Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 16.167
Filter
1.
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
2.
Front Immunol ; 15: 1396122, 2024.
Article in English | MEDLINE | ID: mdl-38817601

ABSTRACT

As the world population ages, osteoporosis, the most common disease of bone metabolism, affects more than 200 million people worldwide. The etiology is an imbalance in bone remodeling process resulting in more significant bone resorption than bone remodeling. With the advent of the osteoimmunology field, the immune system's role in skeletal pathologies is gradually being discovered. The cytokine interferon-gamma (IFN-γ), a member of the interferon family, is an important factor in the etiology and treatment of osteoporosis because it mediates bone remodeling. This review starts with bone remodeling process and includes the cellular and key signaling pathways of bone remodeling. The effects of IFN-γ on osteoblasts, osteoclasts, and bone mass are discussed separately, while the overall effects of IFN-γ on primary and secondary osteoporosis are summarized. The net effect of IFN-γ on bone appears to be highly dependent on the environment, dose, concentration, and stage of cellular differentiation. This review focuses on the mechanisms of bone remodeling and bone immunology, with a comprehensive discussion of the relationship between IFN-γ and osteoporosis. Finding the paradoxical balance of IFN-γ in bone immunology and exploring the potential of its clinical application provide new ideas for the clinical treatment of osteoporosis and drug development.


Subject(s)
Bone Remodeling , Interferon-gamma , Osteoporosis , Humans , Bone Remodeling/drug effects , Osteoporosis/immunology , Osteoporosis/etiology , Interferon-gamma/metabolism , Interferon-gamma/immunology , Animals , Osteoclasts/immunology , Osteoclasts/metabolism , Osteoblasts/immunology , Osteoblasts/metabolism , Signal Transduction , Bone and Bones/immunology , Bone and Bones/metabolism , Bone and Bones/pathology
3.
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
4.
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
5.
J Histochem Cytochem ; 72(5): 309-327, 2024 May.
Article in English | MEDLINE | ID: mdl-38725403

ABSTRACT

To clarify the cellular mechanism of cortical porosity induced by intermittent parathyroid hormone (PTH) administration, we examined the femoral cortical bone of mice that received 40 µg/kg/day (four times a day) human PTH (hPTH) (1-34). The PTH-driven cortical porosity initiated from the metaphyseal region and chronologically expanded toward the diaphysis. Alkaline phosphatase (ALP)-positive osteoblasts in the control mice covered the cortical surface, and endomucin-positive blood vessels were distant from these osteoblasts. In PTH-administered mice, endomucin-reactive blood vessels with TRAP-positive penetrated the ALP-positive osteoblast layer, invading the cortical bone. Statistically, the distance between endomucin-positive blood vessels and the cortical bone surface abated after PTH administration. Transmission electron microscopic observation demonstrated that vascular endothelial cells often pass through the flattened osteoblast layer and accompanied osteoclasts in the deep region of the cortical bone. The cell layers covering mature osteoblasts thickened with PTH administration and exhibited ALP, α-smooth muscle actin (αSMA), vascular cell adhesion molecule-1 (VCAM1), and receptor activator of NF-κB ligand (RANKL). Within these cell layers, osteoclasts were found near endomucin-reactive blood vessels. In PTH-administered femora, osteocytes secreted Dkk1, a Wnt inhibitor that affects angiogenesis, and blood vessels exhibited plasmalemma vesicle-associated protein, an angiogenic molecule. In summary, endomucin-positive blood vessels, when accompanied by osteoclasts in the ALP/αSMA/VCAM1/RANKL-reactive osteoblastic cell layers, invade the cortical bone, potentially due to the action of osteocyte-derived molecules such as DKK1.


