[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,318)

Search Parameters:
Keywords = osteoblast differentiation

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
14 pages, 9728 KiB  
Article
Exploring the Anti-Osteoporotic Effects of n-Hexane Fraction from Cotoneaster wilsonii Nakai: Activation of Runx2 and Osteoblast Differentiation In Vivo
by Soyeon Hong, Hee Ju Lee, Da Seul Jung, Saruul Erdenebileg, Hoseong Hwang, Hak Cheol Kwon, Jaeyoung Kwon and Gyhye Yoo
Pharmaceuticals 2025, 18(1), 45; https://doi.org/10.3390/ph18010045 - 3 Jan 2025
Viewed by 321
Abstract
Background: Osteoporosis is characterized by the microstructural depletion of bone tissue and decreased bone density, leading to an increased risk of fractures. Cotoneaster wilsonii Nakai, an endemic species of the Korean Peninsula, grows wild in Ulleungdo. In this study, we aimed to investigate [...] Read more.
Background: Osteoporosis is characterized by the microstructural depletion of bone tissue and decreased bone density, leading to an increased risk of fractures. Cotoneaster wilsonii Nakai, an endemic species of the Korean Peninsula, grows wild in Ulleungdo. In this study, we aimed to investigate the effects of C. wilsonii and its components on osteoporosis. Methods and Results: The alkaline phosphatase (ALP) activity of C. wilsonii extracts and fractions was evaluated in MC3T3-E1 pre-osteoblasts, and the n-hexane fraction (CWH) showed the best properties for ALP activity. The effects of the CWH on bone formation were assessed in MC3T3-E1 cells and ovariectomized mice. Biochemical assays and histological analyses focused on the signaling activation of osteoblast differentiation and osteogenic markers, such as ALP, collagen, and osterix. The CWH significantly activated TGF-β and Wnt signaling, enhancing osteoblast differentiation and bone matrix formation. Notably, CWH treatment improved micro-CT indices, such as femoral bone density, and restored serum osteocalcin levels compared to OVX controls. Conclusions: These results highlight the potential of the C. wilsonii Nakai n-hexane fraction as a promising therapeutic agent for managing osteoporosis. Full article
(This article belongs to the Special Issue The Role of Phytochemicals in Aging and Aging-Related Diseases)
Show Figures

Figure 1

Figure 1
<p>Effects of <span class="html-italic">C. wilsonii</span> (CW) on osteoblastic differentiation in vitro. (<b>A</b>) Comparison of ALP activity between <span class="html-italic">C. wilsonii</span> ethanol extract (CW-EtOH) and its fractions (CWH; <span class="html-italic">n</span>-Hexane fraction, CW-BuOH; buthanol fraction, CW-H2O; water-soluble layers). (<b>B</b>) ALP activity of CWH. (<b>C</b>) Protein expression analysis of osteoblastic markers using Western blot. (<b>D</b>) mRNA expression levels of osteoblastic markers determined by qRT-PCR. Data are presented as mean ± SEM (n = 6). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 are significantly different from the DIFF group. UN: undifferentiated MC3T3-E1 cell; DIFF: differentiated MC3T3-E1 cell; CWH10 and CWH20: differentiated MC3T3-E1 cells treated with 10 μg/mL and 20 μg/mL CWH, respectively.</p>
Full article ">Figure 2
<p>Effects of of <span class="html-italic">n</span>-haxane fraction of <span class="html-italic">C. wilsonii</span> (CWH) on physiological markers in ovariectomized mice. (<b>A</b>) Experimental design for animal study. (<b>B</b>) Measurements of body weight, uterus weight, and serum osteocalcin levels in OVX mice treated with CWH. (<b>C</b>) Histological analysis using H&amp;E staining and type I collagen (COL1) immunohistochemical staining in the distal femoral region of OVX mice. Data are presented as mean ± SEM (n = 6). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 are significantly different from the OVX group. Blue arrows: lipid droplets; Red arrows: collagen.</p>
Full article ">Figure 3
<p>Effects of <span class="html-italic">n</span>-hexane fraction of <span class="html-italic">C. wilsonii</span> (CWH) on bone markers in ovariectomized mice. (<b>A</b>) Micro-computed tomography images of the distal femoral region of mice. (<b>B</b>) Tomographic measurements of BMD, Tb.N, BV/TV, Tb.Sp, BS/BV, and BS/TV. Data are presented as mean ± SEM (n = 6). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 are significantly different from the OVX group.</p>
Full article ">Figure 4
<p>Effects <span class="html-italic">n</span>-hexane fraction of <span class="html-italic">C. wilsonii</span> (CWH) on the osteoblast population of bone marrow. (<b>A</b>) ALP activity, (<b>B</b>) protein expression, and (<b>C</b>) mRNA expression levels of osteoblastic markers in primary bone marrow cells. Data are presented as mean ± SEM (n = 6). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 are significantly different from the OVX group.</p>
Full article ">Figure 5
<p>HPLC chromatogram of the <span class="html-italic">n</span>-hexane fraction of <span class="html-italic">C. wilsonii</span>. Peaks: CWH_C1, lupeol; CWH_C2, β-sitosterol; CWH_C3, betulin; CWH_C4, ethyl caffeate.</p>
Full article ">Figure 6
<p>Effects of compounds of the <span class="html-italic">C. wilsonii n</span>-hexane fraction on osteoblastic differentiation in vitro. (<b>A</b>) ALP activity (<b>B</b>) of osteoblastic markers determined by Western blot. mRNA (<b>C</b>) expression levels of osteoblastic markers determined by qRT-PCR. Data are presented as mean ± SEM (n = 6). ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001, significantly different from the DIFF group. UN: undifferentiated MC3T3-E1 cells; DIFF: differentiated MC3T3-E1 cells; C1, Lupeol; C2, β-sitosterol; C3, botulin; C4, ethyl caffeate.</p>
Full article ">
16 pages, 7192 KiB  
Article
Osteoblastic Differentiation of Human Adipose-Derived Mesenchymal Stem Cells on P3HT Thin Polymer Film
by Paola Campione, Maria Giovanna Rizzo, Luana Vittoria Bauso, Ileana Ielo, Grazia Maria Lucia Messina and Giovanna Calabrese
J. Funct. Biomater. 2025, 16(1), 10; https://doi.org/10.3390/jfb16010010 - 2 Jan 2025
Viewed by 419
Abstract
Bone defects restoration has always been an arduous challenge in the orthopedic field due to the limitations of conventional grafts. Bone tissue engineering offers an alternative approach by using biomimetic materials, stem cells, and growth factors that are able to improve the regeneration [...] Read more.
Bone defects restoration has always been an arduous challenge in the orthopedic field due to the limitations of conventional grafts. Bone tissue engineering offers an alternative approach by using biomimetic materials, stem cells, and growth factors that are able to improve the regeneration of bone tissue. Different biomaterials have attracted great interest in BTE applications, including the poly(3-hexylthiofene) (P3HT) conductive polymer, whose primary advantage is its capability to provide a native extracellular matrix-like environment. Based on this evidence, in this study, we evaluated the biological response of human adipose-derived mesenchymal stem cells cultured on P3HT thin polymer film for 14 days. Our results suggest that P3HT represents a good substrate to induce osteogenic differentiation of osteoprogenitor cells, even in the absence of specific inductive growth factors, thus representing a promising strategy for bone regenerative medicine. Therefore, the system provided may offer an innovative platform for next-generation biocompatible materials for regenerative medicine. Full article
(This article belongs to the Special Issue Mesoporous Nanomaterials for Bone Tissue Engineering)
Show Figures

Figure 1

Figure 1
<p>Morphological analysis (<b>a</b>) and Young’s modulus (<b>b</b>) of P3HT thin film.</p>
Full article ">Figure 2
<p>Cell viability analysis of hADMSCs cultured on P3HT thin polymer films, in GM and OM, for 1, 7, and 14 days. Data are reported as percentage of mean ± standard deviation obtained from three different samples for type. ** <span class="html-italic">p</span> &lt; 0.01 show significant differences between the different groups, as reported by the Holm post hoc test.</p>
Full article ">Figure 3
<p>(<b>A</b>) Representative images of Alizarin Red S staining on hADMSCs cultured on P3HT thin polymer film in presence of GM or OM, for 1, 7, and 14 days. Scale bars: 100 μm. (<b>B</b>) Quantitative evaluation of the Alizarin Red S images by measuring the integrated density (IntDen). The multi-way ANOVA test <span class="html-italic">p</span>-value is reported, and ** <span class="html-italic">p</span> &lt; 0.01 and **** <span class="html-italic">p</span> &lt; 0.0001 show significant differences between the different groups, as reported by the Holm post hoc test.</p>
Full article ">Figure 4
<p>(<b>a</b>) Representative diagram of osteogenic markers expression during the osteoblastic differentiation of hADMSCs. Gene expression profile of (<b>b</b>) Osteopontin, (<b>c</b>) Osteocalcin, and (<b>d</b>) Osteonectin in hADMSCs cultured on uncoated (CTRL) and P3HT-coated slides, in presence of GM or OM for 1, 7, and 14 days. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used as an endogenous control. The multi-way ANOVA test <span class="html-italic">p</span>-value is reported, and ** <span class="html-italic">p</span> &lt; 0.01 show significant differences between the different groups, as reported by the Holm post hoc test. ns = not significant.</p>
Full article ">Figure 5
<p>(<b>A</b>) Immunofluorescence analysis of hADMSCs cultured on P3HT thin polymer film, in presence of GM or OM for 1, 7 and 14 days, showing the expression of Osterix. Scale bars: 50 μm. (<b>B</b>) Quantitative evaluation of Osterix-positive cells vs. DAPI-positive cells per field. Multi-way ANOVA test <span class="html-italic">p</span>-value is reported and * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001 show significant differences between the different groups, as reported by the Holm post hoc test.</p>
Full article ">Figure 6
<p>Morphological analysis of hADMSCs at D14 on glass with growth medium (GM) (<b>a</b>) and with osteogenic differentiation medium (OM) (<b>b</b>), on P3HT thin film in GM (<b>c</b>) and in OM (<b>d</b>). 50 µm × 50 µm acquisition (<b>i</b>) and 100 µm × 100 µm acquisition (<b>ii</b>).</p>
Full article ">Figure 7
<p>Young’s modulus of hADMSCs at D14 on glass (CTRL) and P3HT in presence of normal growth medium (GM) osteogenic differentiation medium (OM). The one-way ANOVA test <span class="html-italic">p</span>-value is reported and ** <span class="html-italic">p</span> &lt; 0.01 show significant differences between the different groups, as reported by the Holm post hoc test. ns = not significant.</p>
Full article ">
14 pages, 2929 KiB  
Article
Small Extracellular Vesicles Derived from Lipopolysaccharide-Treated Stem Cells from the Apical Papilla Modulate Macrophage Phenotypes and Inflammatory Interactions in Pulpal and Periodontal Tissues
by Solène Tessier, Boris Halgand, Davy Aubeux, Joëlle Véziers, Angélique Galvani, Juliette Jamoneau, Fabienne Pérez, Valérie Geoffroy and Alexis Gaudin
Int. J. Mol. Sci. 2025, 26(1), 297; https://doi.org/10.3390/ijms26010297 - 31 Dec 2024
Viewed by 250
Abstract
Inflammation significantly influences cellular communication in the oral environment, impacting tissue repair and regeneration. This study explores the role of small extracellular vesicles (sEVs) derived from lipopolysaccharide (LPS)-treated stem cells from the apical papilla (SCAP) in modulating macrophage polarization and osteoblast differentiation. SCAPs [...] Read more.
Inflammation significantly influences cellular communication in the oral environment, impacting tissue repair and regeneration. This study explores the role of small extracellular vesicles (sEVs) derived from lipopolysaccharide (LPS)-treated stem cells from the apical papilla (SCAP) in modulating macrophage polarization and osteoblast differentiation. SCAPs were treated with LPS for 24 h, and sEVs from untreated (SCAP-sEVs) and LPS-treated SCAP (LPS-SCAP-sEVs) were isolated via ultracentrifugation and characterized using transmission electron microscopy, Western blot, and Tunable Resistive Pulse Sensing. LPS-SCAP-sEVs exhibited characteristic exosome morphology (~100 nm diameter) and expressed vesicular markers (CD9, CD63, CD81, and HSP70). Functional analysis revealed that LPS-SCAP-sEVs promoted M1 macrophage polarization, as evidenced by the increased pro-inflammatory cytokines (IL-6 and IL-1β) and the reduced anti-inflammatory markers (IL-10 and CD206), while impairing the M2 phenotype. Additionally, LPS-SCAP-sEVs had a minimal impact on SCAP metabolic activity or osteogenic gene expression but significantly reduced mineralization capacity in osteogenic conditions. These findings suggest that sEVs mediate the inflammatory interplay between SCAP and macrophages, skewing macrophage polarization toward a pro-inflammatory state and hindering osteoblast differentiation. Understanding this sEV-driven communication axis provides novel insights into the cellular mechanisms underlying inflammation in oral tissues and highlights potential therapeutic targets for modulating extracellular vesicle activity during acute inflammatory episodes. Full article
(This article belongs to the Special Issue Molecular Advances in Dental Pulp Tissue Engineering, 2nd Edition)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Characterization of sEVs derived from control SCAP (sEVs-CT) (<b>a</b>) or from LPS-treated SCAP (sEVs-LPS) (<b>b</b>). Size distribution and concentration of sEVs-CT (<b>a</b>) and sEVs-LPS (<b>b</b>) assessed using Tunable Resistive Pulse Sensing (TRPS) technology. (<b>c</b>) TRPS analysis showing the mean size of sEVs-CT and sEVs-LPS. (<b>d</b>) TRPS analysis showing the quantity of sEVs-CT and sEVs-LPS secreted per cell. (<b>e</b>) Ultrastructure of sEVs-CT and sEVs-LPS observed using electron microscopy. (<b>f</b>) Protein concentrations of sEVs-CT and sEVs-LPS determined by micro-BCA assay. (<b>g</b>) Expression of sEV-associated protein positive markers (HSP70, CD63, CD9, and CD81). GM130, a cis-Golgi matrix protein, was used as a negative control to confirm the absence of Golgi apparatus contamination by Western blot. Data are expressed as mean ± SEM. <span class="html-italic">N</span> = 3. * <span class="html-italic">p</span> &lt; 0.05 (Student’s <span class="html-italic">t</span>-test).</p>
Full article ">Figure 2
<p>Effect of sEVs-CT and sEVs-LPS on SCAP osteoblastic differentiation and mineralization capacity. (<b>a</b>–<b>c</b>) Expression of representative genes of osteoblastic differentiation, including <span class="html-italic">ALP</span> (<b>a</b>)<span class="html-italic">, Col1a1</span> (<b>b</b>), and <span class="html-italic">Runx2</span> (<b>c</b>), determined by RT-qPCR. Gene expression (2<sup>−ΔΔ</sup> CT) was normalized to SCAP at day 0 and presented as fold change. In total, <span class="html-italic">18S</span> and <span class="html-italic">PPIA</span> were used as housekeeping genes. (<b>d</b>) Alizarin Red S staining performed on SCAP cultured in an osteogenic induction medium for 17 days. The effects of untreated and LPS-treated SCAP conditioned medium were compared. (CT +: positive control (osteogenic medium); CT −: negative control (the osteogenic medium with exosome-depleted fetal bovine serum); CT-CM: Conditioned medium; LPS-CM: Conditioned medium with LPS). (<b>e</b>) Quantification of the degree of mineralization assessed by dissolution of mineralization crystals with methanol and acetic acid. The optical density was measured at 450 nm. (<b>f</b>) Effect of 1X (1 × 10<sup>7</sup> sEVs per well) and 5X (5 × 10<sup>7</sup> sEVs per well) of sEVs-CT and sEVs-LPS on SCAP metabolic activity determined by CCK-8 assay. The results are presented as the percentage of metabolic activity relative to untreated SCAP at day 0. The data are expressed as mean ± SEM from three independent experiments (<span class="html-italic">N</span> = 3), each performed in triplicate (<span class="html-italic">n</span> = 3). <span class="html-italic">p</span>-values derived from one-way ANOVA followed by Tukey’s multiple comparison tests is * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p>Effect of sEVs-LPS on polarized macrophages. Expression of representative pro-inflammatory marker genes including <span class="html-italic">IL-6</span> (<b>a</b>), <span class="html-italic">TNF-α</span> (<b>b</b>), and <span class="html-italic">IL-1β</span> (<b>c</b>); and anti-inflammatory marker genes, <span class="html-italic">IL-10</span> (<b>d</b>) and <span class="html-italic">CD206</span> (<b>e</b>), after treatment of polarized macrophages with sEVs derived from SCAP or LPS-induced SCAP for 24 h determined by RTqPCR. Gene expression (2<sup>−ΔΔ</sup> CT) was normalized to untreated M0 macrophages and presented as fold change. The <span class="html-italic">18S</span> and <span class="html-italic">PPIA</span> were used as housekeeping genes. Quantification of pro-inflammatory factors IL-6 (<b>f</b>), TNF-α (<b>g</b>), and IL-1β (<b>h</b>) in the culture supernatant of M0 macrophages using bead-based multiplex assays by flow cytometry. The data are expressed as mean ± SEM. <span class="html-italic">N</span> = 3; <span class="html-italic">n</span> = 3. <span class="html-italic">p</span>-values derived from one-way ANOVA followed by Tukey’s multiple comparison tests are * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">
14 pages, 3714 KiB  
Article
Experimental Validation of Antiobesogenic and Osteoprotective Efficacy of Ginsenoside CK via Targeting Lipid and Atherosclerosis Pathways
by Md. Niaj Morshed, Reshmi Akter, Imran Mahmud, Ah-Yeong Gwon, Jin Woo Jeang, Yeong-Geun Lee, Dae Won Park, Deok Chun Yang, Yeon Ju Kim and Se-Chan Kang
Life 2025, 15(1), 41; https://doi.org/10.3390/life15010041 - 31 Dec 2024
Viewed by 327
Abstract
The present study explored the possible antiobesogenic and osteoprotective properties of the gut metabolite ginsenoside CK to clarify its influence on lipid and atherosclerosis pathways, thereby validating previously published hypotheses. These hypotheses were validated by harvesting and cultivating 3T3-L1 and MC3T3-E1 in adipogenic [...] Read more.
The present study explored the possible antiobesogenic and osteoprotective properties of the gut metabolite ginsenoside CK to clarify its influence on lipid and atherosclerosis pathways, thereby validating previously published hypotheses. These hypotheses were validated by harvesting and cultivating 3T3-L1 and MC3T3-E1 in adipogenic and osteogenic media with varying concentrations of CK. We assessed the differentiation of adipocytes and osteoblasts in these cell lines by applying the most effective doses of CK that we initially selected. Using 3T3-L1 adipocytes in vitro assessments, CK could effectively decrease intracellular lipid accumulation, inhibit α-glucosidase enzyme, increase 2-NBDG glucose uptake, reduce inflammation-associated cytokines (TNFα, and IL-6), adipogenic regulatory genes (PPARγ, FAS, C/EBPα), lipogenic gene LPL, and increase the expression of thermogenic gene UCP1. Additionally, CK treatment induced osteoblast development in MC3T3-E1 cells as shown by increased mineralization and calcium distribution, collagen content, alkaline phosphatase activity, and decreased inflammatory cytokines TNFα, and IL-6 and increased the regulated expressions of osteogenic genes including Runx2, ALP, BGLAP, OCN, and Col1a1. Significantly, as a major inhibitory regulator, the TP53 gene was down-regulated in both 3T3-L1 and MC3T3E1 cells after the treatment of CK. These encouraging results demonstrate the possible use of CK as an innovative treatment for controlling obesity and osteoporosis, targeting the underlying mechanisms of obesogenic and bone loss. Further studies are necessary to explore the clinical implications of these results and the potential of CK in future treatment strategies. This research highlights the promise of CK in addressing significant health issues. Full article
(This article belongs to the Section Pharmaceutical Science)
Show Figures

