabrication (FFF). To overcome this, we propose a one-step surface modification of nHAp with calcium stearate monolayer. Moreover, to build the scaffold with suitable mechanical strength, we tested the addition of nHAp with diverse morphology-spherical, plate- and rod-like nanoparticles. Our analysis showed that the composite of polycaprolactone (PCL) reinforced with nHAp with rod and plate morphologies modified with calcium stearate monolayer exhibited a significant increase in compressive strength. However, composites with spherical nHAp added to PCL showed a significant reduction in compressive modulus and compressive strength, but both parameters were within the applicability range of hard tissue scaffolds. None of the tested composite scaffolds showed cytotoxicity in L929 murine fibroblasts or MG-63 human osteoblast-like cells, supporting the proliferation of the latter. Additionally, PCL/nHAp scaffolds reinforced with spherical nHAp caused osteoactivation of bone marrow human mesenchymal stem cells, as indicated by alkaline phosphatase activity and COL1, RUNX2, and BGLAP expression. These results suggest that the calcium stearate monolayer on the surface of the nHAp particles allows the production of polymer/nHAp composites suitable for hard tissue engineering and personalized implant production in 3D printing using the FFF technique.


Subject(s)
Durapatite , Nanoparticles , Printing, Three-Dimensional , Tissue Engineering , Tissue Scaffolds , Tissue Scaffolds/chemistry , Durapatite/chemistry , Durapatite/pharmacology , Mice , Animals , Humans , Nanoparticles/chemistry , Cell Line , Polyesters/chemistry , Osteoblasts/metabolism , Osteoblasts/cytology , Osteogenesis/drug effects , Materials Testing
18.
Nutrients ; 16(10)2024 May 10.
Article in English | MEDLINE | ID: mdl-38794681

ABSTRACT

Recent interest in preventing the development of osteoporosis has focused on the regulation of redox homeostasis. However, the action of lycopene (LYC), a strong natural antioxidant compound, on osteoporotic bone loss remains largely unknown. Here, we show that oral administration of LYC to OVX rats for 12 weeks reduced body weight gain, improved lipid metabolism, and preserved bone quality. In addition, LYC treatment inhibited ROS overgeneration in serum and bone marrow in OVX rats, and in BMSCs upon H2O2 stimulation, leading to inhibiting adipogenesis and promoting osteogenesis during bone remodeling. At the molecular level, LYC improved bone quality via an increase in the expressions of FoxO1 and Runx2 and a decrease in the expressions of PPARγ and C/EBPα in OVX rats and BMSCs. Collectively, these findings suggest that LYC attenuates osteoporotic bone loss through promoting osteogenesis and inhibiting adipogenesis via regulation of the FoxO1/PPARγ pathway driven by oxidative stress, presenting a novel strategy for osteoporosis management.


Subject(s)
Adipogenesis , Lycopene , Mesenchymal Stem Cells , Osteogenesis , Ovariectomy , PPAR gamma , Rats, Sprague-Dawley , Signal Transduction , Animals , Osteogenesis/drug effects , Adipogenesis/drug effects , Lycopene/pharmacology , PPAR gamma/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Female , Signal Transduction/drug effects , Rats , Osteoporosis/prevention & control , Oxidative Stress/drug effects , Forkhead Box Protein O1/metabolism , Antioxidants/pharmacology , Reactive Oxygen Species/metabolism
19.
J Mech Behav Biomed Mater ; 155: 106579, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38749266

ABSTRACT

Silicon nitride is utilized clinically as a bioceramic for spinal fusion cages, owing to its high strength, osteoconductivity, and antibacterial effects. Nevertheless, silicon nitride exhibits suboptimal damping properties, a critical factor in mitigating traumatic bone injuries and fractures. In fact, there is a scarcity of spinal implants that simultaneously demonstrate proficient damping performance and support osteogenesis. In our study, we fabricated a novel sodium alginate-silicon nitride/poly(vinyl alcohol) (SA-SiN/PVA) composite scaffold, enabling enhanced energy absorption and rapid elastic recovery under quasi-static and impact loading scenarios. Furthermore, the study demonstrated that the incorporation of physical and chemical cross-linking significantly improved stiffness and recoverable energy dissipation. Concerning the interaction between cells and materials, our findings suggest that the addition of silicon nitride stimulated osteogenic differentiation while inhibiting Staphylococcus aureus growth. Collectively, the amalgamation of ceramics and tough hydrogels facilitates the development of advanced composites for spinal implants, manifesting superior damping, osteogenic potential, and antibacterial properties. This approach holds broader implications for applications in bone tissue engineering.


Subject(s)
Alginates , Biocompatible Materials , Materials Testing , Polyvinyl Alcohol , Silicon Compounds , Staphylococcus aureus , Alginates/chemistry , Alginates/pharmacology , Polyvinyl Alcohol/chemistry , Silicon Compounds/chemistry , Silicon Compounds/pharmacology , Staphylococcus aureus/drug effects , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Osteogenesis/drug effects , Mechanical Phenomena , Tissue Scaffolds/chemistry , Humans
20.
Int J Mol Sci ; 25(10)2024 May 08.
Article in English | MEDLINE | ID: mdl-38791156

ABSTRACT

The deterioration of osteoblast-led bone formation and the upregulation of osteoclast-regulated bone resorption are the primary causes of bone diseases, including osteoporosis. Numerous circulating factors play a role in bone homeostasis by regulating osteoblast and osteoclast activity, including the sphingolipid-sphingosine-1-phosphate (S1P). However, to date no comprehensive studies have investigated the impact of S1P activity on human and murine osteoblasts and osteoclasts. We observed species-specific responses to S1P in both osteoblasts and osteoclasts, where S1P stimulated human osteoblast mineralisation and reduced human pre-osteoclast differentiation and mineral resorption, thereby favouring bone formation. The opposite was true for murine osteoblasts and osteoclasts, resulting in more mineral resorption and less mineral deposition. Species-specific differences in osteoblast responses to S1P were potentially explained by differential expression of S1P receptor 1. By contrast, human and murine osteoclasts expressed comparable levels of S1P receptors but showed differential expression patterns of the two sphingosine kinase enzymes responsible for S1P production. Ultimately, we reveal that murine models may not accurately represent how human bone cells will respond to S1P, and thus are not a suitable model for exploring S1P physiology or potential therapeutic agents.


Subject(s)
Cell Differentiation , Lysophospholipids , Osteoblasts , Osteoclasts , Species Specificity , Sphingosine , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Lysophospholipids/metabolism , Humans , Animals , Mice , Osteoclasts/metabolism , Osteoclasts/cytology , Osteoblasts/metabolism , Osteoblasts/drug effects , Osteogenesis/drug effects , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Sphingosine-1-Phosphate Receptors/metabolism , Bone and Bones/metabolism , Bone Resorption/metabolism , Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...