Subject(s)
Cortical Bone , Endothelial Cells , Parathyroid Hormone , Animals , Mice , Parathyroid Hormone/pharmacology , Parathyroid Hormone/administration & dosage , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Cortical Bone/drug effects , Cortical Bone/metabolism , Porosity , Male , Osteoblasts/drug effects , Osteoblasts/metabolism , Immunohistochemistry , Femur/drug effects , Femur/blood supply , Femur/metabolism , Humans
6.
Front Immunol ; 15: 1397432, 2024.
Article in English | MEDLINE | ID: mdl-38751427

ABSTRACT

Introduction: The release of mature interleukin (IL-) 1ß from osteoblasts in response to danger signals is tightly regulated by the nucleotide-binding oligomerization domain leucine-rich repeat and pyrin-containing protein 3 (NLRP3) inflammasome. These danger signals include wear products resulting from aseptic loosening of joint arthroplasty. However, inflammasome activation requires two different signals: a nuclear factor-kappa B (NF-κB)-activating priming signal and an actual inflammasome-activating signal. Since human osteoblasts react to wear particles via Toll-like receptors (TLR), particles may represent an inflammasome activator that can induce both signals. Methods: Temporal gene expression profiles of TLRs and associated intracellular signaling pathways were determined to investigate the period when human osteoblasts take up metallic wear particles after initial contact and initiate a molecular response. For this purpose, human osteoblasts were treated with metallic particles derived from cobalt-chromium alloy (CoCr), lipopolysaccharides (LPS), and tumor necrosis factor-alpha (TNF) alone or in combination for incubation times ranging from one hour to three days. Shortly after adding the particles, their uptake was observed by the change in cell morphology and spectral data. Results: Exposure of osteoblasts to particles alone increased NLRP3 inflammasome-associated genes. The response was not significantly enhanced when cells were treated with CoCr + LPS or CoCr + TNF, whereas inflammation markers were induced. Despite an increase in genes related to the NLRP3 inflammasome, the release of IL-1ß was unaffected after contact with CoCr particles. Discussion: Although CoCr particles affect the expression of NLRP3 inflammasome-associated genes, a single stimulus was not sufficient to prime and activate the inflammasome. TNF was able to prime the NLRP3 inflammasome of human osteoblasts.


Subject(s)
Gene Expression Regulation , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Osteoblasts , Tumor Necrosis Factor-alpha , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Osteoblasts/metabolism , Osteoblasts/drug effects , Osteoblasts/immunology , Inflammasomes/metabolism , Tumor Necrosis Factor-alpha/metabolism , Gene Expression Regulation/drug effects , Cells, Cultured , Signal Transduction/drug effects
7.
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
8.
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
9.
Biol Open ; 13(5)2024 May 15.
Article in English | MEDLINE | ID: mdl-38742438

ABSTRACT

Bone is increasingly recognized as a target for diabetic complications. In order to evaluate the direct effects of high glucose on bone, we investigated the global transcriptional changes induced by hyperglycemia in osteoblasts in vitro. Rat bone marrow-derived mesenchymal stromal cells were differentiated into osteoblasts for 10 days, and prior to analysis, they were exposed to hyperglycemia (25 mM) for the short-term (1 or 3 days) or long-term (10 days). Genes and pathways regulated by hyperglycemia were identified using mRNA sequencing and verified with qPCR. Genes upregulated by 1-day hyperglycemia were, for example, related to extracellular matrix organization, collagen synthesis and bone formation. This stimulatory effect was attenuated by 3 days. Long-term exposure impaired osteoblast viability, and downregulated, for example, extracellular matrix organization and lysosomal pathways, and increased intracellular oxidative stress. Interestingly, transcriptional changes by different exposure times were mostly unique and only 89 common genes responding to glucose were identified. In conclusion, short-term hyperglycemia had a stimulatory effect on osteoblasts and bone formation, whereas long-term hyperglycemia had a negative effect on intracellular redox balance, osteoblast viability and function.


