Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 11.244
Filter
1.
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
2.
PLoS One ; 19(6): e0301618, 2024.
Article in English | MEDLINE | ID: mdl-38843277

ABSTRACT

Periprosthetic tissue inflammation is a challenging complication arising in joint replacement surgeries, which is often caused by wear debris from polyethylene (PE) components. In this study, we examined the potential biological effects of grafting a [2-(methacryloyloxy)ethyl]dimethyl-(3-sulfopropyl)ammonium hydroxide (MEDSAH) polymer onto the surface of PE through a solvent-evaporation technique. J774A.1 macrophage-like cells and primary cultured mouse osteoblasts were treated with PE powder with or without the MEDSAH coating. MEDSAH grafting on PE substantially reduced the expression of pro-inflammatory cytokines and other mediators in primary cultured mouse osteoblasts, but did not significantly impact macrophage-mediated inflammation. Our findings suggest that a MEDSAH coating on PE-based materials has potential utility in mitigating periprosthetic tissue inflammation and osteolysis and preventing aseptic loosening in total joint replacements. Further research, including large-scale clinical trials and biomechanical analyses, is needed to assess the long-term performance and clinical implications of MEDSAH-coated PE-based materials in total joint arthroplasty.


Subject(s)
Inflammation , Osteoblasts , Polyethylene , Animals , Mice , Inflammation/pathology , Osteoblasts/metabolism , Osteoblasts/drug effects , Macrophages/metabolism , Cell Line , Cytokines/metabolism , Osteolysis/etiology , Osteolysis/pathology , Coated Materials, Biocompatible/chemistry , Methacrylates/chemistry , Arthroplasty, Replacement/adverse effects
3.
PeerJ ; 12: e17488, 2024.
Article in English | MEDLINE | ID: mdl-38827303

ABSTRACT

Epigallocatechin gallate (EGCG), an active constituent of tea, is recognized for its anticancer and anti-inflammatory properties. However, the specific mechanism by which EGCG protects osteoblasts from cadmium-induced damage remains incompletely understood. Here, the action of EGCG was investigated by exposing MC3T3-E1 osteoblasts to EGCG and CdCl2 and examining their growth, apoptosis, and differentiation. It was found that EGCG promoted the viability of cadmium-exposed MC3T3-E1 cells, mitigated apoptosis, and promoted both maturation and mineralization. Additionally, CdCl2 has been reported to inhibit both the phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) and nuclear factor erythroid 2-related factor 2/heme oxygenase-1(Nrf2/HO-1) signaling pathways. EGCG treatment attenuated cadmium-induced apoptosis in osteoblasts and restored their function by upregulating both signaling pathways. The findings provide compelling evidence for EGCG's role in attenuating cadmium-induced osteoblast apoptosis and dysfunction through activating the PI3K/AKT/mTOR and Nrf2/HO-1 pathways. This suggests the potential of using EGCG for treating cadmium-induced osteoblast dysfunction.


Subject(s)
Apoptosis , Catechin , Heme Oxygenase-1 , NF-E2-Related Factor 2 , Osteoblasts , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , TOR Serine-Threonine Kinases , Catechin/analogs & derivatives , Catechin/pharmacology , Apoptosis/drug effects , NF-E2-Related Factor 2/metabolism , Animals , Mice , TOR Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Heme Oxygenase-1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Osteoblasts/drug effects , Osteoblasts/metabolism , Cadmium/toxicity , Cell Differentiation/drug effects , Cell Line , Membrane Proteins
4.
J Nanobiotechnology ; 22(1): 320, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38849820