Figure 1

Figure 1
<p>The MTT assay was used to measure cell viability. CK (3.125–50 μM) was administered to (<b>A</b>) 3T3-L1 pre-adipocyte (Orange-colored columns) and (<b>B</b>) MC3T3-E1 pre-osteoblast cells (1 × 10<sup>4</sup> cells/well) (Tan-colored columns) for 24 h. A two-tailed Student’s <span class="html-italic">t</span>-test was used to determine whether there was a substantial distinction between the groups; ns denotes a non-significant change, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 when compared to a control group (Black-colored column) that was not treated.</p>
Full article ">Figure 2
<p>The inhibitory effect of CK on lipid accumulation in MDI-induced 3T3-L1 adipocytes was assessed as follows: (<b>A</b>) Oil Red O staining was performed to visualize fat droplets, which were then observed under a light microscope at 20× magnification. (<b>B</b>) Lipid accumulation was quantified by measuring the absorbance of Oil Red O dissolved in isopropyl alcohol at 520 nm. Data represent the mean ± SEM from three independent experiments. Statistical significance is indicated as ### <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, compared to the MDI-treated group.</p>
Full article ">Figure 3
<p>(<b>A</b>) The proportion of a-glucosidase inhibition at various CK concentrations. Acarbose, or ACR, was employed as a control. We also analyzed the half-maximum inhibitory concentration (IC<sub>50</sub>) of CK and ACR (<b>B</b>) 3T3L1 cells using the 2-NBDG uptake test. Using a fluorescent derivative of glucose 2-NBDG, the impact of CK on 3T3L1 cells’ glucose absorption was examined for 24 h with and without CK. Insulin was employed as a positive control at 100 nM. Data are presented as a control percentage. *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>The relative expression of inflammatory genes (<span class="html-italic">TNFα</span>, <span class="html-italic">IL-6</span>), adipogenic genes (<span class="html-italic">PPARγ</span>, <span class="html-italic">C/EBPα</span>, <span class="html-italic">LPL</span>), the lipogenic gene <span class="html-italic">FAS</span>, and the thermogenic gene <span class="html-italic">UCP1</span>, along with <span class="html-italic">TP</span><sup>53</sup>, was measured in differentiated 3T3-L1 cells treated with CK at concentrations of 2.5 μM and 10 μM, or RSG at 20 μM. Results are presented as the mean ± standard deviation from three independent experiments. Statistical significance was assessed using a two-tailed Student’s <span class="html-italic">t</span>-test. Significant differences in gene expression between untreated and treated groups are indicated as ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>(<b>A</b>) ALP activity in CK and E<sub>2</sub>-treated differentiated MC3T3-E1 cells. (<b>B</b>) The impact of CK on MC3T3-E1 cell mineralization. A 96-well plate was used to measure the quantity of Alizarin Red S at 562 nm. (<b>C</b>) The calcium-binding Alizarin Red S dye was used to evaluate the calcium deposits in the extracellular matrix for matrix mineralization. Up to Day 28, CK treatment accelerated the mineralization of the extracellular matrix. The images are representative of over three different concentrations of CK and E<sub>2.</sub> The results are presented as the mean ± standard deviation from three independent experiments. Statistical significance was determined using a two-tailed Student’s <span class="html-italic">t</span>-test. Significant differences in lipid production compared to the untreated (control) group are denoted by ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 6
<p>Collagen content in MC3T3-E1 cells is increased by CK extracts. For 12 days, cells were exposed to extracts at doses ranging from 2.5 to 10 μM, either with or without a differentiation medium. E<sub>2</sub> was used at a concentration of 100 nm. (<b>A</b>) Picro-Sirius red staining was carried out and seen using a microscope (magnification of ×100). (<b>B</b>) Absorbance was measured at 550 nm to determine the amount of collagen. The presented data are the mean ± standard deviation (SD) of three studies. Statistical analysis revealed significant differences, denoted as * <span class="html-italic">p</span> &lt; 0.1, *** <span class="html-italic">p</span> &lt; 0.001 when compared with the indicated ascorbic acid and β-glycerophosphate treated group.</p>
Full article ">Figure 7
<p>Relative expression of proinflammatory cytokines TNFα, and IL-6, and osteogenic genes <span class="html-italic">RunX2</span>, <span class="html-italic">ALP</span>, <span class="html-italic">BGLAP</span>, <span class="html-italic">OCN</span>, and <span class="html-italic">COL1a1</span> with <span class="html-italic">TP</span><sup>53</sup> in differentiated MC3T3-E1 cells on treatment with CK at 2.5 μM and E<sub>2</sub> at 100 nm concentrations, respectively. A two-tailed Student’s <span class="html-italic">t</span>-test was used to determine whether the difference was statistically significant. The non-treated and treated groups’ significant differences in gene expression are indicated by * <span class="html-italic">p</span> &lt; 0.1, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. control.</p>
Full article ">
16 pages, 3485 KiB  
Article
Exploring the Biological Impact of β-TCP Surface Polarization on Osteoblast and Osteoclast Activity
by Jingpu Zheng, Kosuke Nozaki, Kazuaki Hashimoto, Kimihiro Yamashita and Noriyuki Wakabayashi
Int. J. Mol. Sci. 2025, 26(1), 141; https://doi.org/10.3390/ijms26010141 - 27 Dec 2024
Viewed by 390
Abstract
β-tricalcium phosphate (β-TCP) is a widely utilized resorbable bone graft material, whose surface charge can be modified by electrical polarization. However, the specific effects of such a charge modification on osteoblast and osteoclast functions remain insufficiently studied. In this work, electrically polarized β-TCP [...] Read more.
β-tricalcium phosphate (β-TCP) is a widely utilized resorbable bone graft material, whose surface charge can be modified by electrical polarization. However, the specific effects of such a charge modification on osteoblast and osteoclast functions remain insufficiently studied. In this work, electrically polarized β-TCP with a high surface charge density was synthesized and evaluated in vitro in terms of its physicochemical properties and biological activity. Polarization was performed to achieve a high surface charge density, which was quantified using a thermally stimulated depolarization current. The proliferation and differentiation of MC3T3-E1 osteoblast-like cells were assessed via WST-8 and alkaline phosphatase assays. Tartrate-resistant acid phosphatase (TRAP) activity and a resorption pit assay were used to evaluate the impact of surface charge on RAW264.7 osteoclast-like cell activity. Polarized β-TCP exhibited a surface charge of 1.3 mC cm−2. Electrically polarized surfaces significantly enhanced osteoblast proliferation and differentiation. TRAP activity assays demonstrated effective osteoclast differentiation of RAW264.7 cells, with enhanced activity observed on charged surfaces. Resorption pit assays further revealed improved osteoclast resorption capacity on β-TCP surfaces with a polarized charge. These findings indicate that β-TCP with a highly dense surface charge promotes osteoblast proliferation and differentiation, as well as osteoclast activity and resorption capacity. Full article
Show Figures

Figure 1

Figure 1
<p>TSDC curves of β-TCP samples before and after electrical polarization. The stored charge density of the polarized β-TCP was calculated to be 1.3 mC cm<sup>−2</sup>.</p>
Full article ">Figure 2
<p>XRD patterns of standard β-TCP, polarized β-TCP, and unpolarized β-TCP.</p>
Full article ">Figure 3
<p>FT-IR spectra of unpolarized β-TCP and polarized β-TCP.</p>
Full article ">Figure 4
<p>MC3T3-E1 proliferation on β-TCP samples measured using the CCK-8 assay after days 1, 3, and 7 (* <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p>Alkaline phosphatase (ALP) activity in MC3T3-E1 cells cultured on β-TCP samples measured after days 7, 11, and 14 in osteogenic media (* <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 6
<p>Effect of surface charge on osteoclast differentiation in RAW264.7 cells. TRAP staining was performed after the cells were cultured with RANKL (60 ng mL<sup>−1</sup>) on β-TCP samples for 3 and 5 days. Representative images show osteoclast formation on (<b>a</b>) β-TCP, (<b>b</b>) P-β-TCP, and (<b>c</b>) N-β-TCP for 3 days and (<b>d</b>) β-TCP, (<b>e</b>) P-β-TCP, and (<b>f</b>) N-β-TCP for 5 days (original magnification, 100×). (<b>g</b>) Statistical comparison of TRAP activity among uncharged and surface-charged groups. Scale bar = 100 μm (* <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 7
<p>Influence of surface charge on osteoclast-mediated bone resorption. Samples were cultured with RAW264.7 cells for 10 days, examined using confocal laser scanning microscopy (CLSM), and imaged at the same magnification. Resorption pit volumes formed by osteoclasts were quantified using Image-Pro Plus 7.0 software (Media Cybernetics, Rockville, MD, USA). Representative SEM images of (<b>a</b>) β-TCP, (<b>b</b>) P-β-TCP, and (<b>c</b>) N-β-TCP, (original magnification, 100×) and (<b>d</b>) β-TCP, (<b>e</b>) P-β-TCP, and (<b>f</b>) N-β-TCP (original magnification, 200×). (<b>g</b>) Statistical comparison of the average pit volumes between uncharged and surface-charged β-TCP groups (* <span class="html-italic">p</span> &lt; 0.05). Scale bar = 100 μm.</p>
Full article ">Scheme 1
<p>Electrical polarization process and surface charge evaluation of β-TCP. The sample was sandwiched between platinum electrodes and alumina plates. Polarization was conducted at 400 °C with a DC electric field of 0.65 V/mm. After cooling to room temperature under the maintained field, TSDC was measured using a galvanometer (6514/J, Tektronix Inc., Tokyo, Japan). The stored charge <span class="html-italic">Q</span> was calculated from the depolarization current density <span class="html-italic">J</span>(<span class="html-italic">T</span>) using the equation <span class="html-italic">Q</span> = (<math display="inline"><semantics> <mrow> <mstyle scriptlevel="0" displaystyle="true"> <mfrac> <mrow> <mn>1</mn> </mrow> <mrow> <mi>β</mi> </mrow> </mfrac> </mstyle> <mrow> <mo stretchy="false">∫</mo> <mrow> <mi>J</mi> <mfenced separators="|"> <mrow> <mi>T</mi> </mrow> </mfenced> <mi>d</mi> <mi>T</mi> </mrow> </mrow> </mrow> </semantics></math>, where <span class="html-italic">T</span> is the temperature and <span class="html-italic">B</span> is the heating rate. This setup allows for the evaluation of the surface charge characteristics in the β-TCP sample.</p>
Full article ">
18 pages, 2278 KiB  
Article
FXR Activation Accelerates Early Phase of Osteoblast Differentiation Through COX-2-PGE2-EP4 Axis in BMP-2-Induced Mouse Mesenchymal Stem Cells
by Ko Fujimori, Yusuke Iguchi, Yukiko Yamashita, Keigo Gohda and Naoki Teno
Molecules 2025, 30(1), 58; https://doi.org/10.3390/molecules30010058 - 27 Dec 2024
Viewed by 313
Abstract
Farnesoid X receptor (FXR), a nuclear receptor, is expressed in calvaria and bone marrow stromal cells and plays a role in bone homeostasis. However, the mechanism of FXR-activated osteoblast differentiation remains unclear. In this study, we investigated the regulatory mechanism underlying FXR-activated osteoblast [...] Read more.
Farnesoid X receptor (FXR), a nuclear receptor, is expressed in calvaria and bone marrow stromal cells and plays a role in bone homeostasis. However, the mechanism of FXR-activated osteoblast differentiation remains unclear. In this study, we investigated the regulatory mechanism underlying FXR-activated osteoblast differentiation using bone morphogenetic protein-2 (BMP-2)-induced mouse ST-2 mesenchymal stem cells. We also synthesized a novel FXR agonist, FLG390, and compared its biological effects in osteoblast differentiation with a known FXR agonist, chenodeoxycholic acid (CDCA). As an FXR agonist, FLG390 accelerated osteoblast differentiation to a comparable extent with CDCA, enhancing alkaline phosphatase (ALP) activity and the expression of osteoblast differentiated-related genes such as ALP, collagen type 1 α1 chain (COL1A1), and runt-related transcription factor 2 (RUNX2). FXR activation elevated the expression of cyclooxygenase (COX)-2 and the production of prostaglandin (PG) E2 in the early phase of osteoblast differentiation. A selective COX-2 inhibitor and an antagonist of EP4 receptors, one of PGE2 receptors, partially suppressed FXR-activated osteoblast differentiation. Moreover, treatment with either inhibitor during the first 6 h after initiating osteoblast differentiation repressed FXR-activated osteoblast differentiation to the same extent as did the treatment for 6 d. Therefore, a novel FXR agonist, FLG390, exhibited potency comparable to CDCA. FXR activation promoted the early phase of osteoblast differentiation via the COX-2-PGE2-EP4 axis, representing a potential target for control of bone metabolism. Full article
Show Figures