Subject(s)
Gene Expression Regulation , Glucose , Osteoblasts , Osteoblasts/metabolism , Osteoblasts/drug effects , Animals , Glucose/metabolism , Rats , Gene Expression Regulation/drug effects , Gene Expression Profiling , Hyperglycemia/metabolism , Hyperglycemia/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Transcriptome , Osteogenesis/drug effects , Osteogenesis/genetics , Cell Survival/drug effects , Transcription, Genetic/drug effects , Cells, Cultured , Oxidative Stress/drug effects
10.
FASEB J ; 38(10): e23646, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38795328

ABSTRACT

Multiple regulatory mechanisms are in place to ensure the normal processes of bone metabolism, encompassing both bone formation and absorption. This study has identified chaperone-mediated autophagy (CMA) as a critical regulator that safeguards bone formation from the detrimental effects of excessive inflammation. By silencing LAMP2A or HSCA8, we observed a hindrance in the osteoblast differentiation of human bone marrow mesenchymal stem cells (hBMSCs) in vitro. To further elucidate the role of LAMP2A, we generated LAMP2A gene knockdown and overexpression of mouse BMSCs (mBMSCs) using adenovirus. Our results showed that LAMP2A knockdown led to a decrease in osteogenic-specific proteins, while LAMP2A overexpression favored the osteogenesis of mBMSCs. Notably, active-ß-catenin levels were upregulated by LAMP2A overexpression. Furthermore, we found that LAMP2A overexpression effectively protected the osteogenesis of mBMSCs from TNF-α, through the PI3K/AKT/GSK3ß/ß-catenin pathway. Additionally, LAMP2A overexpression significantly inhibited osteoclast hyperactivity induced by TNF-α. Finally, in a murine bone defect model, we demonstrated that controlled release of LAMP2A overexpression adenovirus by alginate sodium capsule efficiently protected bone healing from inflammation, as confirmed by imaging and histological analyses. Collectively, our findings suggest that enhancing CMA has the potential to safeguard bone formation while mitigating hyperactivity in bone absorption.


Subject(s)
Chaperone-Mediated Autophagy , Glycogen Synthase Kinase 3 beta , Inflammation , Lysosomal-Associated Membrane Protein 2 , Mesenchymal Stem Cells , Osteogenesis , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , beta Catenin , Animals , Osteogenesis/physiology , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Proto-Oncogene Proteins c-akt/metabolism , Mice , Phosphatidylinositol 3-Kinases/metabolism , beta Catenin/metabolism , Humans , Mesenchymal Stem Cells/metabolism , Inflammation/metabolism , Lysosomal-Associated Membrane Protein 2/metabolism , Lysosomal-Associated Membrane Protein 2/genetics , Signal Transduction , Male , Mice, Inbred C57BL , Osteoblasts/metabolism , Cell Differentiation , Osteoclasts/metabolism
11.
Cells ; 13(10)2024 May 16.
Article in English | MEDLINE | ID: mdl-38786073

ABSTRACT

In this study of the alterations of Glypicans 1 to 6 (GPCs) and Notum in plasma, bone marrow mesenchymal stromal cells (BM-MSCs) and osteoblasts in Osteoarthritis (OA), the levels of GPCs and Notum in the plasma of 25 patients and 24 healthy subjects were measured. In addition, BM-MSCs from eight OA patients and eight healthy donors were cultured over a period of 21 days using both a culture medium and an osteogenic medium. Protein and gene expression levels of GPCs and Notum were determined using ELISA and qPCR at 0, 7, 14 and 21 days. GPC5 and Notum levels decreased in the plasma of OA patients, while the BM-MSCs of OA patients showed downexpression of GPC6 and upregulation of Notum. A decrease in GPC5 and Notum proteins and an increase in GPC3 were found. During osteogenic differentiation, elevated GPCs 2, 4, 5, 6 and Notum mRNA levels and decreased GPC3 were observed in patients with OA. Furthermore, the protein levels of GPC2, GPC5 and Notum decreased, while the levels of GPC3 increased. Glypicans and Notum were altered in BM-MSCs and during osteogenic differentiation from patients with OA. The alterations found point to GPC5 and Notum as new candidate biomarkers of OA pathology.