ABSTRACT

Simultaneously modulating the inflammatory microenvironment and promoting local bone regeneration is one of the main challenges in treating bone defects. In recent years, osteoimmunology has revealed that the immune system plays an essential regulatory role in bone regeneration and that macrophages are critical components. In this work, a mussel-inspired immunomodulatory and osteoinductive dual-functional hydroxyapatite nano platform (Gold/hydroxyapatite nanocomposites functionalized with polydopamine - PDA@Au-HA) is developed to accelerate bone tissues regeneration by regulating the immune microenvironment. PDA coating endows nanomaterials with the ability to scavenge reactive oxygen species (ROS) and anti-inflammatory properties, and it also exhibits an immunomodulatory ability to inhibit M1 macrophage polarization and activate M2 macrophage secretion of osteogenesis-related cytokines. Most importantly, this nano platform promotes the polarization of M2 macrophages and regulates the crosstalk between macrophages and pre-osteoblast cells to achieve bone regeneration. Au-HA can synergistically promote vascularized bone regeneration through sustained release of Ca and P particles and gold nanoparticles (NPs). This nano platform has a synergistic effect of good compatibility, scavenging of ROS, and anti-inflammatory and immunomodulatory capability to accelerate the bone repair process. Thus, our research offers a possible therapeutic approach by exploring PDA@Au-HA nanocomposites as a bifunctional platform for tissue regeneration.


Subject(s)
Bivalvia , Bone Regeneration , Durapatite , Gold , Indoles , Macrophages , Osteogenesis , Bone Regeneration/drug effects , Durapatite/chemistry , Durapatite/pharmacology , Animals , Mice , Gold/chemistry , Gold/pharmacology , Bivalvia/chemistry , RAW 264.7 Cells , Macrophages/drug effects , Indoles/chemistry , Indoles/pharmacology , Osteogenesis/drug effects , Reactive Oxygen Species/metabolism , Polymers/chemistry , Polymers/pharmacology , Nanocomposites/chemistry , Metal Nanoparticles/chemistry , Osteoblasts/drug effects , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Immunologic Factors/pharmacology , Immunologic Factors/chemistry , Cytokines/metabolism
5.
Carbohydr Polym ; 339: 122232, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38823905

ABSTRACT

In this study, new types of hybrid double-network (DN) hydrogels composed of polyvinyl alcohol (PVA), chitosan (CH), and sodium alginate (SA) are introduced, with the hypothesis that this combination and incorporating multi-walled carbon nanotubes (MWCNTs) and graphene nanoplatelets (GNPs) will enhance osteogenetic differentiation and the structural and mechanical properties of scaffolds for bone tissue engineering applications. Initially, the impact of varying mass ratios of the PVA/CH/SA mixture on mechanical properties, swelling ratio, and degradability was examined. Based on this investigation, a mass ratio of 4:6:6 was determined to be optimal. At this ratio, the hydrogel demonstrated a Young's modulus of 47.5 ± 5 kPa, a swelling ratio of 680 ± 6 % after 3 h, and a degradation rate of 46.5 ± 5 % after 40 days. In the next phase, following the determination of the optimal mass ratio, CNTs and GNPs were incorporated into the 4:6:6 composite resulting in a significant enhancement in the electrical conductivity and stiffness of the scaffolds. The introduction of CNTs led to a notable increase of 36 % in the viability of MG63 osteoblast cells. Additionally, the inhibition zone test revealed that GNPs and CNTs increased the diameter of the inhibition zone by 49.6 % and 52.6 %, respectively.


Subject(s)
Alginates , Bone Regeneration , Chitosan , Hydrogels , Polyvinyl Alcohol , Tissue Engineering , Tissue Scaffolds , Chitosan/chemistry , Alginates/chemistry , Alginates/pharmacology , Polyvinyl Alcohol/chemistry , Tissue Scaffolds/chemistry , Humans , Bone Regeneration/drug effects , Hydrogels/chemistry , Hydrogels/pharmacology , Tissue Engineering/methods , Nanotubes, Carbon/chemistry , Osteoblasts/drug effects , Osteoblasts/cytology , Graphite/chemistry , Graphite/pharmacology , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Cell Survival/drug effects , Cell Line
6.
Cell Mol Biol (Noisy-le-grand) ; 70(6): 135-141, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38836669

ABSTRACT

Epigenetic change has been found to play an important role in cell differentiation and regulation and the dental pulp stem cell in tissue engineering is gaining attention due to the ability of cells to differentiate into odontoblast and other cells. This study evaluated the influence of poly L- lactic acid with hydroxyapatite-coated with polyaniline scaffold (PLLA/HA/PANI) on dental pulp stem cell (DPSC) proliferation and differentiation. After scaffold preparation and DPSCs seeding, the cells proliferation and differentiation were evaluated by immunocytochemistry assay and cell viability was measured by cytotoxicity / MTT assay. The results showed (PLLA/HA/PANI) scaffold facilitates DPSC proliferation and differentiation with gene expression. This finding underscores the promise of this biomaterial combination as a scaffold for dental tissue regeneration and application.


