Recombinant Unique Cartilage Matrix-associated Protein Potentiates Osteogenic Differentiation and Mineralization of MC3T3-E1 Cells

2021 ◽  
Vol 21 ◽  
Author(s):  
Yeon-Ju Lee ◽  
Hyeon Yeong Ju ◽  
Seung-Yoon Park ◽  
Hye Jung Ihn ◽  
Eui Kyun Park ◽  
...  

Objective: The relative balance of osteoblasts in bone formation and osteoclasts in bone resorption is crucial for maintaining bone health. With age, this balance between osteoblasts and osteoclasts is broken, resulting in bone loss. Anabolic drugs are continuously being developed to counteract this low bone mass. Recombinant proteins are used as biotherapeutics due to being relatively easy to produce on a large scale and are cost-effective through various expression systems. This study aimed to develop a recombinant protein that would positively impact osteoblast differentiation and mineralized nodule formation using unique cartilage matrix-associated protein (UCMA). Methods: A recombinant glutathione-S-transferase (GST)-UCMA fusion protein was generated in an E.coli system, and purified by affinity chromatography. MC3T3-E1 osteoblast cells and Osterix (Osx)-knockdown stable cells were cultured for 14 days to investigate osteoblast differentiation and nodule formation in the presence of the recombinant GST-UCMA protein. The differentiated cells were assessed by alizarin red S staining and quantitative PCR of the osteoblast differentiation marker osteocalcin. In addition, cell viability in the presence of the recombinant GST-UCMA protein was determined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and cell adhesion assay. Results: The isolation of both purified recombinant GST-only and GST-UCMA proteins were confirmed at 26 kDa and 34 kDa, respectively, by Coomassie staining and western blot analysis. Neither dose-dependent nor time-dependent presence of recombinant GSTUCMA affected MC3T3-E1 cell viability. However, MC3T3-E1 cell adhesion to the recombinant GST-UCMA protein increased dose-dependently. Osteoblast differentiation and nodule formation were promoted in both MC3T3-E1 osteoblast cells and Osxknockdown stable cells when cultured in the presence of recombinant GST-UCMA protein. Conclusion: A recombinant GST-UCMA protein induces osteogenic differentiation and mineralization, suggesting its potential use as an anabolic drug to increase low bone mass in osteoporotic patients.

2015 ◽  
Vol 1721 ◽  
Author(s):  
Maraolina Domínguez-Díaz ◽  
Angelica Meneses-Acosta ◽  
Angel Romo-Uribe

ABSTRACTBiodegradable Normal Human Osteoblast (NHOst) cells were inoculated into the polymer scaffolds of poly(β-hydroxybutyrate) (PHB) obtained from a specially developed strain of Azotobacter vinelandii. Cell adhesion is essential to promote growth on scaffolds for tissue engineering. Thus, in this research we focused on the adhesion of osteoblast cells to PHB scaffolds produced by solution casting and electrospinning. Cell viability was also investigated up to 168 hrs. Water contact angle on the PHB scaffolds was determined prior to the cells inoculation. The contact angle is usually related to the ability of different cell strains to adhere to a given material. The as cast film exhibited a contact angle α=72° whereas for the electrospun membrane α=102°, thus in theory cell adhesion would be greater for the cast film. Biological testing was carried out on plates of 24 wells; cell viability was determined by Trypan Blue, cell morphology by optical microscopy, and cell nuclei integrity by staining with Acridine orange. Parallel studies were carried out on control (empty) wells. Microscopy observations 168 hrs after cell inoculation showed larger quantities of osteoblast cells in the wells containing PHB scaffolds and the cell nuclei were still active. Moreover, it was found that the cells grew inside the PHB scaffolds and the cell viability was slightly greater for the electrospun scaffold. Interestingly, the time to remove the cells from the scaffolds (film and membranes) was increasing function of the cell culture time, therefore suggesting that PHB promotes adhesion of Normal Human Osteoblast cells to its surface.


2020 ◽  
Vol 11 (1) ◽  
Author(s):  
Dan Zhang ◽  
Kim De Veirman ◽  
Rong Fan ◽  
Qiang Jian ◽  
Yuchen Zhang ◽  
...  