Subject(s)
Glypicans , Mesenchymal Stem Cells , Osteoarthritis , Osteoblasts , Humans , Mesenchymal Stem Cells/metabolism , Osteoarthritis/blood , Osteoarthritis/pathology , Osteoarthritis/genetics , Osteoarthritis/metabolism , Osteoblasts/metabolism , Osteoblasts/pathology , Male , Female , Glypicans/metabolism , Glypicans/blood , Glypicans/genetics , Middle Aged , Cell Differentiation , Osteogenesis/genetics , Aged , Case-Control Studies , Cells, Cultured , Bone Marrow Cells/metabolism
12.
Bone ; 184: 117113, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38703937

ABSTRACT

Ca2+/calmodulin-dependent protein kinase kinase 2 (CaMKK2) is a multi-functional, serine/threonine protein kinase with predominant roles in inflammation, systemic energy metabolism, and bone remodeling. We previously reported that global ablation of CaMKK2 or its systemic pharmacological inhibition led to bone mass accrual in mice by stimulating osteoblasts and inhibiting osteoclasts. However, a direct, cell-intrinsic role for the kinase in the osteoblast lineage has not been established. Here we report that conditional deletion of CaMKK2 from osteoprogenitors, using the Osterix 1 (Osx1) - GFP::Cre (tetracycline-off) mouse line, resulted in increased trabecular bone mass due to an acute stimulation of osteoblast function in male and female mice. The acute simulation of osteoblasts and bone formation following conditional ablation of osteoprogenitor-derived CaMKK2 was sustained only in female mice. Periosteal bone formation at the cortical bone was enhanced only in male conditional knockout mice without altering cortical bone mass or strength. Prolonged deletion of CaMKK2 in early osteoblasts was accompanied by a stimulation of osteoclasts in both sexes, indicating a coupling effect. Notably, alterations in trabecular and cortical bone mass were absent in the doxycycline-removed "Cre-only" Osx1-GFP::Cre mice. Thus, the increase in osteoblast function at the trabecular and cortical bone surfaces following the conditional deletion of CaMKK2 in osteoprogenitors is indicative of a direct but sex-divergent role for the kinase in osteoblasts.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Kinase , Osteoblasts , Sp7 Transcription Factor , Animals , Osteoblasts/metabolism , Female , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Male , Sp7 Transcription Factor/metabolism , Sp7 Transcription Factor/genetics , Osteogenesis/physiology , Sex Characteristics , Mice , Mice, Knockout , Osteoclasts/metabolism , Stem Cells/metabolism , Gene Deletion
13.
Int J Biol Macromol ; 268(Pt 2): 132013, 2024 May.
Article in English | MEDLINE | ID: mdl-38697412

ABSTRACT

Osteoporosis is a prevalent condition characterized by bone loss and decreased skeletal strength, resulting in an elevated risk of fractures. Calcium plays a crucial role in preventing and managing osteoporosis. However, traditional calcium supplements have limited bioavailability, poor solubility, and adverse effects. In this study, we isolated a natural soluble biopolymer, calcium polymalate (PMACa), from the fermentation broth of the fungus Aureobasidium pullulans, to investigate its potential as an anti-osteoporosis therapeutic agent. Characterization revealed that linear PMA-Ca chains juxtaposed to form a porous, rod-like state, in the presence of Ca2+. In vivo mouse models demonstrated that PMA-Ca significantly promoted the conversion of serum calcium into bone calcium, and stimulated bone growth and osteogenesis. Additionally, PMA-Ca alleviated exercise fatigue in mice by facilitating the removal of essential metabolites, such as serum lactate (BLA) and blood urea nitrogen (BUN), from their bloodstream. In vitro studies further showed that PMA-Ca strengthened osteoblast cell activity, proliferation, and mineralization. And PMA-Ca upregulated the expression of some genes involved in osteoblast differentiation, indicating a potential correlation between bone formation and PMACa. These findings indicate that soluble PMA-Ca has the potential to be a novel biopolymer-based calcium supplement with sustainable production sourced from the fermentation industry.


