Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 14.897
1.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(4): 697-705, 2024 Apr 20.
Article Zh | MEDLINE | ID: mdl-38708503

OBJECTIVE: To explore the role of zinc finger protein 36(ZFP36) in regulating osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) and preosteoblasts. METHODS: ZFP36 expression was observed in primary mouse BMSCs and mouse preosteoblasts (MC3T3-E1 cells) during induced osteogenic differentiation. Zfp36-deficient cell models were constructed in the two cells using RNA interference technique and the changes in differentiation capacities of the transfected cells into osteoblasts were observed. Transcriptome sequencing was used to investigate the potential mechanisms of ZFP36 for regulating osteoblast differentiation of the two cells. U0126, a ERK/MAPK signal suppressor, was used to verify the regulatory mechanism of Zfp36 in osteogenic differentiation of Zfp36-deficient cells. RESULTS: During the 14-day induction of osteogenic differentiation, both mouse BMSCs and MC3T3-E1 cells exhibited increased expression of ZFP36, and its mRNA expression reached the peak level on Day 7(P < 0.0001). The Zfp36-deficient cell models showed reduced intensity of alkaline phosphatase (ALP) staining and alizarin red staining with significantly lowered expressions of the osteogenic marker genes including Alpl, Sp7, Bglap and Ibsp (P < 0.01). Transcriptome sequencing verified the reduction of bone mineralization-related gene expressions in Zfp36-deficient cells and indicated the involvement of ERK signaling in the potential regulatory mechanism of Zfp36. Immunoblotting showed that pERK protein expression increased significantly in Zfp36-deficient cells compared with the control cells. In Zfp36-deficient MC3T3-E1 cells, inhibition of activated ERK/MAPK signaling with U0126 resulted in obviously enhanced ALP staining and significantly increased expressions of osteoblast differentiation markers Runx2 and Bglap (P < 0.05). CONCLUSIONS: ZFP36 is involved in the regulation of osteoblast differentiation of mouse BMSCs and preosteoblasts, and ZFP36 deficiency causes inhibition of osteoblast differentiation of the cells by activating the ERK/MAPK signaling pathway.


Cell Differentiation , MAP Kinase Signaling System , Mesenchymal Stem Cells , Osteoblasts , Osteogenesis , Animals , Mice , Alkaline Phosphatase/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Butyrate Response Factor 1/metabolism , Butyrate Response Factor 1/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism
2.
Sci Rep ; 14(1): 10345, 2024 05 06.
Article En | MEDLINE | ID: mdl-38710795

Skeletal bone function relies on both cells and cellular niches, which, when combined, provide guiding cues for the control of differentiation and remodeling processes. Here, we propose an in vitro 3D model based on human fetal osteoblasts, which eases the study of osteocyte commitment in vitro and thus provides a means to examine the influences of biomaterials, substances or cells on the regulation of these processes. Aggregates were formed from human fetal osteoblasts (hFOB1.19) and cultivated under proliferative, adipo- and osteoinductive conditions. When cultivated under osteoinductive conditions, the vitality of the aggregates was compromised, the expression levels of the mineralization-related gene DMP1 and the amount of calcification and matrix deposition were lower, and the growth of the spheroids stalled. However, within spheres under growth conditions without specific supplements, self-organization processes occur, which promote extracellular calcium deposition, and osteocyte-like cells develop. Long-term cultivated hFOB aggregates were free of necrotic areas. Moreover, hFOB aggregates cultivated under standard proliferative conditions supported the co-cultivation of human monocytes, microvascular endothelial cells and stromal cells. Overall, the model presented here comprises a self-organizing and easily accessible 3D osteoblast model for studying bone marrow formation and in vitro remodeling and thus provides a means to test druggable molecular pathways with the potential to promote life-long bone formation and remodeling.


