Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 11.359
Filter
1.
J Nanobiotechnology ; 22(1): 539, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39237993

ABSTRACT

Maxillofacial bone defects can severely impact quality of life by impairing physiological functions such as chewing, breathing, swallowing, and pronunciation. Polyether ether ketone (PEEK) is commonly used for the repair of maxillofacial defects due to its mechanical adaptability, while its osteogenic properties still need refinement. Herein, we have utilized the piezoelectric effect exhibited by barium titanate (BTO) under low-intensity pulsed ultrasound (LIPUS) to develop an ultrasound responsive PEEK (PDA@BTO-SPEEK, PBSP) through the mediating effect of polydopamine (PDA), for repairing maxillofacial bone defects. After modification by PDA@BTO, PBSP possesses better hydrophilicity, which is conducive to cell growth and adhesion. Simultaneously, by virtue of the piezoelectric characteristics of BTO, PBSP obtains a piezoelectric coefficient that matches the bone cortex. Notably, when PBSP is stimulated by LIPUS, it can generate stable electricity and effectively accelerate the osteogenic differentiation of osteoblasts through the regulation of the Piezo1-induced calcium (Ca2+) influx and Akt/GSK3ß/ß-catenin pathway. In addition, PBSP presents satisfactory therapeutic effects in rat skull defect models, and its osteogenic efficiency can be further improved under LIPUS stimulation with high tissue penetration. Collectively, PBSP + LIPUS exhibits great potential as a promising alternative strategy for the repair of maxillofacial bone defects.


Subject(s)
Benzophenones , Glycogen Synthase Kinase 3 beta , Ketones , Osteogenesis , Polyethylene Glycols , Polymers , Proto-Oncogene Proteins c-akt , Rats, Sprague-Dawley , beta Catenin , Animals , Glycogen Synthase Kinase 3 beta/metabolism , Polymers/chemistry , Osteogenesis/drug effects , Rats , Polyethylene Glycols/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Ketones/chemistry , Ketones/pharmacology , beta Catenin/metabolism , Cell Differentiation/drug effects , Osteoblasts/drug effects , Ultrasonic Waves , Indoles/chemistry , Indoles/pharmacology , Male , Signal Transduction/drug effects , Skull/drug effects , Titanium/chemistry , Titanium/pharmacology , Bone Regeneration/drug effects
2.
J Orthop Surg Res ; 19(1): 531, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39218922

ABSTRACT

BACKGROUND: Bone loss caused by microgravity exposure presents a serious threat to the health of astronauts, but existing treatment strategies have specific restrictions. This research aimed to investigate whether salidroside (SAL) can mitigate microgravity-induced bone loss and its underlying mechanism. METHODS: In this research, we used hindlimb unloading (HLU) and the Rotary Cell Culture System (RCCS) to imitate microgravity in vivo and in vitro. RESULTS: The results showed that salidroside primarily enhances bone density, microstructure, and biomechanical properties by stimulating bone formation and suppressing bone resorption, thereby preserving bone mass in HLU rats. In MC3T3-E1 cells cultured under simulated microgravity in rotary wall vessel bioreactors, the expression of osteogenic genes significantly increased after salidroside administration, indicating that salidroside can promote osteoblast differentiation under microgravity conditions. Furthermore, the Nrf2 inhibitor ML385 diminished the therapeutic impact of salidroside on microgravity-induced bone loss. Overall, this research provides the first evidence that salidroside can mitigate bone loss induced by microgravity exposure through stimulating the Nrf2/HO-1 pathway. CONCLUSION: These findings indicate that salidroside has great potential for treating space-related bone loss in astronauts and suggest that Nrf2/HO-1 is a viable target for counteracting microgravity-induced bone damage.


Subject(s)
Glucosides , NF-E2-Related Factor 2 , Phenols , Weightlessness Simulation , Glucosides/pharmacology , Glucosides/therapeutic use , Animals , Phenols/pharmacology , Phenols/therapeutic use , NF-E2-Related Factor 2/metabolism , Mice , Weightlessness Simulation/adverse effects , Rats , Male , Heme Oxygenase-1/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Weightlessness/adverse effects , Osteogenesis/drug effects , Osteoblasts/drug effects , Osteoblasts/metabolism , Hindlimb Suspension , Bone Resorption/prevention & control , Bone Resorption/etiology , Bone Resorption/metabolism , Bone Density/drug effects , Membrane Proteins
3.
Carbohydr Polym ; 344: 122496, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39218539

ABSTRACT

Chitosan is a promising natural polymer for coatings, it combines intrinsic antibacterial and pro-osteoblastic properties, but the literature still has a gap from the development to behavior of these coatings, so this systematic review aimed to answer, "What is the relationship between the physical and chemical properties of polymeric chitosan coatings on titanium implants on antibacterial activity and osteoblast viability?". PRISMA guidelines was followed, the search was applied into 4 databases and grey literature, without the restriction of time and language. The selection process occurred in 2 blinded steps by the authors. The criteria of eligibility were in vitro studies that evaluated the physical, chemical, microbiological, and biological properties of chitosan coatings on titanium surfaces. The risk of bias was analyzed by the specific tool. Of 734 potential articles 10 were included; all had low risk of bias. The coating was assessed according to the technique of fabrication, FT-IR, thickness, adhesion, roughness, wettability, antibacterial activity, and osteoblast viability. The analyzed coatings showed efficacy on antibacterial activity and cytocompatibility dependent on the class of material incorporated. Thus, this review motivates the development of time-dependent studies to optimize manufacturing and allow for an increase in patents and availability on the market.