Subject(s)
Aureobasidium , Calcium , Fermentation , Osteoporosis , Solubility , Animals , Mice , Osteoporosis/drug therapy , Osteoporosis/metabolism , Calcium/metabolism , Biopolymers/chemistry , Biopolymers/pharmacology , Aureobasidium/metabolism , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteogenesis/drug effects , Fatigue/drug therapy , Water/chemistry , Cell Proliferation/drug effects , Disease Models, Animal , Female , Cell Differentiation/drug effects
14.
Biomed Mater ; 19(4)2024 May 22.
Article in English | MEDLINE | ID: mdl-38740037

ABSTRACT

The purpose of this study was to construct a rutin-controlled release system on the surface of Ti substrates and investigate its effects on osteogenesis and osseointegration on the surface of implants. The base layer, polyethylenimine (PEI), was immobilised on a titanium substrate. Then, hyaluronic acid (HA)/chitosan (CS)-rutin (RT) multilayer films were assembled on the PEI using layer-by-layer (LBL) assembly technology. We used scanning electron microscopy (SEM), Fourier transform infrared (FTIR) spectroscopy and contact angle measurements to examine all Ti samples. The drug release test of rutin was also carried out to detect the slow-release performance. The osteogenic abilities of the samples were evaluated by experiments on an osteoporosis rat model and MC3T3-E1 cells. The results (SEM, FTIR and contact angle measurements) all confirmed that the PEI substrate layer and HA/CS-RT multilayer film were effectively immobilised on titanium. The drug release test revealed that a rutin controlled release mechanism had been successfully established. Furthermore, thein vitrodata revealed that osteoblasts on the coated titanium matrix had greater adhesion, proliferation, and differentiation capacity than the osteoblasts on the pure titanium surface. When MC3T3-E1 cells were exposed to H2O2-induced oxidative stressin vitro, cell-based tests revealed great tolerance and increased osteogenic potential on HA/CS-RT substrates. We also found that the HA/CS-RT coating significantly increased the new bone mass around the implant. The LBL-deposited HA/CS-RT multilayer coating on the titanium base surface established an excellent rutin-controlled release system, which significantly improved osseointegration and promoted osteogenesis under oxidative stress conditions, suggesting a new implant therapy strategy for patients with osteoporosis.


Subject(s)
Coated Materials, Biocompatible , Hyaluronic Acid , Osseointegration , Osteoblasts , Osteogenesis , Osteoporosis , Prostheses and Implants , Rutin , Surface Properties , Titanium , Animals , Titanium/chemistry , Rutin/chemistry , Rutin/pharmacology , Osteogenesis/drug effects , Rats , Osteoporosis/drug therapy , Mice , Osteoblasts/drug effects , Osteoblasts/cytology , Osteoblasts/metabolism , Osseointegration/drug effects , Hyaluronic Acid/chemistry , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Oxidation-Reduction , Chitosan/chemistry , Female , Rats, Sprague-Dawley , Cell Adhesion/drug effects , Spectroscopy, Fourier Transform Infrared , Cell Differentiation/drug effects , Microscopy, Electron, Scanning , Cell Proliferation/drug effects , Polyethyleneimine/chemistry , 3T3 Cells , Oxidative Stress/drug effects , Layer-by-Layer Nanoparticles
15.
J Orthop Surg Res ; 19(1): 294, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38745231

ABSTRACT

BACKGROUND: Osteonecrosis of the femoral head caused by glucocorticoids (GIONFH) is a significant issue resulting from prolonged or excessive clinical glucocorticoid use. Astaxanthin, an orange-red carotenoid present in marine organisms, has been the focus of this study to explore its impact and mechanism on osteoblast apoptosis induced by dexamethasone (Dex) and GIONFH. METHODS: In this experiment, bioinformatic prediction, molecular docking and dynamics simulation, cytotoxicity assay, osteogenic differentiation, qRT-PCR analysis, terminal uridine nickend labeling (TUNEL) assay, determination of intracellular ROS, mitochondrial function assay, immunofluorescence, GIONFH rat model construction, micro-computed tomography (micro-CT) scans were performed. RESULTS: Our research demonstrated that a low dose of astaxanthin was non-toxic to healthy osteoblasts and restored the osteogenic function of Dex-treated osteoblasts by reducing oxidative stress, mitochondrial dysfunction, and apoptosis. Furthermore, astaxanthin rescued the dysfunction in poor bone quality, bone metabolism and angiogenesis of GIONFH rats. The mechanism behind this involves astaxanthin counteracting Dex-induced osteogenic damage by activating the Nrf2 pathway. CONCLUSION: Astaxanthin shields osteoblasts from glucocorticoid-induced oxidative stress and mitochondrial dysfunction via Nrf2 pathway activation, making it a potential therapeutic agent for GIONFH treatment.