Cell Differentiation , Coculture Techniques , Osteoblasts , Humans , Osteoblasts/metabolism , Osteoblasts/cytology , Cellular Microenvironment , Bone Marrow Cells/metabolism , Bone Marrow Cells/cytology , Osteogenesis , Cell Aggregation , Cells, Cultured
3.
Biomed Eng Online ; 23(1): 44, 2024 May 05.
Article En | MEDLINE | ID: mdl-38705993

BACKGROUND: Osteocytes are critical mechanosensory cells in bone, and mechanically stimulated osteocytes produce exosomes that can induce osteogenesis. MicroRNAs (miRNAs) are important constituents of exosomes, and some miRNAs in osteocytes regulate osteogenic differentiation; previous studies have indicated that some differentially expressed miRNAs in mechanically strained osteocytes likely influence osteoblastic differentiation. Therefore, screening and selection of miRNAs that regulate osteogenic differentiation in exosomes of mechanically stimulated osteocytes are important. RESULTS: A mechanical tensile strain of 2500 µÎµ at 0.5 Hz 1 h per day for 3 days, elevated prostaglandin E2 (PGE2) and insulin-like growth factor-1 (IGF-1) levels and nitric oxide synthase (NOS) activity of MLO-Y4 osteocytes, and promoted osteogenic differentiation of MC3T3-E1 osteoblasts. Fourteen miRNAs differentially expressed only in MLO-Y4 osteocytes which were stimulated with mechanical tensile strain, were screened, and the miRNAs related to osteogenesis were identified. Four differentially expressed miRNAs (miR-1930-3p, miR-3110-5p, miR-3090-3p, and miR-3058-3p) were found only in mechanically strained osteocytes, and the four miRNAs, eight targeted mRNAs which were differentially expressed only in mechanically strained osteoblasts, were also identified. In addition, the mechanically strained osteocyte-derived exosomes promoted the osteoblastic differentiation of MC3T3-E1 cells in vitro, the exosomes were internalized by osteoblasts, and the up-regulated miR-3110-5p and miR-3058-3p in mechanically strained osteocytes, were both increased in the exosomes, which was verified via reverse transcription quantitative polymerase chain reaction (RT-qPCR). CONCLUSIONS: In osteocytes, a mechanical tensile strain of 2500 µÎµ at 0.5 Hz induced the fourteen differentially expressed miRNAs which probably were in exosomes of osteocytes and involved in osteogenesis. The mechanically strained osteocyte-derived exosomes which contained increased miR-3110-5p and miR-3058-3p (two of the 14 miRNAs), promoted osteoblastic differentiation.


Exosomes , MicroRNAs , Osteocytes , Osteogenesis , Stress, Mechanical , Animals , Mice , Cell Line , Exosomes/metabolism , Gene Expression Regulation , MicroRNAs/genetics , MicroRNAs/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Osteocytes/cytology , Osteocytes/metabolism , Osteogenesis/genetics
4.
Int J Mol Sci ; 25(9)2024 Apr 29.
Article En | MEDLINE | ID: mdl-38732057

Implant therapy is a common treatment option in dentistry and orthopedics, but its application is often associated with an increased risk of microbial contamination of the implant surfaces that cause bone tissue impairment. This study aims to develop two silver-enriched platelet-rich plasma (PRP) multifunctional scaffolds active at the same time in preventing implant-associated infections and stimulating bone regeneration. Commercial silver lactate (L) and newly synthesized silver deoxycholate:ß-Cyclodextrin (B), were studied in vitro. Initially, the antimicrobial activity of the two silver soluble forms and the PRP enriched with the two silver forms has been studied on microbial planktonic cells. At the same time, the biocompatibility of silver-enriched PRPs has been assessed by an MTT test on human primary osteoblasts (hOBs). Afterwards, an investigation was conducted to evaluate the activity of selected concentrations and forms of silver-enriched PRPs in inhibiting microbial biofilm formation and stimulating hOB differentiation. PRP-L (0.3 µg/mm2) and PRP-B (0.2 µg/mm2) counteract Staphylococcus aureus, Staphylococcus epidermidis and Candida albicans planktonic cell growth and biofilm formation, preserving hOB viability without interfering with their differentiation capability. Overall, the results obtained suggest that L- and B-enriched PRPs represent a promising preventive strategy against biofilm-related implant infections and demonstrate a new silver formulation that, together with increasing fibrin binding protecting silver in truncated cone-shaped cyclic oligosaccharides, achieved comparable inhibitory results on prokaryotic cells at a lower concentration.