Figure 1

Figure 1
<p>Selectivity screening of <b>FLG390</b> against human nuclear receptors and TGR5. (<b>A</b>) Structure of FXR agonists. (<b>B</b>) Specificity of <b>FLG390</b> to nuclear receptors. The agonist activity of <b>FLG390</b> for each nuclear receptor was determined at a concentration of 1 μM in the luciferase reporter assay. The vertical axis shows the relative luciferase activity to the maximum activity of the following full agonists for each receptor: FXR—GW4064; RXRα—9-<span class="html-italic">cis</span>-retinoic acid; RARα—all-<span class="html-italic">trans</span> retinoic acid; VDR—1α,25-dihydroxyvitamin D<sub>3</sub>; PPARα—GW7647; PPARδ—GW501516; PPARγ—GW1929; LXRα and LXRβ—T0901317; and TGR5—lithocholic acid.</p>
Full article ">Figure 2
<p>ALP activity and mRNA expression in the FXR agonist-treated ST-2 MSCs. (<b>A</b>) Cell viability of ST-2 MSCs. The cells were cultured for 24 h in the medium in the absence (V: vehicle; white column) or presence of BMP-2 (50 ng/mL; gray column) and/or CDCA (10 µM; hatched column) or <b>FLG390</b> (1, 5, 10 µM; black columns). The data are presented as the mean ± S.D. (<span class="html-italic">n</span> = 8). (<b>B</b>) ALP staining. ST-2 MSCs were differentiated into osteoblasts for 6 d in the medium containing BMP-2 (50 ng/mL) with or without CDCA (10 µM) or <b>FLG390</b> (5, 10 µM). (<b>C</b>) ALP activity in BMP-2- and FXR agonist-treated ST-2 MSCs. The cells were differentiated into osteoblasts for 6 d in the medium with BMP-2 (50 ng/mL; gray column) and/or CDCA (10 µM; hatched column) or <b>FLG390</b> (1, 5, 10 µM; black columns). The data are presented as the mean ± S.D. from three independent experiments. * <span class="html-italic">p</span> &lt; 0.05. (<b>D</b>) Expression of the ALP mRNA in BMP-2- and FXR agonist-treated ST-2 MSCs. The cells were cultured as stated in the legend of <a href="#molecules-30-00058-f002" class="html-fig">Figure 2</a>C. The data are shown as the mean ± S.D. from three independent experiments. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p>Activation by FXR agonists through FXR in ST-2 MSCs. (<b>A</b>) ALP staining. ST-2 MSCs were differentiated into osteoblasts for 6 d in the presence of BMP-2 (50 ng/mL) and/or CDCA (10 µM) or <b>FLG390</b> (10 µM) with or without guggulsterone (GS; 25 μM). (<b>B</b>) ALP activity. ST-2 MSCs were differentiated into osteoblasts for 6 d in the medium containing BMP-2 (50 ng/mL; gray column) and/or CDCA (10 µM; hatched column) or <b>FLG390</b> (10 µM; black columns) with or without guggulsterone (GS; 25 μM; red columns). The data are shown as the mean ± S.D. from three independent experiments. * <span class="html-italic">p</span> &lt; 0.05. (<b>C</b>) Expression of the osteogenesis-related genes by BMP-2- and FXR agonist-treated ST-2 MSCs. The cells were cultured as described in the legend of <a href="#molecules-30-00058-f003" class="html-fig">Figure 3</a>B. The data are represented as the mean ± S.D. from three independent experiments. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>Expression of FXR and osteogenesis-related genes during osteoblast differentiation in BMP-2-induced ST-2 MSCs. (<b>A</b>) Expression of the FXR gene during osteoblast differentiation of BMP-2-induced ST-2 MSCs in the presence or absence of the FXR agonist. The cells were differentiated into osteoblasts for 6 d in the medium containing BMP-2 (50 ng/mL; gray columns), and/or CDCA (10 µM; hatched columns) or <b>FLG390</b> (10 µM; black columns). The data are shown as the mean ± S.D. from three independent experiments. (<b>B</b>) Expression of the osteogenesis-related genes during osteoblast differentiation of BMP-2-induced ST-2 MSCs in the presence or absence of the FXR agonist. The cells were cultured as described in the legend of <a href="#molecules-30-00058-f004" class="html-fig">Figure 4</a>A. The data are represented as the mean ± S.D. from three independent experiments. # <span class="html-italic">p</span> &lt; 0.05, vs. 0 h; * <span class="html-italic">p</span> &lt; 0.05, vs. BMP-2 alone-treated cells at each time point.</p>
Full article ">Figure 5
<p>Expression of PGE<sub>2</sub> synthases in BMP-2-induced ST-2 MSCs. (<b>A</b>) Expression levels of the COX-1, COX-2, and mPGES-1 genes during osteoblast differentiation of BMP-2-induced ST-2 MSCs in the presence or absence of the FXR agonist. The cells were differentiated into osteoblasts for 6 d in the medium containing BMP-2 (50 ng/mL; gray columns) with or without CDCA (10 µM; hatched columns) or <b>FLG390</b> (10 µM; black columns). The data are presented as the mean ± S.D. from three independent experiments. # <span class="html-italic">p</span> &lt; 0.05, vs. 0 h; * <span class="html-italic">p</span> &lt; 0.05, vs. BMP-2 alone-treated cells at each time point. (<b>B</b>) PGE<sub>2</sub> production during the osteoblast differentiation of BMP-2-induced ST-2 MSCs in the presence of the FXR agonist. The cells were cultured as stated in the legend of <a href="#molecules-30-00058-f005" class="html-fig">Figure 5</a>A and further incubated with A23187 (5 µM) for 15 min at 37 °C. The data are shown as the mean ± S.D. from three independent experiments. # <span class="html-italic">p</span> &lt; 0.05, vs. 0 h; * <span class="html-italic">p</span> &lt; 0.05, vs. BMP-2 alone-treated cells at each time point.</p>
Full article ">Figure 6
<p>Change in expression of osteogenesis-related genes by COX inhibitors in BMP-2-induced ST-2 MSCs. (<b>A</b>) ALP activity. ST2-MSCs were differentiated into osteoblasts for 6 d in the medium containing BMP-2 (50 ng/mL; gray columns) with or without CDCA (10 µM; hatched columns) or <b>FLG390</b> (10 µM; black columns) in the presence of each of indomethacin (1 µM; green columns), SC-560 (100 nM; yellow columns), and NS398 (1 µM; blue columns). The cells were pre-incubated with each COX inhibitor for 30 min before initiation of osteoblast differentiation. The data are shown as the mean ± S.D. from three independent experiments. # <span class="html-italic">p</span> &lt; 0.05, vs. vehicle; * <span class="html-italic">p</span> &lt; 0.05, vs. BMP-2 alone-treated cells in each treatment. <sup><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.05, vs. BMP-2- and <b>FLG390</b>-treated cells. (<b>B</b>) Expression of the osteogenesis-related genes. ST-2 MSCs were cultured as stated in the legend of <a href="#molecules-30-00058-f006" class="html-fig">Figure 6</a>A. The data are presented as the mean ± S.D. from three independent experiments. # <span class="html-italic">p</span> &lt; 0.05, vs. vehicle; * <span class="html-italic">p</span> &lt; 0.05, vs. BMP-2 alone-treated cells in each treatment. <sup><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.05, vs. BMP-2- and <b>FLG390</b>-treated cells.</p>
Full article ">Figure 7
<p>Expression of EP receptors during osteoblast differentiation in BMP-2-induced ST-2 MSCs. (<b>A</b>) Expression of the EP1 and EP4 receptor genes during osteoblast differentiation in BMP-2-induced ST-2 MSCs. The cells were differentiated into osteoblasts for 6 d in the medium containing BMP-2 (50 ng/mL; gray columns) with or without CDCA (10 µM; hatched columns) or <b>FLG390</b> (10 µM; black columns). The data are presented as the mean ± S.D. from three independent experiments. # <span class="html-italic">p</span> &lt; 0.05, vs. 0 h; * <span class="html-italic">p</span> &lt; 0.05, vs. BMP-2 alone-treated cells. (<b>B</b>) ALP activity. ST2-MSCs were differentiated into osteoblasts for 6 d in the medium containing BMP-2 (50 ng/mL; gray columns) with or without CDCA (10 µM; hatched columns) or <b>FLG390</b> (10 µM; black columns) in the presence or absence of GW848687X, an EP1 receptor antagonist (20 nM; purple columns) or ONO-AE3-208, an EP4 receptor antagonist (10 nM; orange columns). The cells were pre-incubated with each EP receptor antagonist for 30 min before the initiation of osteoblast differentiation. The data are shown as the mean ± S.D. from three independent experiments. # <span class="html-italic">p</span> &lt; 0.05, vs. 0 h; * <span class="html-italic">p</span> &lt; 0.05, vs. BMP-2 alone-treated cells; <sup><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.05, vs. BMP-2- and CDCA- or <b>FLG390</b>-treated cells in each treatment. (<b>C</b>) Expression level of the osteogenesis-related genes in the EP1 or EP4 receptor antagonist-treated BMP-2-induced ST-2 MSCs. The cells were cultured as described in the legend of <a href="#molecules-30-00058-f007" class="html-fig">Figure 7</a>B. The data are shown as the mean ± S.D. from three independent experiments. # <span class="html-italic">p</span> &lt; 0.05, vs. 0 h; * <span class="html-italic">p</span> &lt; 0.05, vs. BMP-2 alone-treated cells; <sup><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.05, vs. BMP-2- and CDCA- or <b>FLG390</b>-treated cells in each treatment.</p>
Full article ">Figure 8
<p>Phase-specific effect of inhibition of COX-2 or EP4 receptors during osteoblast differentiation in BMP-2- and FXR agonist-induced ST-2 MSCs. (<b>A</b>) Inhibition of PGE<sub>2</sub> production by NS398 in the presence or absence of BMP-2 and/or CDCA or <b>FLG390</b> in ST-2 MSCs. The cells were incubated for 6 h in the medium containing BMP-2 (50 ng/mL) with or without CDCA (10 µM), <b>FLG390</b> (10 µM), and NS398 (1 µM), and further incubated with A23187 (5 µM) for 15 min at 37 °C. # <span class="html-italic">p</span> &lt; 0.05, vs. 0 h; * <span class="html-italic">p</span> &lt; 0.05, vs. BMP-2 alone-treated cells; <sup><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.05. (<b>B</b>) Phase-specific effect of COX-2 inhibitor or EP4 receptor antagonist during the osteoblast differentiation of BMP-2- and FXR agonist-treated ST-2 MSCs. The cells (white columns) were differentiated into osteoblasts for 6 d in the medium containing BMP-2 (50 ng/mL; gray columns) with or without CDCA (10 μM) or <b>FLG390</b> (10 μM; black columns) in the presence or absence of NS398 (1 µM) or ONO-AE3-208 (10 nM) during 0–6 h or 0–6 d of 6-day osteoblast differentiation. The cells were pre-incubated with NS398 or ONO-AE3-208 for 30 min prior to the initiation of osteoblast differentiation. The data are shown as the mean ± S.D. from three independent experiments. # <span class="html-italic">p</span> &lt; 0.05, vs. 0 h; * <span class="html-italic">p</span> &lt; 0.05, vs. BMP-2 alone-treated cells; <sup><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.05, vs. BMP-2- and CDCA-treated cells, <sup>§</sup> <span class="html-italic">p</span> &lt; 0.05, vs. BMP-2- and <b>FLG390</b>-treated cells.</p>
Full article ">Scheme 1
<p>Reagents and conditions: (<b>a</b>) 10% Pd/C, H<sub>2</sub>, and methanol at room temperature for 1 h; (<b>b</b>) cyclopropanecarboxylic acid, 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (WSCI-HCl), HOAt (<sup>3</sup>H-[1,2,3]triazolo [4,5-b]pyridin-3-ol), and <span class="html-italic">N</span>,<span class="html-italic">N</span>-dimethylformamide (DMF) at room temperature for 15 h; (<b>c</b>) CH<sub>3</sub>COOH at 80 °C for 4 h; (<b>d</b>) 6 M HCl and dioxane at 90 °C for 1 h; (<b>e</b>) monomethyl terephthalate, HOAt, WSCI-HCl, and DMF at room temperature for 15 h; (<b>f</b>) 1 M NaOH and tetrahydrofuran (THF)/methanol at room temperature for 15 h.</p>
Full article ">
16 pages, 3960 KiB  
Article
Hypoxia Promotes Osteoclast Differentiation by Weakening USP18-Mediated Suppression on the NF-κB Signaling Pathway
by Xiaoxia Fan, Botong Li, Shengjun Chai, Rong Zhang, Chunmei Cai and Rili Ge
Int. J. Mol. Sci. 2025, 26(1), 10; https://doi.org/10.3390/ijms26010010 - 24 Dec 2024
Viewed by 244
Abstract
Osteoporosis, a prevalent metabolic bone disorder, is characterized by reduced bone density and increased fracture risk. The pathogenesis of osteoporosis is closely associated with an imbalance in bone remodeling, in which the resorption function of osteoclasts exceeds the formation function of osteoblasts. Hypoxia [...] Read more.
Osteoporosis, a prevalent metabolic bone disorder, is characterized by reduced bone density and increased fracture risk. The pathogenesis of osteoporosis is closely associated with an imbalance in bone remodeling, in which the resorption function of osteoclasts exceeds the formation function of osteoblasts. Hypoxia has been implicated in the promotion of osteoclast differentiation and the subsequent development of osteoporosis. The ubiquitin–proteasome system (UPS) and its regulatory enzymes, deubiquitinating enzymes (DUBs), play a significant role in bone homeostasis. In this study, we investigated the contribution and mechanism of Ubiquitin-specific protease 18 (USP18), a DUB, in osteoclast differentiation under hypoxic conditions. BMDMs and RAW264.7 cells were treated with RANKL to induce osteoclastogenesis and were subjected to overexpression or knockdown of USP18 under normoxic or hypoxia conditions. Osteoclast formation was assessed using TRAP staining, and the expression of osteoclast marker genes was determined using qRT-PCR. The activation of the NF-κB signaling pathway was evaluated using immunoblotting. We found that hypoxia significantly enhanced the differentiation of BMDMs and RAW264.7 cells into osteoclasts, accompanied by a notable downregulation of USP18 expression. The overexpression of USP18 inhibited RANKL-induced osteoclast differentiation, while the knockdown of USP18 promoted that process, unveiling the inhibitory effect of USP18 in osteoclastogenesis. Furthermore, the overexpression of USP18 rescued the hypoxia-induced increase in osteoclast differentiation. Mechanistic insights revealed that USP18 inhibits osteoclastogenesis by suppressing the NF-κB signaling pathway, with a potential target on TAK1 or its upstream molecules. This study indicates that hypoxia promotes osteoclast differentiation through the downregulation of USP18, which, in turn, relieves the suppression of the activation of the NF-κB signaling pathway. The USP18 emerges as a potential therapeutic target for osteoporosis treatment, highlighting the importance of the hypoxia–DUB axis in the pathogenesis of the disease. Full article
(This article belongs to the Section Molecular Endocrinology and Metabolism)
Show Figures