Subject(s)
Femur Head Necrosis , Glucocorticoids , Mitochondria , NF-E2-Related Factor 2 , Osteoblasts , Osteogenesis , Oxidative Stress , Xanthophylls , Animals , Xanthophylls/pharmacology , Oxidative Stress/drug effects , NF-E2-Related Factor 2/metabolism , Glucocorticoids/adverse effects , Glucocorticoids/toxicity , Femur Head Necrosis/chemically induced , Femur Head Necrosis/metabolism , Osteogenesis/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Rats , Osteoblasts/drug effects , Osteoblasts/metabolism , Male , Dexamethasone/pharmacology , Dexamethasone/adverse effects , Rats, Sprague-Dawley , Apoptosis/drug effects , Disease Models, Animal
16.
Biol Open ; 13(5)2024 May 15.
Article in English | MEDLINE | ID: mdl-38809145

ABSTRACT

Bone is increasingly recognized as a target for diabetic complications. In order to evaluate the direct effects of high glucose on bone, we investigated the global transcriptional changes induced by hyperglycemia in osteoblasts in vitro. Rat bone marrow-derived mesenchymal stromal cells were differentiated into osteoblasts for 10 days, and prior to analysis, they were exposed to hyperglycemia (25 mM) for the short-term (1 or 3 days) or long-term (10 days). Genes and pathways regulated by hyperglycemia were identified using mRNA sequencing and verified with qPCR. Genes upregulated by 1-day hyperglycemia were, for example, related to extracellular matrix organization, collagen synthesis and bone formation. This stimulatory effect was attenuated by 3 days. Long-term exposure impaired osteoblast viability, and downregulated, for example, extracellular matrix organization and lysosomal pathways, and increased intracellular oxidative stress. Interestingly, transcriptional changes by different exposure times were mostly unique and only 89 common genes responding to glucose were identified. In conclusion, short-term hyperglycemia had a stimulatory effect on osteoblasts and bone formation, whereas long-term hyperglycemia had a negative effect on intracellular redox balance, osteoblast viability and function.


Subject(s)
Gene Expression Regulation , Glucose , Osteoblasts , Osteoblasts/metabolism , Osteoblasts/drug effects , Animals , Glucose/metabolism , Rats , Gene Expression Regulation/drug effects , Gene Expression Profiling , Hyperglycemia/metabolism , Hyperglycemia/genetics , Cell Differentiation/drug effects , Cell Differentiation/genetics , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Transcriptome , Osteogenesis/drug effects , Osteogenesis/genetics , Cell Survival/drug effects , Transcription, Genetic/drug effects , Cells, Cultured , Oxidative Stress/drug effects
17.
Bone Res ; 12(1): 33, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38811544

ABSTRACT

Wnt/ß-catenin signaling is critical for various cellular processes in multiple cell types, including osteoblast (OB) differentiation and function. Exactly how Wnt/ß-catenin signaling is regulated in OBs remain elusive. ATP6AP2, an accessory subunit of V-ATPase, plays important roles in multiple cell types/organs and multiple signaling pathways. However, little is known whether and how ATP6AP2 in OBs regulates Wnt/ß-catenin signaling and bone formation. Here we provide evidence for ATP6AP2 in the OB-lineage cells to promote OB-mediated bone formation and bone homeostasis selectively in the trabecular bone regions. Conditionally knocking out (CKO) ATP6AP2 in the OB-lineage cells (Atp6ap2Ocn-Cre) reduced trabecular, but not cortical, bone formation and bone mass. Proteomic and cellular biochemical studies revealed that LRP6 and N-cadherin were reduced in ATP6AP2-KO BMSCs and OBs, but not osteocytes. Additional in vitro and in vivo studies revealed impaired ß-catenin signaling in ATP6AP2-KO BMSCs and OBs, but not osteocytes, under both basal and Wnt stimulated conditions, although LRP5 was decreased in ATP6AP2-KO osteocytes, but not BMSCs. Further cell biological studies uncovered that osteoblastic ATP6AP2 is not required for Wnt3a suppression of ß-catenin phosphorylation, but necessary for LRP6/ß-catenin and N-cadherin/ß-catenin protein complex distribution at the cell membrane, thus preventing their degradation. Expression of active ß-catenin diminished the OB differentiation deficit in ATP6AP2-KO BMSCs. Taken together, these results support the view for ATP6AP2 as a critical regulator of both LRP6 and N-cadherin protein trafficking and stability, and thus regulating ß-catenin levels, demonstrating an un-recognized function of osteoblastic ATP6AP2 in promoting Wnt/LRP6/ß-catenin signaling and trabecular bone formation.