Biofilms , Osteoblasts , Platelet-Rich Plasma , Silver , Humans , Biofilms/drug effects , Silver/chemistry , Silver/pharmacology , Osteoblasts/drug effects , Osteoblasts/cytology , Staphylococcus aureus/drug effects , Candida albicans/drug effects , Prosthesis-Related Infections/prevention & control , Prosthesis-Related Infections/microbiology , Staphylococcus epidermidis/drug effects
5.
Biomed Mater ; 19(4)2024 May 22.
Article En | MEDLINE | ID: mdl-38740037

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.


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
6.
Nanoscale ; 16(20): 9861-9874, 2024 May 23.
Article En | MEDLINE | ID: mdl-38712977

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


Anti-Bacterial Agents , Bone Regeneration , Chitosan , Durapatite , Nanofibers , Osteoblasts , Osteogenesis , Polyesters , Chitosan/chemistry , Chitosan/pharmacology , Durapatite/chemistry , Durapatite/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bone Regeneration/drug effects , Nanofibers/chemistry , Polyesters/chemistry , Polyesters/pharmacology , Animals , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteogenesis/drug effects , Mice , Tissue Scaffolds/chemistry , Gels/chemistry , Staphylococcus aureus/drug effects , Fibroblasts/drug effects , Fibroblasts/cytology
7.
Bull Exp Biol Med ; 176(5): 620-625, 2024 Mar.
Article En | MEDLINE | ID: mdl-38733480

We studied the interaction of human buccal mesenchymal stem cells (MSCs) and osteoblasts differentiated from them with the surface of titanium samples. MSCs were isolated by enzymatic method from buccal fat pads. The obtained cell culture was presented by MSCs, which was confirmed by flow cytometry and differentiation into adipocytes and osteoblasts. Culturing of buccal MSCs on titanium samples was accompanied by an increase in the number of cells for 15 days and the formation of a developed network of F-actin fibers in the cells. The viability of buccal MSCs decreased by 8 days, but was restored by 15 days. Culturing of osteoblasts obtained as a result of buccal MSC differentiation on the surface of titanium samples was accompanied by a decrease in their viability and proliferation. Thus, MSCs from buccal fat pads can be used to coat implants to improve osseointegration during bone reconstruction in craniofacial surgery and dentistry. To improve the integration of osteoblasts, modification of the surface of titanium samples is required.


Cell Differentiation , Mesenchymal Stem Cells , Osseointegration , Osteoblasts , Titanium , Titanium/chemistry , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Humans , Osseointegration/physiology , Osteoblasts/cytology , Osteoblasts/physiology , Cells, Cultured , Cell Proliferation , Dental Implants , Cell Survival , Adipocytes/cytology , Adipocytes/physiology , Mouth Mucosa/cytology , Osteogenesis/physiology
8.
J Biomed Mater Res B Appl Biomater ; 112(6): e35409, 2024 Jun.
Article En | MEDLINE | ID: mdl-38786580

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


Durapatite , Nanoparticles , Printing, Three-Dimensional , Tissue Engineering , Tissue Scaffolds , Tissue Scaffolds/chemistry , Durapatite/chemistry , Durapatite/pharmacology , Mice , Animals , Humans , Nanoparticles/chemistry , Cell Line , Polyesters/chemistry , Osteoblasts/metabolism , Osteoblasts/cytology , Osteogenesis/drug effects , Materials Testing
9.
Int J Biol Macromol ; 268(Pt 1): 131688, 2024 May.
Article En | MEDLINE | ID: mdl-38642688

Large bone defects, often resulting from trauma and disease, present significant clinical challenges. Electrospun fibrous scaffolds closely resembling the morphology and structure of natural ECM are highly interested in bone tissue engineering. However, the traditional electrospun fibrous scaffold has some limitations, including lacking interconnected macropores and behaving as a 2D scaffold. To address these challenges, a sponge-like electrospun poly(L-lactic acid) (PLLA)/polycaprolactone (PCL) fibrous scaffold has been developed by an innovative and convenient method (i.e., electrospinning, homogenization, progen leaching and shaping). The resulting scaffold exhibited a highly porous structure (overall porosity = 85.9 %) with interconnected, regular macropores, mimicking the natural extracellular matrix. Moreover, the incorporation of bioactive glass (BG) particles improved the hydrophilicity (water contact angle = 79.7°) and biocompatibility and promoted osteoblast cell growth. In-vitro 10-day experiment revealed that the scaffolds led to high cell viability. The increment of the proliferation rates was 195.4 % at day 7 and 281.6 % at day 10. More importantly, Saos-2 cells could grow, proliferate, and infiltrate into the scaffold. Therefore, this 3D PLLA/PCL with BG sponge holds great promise for bone defect repair in tissue engineering applications.


