scholarly journals Effect of Azithromycin on Mineralized Nodule Formation in MC3T3-E1 Cells

2021 ◽  
Vol 43 (3) ◽  
pp. 1451-1459
Author(s):  
Kengo Kato ◽  
Manami Ozaki ◽  
Kumiko Nakai ◽  
Maki Nagasaki ◽  
Junya Nakajima ◽  
...  

Azithromycin displays immunomodulatory and anti-inflammatory effects in addition to broad-spectrum antimicrobial activity and is used to treat inflammatory diseases, including respiratory and odontogenic infections. Few studies have reported the effect of azithromycin therapy on bone remodeling processes. The aim of this study was to examine the effects of azithromycin on the osteogenic function of osteoblasts using osteoblast-like MC3T3-E1 cells. Cells were cultured in the presence of 0, 0.1, 1, and 10 µg/mL azithromycin, and cell proliferation and alkaline phosphatase (ALPase) activity were determined. In vitro mineralized nodule formation was detected with alizarin red staining. The expression of collagenous and non-collagenous bone matrix protein was determined using real-time PCR or enzyme-linked immunosorbent assays. In cells cultured with 10 µg/mL azithromycin, the ALPase activity and mineralized nodule formation decreased, while the type I collagen, bone sialoprotein, osteocalcin, and osteopontin mRNA expression as well as osteopontin and phosphorylated osteopontin levels increased. These results suggest that a high azithromycin concentration (10 µg/mL) suppresses mineralized nodule formation by decreasing ALPase activity and increasing osteopontin production, whereas low concentrations (≤l.0 µg/mL) have no effect on osteogenic function in osteoblastic MC3T3-E1 cells.

1992 ◽  
Vol 12 (5) ◽  
pp. 407-411 ◽  
Author(s):  
Östen Ljunggren ◽  
Sverker Ljunghall

Bone resorption, in vitro, is often measured as the release of prelabelled45Ca from neonatal mouse calvarial bones, or from fetal rat long bones. In this report we describe a technique to measure the breakdown of bone-matrix, in vitro. We also describe a new way to dissect neonatal mouse calvarial bones, in order to obtain large amounts of bone samples. Twelve bone fragments were dissected out from each mouse calvaria and were thereafter cultured in CMRL 1066 culture medium in serum-free conditions in 0.5 cm2 multiwell culture dishes. Matrix degradation after treatment with parathyroid hormone was assessed by measuring the amount of carboxyterminal telopeptide of type I collagen (ICTP) by RIA. The data on matrix degradation was compared to the release of prelabelled45Ca from neonatal mouse calvarial bones. We found that the dose-responses for parathyroid hormone-induced release of prelabelled45Ca and ICTP were identical. In conclusion: RIA-analysis of the ICTP-release is an easy and accurate method to measure degradation of bone-matrix, in vitro. Furthermore, the new dissection technique, described in this report, makes it easy to obtain large amounts of bone samples and thus to perform extensive experiments, e.g. dose-responses for agents that enhance bone resorption.


2001 ◽  
Vol 281 (1) ◽  
pp. L164-L171 ◽  
Author(s):  
Tadashi Mio ◽  
Xiangde Liu ◽  
Myron L. Toews ◽  
Yuichi Adachi ◽  
Debra J. Romberger ◽  
...  

Bradykinin is a multifunctional mediator of inflammation believed to have a role in asthma, a disorder associated with remodeling of extracellular connective tissue. Using contraction of collagen gels as an in vitro model of wound contraction, we assessed the effects of bradykinin tissue on remodeling. Human fetal lung fibroblasts were embedded in type I collagen gels and cultured for 5 days. After release, the floating gels were cultured in the presence of bradykinin. Bradykinin significantly stimulated contraction in a concentration- and time-dependent manner. Coincubation with phosphoramidon augmented the effect of 10−9 and 10−8 M bradykinin. A B2 receptor antagonist attenuated the effect of bradykinin, whereas a B1 receptor antagonist had no effect, suggesting that the effect is mediated by the B2 receptor. An inhibitor of intracellular Ca2+mobilization abolished the response; addition of EGTA to the culture medium attenuated the contraction of control gels but did not modulate the response to bradykinin. In contrast, the phospholipase C inhibitor U-73122 and the protein kinase C inhibitors staurosporine and GF-109203X attenuated the responses. These data suggest that by augmenting the contractility of fibroblasts, bradykinin may have an important role in remodeling of extracellular matrix that may result in tissue dysfunction in chronic inflammatory diseases, such as asthma.