Subject(s)
Low Density Lipoprotein Receptor-Related Protein-6 , Mice, Knockout , Osteoblasts , Osteogenesis , Vacuolar Proton-Translocating ATPases , Wnt Signaling Pathway , beta Catenin , Animals , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Wnt Signaling Pathway/physiology , beta Catenin/metabolism , beta Catenin/genetics , Osteoblasts/metabolism , Osteogenesis/physiology , Mice , Vacuolar Proton-Translocating ATPases/metabolism , Vacuolar Proton-Translocating ATPases/genetics , Protein Transport , Cell Differentiation , Osteocytes/metabolism , Prorenin Receptor
18.
JCI Insight ; 9(10)2024 May 22.
Article in English | MEDLINE | ID: mdl-38713511

ABSTRACT

While sclerostin-neutralizing antibodies (Scl-Abs) transiently stimulate bone formation by activating Wnt signaling in osteoblast lineage cells, they exert sustained inhibition of bone resorption, suggesting an alternate signaling pathway by which Scl-Abs control osteoclast activity. Since sclerostin can activate platelet-derived growth factor receptors (PDGFRs) in osteoblast lineage cells in vitro and PDGFR signaling in these cells induces bone resorption through M-CSF secretion, we hypothesized that the prolonged anticatabolic effect of Scl-Abs could result from PDGFR inhibition. We show here that inhibition of PDGFR signaling in osteoblast lineage cells is sufficient and necessary to mediate prolonged Scl-Ab effects on M-CSF secretion and osteoclast activity in mice. Indeed, sclerostin coactivates PDGFRs independently of Wnt/ß-catenin signaling inhibition, by forming a ternary complex with LRP6 and PDGFRs in preosteoblasts. In turn, Scl-Ab prevents sclerostin-mediated coactivation of PDGFR signaling and consequent M-CSF upregulation in preosteoblast cultures, thereby inhibiting osteoclast activity in preosteoblast/osteoclast coculture assays. These results provide a potential mechanism explaining the dissociation between anabolic and antiresorptive effects of long-term Scl-Ab.


Subject(s)
Adaptor Proteins, Signal Transducing , Bone Resorption , Osteoblasts , Osteoclasts , Receptors, Platelet-Derived Growth Factor , Signal Transduction , Animals , Osteoblasts/metabolism , Mice , Adaptor Proteins, Signal Transducing/metabolism , Bone Resorption/metabolism , Osteoclasts/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Wnt Signaling Pathway/drug effects , Antibodies, Neutralizing/pharmacology , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Cell Lineage , Osteogenesis/drug effects , Cell Differentiation
19.
Cell Biochem Funct ; 42(4): e4068, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38817105

ABSTRACT

Evidence is accumulating that osteal macrophages, in addition to bone-resorbing osteoclasts and bone-forming osteoblasts, participate vitally in bone remodeling process. Oncostatin M (OSM), an inflammatory cytokine belonging to interleukin-6 superfamily, is recognized as an essential factor secreted by osteal macrophages to orchestrate bone remodeling. Osteoprotegerin (OPG) produced by osteoblasts regulates osteoclastogenesis. We have reported that bone morphogenetic protein-4 (BMP-4) stimulates OPG synthesis in MC3T3-E1 osteoblast-like cells, and that SMAD1/5/8(9), p38 mitogen-activated protein kinase (MAPK), and p70 S6 kinase are involved in the OPG synthesis. The present study aims to investigate the effect of OSM on the synthesis of OPG stimulated by BMP-4 in osteoblasts. OSM suppressed the release and the mRNA expression of OPG upregulated by BMP-4 in MC3T3-E1 cells. Neither the BMP-4-induced phosphorylation of SMAD1/5/9 nor that of p38 MAPK was affected by OSM. On the other hand, the phosphorylation of p70 S6 kinase stimulated by BMP-4 was considerably suppressed by OSM. These results strongly suggest that OSM suppresses the BMP-4-stimulated OPG synthesis via inhibition of the p70 S6 kinase-mediated pathway in osteoblast-like cells.