Figure 1

Figure 1
<p>Hypoxia significantly enhances the osteoclast differentiation of RAW264.7. (<b>A</b>–<b>C</b>) RAW264.7 cells were treated with 75 ng/mL RANKL for 0–4 days under normoxic (21% O<sub>2</sub>) or hypoxic (1% O<sub>2</sub>) conditions. Osteoclastogenesis was measured using tartrate-resistant acid phosphatase (TRAP) staining, and TRAP-positive multinucleated cells were considered mature osteoclasts. Scale bar, 200 μm. <span class="html-italic">n</span> = 5. (<b>D</b>–<b>G</b>) qRT-PCR detected the mRNA expression of osteoclast markers using RNA isolated from RAW264.7 cells cultured with or without 75 ng/mL RANKL for 2 days under normoxic (21% O<sub>2</sub>) or hypoxic (1% O<sub>2</sub>) conditions. The relative mRNA level of individual genes was expressed as the fold induction compared with no treatment (<span class="html-italic">n</span> = 3). Data are presented as mean ± SD. (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001, ns: not significant).</p>
Full article ">Figure 2
<p>Hypoxia dramatically suppresses the USP18 expression during osteoclast differentiation. RAW264.7 cells were treated with 75 ng/mL RANKL for 0 h, 24 h, 48 h, and 72 h under normoxic (21% O<sub>2</sub>) or hypoxic (1% O<sub>2</sub>) conditions. (<b>A</b>,<b>B</b>) A qRT-PCR analysis was performed for USP18 expression. (<b>C</b>) The protein level of USP18 was detected using Western blot. Data are presented as mean ± SD (<span class="html-italic">n</span> = 3) (** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001, ns: not significant).</p>
Full article ">Figure 3
<p>Overexpression of USP18 obviously inhibits RANKL-induced osteoclast differentiation. RAW264.7 cells were transfected with USP18 plasmid for overexpression, followed by treatment with 75 ng/mL RANKL for 0–4 days. (<b>A</b>–<b>C</b>) Osteoclastogenesis was measured using TRAP staining. Scale bar, 200 μm. <span class="html-italic">n</span> = 5. (<b>D</b>–<b>G</b>) qRT-PCR detected the mRNA expression of osteoclast markers using RNA isolated from RAW264.7 cells cultured in the presence of 75 ng/mL RANKL for 2 days (<span class="html-italic">n</span> = 3). Data are presented as mean ± SD (*** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 4
<p>Knockdown of USP18 remarkably promotes RANKL-induced osteoclast differentiation. RAW264.7 cells were transfected with siRNA for silencing USP18, followed by treatment with 75 ng/mL RANKL for 0–4 days. (<b>A</b>) Whole-cell lysates were immunoblotted with the indicated antibodies (<span class="html-italic">n</span> = 3). (<b>B</b>) qRT-PCR analysis was performed for USP18 expression (<span class="html-italic">n</span> = 3). (<b>C</b>–<b>E</b>) Osteoclastogenesis was measured using TRAP staining. Scale bar, 200 μm. <span class="html-italic">n</span> = 5. (<b>F</b>–<b>I</b>) qRT-PCR detected the mRNA expression of osteoclast markers using RNA isolated from RAW264.7 cells cultured in the presence of RANKL for 2 days (<span class="html-italic">n</span> = 3). Data are presented as mean ± SD (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">Figure 5
<p>USP18 rescues hypoxia-induced promotion of osteoclast differentiation. RAW264.7 cells were transfected with USP18 plasmid for overexpression and then treated with RANKL for 2 (qRT-PCR) or 3 (TRAP staining) days under normoxic (21% O<sub>2</sub>) or hypoxic (1% O<sub>2</sub>) conditions. (<b>A</b>–<b>C</b>) Osteoclastogenesis was measured using TRAP staining. Scale bar, 200 μm. <span class="html-italic">n</span> = 5. (<b>D</b>–<b>G</b>) qRT-PCR detected the mRNA expression of osteoclast markers (<span class="html-italic">n</span> = 3). Data are presented as mean ± SD (*** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001, ns: not significant).</p>
Full article ">Figure 6
<p>USP18 inhibits osteoclast differentiation by suppressing the NF-κB signaling pathway. (<b>A</b>–<b>C</b>) RAW264.7 cells were transfected with USP18 plasmid and siRNA for overexpression and silencing, respectively, followed by treatment with 75 ng/mL RANKL for 2 (<b>A</b>), 24 (<b>B</b>), and 48 (<b>C</b>) hours. Whole-cell lysates were immunoblotted with the indicated antibodies. Data are presented as mean ± SD, (<span class="html-italic">n</span> = 3) (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001).</p>
Full article ">
20 pages, 10526 KiB  
Article
Evaluation of Genotoxic Effects of N-Methyl-N-Nitroso-Urea and Etoposide on the Differentiation Potential of MSCs from Umbilical Cord Blood and Bone Marrow
by Meryem Ouzin, Sebastian Wesselborg, Gerhard Fritz and Gesine Kogler
Cells 2024, 13(24), 2134; https://doi.org/10.3390/cells13242134 - 23 Dec 2024
Viewed by 279
Abstract
The present study investigates the influence of nitrosamines and etoposide on mesenchymal stromal cells (MSCs) in a differentiation state- and biological age-dependent manner. The genotoxic effects of the agents on both neonatal and adult stem cell populations after treatment, before, or during the [...] Read more.
The present study investigates the influence of nitrosamines and etoposide on mesenchymal stromal cells (MSCs) in a differentiation state- and biological age-dependent manner. The genotoxic effects of the agents on both neonatal and adult stem cell populations after treatment, before, or during the course of differentiation, and the sensitivity of the different MSC types to different concentrations of MNU or etoposide were assessed. Hereby, the multipotent differentiation capacity of MSCs into osteoblasts, adipocytes, and chondrocytes was analyzed. Our findings reveal that while all cell types exhibit DNA damage upon exposure, neonatal CB-USSCs demonstrate enhanced resistance to genotoxic damage compared with their adult counterparts. Moreover, the osteogenic differentiation of MSCs was more susceptible to genotoxic damage, whereas the adipogenic and chondrogenic differentiation potentials did not show any significant changes upon treatment with genotoxin. Furthermore, we emphasize the cell-specific variability in responses to genotoxic damage and the differences in sensitivity and reaction across different cell types, thus advocating the consideration of these variabilities during drug testing and developmental biological research. Full article
(This article belongs to the Section Stem Cells)
Show Figures

Figure 1

Figure 1
<p>Cell viability and corresponding IC<sub>50</sub> values after MNU or etoposide treatment measured by resazurin reduction assay. Representative data from an MSC from each source and an iPSC cell line is shown (Mean ± SD from N = 2, n = 3). Abbreviations: CB, cord blood; BM, bone marrow; iPSC, induced pluripotent stem cell; MNU, N-methyl-N-nitroso-urea; USSC; unrestricted somatic stem cells.</p>
Full article ">Figure 2
<p>(<b>A</b>) Morphological changes in MSCs from different sources 72 h after treatment with 5 mM MNU for 1 h or 10 µM etoposide for 24 h in comparison with the untreated cells. Morphology was representatively analyzed 72 h after 5 mM MNU or 10 µM etoposide treatment. Scale bars = 500 µm. (<b>B</b>) Dose-dependent impact of genotoxic stress induced by MNU or etoposide on proliferation kinetics of CB-USSC, CB-MSC, and BM-MSC. To determine long-term growth kinetics, cells were exposed to different doses of MNU (untreated, 1 mM, 3 mM, and 5 mM) or etoposide (untreated, 1 µM, 5 µM, 10 µM), and cell numbers were counted after each passage. CPDs are shown (Mean ± SD from N = 3). (<b>C</b>) Analysis of DNA damage in MSCs after MNU or etoposide treatment by alkaline comet assay. For each condition, the intensity of 50 comets was assessed. CB-USSC2, CB-USSC3, CB-USSC4, CB-MSC1, CB-MSC2, CB-MSC3, CB-MSC4, BM-MSC2, BM-MSC3. Quantitative data are presented as means ± SD. * denotes <span class="html-italic">p</span> ≤ 0.05 and **** denotes <span class="html-italic">p</span> ≤ 0.0001. Abbreviations: BM, bone marrow; CB, cord blood; CPD, cumulative population doublings; MNU, N-methyl-N-nitroso-urea; ns, no significance; USSC; unrestricted somatic stem cells.</p>
Full article ">Figure 3
<p>(<b>A</b>) Osteogenic differentiation of MSCs after MNU treatment on d0. Representative data (CB-USSC3-CB-MSC3-BM-MSC1) after Alizarin Red S staining on d14 of osteogenic differentiation is shown. Scale bars = 500 µm. (<b>B</b>) Osteogenic differentiation of MSCs after 1 h MNU treatment with different doses (untreated, 1 mM, 3 mM, and 5 mM) on d0. Quantitative determination of Alizarin Red S staining on d14 of osteogenic differentiation is shown. Quantitative data are presented as means ± SD of N = 3, n = 3. Differences were considered significant at <span class="html-italic">p</span> ≤ 0.05. * denotes <span class="html-italic">p</span> ≤ 0.05, ** denotes <span class="html-italic">p</span> ≤ 0.01 and **** denotes <span class="html-italic">p</span> ≤ 0.0001. Abbreviations: BM, bone marrow; CB, cord blood; d, day; MNU, N-methyl-N-nitroso-urea; USSC; unrestricted somatic stem cells.</p>
Full article ">Figure 4
<p>(<b>A</b>) Adipogenic differentiation of MSCs after MNU treatment on d0. Representative data (CB-MSC3-BM-MSC4) after Oil Red O staining on d21 of adipogenic differentiation is shown. The induced samples successfully underwent adipogenesis, while the uninduced sample was devoid of characteristic lipid droplet formation after 21 days of differentiation. Scale bars = 500 µm. (<b>B</b>) Adipogenic differentiation of MSCs after 1 h MNU treatment with different doses (untreated, 1 mM, 3 mM, and 5 mM) on d0. Quantitative determination of Oil Red O staining on d21 of adipogenic differentiation is shown. Quantitative data are presented as means ± SD of N = 3, n = 3. Differences were considered significant at <span class="html-italic">p</span> ≤ 0.05. ** denotes <span class="html-italic">p</span> ≤ 0.01, and ns denotes non-significant. Abbreviations: BM, bone marrow; CB, cord blood; d; day; MNU, N-methyl-N-nitroso-urea.</p>
Full article ">Figure 5
<p>Chondrogenic differentiation of MSCs after 1 h MNU treatment on d0 of differentiation. Areas and diameters of the chondrogenic pellets were measured on d7, d14, and d21 of chondrogenic differentiation (Mean + SD N = 3, n = 3). Abbreviations: BM, bone marrow; CB, cord blood; d; day; MNU, N-methyl-N-nitroso-urea; USSC; unrestricted somatic stem cells.</p>
Full article ">Figure 6
<p>(<b>A</b>) Osteogenic differentiation of MSCs after 24 h etoposide treatment on d0. Representative data (CB-USSC4-CB-MSC2- BM-MSC1) after Alizarin Red S staining on d14 of osteogenic differentiation is shown. Scale bars = 500 µm. (<b>B</b>) Osteogenic differentiation of MSCs after 24 h etoposide treatment with different doses (untreated, 1 µM, 5 µM, and 10 µM) on d0. Quantitative determination of Alizarin Red S staining on d14 of osteogenic differentiation is shown. Quantitative data are presented as means ± SD of N = 3, n = 3. Differences were considered significant at <span class="html-italic">p</span> ≤ 0.05. * denotes <span class="html-italic">p</span> ≤ 0.05, ** denotes <span class="html-italic">p</span> ≤ 0.01, **** denotes <span class="html-italic">p</span> ≤ 0.0001 and ns denotes non-significant. Abbreviations: BM, bone marrow; CB, cord blood; USSC, unrestricted somatic stem cells.</p>
Full article ">Figure 7
<p>(<b>A</b>) Adipogenic differentiation of MSCs after 24 h etoposide treatment on d0. Representative data (CB-MSC2-BM-MSC2) after Oil Red O staining on d21 of adipogenic differentiation is shown. The induced samples successfully underwent adipogenesis, while the uninduced sample was devoid of characteristic lipid droplet formation after 21 days of differentiation. Scale bars = 1 mm. (<b>B</b>) Adipogenic differentiation of MSCs after 24 h etoposide treatment with different doses (untreated, 1 µM, 5 µM, and 10 µM) on d0. Quantitative determination of Oil Red O staining on d21 of adipogenic differentiation is shown. Quantitative data are presented as means ± SD of N = 3, n = 3. Differences were considered significant at <span class="html-italic">p</span> ≤ 0.05. ** denotes <span class="html-italic">p</span> ≤ 0.01, and ns denotes non-significant. Abbreviations: BM, bone marrow; CB, cord blood.</p>
Full article ">Figure 8
<p>Chondrogenic differentiation of MSCs after 24 h etoposide treatment on d0 of differentiation. Areas and diameters of the chondrogenic pellets were measured on d7, d14, and d21 of chondrogenic differentiation (Mean + SD N = 3, n = 3). Abbreviations: BM, bone marrow; CB, cord blood; d; day; MNU, N-methyl-N-nitroso-urea; USSC; unrestricted somatic stem cells.</p>
Full article ">Figure 9
<p>(<b>A</b>) Osteogenic differentiation of MSCs after 1 h MNU treatment on d7. Representative data (CB-USSC4-CB-MSC4-BM-MSC4) after Alizarin Red S staining on d14 of osteogenic differentiation is shown. Scale bars = 500 µm. (<b>B</b>) Osteogenic differentiation of MSCs after 1 h MNU treatment with different doses (untreated, 1 mM, 3 mM, and 5 mM) on d7. Quantitative determination of Alizarin Red S staining on d14 of osteogenic differentiation is shown. Quantitative data are presented as means ± SD of N = 3, n = 3. Differences were considered significant at <span class="html-italic">p</span> ≤ 0.05. * denotes <span class="html-italic">p</span> ≤ 0.05, ** denotes <span class="html-italic">p</span> ≤ 0.01, *** denotes <span class="html-italic">p</span> ≤ 0.001 and ns denotes non-significant. Abbreviations: BM, bone marrow; CB, cord blood; d, day; MNU, N-methyl-N-nitroso-urea; USSC, unrestricted somatic stem cells.</p>
Full article ">Figure 10
<p>(<b>A</b>) Adipogenic differentiation of MSCs after 1 h MNU treatment on d7. Representative data (CB-MSC3-BM-MSC1) after Oil Red O staining on d21 of adipogenic differentiation is shown. The induced samples successfully underwent adipogenesis, while the uninduced sample was devoid of characteristic lipid droplet formation after 21 days of differentiation. Scale bars = 500 µm or 1 mm. (<b>B</b>) Adipogenic differentiation of MSCs after 1 h MNU treatment with different doses (untreated, 1 mM, 3 mM, and 5 mM) on d7. Quantitative determination of Oil Red O staining on d21 of adipogenic differentiation is shown. Quantitative data are presented as means ± SD of N = 3, n = 3. Differences were considered significant at <span class="html-italic">p</span> ≤ 0.05. ** denotes <span class="html-italic">p</span> ≤ 0.01, and ns denotes non-significant. Abbreviations: BM, bone marrow; CB, cord blood; d, day; MNU, N-methyl-N-nitroso-urea.</p>
Full article ">Figure 11
<p>Chondrogenic differentiation of MSCs after 1 h MNU treatment on d7 of differentiation. Areas and diameters of the chondrogenic pellets were measured on d7, d14, and d21 of chondrogenic differentiation (Mean + SD N = 3, n = 3). Abbreviations: BM, bone marrow; CB, cord blood; d; day; MNU, N-methyl-N-nitroso-urea; USSC; unrestricted somatic stem cells.</p>
Full article ">Figure 12
<p>(<b>A</b>) Osteogenic differentiation of MSCs after 24 h etoposide treatment on d7. Representative data (CB-USSC4-CB-MSC3-BM-MSC6) after Alizarin Red S staining on d14 of osteogenic differentiation is shown. Scale bars = 1 mm. (<b>B</b>) Osteogenic differentiation of MSCs after 24 h etoposide treatment with different doses (untreated, 1 µM, 5 µM, and 10 µM) on d7. Quantitative determination of Alizarin Red S staining on d14 of osteogenic differentiation is shown. Quantitative data are presented as means ± SD of N = 3, n = 3. Differences were considered significant at <span class="html-italic">p</span> ≤ 0.05. ns denotes non-significant. Abbreviations: BM, bone marrow; CB, cord blood; d, day; USSC, unrestricted somatic stem cells.</p>
Full article ">Figure 13
<p>Adipogenic differentiation of MSCs after 24 h etoposide treatment on d7. (<b>A</b>) Representative data (CB-MSC3-BM-MSC2) after Oil Red O staining on d21 of adipogenic differentiation is shown. The induced samples successfully underwent adipogenesis, while the uninduced sample was devoid of characteristic lipid droplet formation after 21 days of differentiation. Scale bars = 500 µm or 1 mm. (<b>B</b>) Adipogenic differentiation of MSCs after 24 h etoposide treatment with different doses (untreated, 1 µM, 5 µM, and 10 µM) on d7. Quantitative determination of Oil Red O staining on d21 of adipogenic differentiation is shown. Quantitative data are presented as means ± SD of N = 3, n = 3. ns denotes non-significant. Abbreviations: BM, bone marrow; CB, cord blood; d, day.</p>
Full article ">Figure 14
<p>Chondrogenic differentiation of MSCs after 24 h etoposide treatment on d7 of differentiation. Areas and diameters of the chondrogenic pellets were measured on d7, d14, and d21 of chondrogenic differentiation (Mean + SD N = 3, n = 3). Abbreviations: BM, bone marrow; CB, cord blood; d, day; USSC, unrestricted somatic stem cell.</p>
Full article ">
22 pages, 2869 KiB  
Article
Investigation of Oxidative-Stress Impact on Human Osteoblasts During Orthodontic Tooth Movement Using an In Vitro Tension Model
by Samira Hosseini, Julia Diegelmann, Matthias Folwaczny, Hisham Sabbagh, Sven Otto, Tamara Katharina Kakoschke, Andrea Wichelhaus, Uwe Baumert and Mila Janjic Rankovic
Int. J. Mol. Sci. 2024, 25(24), 13525; https://doi.org/10.3390/ijms252413525 - 17 Dec 2024
Viewed by 419
Abstract
In recent years, there has been a growing number of adult orthodontic patients with periodontal disease. The progression of periodontal disease is well-linked to oxidative stress (OS). Nevertheless, the impact of OS on orthodontic tooth movement (OTM) is not fully clarified. Therefore, we [...] Read more.
In recent years, there has been a growing number of adult orthodontic patients with periodontal disease. The progression of periodontal disease is well-linked to oxidative stress (OS). Nevertheless, the impact of OS on orthodontic tooth movement (OTM) is not fully clarified. Therefore, we applied an OS in vitro-model utilizing H2O2 to study its effect on tension-induced mechanotransduction in human osteoblasts (hOBs). Experimental parameters were established based on cell viability and proliferation. Apoptosis detection was based on caspase-3/7 activity. Gene expression related to bone-remodeling (RUNX2, P2RX7, TNFRSF11B/OPG), inflammation (CXCL8/IL8, IL6, PTRGS2/COX2), autophagy (MAP1LC3A/LC3, BECN1), and apoptosis (CASP3, CASP8) was analyzed by RT-qPCR. IL6 and PGE2 secretion were determined by ELISA. Tension increased the expression of PTRGS2/COX2 in all groups, especially after stimulation with higher H2O2 concentration. This corresponds also to the measured PGE2 concentrations. CXCL8/IL8 was upregulated in all groups. Cells subjected to tension alone showed a general upregulation of osteogenic differentiation-related genes; however, pre-stimulation with OS did not induce significant changes especially towards downregulation. MAP1LC3A/LC3, BECN1 and CASP8 were generally upregulated in cells without OS pre-stimulation. Our results suggest that OS might have considerable impacts on cellular behavior during OTM. Full article
Show Figures