2003 ◽  
Vol 82 (1) ◽  
pp. 23-27 ◽  
Author(s):  
M. Zhao ◽  
J.E. Berry ◽  
M.J. Somerman

As an approach for improving the outcome and predictability of periodontal regenerative therapies, we have focused on determining the responses of cells within the local environment to putative regenerative factors. This study examined the effects of bone morphogenetic protein-2 (BMP-2) on murine cementoblasts in vitro. Northern blot analysis indicated that BMP-2 decreased mRNA levels of bone sialoprotein and type I collagen dose-dependently (10–300 ng/mL). At low doses, up to 100 ng/mL, BMP-2 had no effect on transcripts for osteocalcin and osteopontin, whereas at 300 ng/mL, BMP-2 greatly increased expression of these two genes. BMP-2 also inhibited cementoblast-mediated mineral nodule formation in a dose-dependent manner (inhibition was noted at 10 ng/mL). Noggin reversed the effects of BMP-2 on gene expression and on mineralization. These findings reflect the diverse responses of periodontal cells to BMP-2 and highlight the need to consider the complexity of factors involved in designing predictable regenerative therapies.


2013 ◽  
Vol 2013 ◽  
pp. 1-13 ◽  
Author(s):  
Yusuke Nakagawa ◽  
Takeshi Muneta ◽  
Kunikazu Tsuji ◽  
Shizuko Ichinose ◽  
Yasuharu Hakamatsuka ◽  
...  

β-Tricalcium phosphate (β-TCP) micron particles whose diameters range from 1 μm to 10 μm have been recently developed, however, their biological effects remain unknown. We investigated the biological effects ofβ-TCP micron particles on proliferation, cytotoxicity, and calcification of human synovial mesenchymal stem cells (MSCs). MSCs were cultured without dexamethasone,β-glycerophosphate, or ascorbic acid. 1.0 mg/mLβ-TCP micron particles inhibited proliferation of MSCs significantly and increased dead cells. In the contact condition, 0.1 mg/mLβ-TCP micron particles promoted calcification of MSCs evaluated by alizarin red staining and enhanced mRNA expressions of runx2, osteopontin, and type I collagen. In the noncontact condition, these effects were not observed. 0.1 mg/mLβ-TCP micron particles increased calcium concentration in the medium in the contact condition, while 1.0 mg/mLβ-TCP micron particles decreased calcium and phosphorus concentrations in the medium in the noncontact condition. By transmission electron microscopy,β-TCP micron particles were localized in the phagosome of MSCs and were dissolved. In conclusion,β-TCP micron particles promoted calcification of MSCs and enhanced osteogenesis-related gene expressionsin vitro.


2021 ◽  
Vol 11 ◽  
Author(s):  
Amy Creecy ◽  
John G. Damrath ◽  
Joseph M. Wallace

Osteocytes make up 90–95% of the cellular content of bone and form a rich dendritic network with a vastly greater surface area than either osteoblasts or osteoclasts. Osteocytes are well positioned to play a role in bone homeostasis by interacting directly with the matrix; however, the ability for these cells to modify bone matrix remains incompletely understood. With techniques for examining the nano- and microstructure of bone matrix components including hydroxyapatite and type I collagen becoming more widespread, there is great potential to uncover novel roles for the osteocyte in maintaining bone quality. In this review, we begin with an overview of osteocyte biology and the lacunar–canalicular system. Next, we describe recent findings from in vitro models of osteocytes, focusing on the transitions in cellular phenotype as they mature. Finally, we describe historical and current research on matrix alteration by osteocytes in vivo, focusing on the exciting potential for osteocytes to directly form, degrade, and modify the mineral and collagen in their surrounding matrix.


2021 ◽  
Vol 22 (16) ◽  
pp. 8445
Author(s):  
Elia Bari ◽  
Fulvio Tartara ◽  
Fabio Cofano ◽  
Giuseppe di Perna ◽  
Diego Garbossa ◽  
...  

Titanium is one of the most frequently used materials in bone regeneration due to its good biocompatibility, excellent mechanical properties, and great osteogenic performance. However, osseointegration with host tissue is often not definite, which may cause implant failure at times. The present study investigates the capacity of the mesenchymal stem cell (MSC)-secretome, formulated as a ready-to-use and freeze-dried medicinal product (the Lyosecretome), to promote the osteoinductive and osteoconductive properties of titanium cages. In vitro tests were conducted using adipose tissue-derived MSCs seeded on titanium cages with or without Lyosecretome. After 14 days, in the presence of Lyosecretome, significant cell proliferation improvement was observed. Scanning electron microscopy revealed the cytocompatibility of titanium cages: the seeded MSCs showed a spread morphology and an initial formation of filopodia. After 7 days, in the presence of Lyosecretome, more frequent and complex cellular processes forming bridges across the porous surface of the scaffold were revealed. Also, after 14 and 28 days of culturing in osteogenic medium, the amount of mineralized matrix detected by alizarin red was significantly higher when Lyosecretome was used. Finally, improved osteogenesis with Lyosecretome was confirmed by confocal analysis after 28 and 56 days of treatment, and demonstrating the production by osteoblast-differentiated MSCs of osteocalcin, a specific bone matrix protein.