Bone and Bones , Polyesters , Tissue Engineering , Tissue Scaffolds , Tissue Scaffolds/chemistry , Polyesters/chemistry , Porosity , Humans , Tissue Engineering/methods , Bone and Bones/drug effects , Osteoblasts/drug effects , Osteoblasts/cytology , Cell Proliferation/drug effects , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cell Survival/drug effects
10.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 55(2): 256-262, 2024 Mar 20.
Article Zh | MEDLINE | ID: mdl-38645858

Runt-related transcription factor (RUNX1) is a transcription factor closely involved in hematopoiesis. RUNX1 gene mutation plays an essential pathogenic role in the initiation and development of hematological tumors, especially in acute myeloid leukemia. Recent studies have shown that RUNX1 is also involved in the regulation of bone development and the pathological progression of bone-related diseases. RUNX1 promotes the differentiation of mesenchymal stem cells into chondrocytes and osteoblasts and modulates the maturation and extracellular matrix formation of chondrocytes. The expression of RUNX1 in mesenchymal stem cells, chondrocytes, and osteoblasts is of great significance for maintaining normal bone development and the mass and quality of bones. RUNX1 also inhibits the differentiation and bone resorptive activities of osteoclasts, which may be influenced by sexual dimorphism. In addition, RUNX1 deficiency contributes to the pathogenesis of osteoarthritis, delayed fracture healing, and osteoporosis, which was revealed by the RUNX1 conditional knockout modeling in mice. However, the roles of RUNX1 in regulating the hypertrophic differentiation of chondrocytes, the sexual dimorphism of activities of osteoclasts, as well as bone loss in diabetes mellitus, senescence, infection, chronic inflammation, etc, are still not fully understood. This review provides a systematic summary of the research progress concerning RUNX1 in the field of bone biology, offering new ideas for using RUNX1 as a potential target for bone related diseases, especially osteoarthritis, delayed fracture healing, and osteoporosis.


Bone Development , Cell Differentiation , Chondrocytes , Core Binding Factor Alpha 2 Subunit , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Humans , Animals , Bone Development/physiology , Bone Development/genetics , Chondrocytes/metabolism , Osteoblasts/metabolism , Osteoblasts/cytology , Osteoclasts/metabolism , Osteoclasts/cytology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mice , Bone Diseases/genetics , Bone Diseases/metabolism , Osteoporosis/genetics , Osteoporosis/metabolism , Osteoarthritis/metabolism , Osteoarthritis/genetics , Osteoarthritis/etiology
11.
J Biomater Appl ; 38(10): 1100-1117, 2024 May.
Article En | MEDLINE | ID: mdl-38580320

The surface topological features of bioimplants are among the key indicators for bone tissue replacement because they directly affect cell morphology, adhesion, proliferation, and differentiation. In this study, we investigated the physical, electrochemical, and biological responses of sandblasted titanium (SB-Ti) surfaces with pore geometries fabricated using a plasma electrolytic oxidation (PEO) process. The PEO treatment was conducted at an applied voltage of 280 V in a solution bath consisting of 0.15 mol L-1 calcium acetate monohydrate and 0.02 mol L-1 calcium glycerophosphate for 3 min. The surface chemistry, wettability, mechanical properties and corrosion behavior of PEO-treated sandblasted Ti implants using hydroxyapatite particles (PEO-SB-Ti) were improved with the distribution of calcium phosphorous porous oxide layers, and showed a homogeneous and hierarchically porous surface with clusters of nanopores in a bath containing calcium acetate monohydrate and calcium glycerophosphate. To demonstrate the efficacy of PEO-SB-Ti, we investigated whether the implant affects biological responses. The proposed PEO-SB-Ti were evaluated with the aim of obtaining a multifunctional bone replacement model that could efficiently induce osteogenic differentiation as well as antibacterial activities. These physical and biological responses suggest that the PEO-SB-Ti may have a great potential for use an artificial bone replacement compared to that of the controls.