Abstract Background Bone destruction is a hallmark of multiple myeloma (MM). It has been reported that proteasome inhibitors (PIs) can reduce bone resorption and increase bone formation in MM patients, but the underlying mechanisms remain unclear. Methods Mesenchymal stem cells (MSCs) were treated with various doses of PIs, and the effects of bortezomib or carfilzomib on endoplasmic reticulum (ER) stress signaling pathways were analyzed by western blotting and real-time PCR. Alizarin red S (ARS) and alkaline phosphatase (ALP) staining were used to determine the osteogenic differentiation in vitro. Specific inhibitors targeting different ER stress signaling and a Tet-on inducible overexpressing system were used to validate the roles of key ER stress components in regulating osteogenic differentiation of MSCs. Chromatin immunoprecipitation (ChIP) assay was used to evaluate transcription factor-promoter interaction. MicroCT was applied to measure the microarchitecture of bone in model mice in vivo. Results We found that both PERK-ATF4 and IRE1α-XBP1s ER stress branches are activated during PI-induced osteogenic differentiation. Inhibition of ATF4 or XBP1s signaling can significantly impair PI-induced osteogenic differentiation. Furthermore, we demonstrated that XBP1s can transcriptionally upregulate ATF4 expression and overexpressing XBP1s can induce the expression of ATF4 and other osteogenic differentiation-related genes and therefore drive osteoblast differentiation. MicroCT analysis further demonstrated that inhibition of XBP1s can strikingly abolish bortezomib-induced bone formation in mouse. Conclusions These results demonstrated that XBP1s is a master regulator of PI-induced osteoblast differentiation. Activation of IRE1α-XBP1s ER stress signaling can promote osteogenesis, thus providing a novel strategy for the treatment of myeloma bone disease.


Author(s):  
Naruemon Panpradit ◽  
Thanapoj Nilmoje ◽  
Julalux Kasetsuwan ◽  
Sujiwan Seubbuk Sangkhamanee ◽  
Rudee Surarit

Abstract Objectives This study aims to evaluate the effect of Porphyromonas gingivalis and nicotine on the in vitro osteogenic differentiation of periodontal ligament (PDL) fibroblasts. Materials and Methods PDLs were cultured in Dulbecco’s modified Eagle’s medium containing 10% fetal bovine serum at 37°C under 5% CO2 and 100% humidified atmosphere. Cells were incubated with various concentrations of nicotine and P. gingivalis extracts, and cell viability was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay. To study cell differentiation, PDLs (5 × 104cells) were treated with the osteogenic differentiation medium containing 10 mM β-glycerophosphate, 10 nM dexamethasone, 50 mg/mL ascorbic acid, 1 μM nicotine, and 50 µg/mL P. gingivalis lysate. mRNA samples were collected at 0, 7, and 14 days. Odontogenic-related gene expression, namely, Runt-related transcription factor 2 (Runx2), collagen type I (COL1A1), and alkaline phosphatase (ALP) was determined by reverse transcription quantitative polymerase chain reaction (RT-qPCR). Calcified nodule formation was determined on day 28 using Alizarin Red S. Analysis of variance and Tukey’s test were used to compare the difference among groups at significant level of p < 0.05. Results It showed that 50 µg/mL of P. gingivalis lysate and 1 µM of nicotine showed no toxicity to PDLs. Runx2, COL1A1, and ALP expression were found to decrease significantly after 7 days of treatment, while osteocalcin expression was found to decrease after 14 days. The nodule formation in the control group was much greater in both number and size of nodules than in experimental groups, which implied a positive sign of calcium deposition in controls. Conclusion The results indicated that nicotine and P. gingivalis showed adverse effect on osteogenic differentiation properties of PDLs.


2020 ◽  
Author(s):  
Meiling Liu ◽  
Haifeng Ding ◽  
Hongzhi Wang ◽  
Manfeng Wang ◽  
Xiaowei Wu ◽  
...  