Subject(s)
Anti-Bacterial Agents , Chitosan , Coated Materials, Biocompatible , Osteoblasts , Titanium , Chitosan/chemistry , Chitosan/pharmacology , Titanium/chemistry , Titanium/pharmacology , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Osteoblasts/drug effects , Osteoblasts/cytology , Surface Properties , Prostheses and Implants , Animals , Cell Survival/drug effects
4.
J Cell Mol Med ; 28(17): e70040, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39219020

ABSTRACT

Bone tissue engineering addresses the limitations of autologous resources and the risk of allograft disease transmission in bone diseases. In this regard, engineered three-dimensional (3D) models emerge as biomimetic alternatives to natural tissues, replicating intracellular communication. Moreover, the unique properties of super-paramagnetic iron oxide nanoparticles (SPIONs) were shown to promote bone regeneration via enhanced osteogenesis and angiogenesis in bone models. This study aimed to investigate the effects of SPION on both osteogenesis and angiogenesis and characterized a co-culture of Human umbilical vein endothelial cells (HUVEC) and MG-63 cells as a model of bone microtissue. HUVECs: MG-63s with a ratio of 4:1 demonstrated the best results among other cell ratios, and 50 µg/mL of SPION was the optimum concentration for maximum survival, cell migration and mineralization. In addition, the data from gene expression illustrated that the expression of osteogenesis-related genes, including osteopontin, osteocalcin, alkaline phosphatase, and collagen-I, as well as the expression of the angiogenesis-related marker, CD-31, and the tube formation, is significantly elevated when the 50 µg/mL concentration of SPION is applied to the microtissue samples. SPION application in a designed 3D bone microtissue model involving a co-culture of osteoblast and endothelial cells resulted in increased expression of specific markers related to angiogenesis and osteogenesis. This includes the design of a novel biomimetic model to boost blood compatibility and biocompatibility of primary materials while promoting osteogenic activity in microtissue bone models. Moreover, this can improve interaction with surrounding tissues and broaden the knowledge to promote superior-performance implants, preventing device failure.


Subject(s)
Bone Regeneration , Coculture Techniques , Human Umbilical Vein Endothelial Cells , Osteogenesis , Tissue Engineering , Humans , Bone Regeneration/drug effects , Osteogenesis/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Tissue Engineering/methods , Magnetite Nanoparticles/chemistry , Neovascularization, Physiologic/drug effects , Cell Movement/drug effects , Magnetic Iron Oxide Nanoparticles/chemistry , Cell Survival/drug effects , Cell Differentiation/drug effects , Osteoblasts/metabolism , Osteoblasts/drug effects , Osteoblasts/cytology
5.
Biotechnol J ; 19(8): e2400288, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39115337

ABSTRACT

Bone tissue engineering offers a promising alternative to stimulate the regeneration of damaged tissue, overcoming the limitations of conventional autografts and allografts. Recently, titanium alloy (Ti) implants have garnered significant attention for treating critical-sized bone defects, especially with the advancement of 3D printing technology. Although Ti alloys have impressive versatility, their lack of cellular adhesion, osteogenic and antibacterial properties are significant factors that contribute to their failure. Hence, to overcome these obstacles, this study aimed to incorporate osteoinductive and antibacterial cue-loaded hydrogels into 3D-printed Ti (3D-Ti) scaffolds. 3D-Ti scaffolds were synthesized using the direct metal laser sintering method and loaded with a gelatin (Gel) hydrogel containing strontium-doped silver nanoparticles (Sr-Ag NPs). Compared with Ag NPs, Sr-doped Ag NPs increased the expression of Runx2 mRNA, which is a key bone transcription factor. We subjected the bioactive 3D-hybrid scaffolds (3D-Ti/Gel/Sr-Ag NPs) to physicochemical and material characterization, followed by cytocompatibility and osteogenic evaluation. The microporous and macroporous topographies of the scaffolds with Sr-Ag NPs showed increased Runx2 expression and matrix mineralization, with potent antibacterial properties. Therefore, the 3D-Ti scaffolds incorporated with Sr-Ag NP-loaded Gel hydrogels favored osteoblast differentiation and antibacterial activity, indicating their potential for orthopedic applications.


Subject(s)
Anti-Bacterial Agents , Cell Differentiation , Gelatin , Hydrogels , Metal Nanoparticles , Osteoblasts , Osteogenesis , Printing, Three-Dimensional , Silver , Strontium , Tissue Engineering , Tissue Scaffolds , Titanium , Silver/chemistry , Silver/pharmacology , Gelatin/chemistry , Strontium/chemistry , Strontium/pharmacology , Titanium/chemistry , Titanium/pharmacology , Tissue Engineering/methods , Osteoblasts/drug effects , Osteoblasts/cytology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Tissue Scaffolds/chemistry , Hydrogels/chemistry , Hydrogels/pharmacology , Metal Nanoparticles/chemistry , Cell Differentiation/drug effects , Osteogenesis/drug effects , Animals , Mice , Bone and Bones/drug effects
6.
Nutrients ; 16(15)2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39125380