Durapatite , Oxidation-Reduction , Surface Properties , Titanium , Titanium/chemistry , Porosity , Durapatite/chemistry , Bone Screws , Animals , Wettability , Materials Testing , Osteogenesis/drug effects , Electrolysis , Plasma Gases/chemistry , Cell Differentiation/drug effects , Corrosion , Biocompatible Materials/chemistry , Osteoblasts/cytology , Mice
12.
Cell Metab ; 36(5): 1144-1163.e7, 2024 May 07.
Article En | MEDLINE | ID: mdl-38574738

Bone secretory proteins, termed osteokines, regulate bone metabolism and whole-body homeostasis. However, fundamental questions as to what the bona fide osteokines and their cellular sources are and how they are regulated remain unclear. In this study, we analyzed bone and extraskeletal tissues, osteoblast (OB) conditioned media, bone marrow supernatant (BMS), and serum, for basal osteokines and those responsive to aging and mechanical loading/unloading. We identified 375 candidate osteokines and their changes in response to aging and mechanical dynamics by integrating data from RNA-seq, scRNA-seq, and proteomic approaches. Furthermore, we analyzed their cellular sources in the bone and inter-organ communication facilitated by them (bone-brain, liver, and aorta). Notably, we discovered that senescent OBs secrete fatty-acid-binding protein 3 to propagate senescence toward vascular smooth muscle cells (VSMCs). Taken together, we identified previously unknown candidate osteokines and established a dynamic regulatory network among them, thus providing valuable resources to further investigate their systemic roles.


Osteoblasts , Animals , Osteoblasts/metabolism , Osteoblasts/cytology , Mice , Bone and Bones/metabolism , Proteomics , Mice, Inbred C57BL , Male , Aging/metabolism , Humans , Cellular Senescence , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/cytology , Multiomics
13.
Sci Rep ; 14(1): 9444, 2024 04 24.
Article En | MEDLINE | ID: mdl-38658667

One of the biggest challenges in tissue engineering and regenerative medicine is to ensure oxygen supply of cells in the (temporary) absence of vasculature. With the vision to exploit photosynthetic oxygen production by microalgae, co-cultivated in close vicinity to oxygen-consuming mammalian cells, we are searching for culture conditions that are compatible for both sides. Herein, we investigated the impact of long-term illumination on mammalian cells which is essential to enable photosynthesis by microalgae: four different cell types-primary human fibroblasts, dental pulp stem cells, and osteoblasts as well as the murine beta-cell line INS-1-were continuously exposed to warm white light, red or blue light over seven days. We observed that illumination with red light has no adverse effects on viability, metabolic activity and growth of the cells whereas exposure to white light has deleterious effects that can be attributed to its blue light portion. Quantification of intracellular glutathione did not reveal a clear correlation of this effect with an enhanced production of reactive oxygen species. Finally, our data indicate that the cytotoxic effect of short-wavelength light is predominantly a direct effect of cell illumination; photo-induced changes in the cell culture media play only a minor role.


Fibroblasts , Light , Reactive Oxygen Species , Humans , Animals , Fibroblasts/metabolism , Fibroblasts/radiation effects , Fibroblasts/cytology , Mice , Reactive Oxygen Species/metabolism , Cell Survival/radiation effects , Dental Pulp/cytology , Dental Pulp/radiation effects , Osteoblasts/metabolism , Osteoblasts/radiation effects , Osteoblasts/cytology , Cells, Cultured , Cell Line , Stem Cells/metabolism , Stem Cells/radiation effects , Stem Cells/cytology , Glutathione/metabolism
14.
Int J Mol Sci ; 25(8)2024 Apr 10.
Article En | MEDLINE | ID: mdl-38673767