2020 ◽  
Vol 79 (Suppl 1) ◽  
pp. 1122.2-1123
Author(s):  
S. H. Jin ◽  
P. R. Park ◽  
M. J. Kim ◽  
Y. J. Lee ◽  
S. Jo ◽  
...  

Background:Ankylosing Spondylitis (AS) is a chronic inflammatory rheumatic disease, which is characterized by the enthesitis, peripheral arthritis, and chronic inflammation of the spine, leading to bony ankylosis. Signal transducer and activator of transcription (STAT) family proteins are latent cytoplasmic transcription factors that convey signals to the nucleus. It is activated by IL-6, IL-23, and IL-22 through JAK-mediated phosphorylation. Moreover, genetic studies implicate interleukin-23 (IL-23) receptor signal, including STAT3 in the development of AS. IL-17A has recently emerged as a potential target that regulates the extensive inflammation and abnormal bone formation observed in AS. It was reported that STAT3 is a regulatory factor that induces Th17 cell development from naive CD4 T cells.Objectives:The aim of this study is to investigate whether the STAT3 phosphorylation (stat3-p) inhibitor has a therapeutic effect on inflammation and new bone formation in AS.Methods:Eight weeks after curdlan injection, SKG mice were treated with stat3-p inhibitor or mock as a control. Clinical and histologic scores for arthritis and enthesitis were evaluated. Synovial fluid mononuclear cells (SFMC) samples were obtained from AS patients. Inflammatory cytokine producing cells were analyzed using flow cytometry. Bone tissue samples were obtained from the facet joints of patients with AS at surgery. Primary bone-derived cells (BdCs) were isolated and cultured. The osteogenic differentiation was assessed in vitro for 3 weeks using ALP activity, Alizarin red S (ARS), Type I collagen, von kossa,and hydroxyapatitestains. Statistical analysis was performed using Prism 5.0 Software. A p < 0.05 was considered statistically significant.Results:The stat3-p inhibitor significantly suppressed peripheral arthritis and enthesitis in SKG mice (figure 1). Inflammatory infiltration around the tendon–bone insertion site and along the tendon, as well as bony involvement were all reduced in stat3-p inhibitor-treated mice compared to control mice. We found that the levels of IFN-±, IL-17, TNF-± were higher in AS Synovial fluid. A significantly decreased frequencies of IFN-±, IL-17, TNF-± producing cells in AS SFMC were shown after stat3-p inhibitor treatment (P < 0.01).In vitro experiment of bone formation, the stat3-p inhibitor suppressed ALP activity. In addition, there were significant decrease in Alizarin red S (ARS), Type I collagen, von kossa staining scores due to stat3-p inhibitor at a concentration of 5 μM.Light intensity of hydroxyapatitestaining was also decreased by stat3-p inhibitor in a dose dependent manner (figure 2). Intriguingly, the stat3-p inhibitor suppressed osteogenesis in both early phase and late phase in AS-BdCs, down-regulating osteoblast-involved genes.Conclusion:The stat3-p inhibitor had beneficial effects on reducing inflammation and new bone formation in AS animal model. In addition, stat3-p inhibitor suppressed bone formation in vitro experiment. These findings suggest that the stat3-p inhibitor could be a potential therapeutic agent for AS.References:[1]Arthritis Res Ther 2018;20:115.[2]Nat Med 2012;18:1069-76.[3]Rheumatology (Oxford) 2017;56:488-493.[4]Nat Rev Immunol. 2011;11:239–50.[5]J Exp Med 2005;201:949–60.Acknowledgments:NoneDisclosure of Interests:None declared


1997 ◽  
Vol 272 (5) ◽  
pp. C1709-C1716 ◽  
Author(s):  
X. Yu ◽  
S. C. Hsieh ◽  
W. Bao ◽  
D. T. Graves