ABSTRACT

BACKGROUND: Excessive fluoride exposure induces skeletal fluorosis, but the specific mechanism responsible is still unclear. Therefore, this study aimed to identify the pathogenesis of fluoride-induced bone injuries. METHODS: We systematically searched fluoride-induced bone injury-related genes from five databases. Then, these genes were subjected to enrichment analyses. A TF (transcription factor)-mRNA-miRNA network and protein-protein interaction (PPI) network were constructed using Cytoscape, and the Human Protein Atlas (HPA) database was used to screen the expression of key proteins. The candidate pharmacological targets were predicted using the Drug Signature Database. RESULTS: A total of 85 studies were included in this study, and 112 osteoblast-, 35 osteoclast-, and 41 chondrocyte-related differential expression genes (DEGs) were identified. Functional enrichment analyses showed that the Atf4, Bcl2, Col1a1, Fgf21, Fgfr1 and Il6 genes were significantly enriched in the PI3K-Akt signaling pathway of osteoblasts, Mmp9 and Mmp13 genes were enriched in the IL-17 signaling pathway of osteoclasts, and Bmp2 and Bmp7 genes were enriched in the TGF-beta signaling pathway of chondrocytes. With the use of the TF-mRNA-miRNA network, the Col1a1, Bcl2, Fgfr1, Mmp9, Mmp13, Bmp2, and Bmp7 genes were identified as the key regulatory factors. Selenium methyl cysteine, CGS-27023A, and calcium phosphate were predicted to be the potential drugs for skeletal fluorosis. CONCLUSIONS: These results suggested that the PI3K-Akt signaling pathway being involved in the apoptosis of osteoblasts, with the IL-17 and the TGF-beta signaling pathways being involved in the inflammation of osteoclasts and chondrocytes in fluoride-induced bone injuries.


Subject(s)
Apoptosis , Fluorides , Inflammation , Osteoblasts , Signal Transduction , Humans , Fluorides/adverse effects , Apoptosis/drug effects , Osteoblasts/drug effects , Osteoblasts/metabolism , Inflammation/chemically induced , Signal Transduction/drug effects , MicroRNAs/metabolism , MicroRNAs/genetics , Osteoclasts/drug effects , Osteoclasts/metabolism , Chondrocytes/drug effects , Chondrocytes/metabolism , Protein Interaction Maps , RNA, Messenger/metabolism , RNA, Messenger/genetics , Gene Regulatory Networks , Gene Expression Regulation/drug effects , Bone Diseases/chemically induced , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Bone ; 188: 117242, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39209139

ABSTRACT

As obesity rates continue to rise, the prevalence of metabolic dysfunction and alcohol-associated steatotic liver disease (MetALD), a new term for Nonalcoholic Fatty Liver Disease (NAFLD), also increases. In an aging population, it is crucial to understand the interplay between metabolic disorders, such as MetALD, and bone health. This understanding becomes particularly significant in the context of implant osseointegration. This study introduces an in vitro model simulating high lipogenesis through the use of human Mesenchymal Stroma Cells-derived adipocytes, 3D intrahepatic cholangiocyte organoids (ICO), and Huh7 hepatocytes, to evaluate the endocrine influence on osteoblasts interacting with titanium. We observed a significant increase in intracellular fat accumulation in all three cell types, along with a corresponding elevation in metabolic gene expression compared to the control groups. Notably, osteoblasts undergoing mineralization in this high-lipogenesis environment also displayed lipid vesicle accumulation. The study further revealed that titanium surfaces modulate osteogenic gene expression and impact cell cycle progression, cell survival, and extracellular matrix remodeling under lipogenic conditions. These findings provide new insights into the challenges of implant integration in patients with obesity and MetALD, offering a deeper understanding of the metabolic influences on bone regeneration and implant success.


Subject(s)
Lipogenesis , Osteogenesis , Titanium , Humans , Titanium/pharmacology , Osteogenesis/drug effects , Lipogenesis/drug effects , Adipocytes/metabolism , Mesenchymal Stem Cells/metabolism , Osteoblasts/metabolism , Osteoblasts/drug effects , Organoids/metabolism , Hepatocytes/metabolism
8.
ACS Appl Mater Interfaces ; 16(34): 45389-45398, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39150145

ABSTRACT

Bioactive metal-based nanostructures, particularly zinc oxide (ZnO), are promising materials for bone tissue engineering. However, integrating them into 3D-printed polymers using traditional blending methods reduces the cell performance. Alternative surface deposition techniques often require extreme conditions that are unsuitable for polymers. To address these issues, we propose a metal-assisted hydrothermal synthesis method to modify 3D printed polycaprolactone (PCL) scaffolds with ZnO nanoparticles (NPs), facilitating the growth of ZnO nanoarrays (NAs) at a low-temperature (55 °C). Physicochemical characterizations revealed that the ZnO NPs form both physical and chemical bonds with the PCL surface; chemical bonding occurs between the carboxylate groups of PCL and Zn(OH)2 during seed deposition and hydrothermal synthesis. The ZnO NPs and NAs grown for a longer time (18 h) on the surface of PCL scaffolds exhibit significant proliferation and early differentiation of osteoblast-like cells. The proposed method is suitable for the surface modification of thermally degradable polymers, opening up new possibilities for the deposition of diverse metals.


Subject(s)
Osteoblasts , Polyesters , Printing, Three-Dimensional , Tissue Engineering , Tissue Scaffolds , Zinc Oxide , Zinc Oxide/chemistry , Tissue Scaffolds/chemistry , Polyesters/chemistry , Osteoblasts/cytology , Osteoblasts/drug effects , Surface Properties , Bone and Bones , Cell Proliferation/drug effects , Humans , Cell Differentiation/drug effects
9.
Medicina (Kaunas) ; 60(8)2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39202599