The MC3T3-E1 preosteoblastic cell line is widely utilised as a reliable in vitro system to assess bone formation. However, the experimental growth conditions for these cells hugely diverge, and, particularly, the osteogenic medium (OSM)'s composition varies in research studies. Therefore, we aimed to define the ideal culture conditions for MC3T3-E1 subclone 4 cells with regard to their mineralization capacity and explore if oxidative stress or the cellular metabolism processes are implicated. Cells were treated with nine different combinations of long-lasting ascorbate (Asc) and ß-glycerophosphate (ßGP), and osteogenesis/calcification was evaluated at three different time-points by qPCR, Western blotting, and bone nodule staining. Key molecules of the oxidative and metabolic pathways were also assessed. It was found that sufficient mineral deposition was achieved only in the 150 µg.mL-1/2 mM Asc/ßGP combination on day 21 in OSM, and this was supported by Runx2, Alpl, Bglap, and Col1a1 expression level increases. NOX2 and SOD2 as well as PGC1α and Tfam were also monitored as indicators of redox and metabolic processes, respectively, where no differences were observed. Elevation in OCN protein levels and ALP activity showed that mineralisation comes as a result of these differences. This work defines the most appropriate culture conditions for MC3T3-E1 cells and could be used by other research laboratories in this field.


Energy Metabolism , Osteoblasts , Osteogenesis , Oxidative Stress , Animals , Mice , Osteogenesis/drug effects , Osteoblasts/metabolism , Osteoblasts/cytology , Cell Line , Glycerophosphates/metabolism , Glycerophosphates/pharmacology , Calcification, Physiologic , Cell Differentiation , Cell Culture Techniques/methods , Ascorbic Acid/pharmacology , Ascorbic Acid/metabolism , Culture Media/chemistry , Culture Media/pharmacology
15.
Int J Mol Sci ; 25(8)2024 Apr 11.
Article En | MEDLINE | ID: mdl-38673812

Here, we report on the development of a cost-effective, well-characterized three-dimensional (3D) model of bone homeostasis derived from commonly available stocks of immortalized murine cell lines and laboratory reagents. This 3D murine-cell-derived bone organoid model (3D-mcBOM) is adaptable to a range of contexts and can be used in conjunction with surrogates of osteoblast and osteoclast function to study cellular and molecular mechanisms that affect bone homeostasis in vitro or to augment in vivo models of physiology or disease. The 3D-mcBOM was established using a pre-osteoblast murine cell line, which was seeded into a hydrogel extracellular matrix (ECM) and differentiated into functional osteoblasts (OBs). The OBs mineralized the hydrogel ECM, leading to the deposition and consolidation of hydroxyapatite into bone-like organoids. Fourier-transform infrared (FTIR) spectroscopy confirmed that the mineralized matrix formed in the 3D-mcBOM was bone. The histological staining of 3D-mcBOM samples indicated a consistent rate of ECM mineralization. Type I collagen C-telopeptide (CTX1) analysis was used to evaluate the dynamics of OC differentiation and activity. Reliable 3D models of bone formation and homeostasis align with current ethical trends to reduce the use of animal models. This functional model of bone homeostasis provides a cost-effective model system using immortalized cell lines and easily procured supplemental compounds, which can be assessed by measuring surrogates of OB and OC function to study the effects of various stimuli in future experimental evaluations of bone homeostasis.


Cell Differentiation , Extracellular Matrix , Organoids , Osteoblasts , Osteogenesis , Animals , Mice , Organoids/cytology , Organoids/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Extracellular Matrix/metabolism , Bone and Bones/cytology , Bone and Bones/metabolism , Cell Line , Collagen Type I/metabolism , Hydrogels/chemistry , Calcification, Physiologic , Cell Culture Techniques, Three Dimensional/methods , Models, Biological
16.
Int J Biol Macromol ; 267(Pt 2): 131412, 2024 May.
Article En | MEDLINE | ID: mdl-38593894