Subject(s)
Biocompatible Materials , Cell Differentiation , Cell Proliferation , Dental Pulp , Durapatite , Odontoblasts , Osteoblasts , Stem Cells , Tissue Scaffolds , Dental Pulp/cytology , Humans , Cell Differentiation/drug effects , Odontoblasts/cytology , Odontoblasts/drug effects , Odontoblasts/metabolism , Tissue Scaffolds/chemistry , Stem Cells/cytology , Stem Cells/metabolism , Stem Cells/drug effects , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Cell Proliferation/drug effects , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Durapatite/chemistry , Durapatite/pharmacology , Aniline Compounds/pharmacology , Aniline Compounds/chemistry , Polyesters/chemistry , Polyesters/pharmacology , Cell Survival/drug effects , Cells, Cultured , Tissue Engineering/methods
7.
Nat Commun ; 15(1): 5027, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38871693

ABSTRACT

Generating 3D bone cell networks in vitro that mimic the dynamic process during early bone formation remains challenging. Here, we report a synthetic biodegradable microporous hydrogel for efficient formation of 3D networks from human primary cells, analysis of cell-secreted extracellular matrix (ECM) and microfluidic integration. Using polymerization-induced phase separation, we demonstrate dynamic in situ formation of microporosity (5-20 µm) within matrix metalloproteinase-degradable polyethylene glycol hydrogels in the presence of living cells. Pore formation is triggered by thiol-Michael-addition crosslinking of a viscous precursor solution supplemented with hyaluronic acid and dextran. The resulting microporous architecture can be fine-tuned by adjusting the concentration and molecular weight of dextran. After encapsulation in microporous hydrogels, human mesenchymal stromal cells and osteoblasts spread rapidly and form 3D networks within 24 hours. We demonstrate that matrix degradability controls cell-matrix remodeling, osteogenic differentiation, and deposition of ECM proteins such as collagen. Finally, we report microfluidic integration and proof-of-concept osteogenic differentiation of 3D cell networks under perfusion on chip. Altogether, this work introduces a synthetic microporous hydrogel to efficiently differentiate 3D human bone cell networks, facilitating future in vitro studies on early bone development.


Subject(s)
Cell Culture Techniques, Three Dimensional , Cell Differentiation , Extracellular Matrix , Hydrogels , Mesenchymal Stem Cells , Osteoblasts , Osteogenesis , Humans , Hydrogels/chemistry , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Osteogenesis/drug effects , Cell Differentiation/drug effects , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Extracellular Matrix/metabolism , Porosity , Cell Culture Techniques, Three Dimensional/methods , Polyethylene Glycols/chemistry , Tissue Engineering/methods , Hyaluronic Acid/chemistry , Cells, Cultured , Tissue Scaffolds/chemistry , Dextrans/chemistry
8.
ACS Appl Bio Mater ; 7(6): 3900-3914, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38840339

ABSTRACT

The poor clinical performance of titanium and its alloy implants is mainly attributed to their lack of antibacterial ability and poor osseointegration. The key and challenge lie in how to enhance their osteoinductivity while imparting antibacterial capability. In this study, a titanium oxide metasurface with light-responsive behavior was constructed on the surface of titanium alloy using an alkaline-acid bidirectional hydrothermal method. The effects of the acid type, acid concentration, hydrothermal time, hydrothermal temperature, and subsequent heat treatments on the optical behavior of the metasurface were systematically investigated with a focus on exploring the influence of the metasurface and photodynamic reaction on the osteogenic activity of osteoblasts. Results show that the type of acid and heat treatment significantly affect the light absorption of the titanium alloy surface, with HCl and post-heat-treatment favoring redshift in the light absorption. Under 808 nm near-infrared (NIR) irradiation for 10 min, in vitro antibacterial experiments demonstrate that the antibacterial rate of the metasurface titanium alloy against Staphylococcus aureus and Escherichia coli were 96.87% and 99.27%, respectively. In vitro cell experiments demonstrate that the nanostructure facilitates cell adhesion, proliferation, differentiation, and expression of osteogenic-related genes. Surprisingly, the nanostructure promoted the expression of relevant osteogenic genes of MC3T3-E1 under 808 nm NIR irradiation. This study provides a method for the surface modification of titanium alloy implants.