ABSTRACT

Background and Objectives: Enhanced osteoblast differentiation may be leveraged to prevent and treat bone-related diseases such as osteoporosis. No-ozone cold plasma (NCP) treatment is a promising and safe strategy to enhance osteoblast differentiation. Therefore, this study aimed to determine the effectiveness of direct and indirect NCP treatment methods on osteoblast differentiation. Mouse osteoblastic cells (MC3T3-E1) were treated with NCP using different methods, i.e., no NCP treatment (NT group; control), direct NCP treatment (DT group), direct NCP treatment followed by media replacement (MC group), and indirect treatment with NCP-treated media only (PAM group). Materials and Methods: The MC3T3-E1 cells were subsequently assessed for cell proliferation, alkaline phosphatase (ALP) activity, calcium deposition, and ALP and osteocalcin mRNA expression using real-time polymerase chain reaction. Results: Cell proliferation significantly increased in the NCP-treated groups (DT and PAM; MC and PAM) compared to the NT group after 24 h (p < 0.038) and 48 h (p < 0.000). ALP activity was increased in the DT and PAM groups at 1 week (p < 0.115) and in the DT, MC, and PAM groups at 2 weeks (p < 0.000) compared to the NT group. Calcium deposition was higher in the NCP-treated groups than in NT group at 2 and 3 weeks (p < 0.000). ALP mRNA expression peaked in the MC group at 2 weeks compared to the NP group (p < 0.014). Osteocalcin mRNA expression increased in the MC group at 2 weeks (p < 0.000) and was the highest in the PAM group at 3 weeks (p < 0.000). Thus, the effects of direct (DT and MC) and indirect (PAM) treatment varied, with MC direct treatment showing the most significant impact on osteoblast activity. Conclusions: The MC group exhibited enhanced osteoblast differentiation, indicating that direct NCP treatment followed by media replacement is the most effective method for promoting bone formation.


Subject(s)
Alkaline Phosphatase , Cell Differentiation , Cell Proliferation , Osteoblasts , Plasma Gases , Animals , Osteoblasts/drug effects , Mice , Cell Proliferation/drug effects , Cell Differentiation/drug effects , Plasma Gases/pharmacology , Plasma Gases/therapeutic use , Alkaline Phosphatase/analysis , Alkaline Phosphatase/metabolism , Ozone/pharmacology , Ozone/therapeutic use , Osteocalcin/analysis
10.
Bone Res ; 12(1): 45, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39164234

ABSTRACT

Bone marrow stromal/stem cells (BMSCs) are generally considered as common progenitors for both osteoblasts and adipocytes in the bone marrow, but show preferential differentiation into adipocytes rather than osteoblasts under aging, thus leading to senile osteoporosis. Accumulated evidences indicate that rejuvenation of BMSCs by autophagic enhancement delays bone aging. Here we synthetized and demonstrated a novel autophagy activator, CXM102 that could induce autophagy in aged BMSCs, resulting in rejuvenation and preferential differentiation into osteoblasts of BMSCs. Furthermore, CXM102 significantly stimulated bone anabolism, reduced marrow adipocytes, and delayed bone loss in middle-age male mice. Mechanistically, CXM102 promoted transcription factor EB (TFEB) nuclear translocation and favored osteoblasts formation both in vitro and in vivo. Moreover, CXM102 decreased serum levels of inflammation and reduced organ fibrosis, leading to a prolonger lifespan in male mice. Our results indicated that CXM102 could be used as an autophagy inducer to rejuvenate BMSCs and shed new lights on strategies for senile osteoporosis and healthyspan improvement.


Subject(s)
Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Mesenchymal Stem Cells , Osteoporosis , Animals , Autophagy/drug effects , Male , Mesenchymal Stem Cells/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Mice , Osteoporosis/pathology , Osteoporosis/metabolism , Longevity , Cell Differentiation , Aging/physiology , Mice, Inbred C57BL , Cellular Senescence/drug effects , Rejuvenation , Osteoblasts/metabolism , Osteoblasts/drug effects , Osteogenesis/drug effects
11.
ACS Appl Mater Interfaces ; 16(33): 43227-43243, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39121390

ABSTRACT

Implant-associated infections and delayed osseointegration are major challenges for the clinical success of titanium implants. To enhance antibacterial effects and promote early osseointegration, we developed a synergistic photothermal (PTT)/photodynamic (PDT) therapy strategy based on near-infrared (NIR) responsive biomimetic micro/nano titanate/TiO2-X heterostructure coatings (KMNW and NaMNS) in situ constructed on the surface of titanium implants. Specifically, KMNW and NaMNS significantly enhanced photothermal conversion capabilities, achieving localized high temperatures of 48-51 °C and promoting substantial amounts of reactive oxygen species production under 808 nm irradiation. In vitro antibacterial experiments demonstrated that KMNW achieved the highest antibacterial rates against Staphylococcus aureus and Escherichia coli, at 98.78 and 98.33% respectively. Moreover, by mimicking the three-dimensional fibrous network of the extracellular matrix during bone healing, both KMNW and NaMNS markedly promoted the proliferation and osteogenic differentiation of osteoblasts. In vivo implantation studies further confirmed these findings, with KMNW and NaMNS exhibiting superior antibacterial performance under NIR irradiation─94.45% for KMNW and 92.66% for NaMNS. Moreover, KMNW and NaMNS also significantly promoted new bone formation and improved osseointegration in vivo. This study presents a promising PTT/PDT therapeutic strategy for dentistry and orthopedics by employing NIR-responsive biomimetic coatings to combat implant-associated infection and accelerate osseointegration.


Subject(s)
Anti-Bacterial Agents , Escherichia coli , Infrared Rays , Osseointegration , Staphylococcus aureus , Titanium , Titanium/chemistry , Titanium/pharmacology , Osseointegration/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Escherichia coli/drug effects , Animals , Staphylococcus aureus/drug effects , Biomimetic Materials/chemistry , Biomimetic Materials/pharmacology , Biomimetic Materials/radiation effects , Prostheses and Implants , Photochemotherapy , Mice , Photothermal Therapy , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Reactive Oxygen Species/metabolism , Microbial Sensitivity Tests , Osteoblasts/drug effects , Osteoblasts/cytology , Osteogenesis/drug effects
12.
Biomed Mater ; 19(5)2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39121887