The synthesis of ideal bioceramics to guide the fate of cells and subsequent bone regeneration within the chemical, biological, and physical microenvironment is a challenging long-term task. This study developed amorphous calcium magnesium phosphate (ACMP) bioceramics via a simple co-precipitation method. The role of Mg2+ in the formation of ACMP is investigated using physicochemical and biological characterization at different Ca/Mg molar ratio of the initial reaction solution. Additionally, ACMP bioceramics show superior cytocompatibility and improved osteogenic differentiation of co-cultured MC3T3-E1 cells. Regulation of the microenvironment with Mg2+ can promote early-stage bone regeneration. For this, bioprinting technology is employed to prepare ACMP-modified 3D porous structures. Our hypothesis is that the incorporation of ACMP into methacrylated gelatin (GelMA) bioink can trigger the osteogenic differentiation of encapsulated preosteoblast and stimulate bone regeneration. The cell-laden ACMP composite structures display stable printability and superior cell viability and cell proliferation. Also, constructs loading the appropriate amount of ACMP bioceramic showed significant osteogenic differentiation activity compared to the pure GelMA. We demonstrate that the dissolved Mg2+ cation microenvironment in ACMP-modified composite constructs plays an effective biochemical role, and can regulate cell fate. Our results predict that GelMA/ACMP bioink has significant potential in patient-specific bone tissue regeneration.


Bioprinting , Bone Regeneration , Calcium Phosphates , Cell Differentiation , Osteogenesis , Printing, Three-Dimensional , Tissue Scaffolds , Bone Regeneration/drug effects , Mice , Animals , Osteogenesis/drug effects , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacology , Cell Differentiation/drug effects , Bioprinting/methods , Tissue Scaffolds/chemistry , Cell Proliferation/drug effects , Magnesium Compounds/chemistry , Magnesium Compounds/pharmacology , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cell Survival/drug effects , Cell Line , Tissue Engineering/methods , Osteoblasts/drug effects , Osteoblasts/cytology , Phosphates/chemistry , Phosphates/pharmacology
17.
Int J Biol Macromol ; 267(Pt 2): 131519, 2024 May.
Article En | MEDLINE | ID: mdl-38608985

Hydrogel has attracted tremendous attentions due to its excellent biocompatibility and adaptability in biomedical field. However, it is challenging by the conflicts between inadequate mechanical properties and service requirements. Herein, a rapid and robust hydrogel was developed by interpenetrating networks between chitosan and silk fibroin macromolecules. Thanks to these unique networks, the chitosan-based hydrogel exhibited superior mechanical performances. The maximum breaking strength, Young's modulus and swelling ratio of the hydrogel were 1187.8 kPa, 383.1 MPa and 4.5 % respectively. The hydrogel also supported the proliferation of human umbilical vein endothelial cells for 7 days. Notably, the hydrogel was easily molded into bone screw, and demonstrated compressive strengths of 45.7 MPa, Young's modulus of 675.6 MPa, respectively. After 49-day biodegradation, the residual rate of the screw in collagenase I solution was up to 89.6 % of the initial weight. In vitro, the screws not only had high resistance to biodegradation, but also had outstanding biocompatibility of osteoblast. This study provided a promising physical-chemical double crosslinking strategy to build orthopedic materials, holding a great potential in biomedical devices.


Biocompatible Materials , Bone Screws , Chitosan , Fibroins , Human Umbilical Vein Endothelial Cells , Materials Testing , Chitosan/chemistry , Chitosan/pharmacology , Fibroins/chemistry , Humans , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Hydrogels/chemistry , Cell Proliferation/drug effects , Osteoblasts/drug effects , Osteoblasts/cytology , Compressive Strength , Elastic Modulus
18.
Biomed Mater ; 19(3)2024 Apr 26.
Article En | MEDLINE | ID: mdl-38626780

Wool derived keratin, due to its demonstrated ability to promote bone formation, has been suggested as a potential bioactive material for implant surfaces. The aim of this study was to assess the effects of keratin-coated titanium on osteoblast functionin vitroand bone healingin vivo. Keratin-coated titanium surfaces were fabricated via solvent casting and molecular grafting. The effect of these surfaces on the attachment, osteogenic gene, and osteogenic protein expression of MG-63 osteoblast-like cells were quantifiedin vitro. The effect of these keratin-modified surfaces on bone healing over three weeks using an intraosseous calvaria defect was assessed in rodents. Keratin coating did not affect MG-63 proliferation or viability, but enhanced osteopontin, osteocalcin and bone morphogenetic expressionin vitro. Histological analysis of recovered calvaria specimens showed osseous defects covered with keratin-coated titanium had a higher percentage of new bone area two weeks after implantation compared to that in defects covered with titanium alone. The keratin-coated surfaces were biocompatible and stimulated osteogenic expression in adherent MG-63 osteoblasts. Furthermore, a pilot preclinical study in rodents suggested keratin may stimulate earlier intraosseous calvaria bone healing.