Subject(s)
Alloys , Anti-Bacterial Agents , Biocompatible Materials , Escherichia coli , Infrared Rays , Materials Testing , Nanostructures , Staphylococcus aureus , Surface Properties , Titanium , Titanium/chemistry , Titanium/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Staphylococcus aureus/drug effects , Alloys/chemistry , Alloys/pharmacology , Escherichia coli/drug effects , Nanostructures/chemistry , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Microbial Sensitivity Tests , Particle Size , Animals , Mice , Osteogenesis/drug effects , Osteoblasts/drug effects , Osteoblasts/cytology , Cell Proliferation/drug effects , Osseointegration/drug effects
9.
J Histochem Cytochem ; 72(5): 309-327, 2024 05.
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
10.
Biomater Adv ; 161: 213894, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38796956

ABSTRACT

Engineering of scaffolds for bone regeneration is often inspired by the native extracellular matrix mimicking its composite fibrous structure. In the present study, we used low loadings of diatomite earth (DE) biosilica to improve the bone regeneration potential of gelatin electrospun fibrillar microenvironments. We explored the effect of increasing the DE content from 1 % to 3 % and 5 %, respectively, on the physico-chemical properties of the fibrous scaffolds denoted FG_DE1, FG_DE3, FG_DE5, regarding the aqueous media affinity, stability under simulated physiological conditions, morphology characteristics, and local mechanical properties at the surface. The presence of biosilica generated composite structures with lower swelling degrees and higher stiffness when compared to gelatin fibers. Increasing DE content led to higher Young modulus, while the stability of the protein matrix in PBS, at 37 °C, over 21 was significantly decreased by the presence of diatomite loadings. The best preosteoblast response was obtained for FG_DE3, with enhanced mineralization during the osteogenic differentiation when compared to the control sample without diatomite. 5 % DE in FG_DE5 proved to negatively influence cells' metabolic activity and morphology. Hence, the obtained composite microfibrillar scaffolds might find application as osteoblast-responsive materials for bone tissue engineering.


Subject(s)
Gelatin , Osteoblasts , Tissue Engineering , Tissue Scaffolds , Gelatin/chemistry , Osteoblasts/drug effects , Osteoblasts/metabolism , Tissue Scaffolds/chemistry , Tissue Engineering/methods , Animals , Diatomaceous Earth/chemistry , Osteogenesis/drug effects , Cell Differentiation/drug effects , Mice , Bone Regeneration/drug effects , Cell Line , Cellular Microenvironment/drug effects , Microfibrils/chemistry , Microfibrils/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix/chemistry , Extracellular Matrix/drug effects
11.
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
12.
Biomed Mater ; 19(4)2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38815600

ABSTRACT

Excessive reactive oxygen species (ROS) in the microenvironment of osteoporosis (OP) not only accelerate the bone absorption, but also affect the osteogenic and mineralized effect of osteoblasts. Procyanidins (PC) have been reported to have anti-oxidation effects, but low bioavailability. This study aimed to explore the effect of magnesium oxide nanoparticles (MgO-PC NPs)-loaded PC on the osteogenesis and mineralization of osteoblasts that stimulated by H2O2. PC was loaded onto MgO NPs and characterized by transmission electron microscopy, energy dispersive spectroscopy, dynamic light scattering, and Fourier transform infrared spectroscopy. After primary screening by cytotoxicity assay, MgO-PC NPs containing 20 µM of PC were chosen for further studies. In H2O2-stimulated osteoblasts, dichlorodihydrofluorescein diacetate probe, Cell Counting Kit-8, quantitative real-time polymerase chain reaction, alkaline phosphatase staining/activity and Alizarin red staining were used to detect the ROS production, cell viability and osteogenic and mineralized markers of osteoblasts. PC was loaded onto MgO NPs to successfully receive MgO-PC NPs with a diameter of about 144 nm and negative potential. PC can sustain release from MgO-PC NPs for at least 16 d. The controlled release of PC from MgO-PC NPs can effectively eliminate ROS and thereby promoted the cell activity. Most importantly, the osteogenesis and mineralization of osteoblasts under oxidative stress were also significantly reversed by MgO-PC NPS. Thus, these findings indicate that MgO-PC NPs may be developed as a potential therapeutic strategy for OP.