ABSTRACT

Guided bone regeneration (GBR) membranes play an important role in oral bone regeneration. However, enhancing their bone regeneration potential and antibacterial properties is crucial. Herein, silk fibroin (SF)/polycaprolactone (PCL) core-shell nanofibers loaded with epigallocatechin gallate (EGCG) were prepared using emulsion electrospinning. The nanofibrous membranes were characterized via scanning electron microscopy, transmission electron microscopy, Fourier transform infrared spectroscopy, thermogravimetric analysis, water contact angle (CA) measurement, mechanical properties testing, drug release kinetics, and 1,1-diphenyl-2-picryl-hydrazyl radical (DPPH) free radical scavenging assay. Mouse pre-osteoblast MC3T3-E1 cells were used to assess the biological characteristics, cytocompatibility, and osteogenic differentiation potential of the nanofibrous membrane. Additionally, the antibacterial properties againstStaphylococcus aureus (S. aureus)andEscherichia coli (E. coli)were evaluated. The nanofibers prepared by emulsion electrospinning exhibited a stable core-shell structure with a smooth and continuous surface. The tensile strength of the SF/PCL membrane loaded with EGCG was 3.88 ± 0.15 Mpa, the water CA was 50°, and the DPPH clearance rate at 24 h was 81.73% ± 0.07%. The EGCG release rate of membranes prepared by emulsion electrospinning was reduced by 12% within 72 h compared to that of membranes prepared via traditional electrospinning.In vitroexperiments indicate that the core-shell membranes loaded with EGCG demonstrated good cell compatibility and promoted adhesion, proliferation, and osteogenic differentiation of MC3T3-E1 cells. Furthermore, the EGCG-loaded membranes exhibited inhibitory effects onE. coliandS. aureus. These findings indicate that core-shell nanofibrous membranes encapsulated with EGCG prepared using emulsion electrospinning possess good antioxidant, osteogenic, and antibacterial properties, making them potential candidates for research in GBR materials.


Subject(s)
Anti-Bacterial Agents , Bone Regeneration , Catechin , Emulsions , Escherichia coli , Fibroins , Nanofibers , Osteogenesis , Polyesters , Staphylococcus aureus , Animals , Fibroins/chemistry , Polyesters/chemistry , Mice , Bone Regeneration/drug effects , Catechin/analogs & derivatives , Catechin/chemistry , Nanofibers/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Emulsions/chemistry , Staphylococcus aureus/drug effects , Osteogenesis/drug effects , Escherichia coli/drug effects , Osteoblasts/cytology , Osteoblasts/drug effects , Guided Tissue Regeneration/methods , Tissue Scaffolds/chemistry , Biocompatible Materials/chemistry , Tissue Engineering/methods , Cell Differentiation/drug effects , Materials Testing , Membranes, Artificial , Tensile Strength , Drug Liberation , Spectroscopy, Fourier Transform Infrared , 3T3 Cells , Cell Line
13.
Sci Rep ; 14(1): 19973, 2024 08 28.
Article in English | MEDLINE | ID: mdl-39198677

ABSTRACT

Osteoblasts and osteoclasts play an important role in maintaining the structural integrity of bone tissue, in which osteoclasts degrade bone structure and osteoblasts restore bone tissue. The imbalance of osteoblast and osteoclast function can lead to many bone-related diseases, such as osteoporosis and inflammatory osteolysis. The drug that can both promote bone formation and inhibit bone loss will be able to treat those diseases. In this study, it was found that LMK-235, an selective HDAC4/5 inhibitor, inhibited the differentiation and maturation of osteoclasts by regulating NF-κB and p-Smad2/3 signaling pathways via inhibition of HDAC4. At the same time, we found that LMK-235 promoted osteoblast mineralization by upregulating Runx2 expression via inhibition of HDAC4. In vivo, LMK-235 was able to alleviate lipopolysaccharide (LPS)-induced calvarial osteolysis and promote the repair of bone defects. Taken together, LMK-235 suppresses osteoclast differentiation and promotes osteoblast formation by inhibiting HDAC4. This may provide a valuable treatment for bone diseases caused by abnormal osteoclast bone resorption and osteoblast bone regeneration.


Subject(s)
Cell Differentiation , Histone Deacetylases , Osteoblasts , Osteoclasts , Osteogenesis , Animals , Mice , Cell Differentiation/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Hydroxamic Acids , Lipopolysaccharides/pharmacology , Mice, Inbred C57BL , NF-kappa B/metabolism , Osteoblasts/metabolism , Osteoblasts/drug effects , Osteoclasts/metabolism , Osteoclasts/drug effects , Osteogenesis/drug effects , Osteolysis/metabolism , Osteolysis/pathology , Pyrimidines , RAW 264.7 Cells , Signal Transduction/drug effects
14.
FASEB J ; 38(17): e70017, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39213037