Abstract Objective: We aimed to investigate the therapeutic effects of Moringa oleifera leaf extracts on osteogenic induction of rat bone marrow mesenchymal stem cells (BMSCs) following peroxidative damage and to explore the underlying mechanisms. Methods: Conditioned medium was used to induce osteogenic differentiation of BMSCs, which were treated with H2O2, Moringa oleifera leaf extracts-containing serum, or the phosphatidyl inositol-3 kinase (PI3K) inhibitor Wortmannin, alone or in combination. Cell viability was measured using the MTT assay. Cell cycle was assayed using flow cytometry. Expression levels of Akt, phosphorylated (p)Akt, Foxo1, and cleaved caspase-3 were analyzed using Western blot analysis. The mRNA levels of osteogenesis-associated genes, including alkaline phosphatase (ALP), collagen І, osteopontin (OPN), and Runx2, were detected using qRT-PCR. Reactive oxygen species (ROS) and malondialdehyde (MDA) levels, as well as superoxide dismutase (SOD), glutathione peroxidase (GSH-PX), and ALP activity were detected using commercially available kits. Osteogenic differentiation capability was determined using alizarin red staining. Results: During osteogenic induction of rat BMSCs, H2O2 reduced cell viability and proliferation, inhibited osteogenesis, increased ROS and MDA levels, and decreased SOD and GSH-PX activity. H2O2 significantly reduced pAkt and Foxo1 expression, and increased cleaved caspase-3 levels in BMSCs. Additional treatments with Moringa oleifera leaf extracts partially reversed the H2O2-induced changes. Wortmannin partially attenuated the effects of Moringa oleifera leaf extracts on protein expression of Foxo1, pAkt, and cleaved caspase-3, as well as mRNA levels of osteogenesis-associated genes.Conclusion: Moringa oleifera leaf extracts ameliorate peroxidative damage and enhance osteogenic induction of rat BMSCs by activating the PI3K/Akt/Foxo1 pathway.


2021 ◽  
Vol 12 ◽  
Author(s):  
Jae-Hyun Kim ◽  
Minsun Kim ◽  
SooYeon Hong ◽  
Eun-Young Kim ◽  
Hyangsook Lee ◽  
...  

Fracture healing is related to osteogenic differentiation and mineralization. Recently, due to the unwanted side effects and clinical limitations of existing treatments, various natural product-based chemical studies have been actively conducted. Albiflorin is a major ingredient in Paeonia lactiflora, and this study investigated its ability to promote osteogenic differentiation and fracture healing. To demonstrate the effects of albiflorin on osteoblast differentiation and calcified nodules, alizarin red S staining and von Kossa staining were used in MC3T3-E1 cells. In addition, BMP-2/Smad and Wnt/β-catenin mechanisms known as osteoblast differentiation mechanisms were analyzed through RT-PCR and western blot. To investigate the effects of albiflorin on fracture healing, fractures were induced using a chainsaw in the femur of Sprague Dawley rats, and then albiflorin was intraperitoneally administered. After 1, 2, and 3 weeks, bone microstructure was analyzed using micro-CT. In addition, histological analysis was performed by staining the fractured tissue, and the expression of osteogenic markers in serum was measured. The results demonstrated that albiflorin promoted osteoblastogenesis and the expression of RUNX2 by activating BMP-2/Smad and Wnt/β-catenin signaling in MC3T3-E1 cells. In addition, albiflorin upregulated the expression of various osteogenic genes, such as alkaline phosphatase, OCN, bone sialoprotein, OPN, and OSN. In the femur fracture model, micro-CT analysis showed that albiflorin played a positive role in the formation of callus in the early stage of fracture recovery, and histological examination proved to induce the expression of osteogenic genes in femur tissue. In addition, the expression of bone-related genes in serum was also increased. This suggests that albiflorin promotes osteogenesis, bone calcification and bone formation, thereby promoting the healing of fractures in rats.