Figure 1

Figure 1
<p>Oxidative stress in vitro-model utilizing H<sub>2</sub>O<sub>2</sub> [<a href="#B21-ijms-25-13525" class="html-bibr">21</a>] and tensile strain [<a href="#B20-ijms-25-13525" class="html-bibr">20</a>] to study its effect on mechanotransduction in primary human alveolar osteoblasts.</p>
Full article ">Figure 2
<p>Effects of different hydrogen peroxide concentrations ranging from 0 µM (i.e., control) to 500 µM on apoptosis induction (<b>upper row</b>) and cell viability (<b>lower row</b>) on primary human osteoblasts. <b>Upper row:</b> Apoptosis induction was detected using the CellEvent™ Caspase-3/7 Detection Reagent (R37111; Life Technologies, Carlsbad, CA, USA). Caspase-3/7-positive cells were stained green (overlay of fluorescence and phase contrast). <b>Lower row:</b> Cell viability assessment using live/dead cell staining. Green cells represent living cells. Dead cells are either detached and washed away or stained with the red color. (Scale: 200 µm).</p>
Full article ">Figure 3
<p>Percentage reduction of resazurin: (<b>a</b>) Cytotoxic effect of H<sub>2</sub>O<sub>2</sub>, (<b>b</b>) cell viability calculated as normalized resazurin reduction relative to the control group. 50 µM and 100 µM were identified as the lowest concentrations of H<sub>2</sub>O<sub>2</sub> showing a cytotoxic effect; however, these did not have pronounced effects on cell viability.</p>
Full article ">Figure 4
<p>Effect of oxidative stress induction alone on gene expression immediately after H₂O₂ incubation (“direct”) and 24 h post-incubation (“recovery”). (<b>a</b>) Experimental design. (<b>b</b>–<b>k</b>) RT-qPCR results for genes related to autophagy (<b>b</b>,<b>c</b>, <span class="html-italic">MAP1LC3A</span>/<span class="html-italic">LC3</span>, <span class="html-italic">BECN1</span>), apoptosis (<b>d</b>,<b>e</b>, <span class="html-italic">CASP3</span>, <span class="html-italic">CASP8</span>), inflammation (<b>f</b>–<b>h</b>, <span class="html-italic">CXCL2/IL8</span>, <span class="html-italic">IL6</span>, <span class="html-italic">PTGS2/COX2</span>), and bone remodeling (<b>i</b>–<b>k</b>, <span class="html-italic">RUNX2</span>, <span class="html-italic">P2RX7</span>, <span class="html-italic">TNFRSF11B</span>/<span class="html-italic">OPG</span>). For each genetic locus, gene expression directly after H<sub>2</sub>O<sub>2</sub> exposure (left panel, “direct”) and after an additional 24 h cultivation in H<sub>2</sub>O<sub>2</sub>-free cell culture medium (right panel, “recovery”) is depicted. Adjusted <span class="html-italic">p</span>-values based on multiple comparisons within each experimental group are reported: *, <span class="html-italic">p</span><sub>adj</sub>. &lt; 0.05; **, <span class="html-italic">p</span><sub>adj</sub>. &lt; 0.01; ***, <span class="html-italic">p</span><sub>adj</sub>. &lt; 0.001.</p>
Full article ">Figure 5
<p>Expressions of genes and metabolites related to inflammation and bone remodeling in mechanically stimulated cells with and without previous H<sub>2</sub>O<sub>2</sub> stimulation. (<b>a</b>) Experimental setup: the control group (ctrl) received neither H<sub>2</sub>O<sub>2</sub> nor tension stimulation. The tension (T10%, T15%) group was stimulated by static tension after 24 h non-stimulation. The H<sub>2</sub>O<sub>2</sub>/tension group was stimulated for 24 h with 50 µM or 100 µM H<sub>2</sub>O<sub>2</sub> followed by 24 h static tension at 10% or 15% stretching. Shown here are the results from 15% tension stimulation. (<b>b</b>–<b>f</b>) The expression of inflammation-related genes and metabolites and (<b>g</b>–<b>i</b>) genes related to bone remodeling are reported. Shown are the adjusted <span class="html-italic">p</span>-values based on multiple comparisons between each experimental treatment (*, <span class="html-italic">p</span><sub>adj</sub>. &lt; 0.05; **, <span class="html-italic">p</span><sub>adj</sub>. &lt; 0.01; ***, <span class="html-italic">p</span><sub>adj</sub>. &lt; 0.001). Results derived from 10% tension are reported in <a href="#app1-ijms-25-13525" class="html-app">Supplementary Materials S1</a>.</p>
Full article ">Figure 6
<p>RT-qPCR results for autophagy- (<b>a</b>,<b>b</b>) and apoptosis (<b>c</b>,<b>d</b>)-related genes. Shown are the adjusted <span class="html-italic">p</span>-values based on multiple comparisons between each experimental treatment. The groups are the same as in <a href="#ijms-25-13525-f005" class="html-fig">Figure 5</a>. *, <span class="html-italic">p</span><sub>adj</sub>. &lt; 0.05; **, <span class="html-italic">p</span><sub>adj</sub>. &lt; 0.01; ***, <span class="html-italic">p</span><sub>adj</sub>. &lt; 0.001.</p>
Full article ">Figure 7
<p>Establishment of a standard curve to assess cell growth of the hOBs used in the experiments. The resazurin standard curve was prepared as described in materials and methods. hOBs of the 5th passage were seeded in triplicate (70,000; 100,000; 200,000; 300,000; 400,000; 500,000) or duplicate (800,000 and 900,000 cells per well). Exponential regression was used to calculate the standard curve (red line) (Microsoft Excel). The cellular growth of hOBs in the different experimental setups (legend: lower right) is shown with violet diamonds (♦) on the fitted curve.</p>
Full article ">Figure 8
<p>Qualitative assessment of cell viability of cells belonging to the different experimental groups using live/dead cell staining. Independent of the experimental group, cells proved to be viable (green staining). Dead cells were rarely observed (red staining). (Scale bar: 200 μm). (Data from experiments with 10% tension are provided in <a href="#app1-ijms-25-13525" class="html-app">Supplementary Materials S1</a>.</p>
Full article ">Figure 9
<p>Reference gene selection was obtained with RefFinder. (<b>a</b>) Cq values for the panel of reference genes. Six quantitative real-time polymerase chain reaction (qPCR) runs were analyzed representing three biological replicates and two technical replicates each (<a href="#app1-ijms-25-13525" class="html-app">Supplementary Materials S3</a>). (<b>b</b>) Analysis of comprehensive gene stability for the panel of reference genes. Lower values indicate higher gene stability (<a href="#app1-ijms-25-13525" class="html-app">Supplementary Materials S3</a>).</p>
Full article ">
11 pages, 1775 KiB  
Article
The Role of Formononetin in Osteoblast Function and Mineralization Potential with Deproteinized Bovine Bone Material
by Ebru Haciosmanoglu Aldogan, Deniz Başaran, Bilgin Öner and Başak Günçer
Curr. Issues Mol. Biol. 2024, 46(12), 14215-14225; https://doi.org/10.3390/cimb46120851 - 17 Dec 2024
Viewed by 390
Abstract
Objectives: Dental bone formation involves various cellular and molecular mechanisms, and phytoestrogens such as formononetin (FORM) are promising because of their estrogenic, anti-inflammatory, and antioxidant effects. This study investigated the effect of FORM on osteoblast proliferation, differentiation, and mineralization in combination with spongiosa [...] Read more.
Objectives: Dental bone formation involves various cellular and molecular mechanisms, and phytoestrogens such as formononetin (FORM) are promising because of their estrogenic, anti-inflammatory, and antioxidant effects. This study investigated the effect of FORM on osteoblast proliferation, differentiation, and mineralization in combination with spongiosa granulates (BO) in vitro. Materials and Methods: Human fetal osteoblast cells (hFOB1.19) were treated with increasing concentrations of FORM (1, 10, and 100 µg/mL), BO, or their combination. Cell proliferation was assessed using a MTT assay. Alkaline phosphatase (ALP) activity, intracellular Ca2+, and Pi levels were measured using ELISA. Vascular endothelial growth factor (VEGF) and osteocalcin expression levels were analyzed by western blotting. Results: Cell proliferation increased with FORM, with or without BO, after 6 days (p < 0.001). FORM and BO had a synergistic effect on ALP activity (p < 0.001). Intracellular Ca2+ and Pi levels were highest in the BO-FORM group, suggesting superior mineralization (p < 0.05). VEGF and osteocalcin expression was significantly upregulated with FORM, alone and with BO (p < 0.05), indicating improved angiogenesis and bone maturation over 9 days. Conclusions: FORM enhances osteoblast proliferation, differentiation, and mineralization potential, particularly in BO spongiosa granulates. These data support the in vitro potential of formononetin-phytoestrogen in promoting osteoblast differentiation and mineralization potential with BO. These findings suggest that FORM, combined with BO, could improve bone augmentation in clinical applications such as maxillofacial surgery. FORM shows valuable potential for clinical applications, such as maxillofacial surgery, by promoting faster and more effective healing. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