Bone Regeneration , Cell Proliferation , Coated Materials, Biocompatible , Keratins , Osteoblasts , Osteogenesis , Skull , Titanium , Titanium/chemistry , Osteoblasts/drug effects , Osteoblasts/cytology , Osteoblasts/metabolism , Bone Regeneration/drug effects , Animals , Keratins/chemistry , Keratins/metabolism , Humans , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Cell Proliferation/drug effects , Skull/drug effects , Skull/injuries , Osteogenesis/drug effects , Rats , Surface Properties , Male , Cell Line , Cell Adhesion/drug effects , Materials Testing , Cell Survival/drug effects , Rats, Sprague-Dawley
19.
Int J Mol Sci ; 25(8)2024 Apr 21.
Article En | MEDLINE | ID: mdl-38674127

Platelet-rich fibrin (PRF) is a widely used autologous blood concentrate in regenerative medicine. This study aimed to characterize the cellular composition and distribution of different PRF matrices generated by high (710 g) and low (44 g) relative centrifugal forces (RCFs) and to analyze their bioactivity on human primary osteoblasts (pOBs). PRF was separated into upper layer (UL) and buffy coat (BC) fractions, and their cellular contents were assessed using histological and immunohistochemical staining. The release of platelet-derived growth factor (PDGF) and transforming growth factor (TGF-ß) was quantified using an ELISA. Indirect PRF treatment on pOBs was performed to evaluate cell viability and morphology. A histological analysis revealed higher quantities of leukocytes and platelets in the low-RCF PRF. TGF-ß release was significantly higher in the low-RCF PRF compared to the high-RCF PRF. All PRF fractions promoted pOB proliferation regardless of the centrifugation protocol used. The low-RCF PRF showed higher TGF-ß levels than the high-RCF PRF. These findings contribute to understanding the cellular mechanisms of PRF and provide insights into optimizing PRF protocols for bone regeneration, advancing regenerative medicine, and improving patient outcomes.


Cell Proliferation , Leukocytes , Osteoblasts , Platelet-Rich Fibrin , Humans , Osteoblasts/cytology , Osteoblasts/metabolism , Platelet-Rich Fibrin/metabolism , Leukocytes/metabolism , Leukocytes/cytology , Cells, Cultured , Transforming Growth Factor beta/metabolism , Cell Survival , Platelet-Derived Growth Factor/metabolism
20.
Bone ; 184: 117090, 2024 Jul.
Article En | MEDLINE | ID: mdl-38579924

Mechanical properties are becoming fundamental for advancing the comprehension of cellular processes. This study addresses the relationship between viscoelastic properties and the cellular mineralization process. Osteoblast-like cells treated with an osteogenic medium were employed for this purpose. Additionally, the study explores the impact of hydroxyapatite (HA) and hydroxyapatite/silver (HA/Ag) composite on this process. AFM relaxation experiments were conducted to extract viscoelastic parameters using the Fractional Zener (FZ) and Fractional Kelvin (FK) models. Our findings revealed that the main phases of mineralization are associated with alterations in the viscoelastic properties of osteoblast-like cells. Furthermore, HA and HA/Ag treatments significantly influenced changes in the viscoelastic properties of these cells. In particular, the HA/Ag treatment demonstrated a marked enhancement in cell fluidity, suggesting a possible role of silver in accelerating the mineralization process. Moreover, the study underscores the independence observed between fluidity and stiffness, indicating that modifications in one parameter may not necessarily correspond to changes in the other. These findings shed light on the factors involved in the cellular mineralization process and emphasize the importance of using viscoelastic properties to discern the impact of treatments on cells.


Calcification, Physiologic , Durapatite , Elasticity , Osteoblasts , Silver , Durapatite/chemistry , Osteoblasts/metabolism , Osteoblasts/drug effects , Osteoblasts/cytology , Silver/chemistry , Calcification, Physiologic/physiology , Calcification, Physiologic/drug effects , Viscosity , Cell Line , Humans , Microscopy, Atomic Force , Animals
...