Subject(s)
Bone Morphogenetic Protein 4 , Oncostatin M , Osteoblasts , Osteoprotegerin , Ribosomal Protein S6 Kinases, 70-kDa , Animals , Mice , Oncostatin M/pharmacology , Oncostatin M/metabolism , Osteoblasts/metabolism , Osteoblasts/drug effects , Osteoblasts/cytology , Osteoprotegerin/metabolism , Osteoprotegerin/biosynthesis , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 4/pharmacology , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Phosphorylation/drug effects , Cell Line
20.
Mol Med Rep ; 30(2)2024 Aug.
Article in English | MEDLINE | ID: mdl-38818814

ABSTRACT

C1q/tumor necrosis factor­related protein 3 (CTRP3) expression is markedly reduced in the serum of patients with osteoporosis. The present study aimed to investigate whether CTRP3 reduces bone loss in oophorectomy (OVX)­induced mice via the AMP­activated protein kinase (AMPK)/sirtuin 1 (SIRT1)/nuclear factor E2­related factor 2 (Nrf2) signaling pathway. Female C57BL/6J mice and MC3T3­E1 cells were used to construct in vivo and in vitro models of osteoporosis, respectively. The left femurs of mice were examined using micro­computed tomography scans and bone­related quantitative morphological evaluation was performed. Pathological changes and the number of osteoclasts in the left femurs of mice were detected using hematoxylin and eosin, and tartrate­resistant acid phosphatase (TRAP) staining. Runt­related transcription factor­2 (RUNX2) expression in the left femurs was detected using immunofluorescence analysis, and the serum levels of bone resorption markers (C­telopeptide of type I collagen and TRAP) and bone formation markers [osteocalcin (OCN) and procollagen type 1 N­terminal propeptide] were detected. In addition, osteoblast differentiation and calcium deposits were examined in MC3T3­E1 cells using alkaline phosphatase (ALP) and Alizarin red staining, respectively. Moreover, RUNX2, ALP and OCN expression levels were detected using reverse transcription­quantitative PCR, and the expression levels of proteins associated with the AMPK/SIRT1/Nrf2 signaling pathway were detected using western blot analysis. The results revealed that globular CTRP3 (gCTRP3) alleviated bone loss and promoted bone formation in OVX­induced mice. gCTRP3 also facilitated the osteogenic differentiation of MC3T3­E1 cells through the AMPK/SIRT1/Nrf2 signaling pathway. The addition of an AMPK inhibitor (Compound C), SIRT1 inhibitor (EX527) or Nrf2 inhibitor (ML385) reduced the osteogenic differentiation of MC3T3­E1 cells via inhibition of gCTRP3. In conclusion, gCTRP3 inhibits OVX­induced osteoporosis by activating the AMPK/SIRT1/Nrf2 signaling pathway.


Subject(s)
AMP-Activated Protein Kinases , NF-E2-Related Factor 2 , Osteoporosis , Ovariectomy , Signal Transduction , Sirtuin 1 , Animals , Sirtuin 1/metabolism , Sirtuin 1/genetics , Female , Mice , Osteoporosis/metabolism , Osteoporosis/etiology , Osteoporosis/pathology , NF-E2-Related Factor 2/metabolism , Ovariectomy/adverse effects , AMP-Activated Protein Kinases/metabolism , Mice, Inbred C57BL , Osteoblasts/metabolism , Cell Line , Osteoclasts/metabolism , Disease Models, Animal , Femur/metabolism , Femur/pathology , Femur/diagnostic imaging , Osteogenesis/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...