Subject(s)
Biflavonoids , Catechin , Cell Survival , Hydrogen Peroxide , Magnesium Oxide , Nanoparticles , Osteoblasts , Osteogenesis , Oxidative Stress , Proanthocyanidins , Reactive Oxygen Species , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoblasts/cytology , Proanthocyanidins/pharmacology , Proanthocyanidins/chemistry , Catechin/chemistry , Catechin/pharmacology , Oxidative Stress/drug effects , Magnesium Oxide/chemistry , Reactive Oxygen Species/metabolism , Animals , Cell Survival/drug effects , Biflavonoids/pharmacology , Biflavonoids/chemistry , Osteogenesis/drug effects , Hydrogen Peroxide/chemistry , Nanoparticles/chemistry , Delayed-Action Preparations/chemistry , Mice , Calcification, Physiologic/drug effects , Spectroscopy, Fourier Transform Infrared
13.
Colloids Surf B Biointerfaces ; 239: 113971, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38759296

ABSTRACT

The optimal material for repairing skull defects should exhibit outstanding biocompatibility and mechanical properties. Specifically, hydrogel scaffolds that emulate the microenvironment of the native bone extracellular matrix play a vital role in promoting osteoblast adhesion, proliferation, and differentiation, thereby yielding superior outcomes in skull reconstruction. In this study, a composite network hydrogel comprising sodium alginate (SA), epigallocatechin gallate (EGCG), and zinc ions (Zn2+) was developed to establish an ideal osteogenic microenvironment for bone regeneration. Initially, physical entanglement and hydrogen bonding between SA and EGCG resulted in the formation of a primary network hydrogel known as SA-EGCG. Subsequently, the inclusion of Zn2+ facilitated the creation of a composite network hydrogels named SA-EGCG-Zn2+ via dynamic coordination bonds with SA and EGCG. The engineered SA-EGCG2 %-Zn2+ hydrogels offered an environment mimicking the native extracellular matrix (ECM). Moreover, the sustained release of Zn2+ from the hydrogel effectively enhanced cell adhesion, promoted proliferation, and stimulated osteoblast differentiation. In vitro experiments have shown that SA-EGCG2 %-Zn2+ hydrogels greatly enhance the attachment and growth of osteoblast precursor cells (MC3T3-E1), while also increasing the expression of genes related to osteogenesis in these cells. Additionally, in vivo studies have confirmed that SA-EGCG2 %-Zn2+ hydrogels promote new bone formation and accelerate the regeneration of bone in situ, indicating promising applications in the realm of bone tissue engineering.


Subject(s)
Alginates , Catechin , Cell Proliferation , Hydrogels , Skull , Tissue Scaffolds , Zinc , Zinc/chemistry , Zinc/pharmacology , Alginates/chemistry , Alginates/pharmacology , Catechin/chemistry , Catechin/analogs & derivatives , Catechin/pharmacology , Skull/drug effects , Skull/injuries , Skull/pathology , Animals , Mice , Hydrogels/chemistry , Hydrogels/pharmacology , Tissue Scaffolds/chemistry , Cell Proliferation/drug effects , Osteoblasts/drug effects , Osteoblasts/cytology , Osteoblasts/metabolism , Cell Differentiation/drug effects , Osteogenesis/drug effects , Bone Regeneration/drug effects , Cell Adhesion/drug effects
14.
Int Immunopharmacol ; 135: 112299, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38776853