Figure 1
<p>Cell proliferation of osteoblasts on days 1, 6, and 9. The control (0 μM) was set at 100%. Each sample was measured in triplicates. Values represent the mean ± SEM of three independent experiments. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared with the control; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with BO.</p>
Full article ">Figure 2
<p>Effects of FORM on Alkaline phosphatase (ALP) activity on days 1, 6, and 9. Values are means ± SEM of three independent experiments. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared with the control; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with BO.</p>
Full article ">Figure 3
<p>Intracellular calcium (Ca<sup>2+</sup>) (<b>A</b>) and phosphate (Pi) (<b>B</b>) levels in osteoblasts treated with FORM and BO+FORM. Values are means ± SEM of three independent experiments. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared with the control; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with BO.</p>
Full article ">Figure 4
<p>Effects of FORM on Vascular endothelial growth factor (VEGF) and osteocalcin expression after 9 days. (<b>A</b>) Western blot analysis. (<b>B</b>) Protein expression levels of VEGF and osteocalcin were analyzed using ImageJ software. Values are expressed as means ± SEM of three experiments. Values are mean ± SEM of three independent experiments. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared with the control; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with BO.</p>
Full article ">
18 pages, 31117 KiB  
Article
Synergistic Effects of Photobiomodulation and Differentiation Inducers on Osteogenic Differentiation of Adipose-Derived Stem Cells in Three-Dimensional Culture
by Daniella Da Silva, Anine Crous and Heidi Abrahamse
Int. J. Mol. Sci. 2024, 25(24), 13350; https://doi.org/10.3390/ijms252413350 (registering DOI) - 12 Dec 2024
Viewed by 403
Abstract
Osteoporosis, a common metabolic bone disorder, leads to increased fracture risk and significant morbidity, particularly in postmenopausal women and the elderly. Traditional treatments often fail to fully restore bone health and may cause side effects, prompting the exploration of regenerative therapies. Adipose-derived stem [...] Read more.
Osteoporosis, a common metabolic bone disorder, leads to increased fracture risk and significant morbidity, particularly in postmenopausal women and the elderly. Traditional treatments often fail to fully restore bone health and may cause side effects, prompting the exploration of regenerative therapies. Adipose-derived stem cells (ADSCs) offer potential for osteoporosis treatment, but their natural inclination toward adipogenic rather than osteogenic differentiation poses a challenge. This study investigates a novel approach combining differentiation inducers (DIs), three-dimensional (3D) hydrogel scaffolds, and photobiomodulation (PBM) to promote osteogenic differentiation of immortalised ADSCs. A dextran-based 3D hydrogel matrix, supplemented with a DI cocktail of dexamethasone, β-glycerophosphate disodium, and ascorbic acid, was used to foster osteogenesis. PBM was applied using near-infrared (825 nm), green (525 nm), and combined wavelengths at fluences of 3 J/cm2, 5 J/cm2, and 7 J/cm2 to enhance osteogenic potential. Flow cytometry identified osteoblast-specific markers, while inverted light microscopy evaluated cellular morphology. Reactive oxygen species assays measured oxidative stress, and quantitative polymerase chain reaction (qPCR) revealed upregulated gene expression linked to osteogenesis. The findings demonstrate that integrating DIs, 3D hydrogels, and PBM effectively drives osteogenic differentiation in immortalised ADSCs. The PBM enhanced osteogenic marker expression, induced morphological changes, and upregulated gene activity, presenting a promising framework for bone regeneration. Future research should assess the stability and functionality of these differentiated cells and explore their applicability in preclinical models of bone injury or degeneration. This integrative approach demonstrated specific efficacy in promoting the osteogenic differentiation of ADSCs, highlighting its potential application in developing targeted treatments for osteoporosis. Full article
(This article belongs to the Special Issue Regenerative Medicine: Biomaterials and Stem Cell Research)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Flow cytometric analysis of CD90 marker expression in immortalised adipose-derived mesenchymal stem cells following photobiomodulation treatment. (<b>a</b>) At 24 h post-treatment, a statistically significant increase in CD90 expression was observed in the G 5 J/cm<sup>2</sup> group (<span class="html-italic">p</span> &lt; 0.0001). (<b>b</b>) At 7 days post-treatment, CD90 expression declined in the G 5 J/cm<sup>2</sup> group, while a statistically significant increase was noted in the NIR 7 J/cm<sup>2</sup> group (<span class="html-italic">p</span> &lt; 0.0001) compared to the control. The data are expressed as mean ± SE. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001. Black stars (*) indicate comparisons between the specified samples and the standard group. Blue stars (<span style="color:#0432FF">*</span>) denote comparisons between the experimental samples and the control group. Comparisons among the experimental PBM groups are marked with orange stars (<span style="color:#ED7D31">*</span>). The sample size was n = 3.</p>
Full article ">Figure 2
<p>Flow cytometric analysis of RUNX2 expression in immortalised adipose-derived mesenchymal stem cells following photobiomodulation treatment. (<b>a</b>) At 24 h post-treatment, RUNX2 expression showed a statistically significant increase in the NIR wavelength group at 3 J/cm<sup>2</sup> and 7 J/cm<sup>2</sup> fluencies (<span class="html-italic">p</span> &lt; 0.05) compared to the NIR 5 J/cm<sup>2</sup>, G 3 J/cm<sup>2</sup>, and G 7 J/cm<sup>2</sup> groups. (<b>b</b>) At 7 days post-treatment, RUNX2 expression significantly increased in the NIR 7 J/cm<sup>2</sup> group compared to the NIR 3 J/cm<sup>2</sup> (<span class="html-italic">p</span> &lt; 0.0001), NIR 5 J/cm<sup>2</sup> (<span class="html-italic">p</span> &lt; 0.05), and G 7 J/cm<sup>2</sup> (<span class="html-italic">p</span> &lt; 0.05) groups. The data are expressed as mean ± SE. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001. Black stars (*) indicate comparisons between the specified samples and the standard group. Comparisons among the experimental PBM groups are marked with orange stars (<span style="color:#ED7D31">*</span>). The sample size was n = 3.</p>
Full article ">Figure 3
<p>Flow cytometric analysis of BGLAP expression in immortalised adipose-derived mesenchymal stem cells following photobiomodulation treatment. (<b>a</b>) At 24 h post-treatment, BGLAP expression was significantly increased in the NIR 5 J/cm<sup>2</sup> group compared to the NIR 3 J/cm<sup>2</sup> and NIR 7 J/cm<sup>2</sup> groups (<span class="html-italic">p</span> &lt; 0.01). (<b>b</b>) At 7 days post-treatment, no statistically significant BGLAP expression was observed in any experimental group. The data are expressed as mean ± SE. ** <span class="html-italic">p</span> &lt; 0.01. Black stars (*) indicate comparisons between the specified samples and the standard group. The sample size was n = 3.</p>
Full article ">Figure 4
<p>Flow cytometric analysis of BGN expression in immortalised adipose-derived mesenchymal stem cells following photobiomodulation treatment. (<b>a</b>) At 24 h post-treatment, BGN expression significantly increased in the G 5 J/cm<sup>2</sup> group compared to the G 3 J/cm<sup>2</sup>, G 7 J/cm<sup>2</sup>, NIR 5 J/cm<sup>2</sup>, and NIR-G 5 J/cm<sup>2</sup> groups (<span class="html-italic">p</span> &lt; 0.0001). (<b>b</b>) At 7 days post-treatment, a significant increase in BGN expression was observed in the NIR 5 J/cm<sup>2</sup> group compared to the G 5 J/cm<sup>2</sup> (<span class="html-italic">p</span> &lt; 0.05) and NIR-G 5 J/cm<sup>2</sup> (<span class="html-italic">p</span> &lt; 0.0001) groups. Additionally, BGN expression in the NIR-G 7 J/cm<sup>2</sup> group was significantly higher compared to the NIR 7 J/cm<sup>2</sup> and G 7 J/cm<sup>2</sup> groups (<span class="html-italic">p</span> &lt; 0.0001). The data are expressed as mean ± SE. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001. Black stars (*) indicate comparisons between the specified samples and the standard group. Blue stars (<span style="color:#0432FF">*</span>) denote comparisons between the experimental samples and the control group. Comparisons among the experimental PBM groups are marked with orange stars (<span style="color:#ED7D31">*</span>). The sample size was n = 3.</p>
Full article ">Figure 5
<p>Flow cytometric analysis of SOST expression in immortalised adipose-derived mesenchymal stem cells following photobiomodulation treatment. (<b>a</b>) At 24 h post-treatment, SOST expression showed a statistically significant increase in the G 5 J/cm<sup>2</sup> and NIR-G 5 J/cm<sup>2</sup> groups compared to the NIR 5 J/cm<sup>2</sup> (<span class="html-italic">p</span> &lt; 0.001), G 3 J/cm<sup>2</sup>, G 7 J/cm<sup>2</sup> (<span class="html-italic">p</span> &lt; 0.001), and NIR-G 3 J/cm<sup>2</sup> and NIR-G 7 J/cm<sup>2</sup> groups (<span class="html-italic">p</span> &lt; 0.0001). (<b>b</b>) At 7 days post-treatment, SOST expression remained significantly elevated in the G 5 J/cm<sup>2</sup> group compared to the NIR-G 5 J/cm<sup>2</sup> (<span class="html-italic">p</span> &lt; 0.05), G 3 J/cm<sup>2</sup> (<span class="html-italic">p</span> &lt; 0.0001), and G 7 J/cm<sup>2</sup> groups (<span class="html-italic">p</span> &lt; 0.0001). The data are expressed as mean ± SE. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001. Black stars (*) indicate comparisons between the specified samples and the standard group. Blue stars (<span style="color:#0432FF">*</span>) denote comparisons between the experimental samples and the control group. Comparisons among the experimental PBM groups are marked with orange stars (<span style="color:#ED7D31">*</span>). The sample size was n = 3.</p>
Full article ">Figure 6
<p>Morphological characterisation of immortalised adipose-derived mesenchymal stem cell differentiation at 24 h and 7 days following photobiomodulation treatment, observed with inverted light microscopy at 10× magnification.</p>
Full article ">Figure 7
<p>Increased intracellular reactive oxygen species production in immortalised adipose-derived mesenchymal stem cells at 24 h and 7 days following photobiomodulation treatment. (<b>a</b>) At 24 h post-treatment, all experimental groups exhibited statistically significant increases in reactive oxygen species levels compared to the control group (<span class="html-italic">p</span> &lt; 0.0001), with the NIR-G combined wavelength group at 7 J/cm<sup>2</sup> showing a notable increase (<span class="html-italic">p</span> &lt; 0.05) compared to the NIR group at 7 J/cm<sup>2</sup>. (<b>b</b>) At 7 days post-treatment, significant increases in reactive oxygen species levels were observed in the NIR group at 5 J/cm<sup>2</sup> (<span class="html-italic">p</span> &lt; 0.05), the G group at 5 J/cm<sup>2</sup> (<span class="html-italic">p</span> &lt; 0.01) and 7 J/cm<sup>2</sup> (<span class="html-italic">p</span> &lt; 0.001), and the NIR-G group at 7 J/cm<sup>2</sup> (<span class="html-italic">p</span> &lt; 0.05), compared to the control group. The data are expressed as mean ± SE. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001. Black stars (*) indicate comparisons between the specified samples and the standard group. Blue stars (<span style="color:#0432FF">*</span>) denote comparisons between the experimental samples and the control group. Comparisons among the experimental PBM groups are marked with orange stars (<span style="color:#ED7D31">*</span>). The sample size was n = 3.</p>
Full article ">Figure 8
<p>Quantitative PCR analysis of osteogenic gene expression. (<b>a</b>) At 24 h post-photobiomodulation treatment, CD90 was significantly downregulated (0.5-fold change) at a combined wavelength of 3 J/cm<sup>2</sup>, indicating a shift towards osteogenic differentiation. RUNX2 exhibited a pronounced upregulation, with a maximum fold change of 4.5 at 825 nm and 7 J/cm<sup>2</sup>, and additional increases of 2.3-fold at 525 nm with 3 J/cm<sup>2</sup> and 3.0-fold at 525 nm with 5 J/cm<sup>2</sup>. BGLAP was significantly upregulated (2.5-fold change) at 5 J/cm<sup>2</sup>, suggesting an early commitment to the osteogenic lineage. (<b>b</b>) At 7 days post-PBM treatment, CD90 showed notable upregulation at various PBM dosages, particularly at 825 nm with 3 J/cm<sup>2</sup> (2.4-fold change) and 7 J/cm<sup>2</sup> (4.4-fold change), as well as at 525 nm with 5 J/cm<sup>2</sup> (4.2-fold change). RUNX2 remained upregulated, with a 3.6-fold change at 825 nm and 3 J/cm<sup>2</sup> and a 3.9-fold change at 825 nm and 7 J/cm<sup>2</sup>. BGLAP continued to increase (2.6-fold change) at 825 nm and 7 J/cm<sup>2</sup>, while BGN showed significant upregulation (3.0-fold change) at 825 nm and 7 J/cm<sup>2</sup>. Additionally, SOST was upregulated (2.1-fold change) at 825 nm and 7 J/cm<sup>2</sup> and (2.8-fold change) at 525 nm and 5 J/cm<sup>2</sup>.</p>
Full article ">Figure 9
<p>Photobiomodulation treatment experimental design.</p>
Full article ">Figure 10
<p>Flow diagram of experimental design and methodology.</p>
Full article ">
16 pages, 2996 KiB  
Article
Lutein Maintains Bone Mass In Vitro and In Vivo Against Disuse-Induced Bone Loss in Hindlimb-Unloaded Mice
by Yuki Tanaka, Tsukasa Tominari, Masaru Takatoya, Daichi Arai, Moe Sugasaki, Ryota Ichimaru, Chisato Miyaura, Chiho Matsumoto, Sihui Ma, Katsuhiko Suzuki, Michiko Hirata, Florian M. W. Grundler and Masaki Inada
Nutrients 2024, 16(24), 4271; https://doi.org/10.3390/nu16244271 - 11 Dec 2024
Viewed by 505
Abstract
Background: Lutein, a carotenoid, exhibits various biological activities such as maintaining the health of the eye, skin, heart, and bone. Recently, we found that lutein has dual roles in suppressing bone resorption and promoting bone formation. In this study, we examined the effects [...] Read more.
Background: Lutein, a carotenoid, exhibits various biological activities such as maintaining the health of the eye, skin, heart, and bone. Recently, we found that lutein has dual roles in suppressing bone resorption and promoting bone formation. In this study, we examined the effects of lutein in a disuse-induced osteoporosis model using hindlimb-unloaded (HLU) mice. Methods: Osteoclast differentiation was assessed by coculturing mouse primary osteoblasts and bone marrow cells or culturing a mouse osteoclast precursor cell line. The bone-resorbing activity was determined by mouse calvarial organ cultures. An in situ docking simulation was conducted to reveal the interaction of lutein and IκB kinase (IKK) β protein. HLU mice were fed a 1% lutein-containing diet for two weeks, and the femoral bone mass was measured by μCT. Results: Osteoclast differentiation is significantly inhibited by lutein, astaxanthin, and β-cryptoxanthin. In contrast, only lutein promoted osteoblastic calcified bone nodule formation. To elucidate the molecular role of lutein, we functionally analyzed the NF-κB complex, a molecule involved in bone metabolism, especially in osteoclasts. Docking simulations showed that lutein binds to IKK, thus inhibiting the activation of NF-κB. In a cell culture analysis, the phosphorylation of p65, the active form of NF-κB in osteoblasts, was suppressed by lutein treatment. In vivo, a μCT analysis of the bone microarchitecture showed that lutein improves several bone parameters while maintaining bone mass. Conclusions: Lutein is effective in maintaining bone mass by controlling both bone resorption and formation, which is applied to prevent disuse-induced osteoporosis. Full article