Platelet-derived growth factor (PDGF) is mitogenic and chemotactic for osteoblastic cells in vitro. It is expressed during osseous wound healing and stimulates formation of new bone in vivo. PDGF stimulates cells by binding to specific cell surface receptors. The purpose of this study was to examine the effects of PDGF on osteoblastic proliferation and differentiation in long-term mineralizing cultures. Utilizing Northern blot analysis, we found that continuous PDGF treatment increased histone expression, indicative of enhanced proliferation, but suppressed osteoblast differentiation, demonstrated by inhibition of alkaline phosphatase, type I collagen, and osteocalcin expression. The inhibitory effect of PDGF on the differentiated function of osteoblasts was further established by findings that PDGF significantly inhibited nodule formation. The expression of PDGF receptors varied at different stages of culture. PDGF receptor mRNA expression increased when the cells had achieved a mature phenotype, during the stage of matrix maturation, and then decreased. However, as demonstrated by thymidine incorporation assays, the capacity of PDGF to stimulate DNA synthesis actually decreased during osteoblast maturation, as receptor expression increased. To investigate this apparent contradiction, tyrosyl phosphorylation and immunoblot assays were performed to assess changes in PDGF activation of their cognate receptors. The pattern of PDGF-induced tyrosyl phosphorylation remained relatively constant. This suggests that the diminished mitogenic activity of PDGF that occurs after osteoblast differentiation is regulated at a postreceptor level.


2017 ◽  
Vol 28 (3) ◽  
pp. 307-316 ◽  
Author(s):  
Luciana B. Alves ◽  
Sérgio L. S. de Souza ◽  
Mario Taba Jr ◽  
Arthur B. Novaes Jr ◽  
Paulo T. de Oliveira ◽  
...  

Abstract This study aimed to investigate the influence of a three-dimensional cell culture model and bioactive glass (BG) particles on the expression of osteoblastic phenotypes in rat calvaria osteogenic cells culture. Cells were seeded on two-dimensional (2D) and three-dimensional (3D) collagen with BG particles for up to 14 days. Cell viability and alkaline phosphatase (ALP) activity was performed. Cell morphology and immunolabeling of noncollagenous bone matrix proteins were assessed by epifluorescence and confocal microscopy. The expressions of osteogenic markers were analyzed using RT-PCR. Mineralized bone-like nodule formation was visualized by microscopy and calcium content was assessed quantitatively by alizarin red assay. Experimental cultures produced a growing cell viability rate up to 14 days. Although ALP activity at 7 days was higher on BG cultures, cells on 3D and 3D+BG had an activity decrease of ALP at 14 days. Three-dimensional conditions favored the immunolabeling for OPN and BSP and the expression of ALP and COL I mRNAs. BG particles influenced positively the OC and OPN mRNAs expression and calcified nodule formation in vitro. The results indicated that the 3D cultures and BG particles contribute to the expression of osteoblastic phenotype and to differentiated and mineralized matrix formation.


2021 ◽  
Vol 13 (582) ◽  
pp. eabe5738
Author(s):  
Tao Wang ◽  
Andrea Wagner ◽  
Renate Gehwolf ◽  
Wenjin Yan ◽  
Fabian S. Passini ◽  
...  

Tendons and tendon interfaces have a very limited regenerative capacity, rendering their injuries clinically challenging to resolve. Tendons sense muscle-mediated load; however, our knowledge on how loading affects tendon structure and functional adaption remains fragmentary. Here, we provide evidence that the matricellular protein secreted protein acidic and rich in cysteine (SPARC) is critically involved in the mechanobiology of tendons and is required for tissue maturation, homeostasis, and enthesis development. We show that tendon loading at the early postnatal stage leads to tissue hypotrophy and impaired maturation of Achilles tendon enthesis in Sparc−/− mice. Treadmill training revealed a higher prevalence of spontaneous tendon ruptures and a net catabolic adaptation in Sparc−/− mice. Tendon hypoplasia was attenuated in Sparc−/− mice in response to muscle unloading with botulinum toxin A. In vitro culture of Sparc−/− three-dimensional tendon constructs showed load-dependent impairment of ribosomal S6 kinase activation, resulting in reduced type I collagen synthesis. Further, functional calcium imaging revealed that lower stresses were required to trigger mechanically induced responses in Sparc−/− tendon fascicles. To underscore the clinical relevance of the findings, we further demonstrate that a missense mutation (p.Cys130Gln) in the follistatin-like domain of SPARC, which causes impaired protein secretion and type I collagen fibrillogenesis, is associated with tendon and ligament injuries in patients. Together, our results demonstrate that SPARC is a key extracellular matrix protein essential for load-induced tendon tissue maturation and homeostasis.


Sign in / Sign up

Export Citation Format

Share Document