ABSTRACT

OBJECTIVE: Periodontitis is a chronic infectious disease, characterized by loss of alveolar bone and supporting tissues. Cistanche deserticola(Cd), a local medicinal herb in Xinjiang, possesses favorable biological characteristics and potential applications. Our aim is to investigate the remodeling properties of Cd extract and elucidate the specific mechanisms underlying its therapeutic effects on periodontitis, by employing a combination of basic experimental and network pharmacology approaches. METHODS: Firstly, UHPLC-QTOF-MS analysis was conducted on Cd extract to identify its main components, with several compounds were identified by standard. Subsequently, in vitro studies were performed using the Cd extract on MC3T3-E1 cells. Cell proliferation viability was assessed using CCK-8 and apoptosis assays, while ALP and ARS staining and quantitative experiments, qRT-PCR, and Western blot assays were employed to evaluate the osteogenic differentiation capability. Network pharmacology analysis was then carried out using the identified compounds to establish a database of Cd components and targets, along with a database of periodontitis. The intersection of these databases revealed the network relationship between Cd components-mapped genes-signaling pathways. KEGG/GO pathway analysis of the targets was performed to filter potential enriched pathways. PPI/CytoHubba protein interaction network analysis was utilized to identify hub genes. Molecular docking and molecular dynamics simulations were employed to analyze the docking and interaction between core gene and Cd components. RESULTS: We detected 38 major components in the Cd extract, with Echinacoside, Acteoside, Tubuloside A, and Cistanoside A undergoing standard substance verification. In vitro studies indicated that the Cd, at concentrations below 100 µg/ mL, did not affect cell proliferation and inhibited apoptosis. Osteogenesis assays demonstrated that Cd at concentrations of 1 µg/ mL, 10 µg/ mL, and 100 µg/ mL significantly promoted the osteogenic differentiation ability of MC3T3-E1 cells. It also notably upregulated the mRNA and protein levels of Alp, Bmp2, Runx2, and Opn, and the optimal concentration was 10 µg/mL. Network pharmacology results revealed the network relationship between Cd's components, crossed targets and signaling pathways. Combined with KEGG/GO pathway analysis and PPI/CytoHubba protein interaction network analysis. The key pathway and hub genes of Cd regulating periodontitis are both related to hypoxia pathway and HIF-1α. Molecular docking results showed a strong binding affinity between Cd compounds and hub genes, and molecular dynamics simulation results indicated the stability of the complexes formed between HIF-1α and several Cd compounds. CONCLUSION: Cistanche deserticola exhibits a notable capacity to promote bone regeneration, and its mechanism of action in regulating periodontitis is associated with the hypoxia signaling pathway. HIF-1α may serve as a potential core gene. Future research will focus on exploring the mechanism of Cd in intervene periodontitis and promoting bone remodeling in hypoxic environment.


Subject(s)
Bone Remodeling , Cistanche , Network Pharmacology , Osteogenesis , Periodontitis , Cistanche/chemistry , Animals , Mice , Periodontitis/drug therapy , Periodontitis/metabolism , Periodontitis/microbiology , Bone Remodeling/drug effects , Osteogenesis/drug effects , Cell Proliferation/drug effects , Molecular Dynamics Simulation , Protein Interaction Maps , Plant Extracts/pharmacology , Plant Extracts/chemistry , Molecular Docking Simulation , Osteoblasts/drug effects , Osteoblasts/metabolism , Signal Transduction/drug effects , Cell Differentiation/drug effects , Apoptosis/drug effects , Cell Line
15.
Cell Rep Med ; 5(5): 101574, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38776873

ABSTRACT

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


Subject(s)
Bone Resorption , Osteoblasts , Osteogenesis , Animals , Humans , Osteoblasts/metabolism , Osteoblasts/drug effects , Osteogenesis/drug effects , Mice , Bone Resorption/pathology , Bone Resorption/metabolism , Anabolic Agents/pharmacology , Anabolic Agents/therapeutic use , Bone Remodeling/drug effects , Osteoporosis/pathology , Osteoporosis/metabolism , Osteoporosis/drug therapy , RANK Ligand/metabolism , Osteoclasts/metabolism , Osteoclasts/drug effects , Bone Development/drug effects , Osteoprotegerin/metabolism , Female , Signal Transduction/drug effects , Peptides/pharmacology , Male , Mice, Inbred C57BL , Bone and Bones/drug effects , Bone and Bones/metabolism , Bone and Bones/pathology
16.
J Mater Chem B ; 12(23): 5661-5677, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38747312