(This article belongs to the Section Micronutrients and Human Health)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Effects of LUT, CRY and AST on osteoclastic bone resorption. (<b>A</b>) Chemical structures of lutein (LUT), β-cryptoxanthin (CRY), and astaxanthin (AST) were described. (<b>B</b>,<b>C</b>) POBs and BMCs were cocultured with IL-1 (2 ng/mL) and LUT, CRY, or AST (20 μM, each) for 7 days. Images show TRAP-stained multinucleated osteoclasts (<b>B</b>). The number of TRAP-stained multinucleated osteoclasts was counted (<b>C</b>). (<b>D</b>) Calvarial bones from neonatal mice were cultured with IL-1 (2 ng/mL) and each carotenoid (20 μM). Bone-resorbing activity was determined by measuring the concentration of calcium leached from bone in conditioned medium. (<b>E</b>) The mRNA expression of <span class="html-italic">Tnfsf11</span> (encoding RANKL) was quantified by RT-qPCR. (<b>F</b>,<b>G</b>) Raw264.7 cells were cultured with or without sRANKL (100 ng/mL) and LUT (3, 10, and 30 μM) for 5 days. Images show TRAP-stained multinucleated osteoclasts (<b>F</b>). The number of TRAP-stained multinucleated osteoclasts was counted (<b>G</b>). (<b>H</b>) The mRNA expression of <span class="html-italic">Ctsk</span> (encoding cathepsin K) was analyzed by RT-qPCR. The data are expressed as the mean ± SEM of 8 cultures (<b>C</b>), 5 bones (<b>D</b>), 4 cultures (<b>G</b>), or triplicate from a representative experiment (<b>E</b>,<b>F</b>). The <span class="html-italic">Actb</span> gene was used for normalization. Asterisks indicate significant differences between 2 groups: ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001 by a one-way ANOVA followed by post hoc Tukey’s test.</p>
Full article ">Figure 2
<p>Effects of LUT, CRY and AST on osteoblastic bone formation. (<b>A</b>,<b>B</b>) POBs were cultured with β-glycerophosphate (β-GP) and ascorbic acid (AA) and LUT, CRY, or AST (20 μM, each) for 14 days. Images of alizarin red S (ARS) and alkaline phosphatase (ALP) double-staining are shown (<b>A</b>). The ARS-positive area was measured as the bone mineralized area (<b>B</b>). (<b>C</b>) The mRNA expression of <span class="html-italic">Bmp2</span> and <span class="html-italic">Sost</span> (encoding sclerostin) was analyzed by RT-qPCR. The data are expressed as the mean ± SEM of 8 cultures (<b>B</b>) or triplicate from a representative experiment (<b>C</b>). The <span class="html-italic">Actb</span> gene was used for normalization. Asterisks indicate a significant difference between 2 groups: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001 by a one-way ANOVA followed by post hoc Tukey’s test (<b>B</b>) and by a two-tailed Welch’s <span class="html-italic">t</span> test (<b>C</b>).</p>
Full article ">Figure 3
<p>Mechanistic analysis of the effects of lutein on osteoclast differentiation and bone mineralization. (<b>A</b>) The overall image of the molecular docking results. (<b>B</b>,<b>C</b>) Three-dimensional (<b>B</b>) and 2-dimensional images (<b>C</b>) of the docking site. The dashed lines indicate interactions between hydrophobic regions and charged areas of the molecules. Numbers indicate the distance (Å) between IKKβ residue and lutein. (<b>D</b>) The effect of lutein on the kinase activity of IKKβ in an in vitro experiment. The data are expressed as the mean ± SEM of 3 wells. Asterisks indicate a significant difference between 2 groups: *** <span class="html-italic">p</span> &lt; 0.001 and **** <span class="html-italic">p</span> &lt; 0.0001 by a one-way ANOVA followed by post hoc Tukey’s test. (<b>E</b>,<b>F</b>) Mouse POBs were treated with IL-1 or β-GP + AA and lutein for 24 h. Whole-cell lysates were collected, and phospho-p65 and β-actin were detected by Western blotting. The blot images are shown in upper images, and the relative blot intensity of phospho-p65 was shown in lower graphs.</p>
Full article ">Figure 4
<p>Lutein intake ameliorates disuse-induced bone loss in HLU mice. (<b>A</b>) Reconstructed images using μCT of a horizontal section and the distal femur from a longitudinal section. The scale bar represents 1 mm. (<b>B</b>–<b>G</b>) The bone microarchitecture parameters, BV/TV (%) (<b>B</b>), Tb.N (1/mm) (<b>C</b>), Tb.Sp (μm) (<b>D</b>), BS/BV (1/mm) (<b>E</b>), Conn.D (1/mm<sup>3</sup>), and TBPf (1/mm) were calculated by μCT. The data are expressed as the mean ± SEM of 5 mice. Asterisks indicate a significant difference between 2 groups: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001 by a one-way ANOVA followed by post hoc Tukey’s test.</p>
Full article ">Figure 5
<p>Model illustrating the effects of lutein on bone resorption and bone formation. Three carotenoids, including astaxanthin, β-cryptoxanthin, and lutein, inhibited osteoclast differentiation and bone resorption, whereas only lutein promoted osteoblastic calcified bone nodule formation. Mechanistically, lutein can directly bind to IKK protein and suppress its kinase activity, attenuating NF-κB transcriptional activation. The inhibition of NF-κB by lutein results in the downregulation of osteoclast markers in osteoclasts and RANKL in osteoblasts, which, in turn, inhibits osteoclastic bone resorption. In contrast, the inhibition of NF-κB by lutein can elevate the expression of <span class="html-italic">Bmp2</span> and suppress the expression of <span class="html-italic">Sost</span>, leading to osteoblastic calcified bone nodule formation.</p>
Full article ">
15 pages, 2093 KiB  
Article
Exploring the In Vitro Effects of Zingerone on Differentiation and Signalling Pathways in Bone Cell Lines
by Brunhildé De Vos, Abe E. Kasonga, Anna M. Joubert and Trevor T. Nyakudya
Metabolites 2024, 14(12), 693; https://doi.org/10.3390/metabo14120693 - 9 Dec 2024
Viewed by 662
Abstract
Objective: Ensuring adequate bone health is crucial for preventing conditions such as osteoporosis and fractures. Zingerone, a phytonutrient isolated from cooked ginger, has gained attention for its potential benefits in bone health. This study evaluated the osteoprotective potential of zingerone and its effects [...] Read more.
Objective: Ensuring adequate bone health is crucial for preventing conditions such as osteoporosis and fractures. Zingerone, a phytonutrient isolated from cooked ginger, has gained attention for its potential benefits in bone health. This study evaluated the osteoprotective potential of zingerone and its effects on differentiation and signalling pathways in vitro using SAOS-2 osteosarcoma and RAW264.7 macrophage cell lines, aiming to elucidate its mechanism of action in bone remodelling. Methods: SAOS-2 osteosarcoma and RAW264.7 macrophage cells were treated with zingerone at concentrations of 200 µM. Osteoblast differentiation was assessed by alkaline phosphatase (ALP) activity, bone mineralisation via Alizarin Red S stain, and gene expression markers (ALP, runt-related transcription factor 2 (Runx2), and osteocalcin) via quantitative polymerase chain reaction (q-PCR). Osteoclast differentiation was evaluated by tartrate-resistant acid phosphatase (TRAP) staining, TRAP activity, and mitogen-activated protein kinase (MAPK) pathways. Results: Treatment with zingerone was non-toxic at 200 µM. Zingerone (200 µM) significantly stimulated the gene expression of ALP and Runx2 in SAOS-2 cells (p < 0.05) without statistically significantly enhancing SAOS-2 mineralisation via calcium deposits. Moreover, zingerone significantly inhibited osteoclast differentiation in RAW264.7 cells as evidenced by reduced TRAP staining and activity (p < 0.05). Conclusions: Zingerone shows promise in reducing osteoclast activity and supporting early osteoblast differentiation, suggesting its potential as a dietary supplement for bone health. Further in vivo and clinical studies are needed to confirm its role in managing osteoporosis. Full article
(This article belongs to the Special Issue Advances in Phytomedicine Intervention on Metabolic Disorders)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Synthesis of zingerone during ginger processing. In the process, 6-Gingerol is formed by cooking or drying ginger root which then produces zingerone through a retro-aldol reaction. Zingerone is not present in fresh ginger. Chemical structures were sketched with PubChem Sketcher V2.4 (<a href="https://pubchem.ncbi.nlm.nih.gov//edit3/index.html" target="_blank">https://pubchem.ncbi.nlm.nih.gov//edit3/index.html</a> accessed on 27 October 2024). MW: molecular weight.</p>
Full article ">Figure 2
<p>Effect of zingerone on cell viability, mineralisation, and ALP activity in SAOS-2 cells. (<b>A</b>) Zingerone (0.1–200 µM) effects on cell viability (%) of undifferentiated SAOS-2 cells following 48 h treatment. Triton X-100 (0.2%) served as the positive control for cytotoxicity. (<b>B</b>) Mineralisation of SAOS-2 cells treated with zingerone (5–200 µM) for 7–21 days, evaluated using Alizarin Red S staining and measured at 540 nm. (<b>C</b>) SAOS-2 cells treated with osMcCoy media with or without zingerone for 21 days. (<b>D</b>) Zingerone’s effect on ALP activity as a marker of osteoblast-like differentiation measured over three stages of differentiation: 7, 14, and 21 days. Resazurin assay was used to evaluate cell viability. Data presented as mean ± SD (n = 3) (<b>A</b>) or mean ± SEM (<b>C</b>,<b>D</b>) containing at least three independent experiments. **** <span class="html-italic">p</span> &lt; 0.001 (vs. DMSO). Zing: zingerone; DMSO ODM–: undifferentiated media; DMSO ODM+: osteogenic media.</p>
Full article ">Figure 3
<p>The gene expression of <span class="html-italic">ALP</span> (<b>i</b>), <span class="html-italic">Runx2</span> (<b>ii</b>), and <span class="html-italic">OC</span> (<b>iii</b>) in SAOS-2 cells treated with 200 µM zingerone for 7 and 14 days of differentiation was assessed by q-PCR. Evaluation of the effects of zingerone on the expression of genes involved in the early and intermediate stages of osteoblast differentiation was conducted by exposing cells to osteogenic media for 7 days (<b>A</b>) and 14 days (<b>B</b>). Data presented as mean ± SD (n = 3), normalised to DMSO ODM–. *<sup>/</sup>** <span class="html-italic">p</span> &lt; 0.05 compared to control (DMSO ODM–). Zing: zingerone; DMSO ODM–: undifferentiated media; DMSO ODM+: osteogenic media.</p>
Full article ">Figure 4
<p>Effect of zingerone on cell viability and osteoclast differentiation in RAW264.7 cells. (<b>A</b>) Zingerone’s (0.1–200 µM) effects on cell viability (%) were evaluated by resazurin assay in undifferentiated RAW264.7 cells after 48 h treatment. Triton X-100 (0.2%) served as the positive control for cytotoxicity. (<b>B</b>) Microscopic images of TRAP-stained RAW264.7 cells (scale bar = 2 mm) treated with zingerone (100–200 µM) and RANKL (5 ng/mL). (<b>C</b>) Quantification of osteoclasts, identified as large multinucleated cells with three or more nuclei, stained pink. (<b>D</b>) TRAP activity measured in conditioned media via <span class="html-italic">p</span>-NPP substrate and displayed relative to the R+ cells. Data presented as mean ± SD (n = 3). (<b>A</b>): **** <span class="html-italic">p</span> &lt; 0.05 vs. DMSO. (<b>C</b>): * <span class="html-italic">p</span> &lt; 0.05 vs. R+. (<b>D</b>): **** <span class="html-italic">p</span> &lt; 0.05 V+ vs. R+. ** <span class="html-italic">p</span> &lt; 0.05 200 µM Zing vs. R+. V+: vehicle control (no RANKL added); R+: RANKL-stimulated cells; Zing: zingerone.</p>
Full article ">Figure 5
<p>The effects of zingerone (200 µM) on osteoclast-specific protein expression via RANKL-stimulated (15 ng/mL) MAPK (JNK, p38, ERK) signalling pathway. (<b>A</b>) Evaluating the effects of 200 µM zingerone on RANKL-stimulated MAPK signalling as represented by membrane images. GAPDH was used as the control (<b>B</b>–<b>D</b>) The expression levels of MAPKs (JNK, p38, and ERK) in RAW264.7s were treated with 200 µM zingerone and quantified by western blot via cytoplasmic extraction protocol. Data presented as mean ± SD of 3 independent repetitions (n = 3). ** <span class="html-italic">p</span> &lt; 0.05 compared to vehicle control (V+). RANKL: receptor activator of nuclear factor kappa beta; JNK: Jun N-terminal kinase; ERK: extracellular signal-regulated kinase; MAPKs: mitogen-activated protein kinases; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; V+: vehicle control (RANKL not present); R+: RANKL-only stimulated control; Zing: zingerone.</p>
Full article ">
26 pages, 7398 KiB  
Article
Extract of Curculigo capitulata Ameliorates Postmenopausal Osteoporosis by Promoting Osteoblast Proliferation and Differentiation
by Ying Wang, Xueru Wang, Kaijin Wang, Weiwei Qin and Ning Li
Cells 2024, 13(23), 2028; https://doi.org/10.3390/cells13232028 - 8 Dec 2024
Viewed by 763
Abstract
Postmenopausal osteoporosis (PMOP) is a bone disease characterized by bone thinning and an increased risk of fractures due to estrogen deficiency. Current PMOP therapies often result in adverse side effects. The traditional medicinal plant Curculigo capitulata is commonly used to strengthen bones and [...] Read more.
Postmenopausal osteoporosis (PMOP) is a bone disease characterized by bone thinning and an increased risk of fractures due to estrogen deficiency. Current PMOP therapies often result in adverse side effects. The traditional medicinal plant Curculigo capitulata is commonly used to strengthen bones and support kidney function, but its role in treating PMOP is not well understood. This study aims to investigate the therapeutic effects of the total extract of Curculigo capitulata (Eocc) on PMOP and to explore the underlying mechanisms. The major components of the extract were identified using HPLC. Transcriptomics was employed to predict potential targets. An osteogenic differentiation model of MC3T3-E1 cells was used in vitro. The osteogenic potential of the Eocc was assessed through CCK-8 cell viability assays, alkaline phosphatase (ALP) staining, Alizarin Red staining, Western blotting, and qPCR. MCF-7 and HEK-293 cells were utilized to evaluate the estrogen-like activity of Eocc. Apoptosis rates were detected by flow cytometry. In vivo, a bilateral ovariectomized mouse model of PMOP was used to further validate the in vitro findings through histopathological analysis and WB results. The results demonstrated that the Eocc promoted the proliferation of MC3T3-E1 cells, increased ALP activity, and stimulated the formation of osteogenic mineralized nodules. It also upregulated the expression of osteogenic markers (Runx2, OCN, OPN, and BSP) at both the protein and mRNA levels. The Eocc induced the activation of ERα both in vitro and in vivo, initiating the Src/PI3K/AKT signaling pathway, leading to the phosphorylation of GSK3β and subsequent osteogenesis. The activation of this pathway also stimulated the phosphorylation of mTOR and p70S6K while downregulating cleaved caspase-3 and caspase-9. Additionally, the Eocc reduced apoptosis during osteogenic differentiation and promoted cell proliferation. These findings suggest that the Eocc facilitates osteoblast proliferation and differentiation, improving bone integrity in PMOP mice, and may represent a promising therapeutic candidate for managing PMOP. Full article
(This article belongs to the Special Issue Natural Products and Their Derivatives Against Human Disease)
Show Figures