ABSTRACT

The use of traditional nicotine delivery products such as tobacco has long been linked to detrimental health effects. However, little work to date has focused on the emerging market of aerosolized nicotine delivery known as electronic nicotine delivery systems (ENDS) or electronic cigarettes, and their potential for new effects on human health. Challenges studying these devices include heterogeneity in the formulation of the common components of most available ENDS, including nicotine and a carrier (commonly composed of propylene glycol and vegetable glycerin, or PG/VG). In the present study, we report on experiments interrogating the effects of major identified components in e-cigarettes. Specifically, the potential concomitant effects of nicotine and common carrier ingredients in commercial "vape" products are explored in vitro to inform the potential health effects on the craniofacial skeleton through novel vectors as compared to traditional tobacco products. MC3T3-E1 murine pre-osteoblast cells were cultured in vitro with clinically relevant liquid concentrations of nicotine, propylene glycol (PG), vegetable glycerin (VG), Nicotine+PG/VG, and the vape liquid of a commercial product (Juul). Cells were treated acutely for 24 h and RNA-Seq was utilized to determine segregating alteration in mRNA signaling. Influential gene targets identified with sparse partial least squares discriminant analysis (sPLS-DA) implemented in mixOmics were assessed using the PANTHER Classification system for molecular functions, biological processes, cellular components, and pathways of effect. Additional endpoint functional analyses were used to confirm cell cycle changes. The initial excitatory concentration (EC50) studied defined a target concentration of carrier PG/VG liquid that altered the cell cycle of the calvarial cells. Initial sPLS-DA analysis demonstrated the segregation of nicotine and non-nicotine exposures utilized in our in vitro modeling. Pathway analysis suggests a strong influence of nicotine exposures on cellular processes including metabolic processes and response to stimuli including autophagic flux. Further interrogation of the individual treatment conditions demonstrated segregation by treatment modality (Control, Nicotine, Carrier (PG+VG), Nicotine+PG/VG) along three dimensions best characterized by: latent variable 1 (PLSDA-1) showing strong segregation based on nicotine influence on cellular processes associated with cellular adhesion to collagen, osteoblast differentiation, and calcium binding and metabolism; latent variable 2 (PLSDA-2) showing strong segregation of influence based on PG+VG and Control influence on cell migration, survival, and cycle regulation; and latent variable 3 (PLSDA-3) showing strong segregation based on Nicotine and Control exposure influence on cell activity and growth and developmental processes. Further, gene co-expression network analysis implicates targets of the major pathway genes associated with bone growth and development, particularly craniofacial (FGF, Notch, TGFß, WNT) and analysis of active subnetwork pathways found these additionally overrepresented in the Juul exposure relative to Nicotine+PG/VG. Finally, experimentation confirmed alterations in cell count, and increased evidence of cell stress (markers of autophagy), but no alteration in apoptosis. These data suggest concomitant treatment with Nicotine+PG/VG drives alterations in pre-osteoblast cell cycle signaling, specifically transcriptomic targets related to cell cycle and potentially cell stress. Although we suspected cell stress and well as cytotoxic effects of Nicotine+PG/VG, no great influence on apoptotic factors was observed. Further RNA-Seq analysis allowed for the direct interrogation of molecular targets of major pathways involved in bone and craniofacial development, each demonstrating segregation (altered signaling) due to e-cigarette-type exposure. These data have implications directed toward ENDS formulation as synergistic effects of Nicotine+PG/VG are evidenced here. Thus, future research will continue to interrogate how varied formulation of Nicotine+PG/VG affects overall cell functions in multiple vital systems.


Subject(s)
Electronic Nicotine Delivery Systems , Nicotine , Osteoblasts , Animals , Mice , Nicotine/pharmacology , Osteoblasts/metabolism , Osteoblasts/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Propylene Glycol , Cell Line
15.
J Ethnopharmacol ; 335: 118690, 2024 Dec 05.
Article in English | MEDLINE | ID: mdl-39142621

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Postmenopausal osteoporosis (PMOP) has been considered as a major causative factor for bone-joint pain and inducing pathologic fractures. Bu-Sui-Dan (BSD), a classic ancient herbal formula, has been shown to exhibit osteoprotective effects by promoting bone marrow development and bone growth. However, the exact mechanism of BSD are still unexplored. AIM OF STUDY: The study aimed to investigate the protective effect of BSD against osteoporotic injury, and to explore whether BSD regulated BMSCs' osteogenic differentiation by targeting VGLL4, which in turn improved PMOP. MATERIALS AND METHODS: The anti-osteoporotic effect of BSD was studied in ovariectomized (OVX) rats and bone marrow mesenchymal stem cells (BMSCs). Micro-CT imaging and HE staining were performed, and the levels of osteogenic protein RUNX2 and osteogenesis-related factor VGLL4 were determined. Co-immunoprecipitation (Co-IP) was further employed to delve into the effects of BSD on the interactions between TEAD4 and RUNX2. The key osteogenic factors 1ALP, COLl1A1, and Osterix expression were detected by RT-qPCR. Co-IP and proximity ligation assay (PLA) were employed to scrutinize the influence of BSD on TEAD4 and RUNX2 inter-binding. Moreover, VGLL4 knockdown in BMSCs was conducted to confirm the role of VGLL4 in the therapeutic mechanism of BSD. RESULTS: BSD showed a dose-dependent protective effect against osteoporotic injury, as evidenced by improvement in bone volume, bone microarchitecture, and histomorphometry. Additionally, BSD treatment increased the levels of RUNX2 and its downstream target genes including ALP, COL1A1, and Osterix. Moreover, BSD upregulated VGLL4 expression and lessened TEAD4-RUNX2 interactions. In BMSCs experiment, BSD-containing serum could promote osteogenic differentiation of BMSCs, boosted the expression of osteogenesis-related factors and VGLL4 level. The knockdown of VGLL4 in BMSCs diminished the promotion effect of BSD in osteoblast differentiation, suggesting that VGLL4 play a vital role in the therapeutic effects exerted by BSD. CONCLUSION: BSD ameliorated osteoporosis injury and promoted osteoblast differentiation through upregulation of VGLL4 levels, which in turn antagonized TEAD4-mediated RUNX2 transcriptional repression. Our study implied that BSD may be an osteoporosis therapeutic agent.