ABSTRACT

Magnesium alloy is currently regarded as the most favourable biodegradable metal; however, obstacles remain to be overcome in terms of managing its corrosion and ensuring its biocompatibility. In this study, a metal-organic complex comprising Ca ions incorporated in tannic acid (TA) was prepared and used to coat magnesium alloy by chemical conversion and dipping processes, followed by modification with stearic acid (SA). This metal-organic complex coating was demonstrated to be homogeneous and compact, and it significantly improved the electrochemical corrosion resistance and long-term degradation behaviour of the coated samples. Consequently, the well-controlled release of Mg and Ca ions, as well as the osteo-compatible TA and SA molecules, promoted the proliferation of osteoblast cells. This metal-organic complex coating offers a promising modifying strategy for magnesium-based orthopaedic implants.


Subject(s)
Alloys , Coated Materials, Biocompatible , Magnesium , Magnesium/chemistry , Alloys/chemistry , Alloys/pharmacology , Corrosion , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Materials Testing , Tannins/chemistry , Tannins/pharmacology , Cell Proliferation/drug effects , Surface Properties , Osteoblasts/drug effects , Osteoblasts/cytology , Absorbable Implants , Humans , Stearic Acids/chemistry , Animals , Calcium/chemistry , Calcium/metabolism , Cell Line
17.
J Ethnopharmacol ; 332: 118366, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-38763371

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Siwu decoction (SWD) is widely used in gynecological diseases, such as peripheral menopause syndrome, premature ovarian failure, and menstrual disorder. However, the mechanism of SWD on postmenopausal osteoporosis (PMOP) remains unclear. AIM OF THE STUDY: To discover the phytoestrogenic osteoprotective effect of SWD on PMOP. MATERIALS AND METHODS: The potential mechanism of SWD on PMOP was filtered through network pharmacology research. The potential mechanism was verified in MC3T3-E1 cell lines in vitro. CCK8 assay was conducted to assess cell proliferation and the expressions of ER/PI3K/AKT pathway were analyzed using Western blot. Female F-344 rats were chosen to set up the PMOP model. The osteoprotective effect of SWD in vivo was evaluated using Hematoxylin-eosin staining, TRAP staining, Goldner staining and DXA. The potential mechanism was verified in vivo through Western blot and immunohistochemistry. RT-qPCR was conducted to unveil the expressions of osteogenesis genes. RESULTS: Network pharmacology research showed that ER/PI3K/AKT pathway may be the potential mechanism of SWD on PMOP. SWD promoted the proliferation of osteoblasts and regulated the protein expressions of ER/PI3K/AKT pathway in vitro. SWD improved the morphological structure, bone mineralization and bone mineral density of femurs and suppressed osteoclastogenesis in PMOP rat model via ER/PI3K/AKT pathway in vivo. In addition, SWD regulated the mRNA expressions of osteogenesis-related genes. CONCLUSIONS: SWD exerts a phytoestrogenic osteoprotective on PMOP by regulating ER/PI3K/AKT pathway, which marks it as a valuable medicine or supplement of PMOP.


Subject(s)
Cell Proliferation , Drugs, Chinese Herbal , Osteoporosis, Postmenopausal , Phytoestrogens , Proto-Oncogene Proteins c-akt , Receptors, Estrogen , Signal Transduction , Animals , Female , Drugs, Chinese Herbal/pharmacology , Osteoporosis, Postmenopausal/drug therapy , Osteoporosis, Postmenopausal/prevention & control , Mice , Signal Transduction/drug effects , Receptors, Estrogen/metabolism , Rats , Proto-Oncogene Proteins c-akt/metabolism , Phytoestrogens/pharmacology , Phytoestrogens/therapeutic use , Cell Proliferation/drug effects , Phosphatidylinositol 3-Kinase/metabolism , Osteogenesis/drug effects , Cell Line , Humans , Bone Density/drug effects , Osteoblasts/drug effects , Osteoblasts/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Disease Models, Animal , Network Pharmacology
18.
J Nanobiotechnology ; 22(1): 261, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38760744