2021 ◽  
Vol 22 (22) ◽  
pp. 12259
Author(s):  
Alireza Valanezhad ◽  
Tetsurou Odatsu ◽  
Shigeaki Abe ◽  
Ikuya Watanabe

Recently, ferroptosis has gained scientists’ attention as an iron-related regulated necrosis. However, not many reports have investigated the effect of ferroptosis on bone. Therefore, with the present study, we assessed the effect of ferroptosis inhibition using ferrostatin-1 on the MC3T3-E1 pre-osteoblast cell. Cell images, cell viability, alkaline phosphatase activity test, alizarin red staining, and RUNX2 gene expression using real-time PCR were applied to investigate the effects of ferrostatin and erastin on MC3T3-E1 osteoblast cells. Erastin was used as a well-known ferroptosis inducer reagent. Erastin with different concentrations ranging from 0 to 50 µmol/L was used for inducing cell death. The 25 µmol/L erastin led to controllable partial cell death on osteoblast cells. Ferrostatin-1 with 0 to 40 µmol/L was used for cell doping and cell death inhibition effect. Ferrostatin-1 also displayed a recovery effect on the samples, which had already received the partially artificial cell death by erastin. Cell differentiation, alizarin red staining, and RUNX2 gene expression confirmed the promotion of the bone formation ability effect of ferrostatin-1 on osteoblast cells. The objective of this study was to assess ferrostatin-1’s effect on the MC3T3-E1 osteoblast cell line based on its ferroptosis inhibitory property.


Author(s):  
Dongdong Fei ◽  
Yanmin Xia ◽  
Qiming Zhai ◽  
Yazheng Wang ◽  
Feng Zhou ◽  
...  

Mesenchymal stem cells (MSCs) are intrinsically heterogeneous and are comprised of distinct subpopulations that differ in their differentiation potential. A deeper understanding of the heterogeneity and intercellular communication within these heterogeneous subpopulations has significant implications for the potential of MSC-based therapy from the bench to the clinic. Here, we focused on the clonal osteogenic heterogeneity of periodontal ligament stem cells (PDLSCs) and explored how interclonal communication affects the osteogenic differentiation among these heterogeneous single-cell colonies (SCCs), and sought to determine the underlying mechanisms. Alkaline phosphatase (ALP) and Alizarin red staining identified the presence of SCCs with high (H-SCCs) and low osteogenic ability (L-SCCs). Conditioned medium derived from H-SCCs (H-CM) promoted mineralized nodule formation to a greater extent than that derived from L-SCCs (L-CM), which served as the target cells (TCs). However, treatment with the exosome biogenesis/release inhibitor GW4869 reduced the H-CM- and L-CM-related osteogenic differentiation-promoting potential. We further found that exosomes secreted by H-SCCs (H-Exo) were superior to those secreted by L-SCCs (L-Exo) in promoting the osteogenic differentiation of TCs. Mechanistically, TCs stimulated with H-CM and H-Exo exhibited higher levels of PINK1/Parkin-mediated mitophagy, while gain- and loss-of-function experiments showed that PINK1/Parkin-mediated mitophagy was positively associated with SCC osteogenic differentiation. Furthermore, PINK1 knock-down in H-Exo- and L-Exo-stimulated TCs inhibited their osteogenic differentiation through inhibiting PINK1/Parkin-mediated mitophagy. Our study uncovers a previously unrecognized mechanism that an exosome-mediated PINK1/Parkin-dependent mitophagy regulates interclonal communication among SCCs with osteogenic heterogeneity.


2020 ◽  
Author(s):  
Zhi Peng ◽  
Zhenkai Lou ◽  
Zhongjie Li ◽  
Shaobo Li ◽  
Kaishun Yang ◽  
...  

Abstract Background: Osteoporosis is the most common bone metabolic disease. Emerging evidence suggests that exosomes are secreted by diverse cells such as bone marrow mesenchymal stem cells (BMSCs), and play important role in cell-to-cell communication and tissue homeostasis. Recently, the discovery of exosomes has attracted attention in the field of bone remodeling. Methods: The exosomes were extracted from BMSCs and labeled by PKH-67, and then incubated with hFOB1.19 cells to investigate the miR-196a function on the osteoblast differentiation of hFOB1.19. The osteoblast differentiation was detected via alizarin red staining and the expression of osteoblast genes were detected by western blot. The cell apoptosis was detected by flow cytometer. The target relationship of miR-196a and Dickkopf-1 (Dkk1) were verified by luciferase assay and western blot. Results: Here we demonstrated that exosomes extracted from BMSCs (BMSC-exo) significantly promoted hFOB1.19 differentiation to osteoblasts. We found that BMSC-exo were enriched with miR-196a and delivered miR-196a to hFOB1.19 cells to inhibit its target Dkk1, which is a negative regulator of Wnt/β-catenin pathway. Conclusion: BMSC-exo activated Wnt/β-catenin pathway to promote osteogenic differentiation, while BMSC-exo failed to exert the effects when miR-196a was deprived. In conclusion, miR-196a delivered by exosomes from BMSCs plays an essential role in enhancing osteoblastic differentiation by targeting Dkk1 to activate Wnt/β-catenin pathway.