Subject(s)
Cell Differentiation , Core Binding Factor Alpha 1 Subunit , Mesenchymal Stem Cells , Osteoblasts , Osteogenesis , Ovariectomy , Rats, Sprague-Dawley , Transcription Factors , Up-Regulation , Animals , Female , Osteoblasts/drug effects , Osteoblasts/metabolism , Core Binding Factor Alpha 1 Subunit/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Cell Differentiation/drug effects , Up-Regulation/drug effects , Osteogenesis/drug effects , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Rats , TEA Domain Transcription Factors , Osteoporosis, Postmenopausal/prevention & control , Cells, Cultured
16.
Biomed Mater ; 19(6)2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39208845

ABSTRACT

Hydrogels have excellent swelling properties and have been widely applied in tissue engineering because of their similarity to the extracellular matrix (ECM). Sodium alginate (SA) and carboxymethyl chitosan (CMCS) were prepared into hydrogel microspheres with Ca2+crosslinking in our study. The morphology, inner structure, mechanical properties, water content, swelling rate and BMP-2 loading and releasing properties were characterized. Our results showed that the composite SA /CMCS hydrogel microspheres were translucent and spherical in shape with uniform particle size. The incorporation of CMCS further increased the diameters of the microspheres, internal pore structure, water content, and mechanical properties of the SA/CMCS hydrogel microspheres. At the same SA concentration, with the increase of CMSC concentration, the diameter of microspheres could be increased by about 0.4 mm, the water content can be increased about 1%-2%. As for the mechanical properties, the compressive strength can be increased by 0.04-0.1 MPa, and the modulus of elasticity can be increased by 0.1-0.15 MPa. BMP-2 was chosen as a model agent and it could be loaded into SA/CMCS microspheres, and the incorporation of CMCS increased BMP-2 loading. The encapsulated BMP-2 was sustainably releasedin vitro. The leaching solutions of the SA/CMCS hydrogel microspheres exhibited good cytocompatibility and could increase ALP activity, ALP expression, and biomineralization on MC3T3-E1 cells. After 7 d of co-culture, ALP activities in S2.5C2 and S2.5C3 groups was increased by 50% and 45% compared with that of the control group. When embedded in the SA/CMCS microspheres, the MC3T3-E1 cells were evenly distributed inside the hydrogel microspheres and remained viable. Transcriptomic studies showed that incorporation of CMCS induced upregulation of 1141 differentially expressed genes (DEGs) and downregulation of 1614 DEGs compared with SA microspheres. The most significantly enriched pathways were the Wnt and MAPK signaling pathways induced by the incorporation of CMCS and BMP-2. In conclusion, our results indicated that the physiochemical characteristics of the SA hydrogel microspheres could be greatly modulated by CMCS to better mimic the ECM microenvironment and induce osteo-inductive activities of MC3T3-E1 cells.


Subject(s)
Alginates , Bone Morphogenetic Protein 2 , Cell Proliferation , Chitosan , Hydrogels , Microspheres , Tissue Engineering , Chitosan/chemistry , Chitosan/analogs & derivatives , Animals , Mice , Hydrogels/chemistry , Cell Proliferation/drug effects , Alginates/chemistry , Bone Morphogenetic Protein 2/metabolism , Tissue Engineering/methods , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Compressive Strength , Particle Size , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoblasts/cytology , Materials Testing , Extracellular Matrix/metabolism , Cell Line , Osteogenesis/drug effects , Water/chemistry
17.
ACS Biomater Sci Eng ; 10(9): 5675-5688, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39108014

ABSTRACT

Diamond-like carbon (DLC) wear debris, which is often composed of different types of structures, is generated from DLC-modified artificial joints in the human body, and its biocompatibility evaluation is especially important to prevent wear-debris-induced implant failure. Here, RAW 264.7 macrophages (inflammatory-reaction assay) and primary mouse osteoblasts (osteoblastogenesis assay) were employed to investigate the toxicity of DLC wear particles (DWPs) by evaluation of cell viability and morphology, enzyme-linked immunosorbent assays, and quantitative reverse-transcription polymerase chain reaction (PCR). Relevant histopathological analysis of rat joints was also performed in vivo. We found that DWPs with a relatively high sp2/sp3 ratio (graphite-phase tendency) manifested a higher cytotoxicity and significant inhibition of osteoblastogenesis. DWPs with a relatively low sp2/sp3 ratio (diamond-phase tendency) showed good biocompatibility in vivo. The DWPs exhibiting a low sp2/sp3 ratio demonstrated reduced secretion of TNF-α and IL-6, along with increased secretion of TIMP-1, resulting in the downregulation of MMP-2 and MMP-9 and upregulation of interleukin-10 (IL-10), thereby attenuating the inflammatory response. Moreover, coculturing osteoblasts with DWPs exhibiting a low sp2/sp3 ratio resulted in an elevated OPG/RANKL ratio and increased expression of OPG mRNA. Because of the absence of electrostatic repulsion, DWPs with a relatively low sp2/sp3 ratio enhanced bovine serum albumin adsorption, which favored cellular activities. Cytotoxicity assessment of DWPs can help establish an evaluation system for particle-related joint disease and can facilitate the clinical application of DLC-coated prostheses.


Subject(s)
Osteoblasts , Animals , Mice , Osteoblasts/metabolism , Osteoblasts/drug effects , RAW 264.7 Cells , Rats , Diamond/chemistry , Cell Survival/drug effects , Male , Joint Prosthesis/adverse effects , Rats, Sprague-Dawley , Arthroplasty, Replacement/adverse effects , Carbon/adverse effects , Biocompatible Materials/chemistry , Materials Testing
18.
Front Endocrinol (Lausanne) ; 15: 1380013, 2024.
Article in English | MEDLINE | ID: mdl-39086902