ABSTRACT

Delayed repair of fractures seriously impacts patients' health and significantly increases financial burdens. Consequently, there is a growing clinical demand for effective fracture treatment. While current materials used for fracture repair have partially addressed bone integrity issues, they still possess limitations. These challenges include issues associated with autologous material donor sites, intricate preparation procedures for artificial biomaterials, suboptimal biocompatibility, and extended degradation cycles, all of which are detrimental to bone regeneration. Hence, there is an urgent need to design a novel material with a straightforward preparation method that can substantially enhance bone regeneration. In this context, we developed a novel nanoparticle, mPPTMP195, to enhance the bioavailability of TMP195 for fracture treatment. Our results demonstrate that mPPTMP195 effectively promotes the differentiation of bone marrow mesenchymal stem cells into osteoblasts while inhibiting the differentiation of bone marrow mononuclear macrophages into osteoclasts. Moreover, in a mouse femur fracture model, mPPTMP195 nanoparticles exhibited superior therapeutic effects compared to free TMP195. Ultimately, our study highlights that mPPTMP195 accelerates fracture repair by preventing HDAC4 translocation from the cytoplasm to the nucleus, thereby activating the NRF2/HO-1 signaling pathway. In conclusion, our study not only proposes a new strategy for fracture treatment but also provides an efficient nano-delivery system for the widespread application of TMP195 in various other diseases.


Subject(s)
Cell Differentiation , Histone Deacetylases , Mesenchymal Stem Cells , Nanoparticles , Animals , Mice , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Nanoparticles/chemistry , Cell Differentiation/drug effects , Histone Deacetylases/metabolism , NF-E2-Related Factor 2/metabolism , Mice, Inbred C57BL , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoblasts/drug effects , Signal Transduction/drug effects , Heme Oxygenase-1/metabolism , Male , Bone Regeneration/drug effects , Osteogenesis/drug effects , Cell Nucleus/metabolism , Fracture Healing/drug effects , Humans , Membrane Proteins
19.
J Pharm Biomed Anal ; 245: 116192, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38703747

ABSTRACT

Calcium sensing receptor (CaSR) has become the novel target of treating osteoporosis with herbal medicine Ligustri Lucidi Fructus (LLF), however, the bioactive compounds responsible for anti-osteoporosis are hard to clarify due to the complexity and diversity of chemical constituents in it. Herein, the immobilized CaSR column was packed with stationary phase materials, which were derived from integrating CLIP-tagged CaSR directly out of crude cell lysates onto the surface of silica gels (5.83 mg/g) in a site-specific covalent manner. The column had a great specificity of recognizing agonists and kept a good stability for at least 3 weeks. The two compounds from LLF extract were screened and identified as olenuezhenoside and ligustroflavone using the immobilized CaSR column in conjunction with mass spectrometry. Molecular docking predicted that both compounds were bound in venus flytrap (VFT) domain of CaSR by the formation of hydrogen bonds. Cellular results showed that both compounds exhibited the distinct osteogenic activity by enhancing the proliferation, differentiation and mineralization of osteoblastic cells. Our study demonstrated that, the immobilized protein column enables to screen the bioactive compounds rapidly from herbal extract, and the newly discovered natural product ligands towards CaSR, including olenuezhenoside and ligustroflavone, will be the candidates for the treatment of osteoporosis.


Subject(s)
Ligustrum , Molecular Docking Simulation , Osteogenesis , Plant Extracts , Receptors, Calcium-Sensing , Receptors, Calcium-Sensing/metabolism , Receptors, Calcium-Sensing/antagonists & inhibitors , Osteogenesis/drug effects , Plant Extracts/pharmacology , Plant Extracts/chemistry , Ligustrum/chemistry , Humans , Osteoblasts/drug effects , Cell Proliferation/drug effects , Cell Differentiation/drug effects , Fruit/chemistry , Animals , Osteoporosis/drug therapy
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...