2019 ◽  
Vol 2019 ◽  
pp. 1-10 ◽  
Author(s):  
Rita Bou Assaf ◽  
Mohammad Fayyad-Kazan ◽  
Fatima Al-Nemer ◽  
Rawan Makki ◽  
Hussein Fayyad-Kazan ◽  
...  

Background. Novel treatments for bone defects, particularly in patients with poor regenerative capacity, are based on bone tissue engineering strategies which include mesenchymal stem cells (MSCs), bioactive factors, and convenient scaffold supports. Objective. In this study, we aimed at comparing the potential for different scaffolds to induce osteogenic differentiation of human maxillary Schneiderian sinus membrane- (hMSSM-) derived cells. Methods. hMSSM-derived cells were seeded on gelatin, collagen, or Hydroxyapatite β-Tricalcium phosphate-Fibrin (Haβ-TCP-Fibrin) scaffolds. Cell viability was determined using an MTT assay. Alizarin red staining method, Alkaline phosphatase (ALP) activity assay, and quantitative real-time PCR analysis were performed to assess hMSSM-derived cells osteogenic differentiation. Results. Cell viability, calcium deposition, ALP activity, and osteoblastic markers transcription levels were most striking in gelatin scaffold-embedded hMSSM-derived cells. Conclusion. Our findings suggest a promising potential for gelatin-hMSSM-derived cell construct for treating bone defects.


2021 ◽  
Vol 27 (1) ◽  
Author(s):  
Li-Rong Ren ◽  
Ru-Bin Yao ◽  
Shi-Yong Wang ◽  
Xiang-Dong Gong ◽  
Ji-Tao Xu ◽  
...  

Abstract Background Osteoporosis seriously disturbs the life of people. Meanwhile, inhibition or weakening of osteogenic differentiation is one of the important factors in the pathogenesis of osteoporosis. It was reported that miR-27a-3p reduced the symptoms of osteoporosis. However, the mechanism by which miR-27a-3p in osteogenic differentiation remains largely unknown. Methods To induce the osteogenic differentiation in MC3T3-E1 cells, cells were treated with osteogenic induction medium (OIM). RT-qPCR was used to evaluate the mRNA expression of miR-27a-3p and CRY2 in cells. The protein levels of CRY2, Runt-related transcription factor 2 (Runx2), osteopontin (OPN), osteocalcin (OCN) and the phosphorylation level of extracellular regulated protein kinases (ERK) 1/2 in MC3T3-E1 cells were evaluated by western blotting. Meanwhile, calcium nodules and ALP activity were tested by alizarin red staining and ALP kit, respectively. Luciferase reporter gene assay was used to analyze the correlation between CRY2 and miR-27a-3p. Results The expression of miR-27a-3p and the phosphorylation level of ERK1/2 were increased by OIM in MC3T3-E1 cells, while CRY2 expression was decreased. In addition, OIM-induced increase of calcified nodules, ALP content and osteogenesis-related protein expression was significantly reversed by downregulation of miR-27a-3p and overexpression of CRY2. In addition, miR-27a-3p directly targeted CRY2 and negatively regulated CRY2. Meanwhile, the inhibitory effect of miR-27a-3p inhibitor on osteogenic differentiation was reversed by knockdown of CRY2 or using honokiol (ERK1/2 signal activator). Furthermore, miR-27a-3p significantly inhibited the apoptosis of MC3T3-E1 cells treated by OIM. Taken together, miR-27a-3p/CRY2/ERK axis plays an important role in osteoblast differentiation. Conclusions MiR-27a-3p promoted osteoblast differentiation via mediation of CRY2/ERK1/2 axis. Thereby, miR-27a-3p might serve as a new target for the treatment of osteoporosis.


Sign in / Sign up

Export Citation Format

Share Document