ABSTRACT

In this study, we used a bioinformatic approach to construct a miRNA-target gene interaction network potentially involved in the anabolic effect of parathyroid hormone analogue teriparatide [PTH (1-34)] on osteoblasts. We extracted a dataset of 26 microRNAs (miRNAs) from previously published studies and predicted miRNA target interactions (MTIs) using four software tools: DIANA, miRWalk, miRDB, and TargetScan. By constructing an interactome of PTH-regulated miRNAs and their predicted target genes, we elucidated signaling pathways regulating pluripotency of stem cells, the Hippo signaling pathway, and the TGF-beta signaling pathway as the most significant pathways in the effects of PTH on osteoblasts. Furthermore, we constructed intersection of MTI networks for these three pathways and added validated interactions. There are 8 genes present in all three selected pathways and a set of 18 miRNAs are predicted to target these genes, according to literature data. The most important genes in all three pathways were BMPR1A, BMPR2 and SMAD2 having the most interactions with miRNAs. Among these miRNAs, only miR-146a-5p and miR-346 have validated interactions in these pathways and were shown to be important regulators of these pathways. In addition, we also propose miR-551b-5p and miR-338-5p for further experimental validation, as they have been predicted to target important genes in these pathways but none of their target interactions have yet been verified. Our wet-lab experiment on miRNAs differentially expressed between PTH (1-34) treated and untreated mesenchymal stem cells supports miR-186-5p from the literature obtained data as another prominent miRNA. The meticulous selection of miRNAs outlined will significantly support and guide future research aimed at discovering and understanding the crucial pathways of osteoanabolic PTH-epigenetic effects on osteoblasts. Additionally, they hold potential for the discovery of new PTH target genes, innovative biomarkers for the effectiveness and safety of osteoporosis-affected treatment, as well as novel therapeutic targets.


Subject(s)
Computational Biology , MicroRNAs , Osteoblasts , Parathyroid Hormone , MicroRNAs/genetics , Osteoblasts/drug effects , Osteoblasts/metabolism , Computational Biology/methods , Parathyroid Hormone/pharmacology , Humans , Gene Regulatory Networks/drug effects , Signal Transduction/drug effects , Animals , Teriparatide/pharmacology
19.
Nutrients ; 16(15)2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39125268

ABSTRACT

Rhodiola rosea, a long-lived herbaceous plant from the Crassulaceae group, contains the active compound salidroside, recognized as an adaptogen with significant therapeutic potential for bone metabolism. Salidroside promotes osteoblast proliferation and differentiation by activating critical signaling pathways, including bone morphogenetic protein-2 and adenosine monophosphate-activated protein kinase, essential for bone formation and growth. It enhances osteogenic activity by increasing alkaline phosphatase activity and mineralization markers, while upregulating key regulatory proteins including runt-related transcription factor 2 and osterix. Additionally, salidroside facilitates angiogenesis via the hypoxia-inducible factor 1-alpha and vascular endothelial growth factor pathway, crucial for coupling bone development with vascular support. Its antioxidant properties offer protection against bone loss by reducing oxidative stress and promoting osteogenic differentiation through the nuclear factor erythroid 2-related factor 2 pathway. Salidroside has the capability to counteract the negative effects of glucocorticoids on bone cells and prevents steroid-induced osteonecrosis. Additionally, it exhibits multifaceted anti-inflammatory actions, notably through the inhibition of tumor necrosis factor-alpha and interleukin-6 expression, while enhancing the expression of interleukin-10. This publication presents a comprehensive review of the literature on the impact of salidroside on various aspects of bone tissue metabolism, emphasizing its potential role in the prevention and treatment of osteoporosis and other diseases affecting bone physiology.


Subject(s)
Bone and Bones , Glucosides , Osteoblasts , Osteogenesis , Osteoporosis , Phenols , Glucosides/pharmacology , Humans , Phenols/pharmacology , Bone and Bones/drug effects , Bone and Bones/metabolism , Osteogenesis/drug effects , Osteoporosis/drug therapy , Animals , Osteoblasts/drug effects , Osteoblasts/metabolism , Rhodiola/chemistry , Signal Transduction/drug effects , Antioxidants/pharmacology , Cell Differentiation/drug effects , Anti-Inflammatory Agents/pharmacology
20.
Bone Res ; 12(1): 43, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39103355

ABSTRACT

Apoptosis is crucial for tissue homeostasis and organ development. In bone, apoptosis is recognized to be a main fate of osteoblasts, yet the relevance of this process remains underexplored. Using our murine model with inducible Caspase 9, the enzyme that initiates intrinsic apoptosis, we triggered apoptosis in a proportion of mature osteocalcin (OCN+) osteoblasts and investigated the impact on postnatal bone development. Osteoblast apoptosis stimulated efferocytosis by osteal macrophages. A five-week stimulation of OCN+ osteoblast apoptosis in 3-week-old male and female mice significantly enhanced vertebral bone formation while increasing osteoblast precursors. A similar treatment regimen to stimulate osterix+ cell apoptosis had no impact on bone volume or density. The vertebral bone accrual following stimulation of OCN+ osteoblast apoptosis did not translate in improved mechanical strength due to disruption of the lacunocanalicular network. The observed bone phenotype was not influenced by changes in osteoclasts but was associated with stimulation of macrophage efferocytosis and vasculature formation. Phenotyping of efferocytic macrophages revealed a unique transcriptomic signature and expression of factors including VEGFA. To examine whether macrophages participated in the osteoblast precursor increase following osteoblast apoptosis, macrophage depletion models were employed. Depletion of macrophages via clodronate-liposomes and the CD169-diphtheria toxin receptor mouse model resulted in marked reduction in leptin receptor+ and osterix+ osteoblast precursors. Collectively, this work demonstrates the significance of osteoblast turnover via apoptosis and efferocytosis in postnatal bone formation. Importantly, it exposes the potential of targeting this mechanism to promote bone anabolism in the clinical setting.


Subject(s)
Apoptosis , Macrophages , Osteoblasts , Osteogenesis , Animals , Osteoblasts/metabolism , Osteoblasts/drug effects , Osteogenesis/physiology , Osteogenesis/drug effects , Macrophages/metabolism , Female , Male , Mice , Phagocytosis/physiology , Mice, Inbred C57BL , Efferocytosis
SELECTION OF CITATIONS
SEARCH DETAIL