Figure 1

Figure 1
<p>Eocc improves bone-related parameters in PMOP mice: (<b>A</b>) Schematic representation of the PMOP mouse model and treatment protocol. (<b>B</b>,<b>C</b>) Representative micro-CT images of the distal femur and bone parameter analysis for different groups. All bar graphs show mean ± SD. All bar graphs show mean ± SD. Comparisons to the OVX group are as follows: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001; comparisons to the Sham group are as follows: ### <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 2
<p>Changes in H&amp;E staining and Masson staining at the distal femur of mice after Eocc treatment. Representative micrographs of H&amp;E and Masson-stained sections, magnified at 40× with a scale bar of 50 µm. The red arrows in the figure indicate bone trabeculae.</p>
Full article ">Figure 3
<p>Eocc promotes the expression of osteogenic marker proteins in PMOP mice: (<b>A</b>,<b>B</b>) Immunohistochemical staining for Runx2, OPN in paraffin-embedded sections of the distal femur, with quantitative analysis of positive staining levels in each tissue region. (<b>C</b>,<b>D</b>) Immunohistochemical staining for OCN, BSP in paraffin-embedded sections of the distal femur, with quantitative analysis of positive staining levels in each tissue region. The area indicated by the red arrows in the image represents the DAB-positive region. Magnified at 40× with a scale bar of 50 µm. All bar graphs show mean ± SD. Comparisons to the OVX group are as follows: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001; comparisons to the sham group are as follows: ## <span class="html-italic">p</span> &lt; 0.01, ### <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3 Cont.
<p>Eocc promotes the expression of osteogenic marker proteins in PMOP mice: (<b>A</b>,<b>B</b>) Immunohistochemical staining for Runx2, OPN in paraffin-embedded sections of the distal femur, with quantitative analysis of positive staining levels in each tissue region. (<b>C</b>,<b>D</b>) Immunohistochemical staining for OCN, BSP in paraffin-embedded sections of the distal femur, with quantitative analysis of positive staining levels in each tissue region. The area indicated by the red arrows in the image represents the DAB-positive region. Magnified at 40× with a scale bar of 50 µm. All bar graphs show mean ± SD. Comparisons to the OVX group are as follows: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001; comparisons to the sham group are as follows: ## <span class="html-italic">p</span> &lt; 0.01, ### <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>The osteogenic effects of the Eocc in vitro: (<b>A</b>) The CCK-8 assay was used to evaluate the cytotoxicity of the Eocc on MC3T3-E1 cells at concentrations ranging from 1 to 1000 μg/mL (<span class="html-italic">n</span> = 3). (<b>B</b>) The CCK-8 assay shows the effect of the Eocc on MC3T3-E1 cell viability at concentrations ranging from 0.001 to 10 µg/mL (<span class="html-italic">n</span> = 5). (<b>C</b>,<b>E</b>) Representative ALP staining images with a scale bar of 200 µm, showing that the Eocc and 17β-estradiol significantly enhance ALP activity (<span class="html-italic">n</span> = 3). (<b>D</b>,<b>F</b>) Representative images of osteogenic mineralized nodule staining, indicating that the Eocc and 17β-estradiol promote mineralized nodule formation (<span class="html-italic">n</span> = 3). Magnified at 40× with a scale bar of 200 µm. All bar graphs show mean ± SD. Comparisons to the control group are as follows: ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001; comparisons to the normal group are as follows: # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01, ### <span class="html-italic">p</span> &lt; 0.001; and comparisons to the E2 group are as follows: &amp;&amp; <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 5
<p>The osteogenic effects of the Eocc in vitro: (<b>A</b>,<b>B</b>) Western blot analysis of Runx2, OCN, OPN, and BSP proteins at different Eocc concentrations (0, 0.1, 1, 10 µg/mL), with quantitative analysis (<span class="html-italic">n</span> = 3). (<b>C</b>) PCR analysis of the osteogenic marker gene expression at various Eocc concentrations (0, 0.1, 1, 10 µg/mL), with quantitative analysis (<span class="html-italic">n</span> = 3). All bar graphs show mean ± SD. Comparisons to the control group are as follows: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 6
<p>The Eocc activates ERα to promote osteogenic activity: (<b>A</b>) Cell viability of MCF-7 cells treated with different concentrations of the Eocc (0, 0.1, 1, 10 µg/mL) for 24 h, assessed by the CCK-8 assay. Treatment with 1 µM of E2 was used as a positive control. (<b>B</b>) The dual luciferase reporter assay measuring the activation of ERα and ERβ. Treatment groups included different concentrations of the Eocc (0, 0.1, 1, 10 µg/mL), positive control E2 (1 µM), co-treatment with E2 and ICI 182,780 (1 µM), and co-treatment with 0.1 µg/mL of the Eocc and ICI 182,780 (1 µM). (<b>C</b>,<b>D</b>) Western blot analysis and quantification of ERα and p-ERα expression levels. (<b>E</b>,<b>F</b>) Representative images of ALP staining (<span class="html-italic">n</span> = 3). Magnified at 40× with a scale bar of 200 µm. (<b>G</b>,<b>H</b>) Representative images of osteogenic mineralization nodule staining (<span class="html-italic">n</span> = 3). Magnified at 40× with a scale bar of 200 µm. (<b>I</b>,<b>J</b>) Western blot analysis and the quantification of osteogenic marker proteins in MC3T3-E1 cells treated with the Eocc, with or without ERα inhibition. All bar graphs represent mean ± SD. Comparisons versus the control group are as follows: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001; versus the normal group are as follows: ## <span class="html-italic">p</span> &lt; 0.01, ### <span class="html-italic">p</span> &lt; 0.001; and versus the E2 or E-1 group are as follows: &amp; <span class="html-italic">p</span> &lt; 0.05, &amp;&amp; <span class="html-italic">p</span> &lt; 0.01, &amp;&amp;&amp; <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 7
<p>PI3K/AKT signaling may represent potential targets of Eocc in PMOP: (<b>A</b>) Transcriptomic analysis of Eocc-treated MC3T3-E1 cells. Volcano plot comparing the Eocc and control groups: blue dots represent downregulated genes, red dots represent upregulated genes, and gray dots indicate genes with no significant change in expression compared to the control group. (<b>B</b>) Heatmap of differentially expressed genes (DEGs): green indicates downregulated genes, and red indicates upregulated genes. (<b>C</b>) GO enrichment analysis of upregulated genes. (<b>D</b>) KEGG pathway enrichment analysis of upregulated genes.</p>
Full article ">Figure 8
<p>The Eocc activates the Src/PI3K/AKT and downstream signaling pathways via ERα: (<b>A</b>,<b>B</b>) Western blot analysis and the quantification of Src, p-Src, PI3K, p-PI3K, AKT, p-AKT, cleaved-caspase-3, and cleaved-caspase-9 expression levels. (<b>C</b>,<b>D</b>) Western blot analysis and quantification of mTOR, p-mTOR, p70S6K, and p-p70S6K expression levels. (<b>E</b>,<b>F</b>) Western blot analysis and quantification of GSK3β and p-GSK3β expression levels. (<b>G</b>,<b>H</b>) Representative images and quantitative analysis of cell apoptosis rates measured by flow cytometry. All bar graphs represent mean ± SD. Comparisons versus the control group: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001; versus the normal group: ### <span class="html-italic">p</span> &lt; 0.001; &amp; <span class="html-italic">p</span> &lt; 0.05, &amp;&amp; <span class="html-italic">p</span> &lt; 0.01, &amp;&amp;&amp; <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 9
<p>Eocc activates ERα expression in vivo: (<b>A</b>,<b>B</b>) Western blot analysis and quantification of p-ERα and ERα expression levels in mouse femur tissue. (<b>C</b>,<b>D</b>) Immunofluorescence detection of p-ERα expression in paraffin-embedded sections of the distal femur. Images are magnified at 200× with a scale bar of 10 µm. All bar graphs show mean ± SD. Comparisons to the OVX group are as follows: ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001; comparisons to the sham group are as follows: # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 10
<p>The Eocc inhibits the expression of apoptotic proteins in vivo: (<b>A</b>,<b>B</b>) Immunohistochemical staining for cleaved-caspase-3 and cleaved-caspase-9 in paraffin-embedded sections of the distal femur, with the quantitative analysis of positive staining levels in each tissue region. Images are magnified at 40× with a scale bar of 50 µm. All bar graphs show mean ± SD. Comparisons to the OVX group are as follows: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001; and comparisons to the sham group are as follows: ## <span class="html-italic">p</span> &lt; 0.01, ### <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 11
<p>The Eocc inhibits osteoblast apoptosis in vivo: (<b>A</b>,<b>B</b>) Effect of the Eocc on TUNEL levels in mouse femur tissue. Images are magnified at 200× with a scale bar of 10 µm. (<b>C</b>) The mechanism diagram of this study. All bar graphs show mean ± SD. Comparisons to the OVX group are as follows: ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001; comparisons to the sham group are as follows: ### <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">
16 pages, 4901 KiB  
Article
A 0.2 T–0.4 T Static Magnetic Field Improves the Bone Quality of Mice Subjected to Hindlimb Unloading and Reloading Through the Dual Regulation of BMSCs via Iron Metabolism
by Jianping Wang, Chenxiao Zhen, Gejing Zhang, Zhouqi Yang and Peng Shang
Int. J. Mol. Sci. 2024, 25(23), 13136; https://doi.org/10.3390/ijms252313136 - 6 Dec 2024
Viewed by 556
Abstract
Osteoporosis is the most prevalent metabolic bone disease, especially when aggravated by aging and long-term bed rest of various causes and also when coupled with astronauts’ longer missions in space. Research on the use of static magnetic fields (SMFs) has been progressing as [...] Read more.
Osteoporosis is the most prevalent metabolic bone disease, especially when aggravated by aging and long-term bed rest of various causes and also when coupled with astronauts’ longer missions in space. Research on the use of static magnetic fields (SMFs) has been progressing as a noninvasive method for osteoporosis due to the complexity of the disease, the inconsistency of the effects of SMFs, and the ambiguity of the mechanism. This paper studied the effects of mice subjected to hindlimb unloading (UL, HLU) and reloading by the 0.2 T–0.4 T static magnetic field (MMF). Primary bone marrow mesenchymal stem cells (BMSCs) were extracted to explore the mechanism. Eight-week-old male C57BL/6 mice were used as an osteoporosis model by HLU for four weeks. The HLU recovery period (reloading, RL) was carried out on all FVEs and recovered in the geomagnetic field (45–64 μT, GMF) and MMF, respectively, for 12 h/d for another 4 weeks. The tibia and femur of mice were taken; also, the primary BMSCs were extracted. MMF promoted the recovery of mechanical properties after HLU, increased the number of osteoblasts, and decreased the number of adipocytes in the bone marrow. MMF decreased the total iron content and promoted the total calcium content in the tibia. In vitro experiments showed that MMF promoted the osteogenic differentiation of BMSCs and inhibited adipogenic differentiation, which is related to iron metabolism, the Wnt/β-catenin pathway, and the PPARγ pathway. MMF accelerated the improvement in bone metabolism and iron metabolism in RL mice to a certain extent, which improved the bone quality of mice. MMF mainly promoted osteogenic differentiation and reduced the adipogenic differentiation of BMSCs, which provides a reliable research direction and transformation basis for the osteoporosis of elderly, bedridden patients and astronauts. Full article
(This article belongs to the Special Issue The Role of Trace Elements in Nutrition and Health)
Show Figures

Figure 1

Figure 1
<p>Effects of MMF on mechanical properties and markers of bone metabolism in mice. (<b>A</b>) mouse tail suspension model, (<b>B</b>) experimental flow chart, (<b>C</b>) mechanical properties of mouse tibia, including stiffness, elastic modulus, ultimate strain, ultimate stress, ultimate load, and ultimate displacement, (<b>D</b>) measurement of serum biochemical indexes. <span class="html-italic">n</span> = 4–6. Data are shown as the mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 2
<p>The number of osteoblasts and adipocytes in the bone marrow of femur were evaluated. (<b>A</b>,<b>B</b>) The femoral sections were subjected to H&amp;E staining to visualize osteoblasts and adipocytes. Black arrows indicate osteoblasts. Red arrows indicate adipocytes. Scale bar = 50 μm. Osteoblastogenesis were evaluated by the osteoblast number per bone surface (N.Ob/BS) in the trabecular bone; (<b>C</b>) Adipogenesis was evaluated by the adipocyte number per bone surface (No. Adipocytes/T.Ar) and adipocyte surface per bone surface (Adipocyte.Ar/T.Ar) in the bone marrow (<b>D</b>). <span class="html-italic">n</span> = 3. All the data are shown as mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>Bone metabolism of MMF on the femur of HLU reloading mice. (<b>A</b>) Total calcium content in the tibia of HLU reloading mice. (<b>B</b>) Immunohistochemical staining of Col1α1, PPARγ, and SOST. Scar bar = 50 μm. Brown color indicates positive expression. (<b>C</b>) Protein expression of osteogenic and osteoclast-related proteins in the tibia of HLU reloading mice; (<b>D</b>) Quantitative statistics of protein expression of (<b>A</b>). <span class="html-italic">n</span> = 4–6. Data are shown as the mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 4
<p>Effects of MMF on iron metabolism in HLU reloading mice. (<b>A</b>) Prussian blue staining of bone tissue in HLU reloading mice, Scar bar = 50 μm; (<b>B</b>) Protein expression related to iron metabolism in the tibia of HLU reloading mice; (<b>C</b>) Quantitative statistics of the protein expression of (<b>B</b>); (<b>D</b>) Protein expression of liver tissue in HLU reloading mice; (<b>E</b>) Quantitative statistics of the protein expression of (<b>D</b>); (<b>F</b>) Protein expression of the duodenum in HLU reloading mice; (<b>G</b>) Quantitative statistics of the protein expression of (<b>F</b>); (<b>H</b>) Total iron content in the tibia of HLU reloading mice. <span class="html-italic">n</span> = 3. Data are shown as the mean ± SEM. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 5
<p>MMF promotes osteogenic differentiation of primary BMSCs and mechanism. (<b>A</b>) ARS and ALP staining after 14 d of osteogenic differentiation of BMSCs, 10×; (<b>B</b>) Protein expression of osteogenic related molecules after 14 d of osteogenic differentiation of BMSCs; (<b>C</b>) Quantitative statistics of the protein expression of (<b>B</b>); (<b>D</b>) Total iron content of BMSCs cells after osteogenic differentiation for 14 d; (<b>E</b>) Expression of iron-metabolism-related proteins of BMSCs after osteogenic differentiation for 14 d; (<b>F</b>) Quantitative statistics of the protein expression of (<b>E</b>); (<b>G</b>) Protein expression of the Wnt/β-catenin signaling pathway after 14 d of osteogenic differentiation of BMSCs; (<b>H</b>) Quantitative statistics of the protein expression of (<b>G</b>). <span class="html-italic">n</span> = 3. Data are shown as the mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 6
<p>MMF inhibits adipogenic differentiation of primary BMSCs derived from HLU model mice. (<b>A</b>) Oil Red O staining after 14 d of adipogenic differentiation of BMSCs, 10×; (<b>B</b>) Protein expression of PPARγ after 14 d of adipogenic differentiation of BMSCs; (<b>C</b>) Quantitative statistics of the protein expression of (<b>B</b>); (<b>D</b>) Expression of iron-metabolism-related proteins of BMSCs after adipogenic differentiation for 14 d; (<b>E</b>) Quantitative statistics of the protein expression of (<b>D</b>). <span class="html-italic">n</span> = 3. Data are shown as the mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 7
<p>Schematic diagram of results summarized in this paper. The use of 0.2 T–0.4 T SMF (MMF) improved the bone quality of HLU reloading mice by affecting bone metabolism and iron metabolism. Through study of the osteogenic and adipogenic differentiation of primary BMSCs, it was proved that MMF promoted osteogenic differentiation and activated the Wnt/β-catenin signaling pathway, inhibiting adipogenic differentiation. Upward arrows represent an increase; downward arrows represent a decrease. All the colors have no real meaning, just for beauty.</p>
Full article ">
Back to TopTop